1
|
Kunzi H, Nsanya MK, Kweka B, Malindisa E, Ngissa NS, Kavishe BB, Jeremiah K, Olsen MF, Krogh-Madsen R, Filteau S, Friis H, Faurholt-Jepsen D, PrayGod G. The association between indoor biofuel use for cooking and glucose metabolism in adults: A cross-sectional study in Tanzania. PLOS GLOBAL PUBLIC HEALTH 2025; 5:e0003816. [PMID: 40367203 PMCID: PMC12077790 DOI: 10.1371/journal.pgph.0003816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 04/15/2025] [Indexed: 05/16/2025]
Abstract
The burden of type 2 diabetes is rapidly increasing in low- and middle-income countries (LMICs), but determinants are not well-characterized. Household air pollution (HAP) from indoor biofuel use for cooking has been associated with non-communicable diseases and could be contributing to the increasing burden of diabetes in LMICs, though data are limited. We assessed the association between indoor biofuel use for cooking and glucose metabolism in HIV-infected and HIV-uninfected Tanzanian adults. This cross-sectional analysis included Tanzanian adults with and without HIV, from whom we collected sociodemographic and non-communicable disease risk factor data. The main predictor variable was indoor biofuel use for cooking, established using self-reported cooking location (indoor or outdoor) and fuel type (electricity/gas or biomass fuel), and categorized as minimal or no exposure, moderate exposure, and high exposure. Blood glucose and insulin were measured during oral glucose tolerance tests, allowing computation of outcome variables including markers of β-cell dysfunction (homeostatic model assessment-β, insulinogenic index, oral disposition index), insulin resistance (HOMA-IR and Matsuda index), and pre-diabetes and diabetes status. Logistic regression was used to assess associations, adjusting for age, sex, physical activity, smoking, socioeconomic status, HIV status, and body mass index. Among 1,871 participants (mean age 40.6 ± 11.9 years; 59.8% female), those with moderate and high exposure to HAP had approximately two-fold higher odds of a lower insulinogenic index (adjusted odds ratio [aOR] = 2.13, 95% CI: 1.27-3.57 and aOR = 2.31, 95% CI: 1.39-3.83, respectively) compared to those with minimal or no exposure. HAP was not associated with other markers of β-cell function, insulin resistance, pre-diabetes, or diabetes. In conclusion, HAP is associated with increased risk of β-cell dysfunction among individuals using biofuel for indoor cooking. Longitudinal studies using objective HAP measurements are needed to confirm these findings.
Collapse
Affiliation(s)
- Happyness Kunzi
- Mwanza Research Centre, National Institute for Medical Research, Mwanza, Tanzania
| | - Mussa K. Nsanya
- Mwanza Research Centre, National Institute for Medical Research, Mwanza, Tanzania
| | - Belinda Kweka
- Mwanza Research Centre, National Institute for Medical Research, Mwanza, Tanzania
| | | | | | | | - Kidola Jeremiah
- Mwanza Research Centre, National Institute for Medical Research, Mwanza, Tanzania
| | - Mette Frahm Olsen
- Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg, Denmark
| | - Rikke Krogh-Madsen
- Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
- Centre for Physical Activity Research, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Suzanne Filteau
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Henrik Friis
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg, Denmark
| | - Daniel Faurholt-Jepsen
- Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - George PrayGod
- Mwanza Research Centre, National Institute for Medical Research, Mwanza, Tanzania
- Muhimbili Medical Research Centre, National Institute for Medical Research, Dar es Salaam, Tanzania
| |
Collapse
|
2
|
Sugimoto H, Hironaka KI, Nakamura T, Yamada T, Miura H, Otowa-Suematsu N, Fujii M, Hirota Y, Sakaguchi K, Ogawa W, Kuroda S. Improved detection of decreased glucose handling capacities via continuous glucose monitoring-derived indices. COMMUNICATIONS MEDICINE 2025; 5:103. [PMID: 40263561 PMCID: PMC12015487 DOI: 10.1038/s43856-025-00819-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Efficiently assessing glucose handling capacity is a critical public health challenge. This study assessed the utility of relatively easy-to-measure continuous glucose monitoring (CGM)-derived indices in estimating glucose handling capacities calculated from resource-intensive clamp tests. METHODS We conducted a prospective study of 64 individuals without prior diabetes diagnosis. The study performed CGM, oral glucose tolerance tests (OGTT), and hyperglycemic and hyperinsulinemic-euglycemic clamp tests. We validated CGM-derived indices characteristics using an independent dataset from another country and mathematical models with simulated data. RESULTS A CGM-derived index reflecting the autocorrelation function of glucose levels (AC_Var) is significantly correlated with clamp-derived disposition index (DI), a well-established measure of glucose handling capacity and predictor of diabetes onset. Multivariate and machine learning models indicate AC_Var's contribution to predicting clamp-derived DI independent from other CGM-derived indices. The model using CGM-measured glucose standard deviation and AC_Var outperforms models using commonly used diabetes diagnostic indices, such as fasting blood glucose, HbA1c, and OGTT measures, in predicting clamp-derived DI. Mathematical simulations also demonstrate the association of AC_Var with DI. CONCLUSIONS CGM-derived indices, including AC_Var, serve as valuable tools for predicting glucose handling capacities in populations without prior diabetes diagnosis. We develop a web application that calculates these CGM-derived indices ( https://cgm-ac-mean-std.streamlit.app/ ).
Collapse
Affiliation(s)
- Hikaru Sugimoto
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Ken-Ichi Hironaka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Tomoaki Nakamura
- Department of Diabetes and Endocrinology, Akashi Medical Center, 743-33 Okubo-cho Yagi, Akashi, Hyogo, 674-0063, Japan
| | - Tomoko Yamada
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Hiroshi Miura
- Department of Diabetes and Endocrinology, Takatsuki General Hospital, 1-3-13 Kosobe-cho, Takatsuki, Osaka, 569-1192, Japan
| | - Natsu Otowa-Suematsu
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Masashi Fujii
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
- Department of Mathematical and Life Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-hiroshima City, Hiroshima, 739-8526, Japan
| | - Yushi Hirota
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Kazuhiko Sakaguchi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Shinya Kuroda
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
3
|
Trinh B, Rasmussen SJ, Brøgger-Jensen ME, Engelhard CA, Lund A, Tavanez AR, Vassilieva A, Janum S, Iepsen UW, Kiens B, Møller K, Pedersen BK, Van Hall G, Ellingsgaard H. Inhibition of basal IL-6 activity promotes subcutaneous fat retention in humans during fasting and postprandial states. Cell Rep Med 2025; 6:102042. [PMID: 40147447 PMCID: PMC12047529 DOI: 10.1016/j.xcrm.2025.102042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/01/2024] [Accepted: 03/04/2025] [Indexed: 03/29/2025]
Abstract
Interleukin-6 (IL-6) knockout mice and humans treated with IL-6 receptor blockade gain adipose tissue mass. This study investigates whether basal IL-6 activity (resting IL-6 levels) influences fat storage during fasting and postprandial states. Using stable-isotope tracer techniques and IL-6 receptor blockade with tocilizumab, we examine fat kinetics in humans. Blocking basal IL-6 activity reduces fasting whole-body lipolysis, decreases hormone-sensitive lipase (HSL) phosphorylation and fatty acid release in adipose tissue, and impairs postprandial fatty acid uptake in the leg. These results suggest diminished fatty acid uptake and oxidation in skeletal muscle, along with enhanced fatty acid entrapment in adipose tissue, which may account for the increased adiposity in the absence of IL-6 activity. Additionally, IL-6 blockade increases the escape of meal-derived fatty acids into the bloodstream. Whether this affects fatty acid storage and lipotoxicity in other tissues warrants further investigation. This study was registered at ClinicalTrials.gov (NCT04687540).
Collapse
Affiliation(s)
- Beckey Trinh
- The Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Endocrinology, Diabetes and Metabolism Clinic, University Hospital of Basel, Basel, Switzerland
| | - Signe Johanne Rasmussen
- The Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Anton Lund
- Department of Neuroanaesthesiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ana Rita Tavanez
- The Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Alexandra Vassilieva
- Department of Neuroanaesthesiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Janum
- Department of Neuroanaesthesiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Winning Iepsen
- The Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Department of Anaesthesia and Intensive Care, Copenhagen University Hospital, Hvidovre, Denmark
| | - Bente Kiens
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Kirsten Møller
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark; The Neuroscience Center and Institute for Clinical Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Bente Klarlund Pedersen
- The Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Gerrit Van Hall
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Clinical Metabolomics Core Facility, Rigshospitalet, Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Helga Ellingsgaard
- The Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
4
|
Malin SK, Remchak ME, Heiston EM, Fabris C, Shah AM. Pancreatic β-cell Function is Higher in Morning Versus Intermediate Chronotypes With Obesity. Obes Sci Pract 2025; 11:e70064. [PMID: 40018087 PMCID: PMC11864105 DOI: 10.1002/osp4.70064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/07/2025] [Accepted: 02/14/2025] [Indexed: 03/01/2025] Open
Abstract
Objectives People with later chronotypes are at greater T2D risk, yet it is unknown if β-cell function differs among chronotypes. Thus we, assessed β-cell function in morning (MORN) and intermediate (INT) chronotypes with obesity. Methods Adults (n = 41, 9M, 55 ± 1.7 y, 36.8 ± 1.0 kg/m2) were grouped as MORN or INT per the Morningness-Eveningness Questionnaire. Glucose, insulin, C-peptide, GIP, and GLP-1(active) were collected every 30 min during a 120 min 75g-OGTT. Insulin secretion rates (ISR) were calculated (regularized deconvolution) to assess early (total area under the curve; tAUC0-30min) and total-phase (tAUC0-120min) glucose-stimulated insulin secretion (GSIS:ISR/Glucose). Skeletal muscle (glucose infusion rate/steady-state insulin) insulin sensitivity and hepatic (HOMA-IR) as well as adipose (Adipose-IR) insulin resistance were assessed during a 120 min euglycemic hyperinsulinemic clamp (40mU/m2/min, 90 mg/dL). β-cell function (disposition index (DI): GSIS adjusted insulin sensitivity) was determined. Body composition (DXA) and fitness (VO2max) were also measured. Results Age, body composition and VO2max were similar between groups, but INT had reduced muscle insulin sensitivity and higher hepatic and adipose IR (p < 0.05). INT had higher C-peptide tAUC0-30min (p = 0.04) and lower hepatic DI (tAUC0-30min p = 0.05 and tAUC0-120min p = 0.07, respectively). Early phase hepatic DI correlated with GLP-1 tAUC0-30min (r = 0.35, p < 0.02) and tAUC0-120min (r = -0.40, p = 0.04). Conclusions β-cell function was higher in MORN versus INT chronotypes. Further work is warranted to discern how chronotype impacts insulin secretion. Trial Registration NCT03355469.
Collapse
Affiliation(s)
- Steven K. Malin
- Department of Kinesiology & HealthRutgers UniversityNew BrunswickNew JerseyUSA
- Division of Endocrinology, Metabolism & NutritionRutgers UniversityNew BrunswickNew JerseyUSA
- New Jersey Institute for FoodNutrition and HealthRutgers UniversityNew BrunswickNew JerseyUSA
- Institute of Translational Medicine and ScienceRutgers UniversityNew BrunswickNew JerseyUSA
| | | | - Emily M. Heiston
- Department of Kinesiology & HealthRutgers UniversityNew BrunswickNew JerseyUSA
| | - Chiara Fabris
- Center for Diabetes TechnologySchool of MedicineUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Ankit M. Shah
- Division of Endocrinology, Metabolism & NutritionRutgers UniversityNew BrunswickNew JerseyUSA
| |
Collapse
|
5
|
Stentebjerg LL, Madsen LR, Støving RK, Hartmann B, Holst JJ, Vinter C, Juhl CB, Hojlund K, Jensen DM. Altered postprandial glucose metabolism and enteropancreatic hormone responses during pregnancy following Roux-en-Y gastric bypass: a prospective cohort study. BMJ Open Diabetes Res Care 2025; 13:e004672. [PMID: 40113260 PMCID: PMC11931895 DOI: 10.1136/bmjdrc-2024-004672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/08/2025] [Indexed: 03/22/2025] Open
Abstract
INTRODUCTION Roux-en-Y gastric bypass (RYGB) increases the risk of postprandial hypoglycemia, whereas pregnancy decreases insulin sensitivity, which could be expected to counteract hypoglycemia. We examined if RYGB performed prior to pregnancy altered the postprandial glucose metabolism and enteropancreatic hormone responses to a mixed meal test (MMT). RESEARCH DESIGN AND METHODS Twenty-three women with RYGB and 23 women matched on prepregnancy body mass index and parity underwent a 4-hour MMT in the first and third trimester of pregnancy with measurement of circulating levels of glucose, insulin, C-peptide, glucose-dependent insulin peptide (GIP), glucagon-like peptide 1 (GLP-1), glucagon, free fatty acids, and lactate. Biochemical hypoglycemia was defined as plasma glucose <3.5 mmol/L. RESULTS Women with RYGB had earlier and higher peak glucose, lower nadir glucose levels, and a higher frequency of biochemical hypoglycemia compared with women without RYGB in both the first and third trimester. The lower glucose levels were preceded by markedly elevated total GLP-1 and insulin levels in women with RYGB, whereas total GIP levels were unaltered. The glucagon levels were lower in women with RYGB. In the first trimester MMT, peak and area under the curve of total plasma GLP-1 and serum insulin levels were negatively associated with nadir plasma glucose, while the early postmeal response of plasma glucagon was positively associated with nadir plasma glucose in the third trimester. CONCLUSIONS These results provide novel insights into the combined effects of RYGB and pregnancy on postmeal glucose metabolism and enteropancreatic hormone responses during pregnancy, and how these changes associate with an increased risk of postprandial hypoglycemia. TRIAL REGISTRATION NUMBER NCT03713060.
Collapse
Affiliation(s)
- Louise Laage Stentebjerg
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Lene Ring Madsen
- Steno Diabetes Center Aarhus, Aarhus Universitetshospital Skejby, Aarhus, Denmark
- Danish Diabetes Academy, Odense University Hospital, Odense, Denmark
| | - René Klinkby Støving
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Endocrinology, Odense University Hospital, Odense, Denmark
| | - Bolette Hartmann
- Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christina Vinter
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
- Department of Gynecology and Obstetrics, Odense University Hospital, Odense, Denmark
| | - Claus Bogh Juhl
- Department of Endocrinology, University Hospital of Southern Denmark, Esbjerg, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Kurt Hojlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Dorte Møller Jensen
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
- Department of Gynecology and Obstetrics, Odense University Hospital, Odense, Denmark
| |
Collapse
|
6
|
Herrerías-García A, Jacobo-Tovar E, Hernández-Robles CM, Guardado-Mendoza R. Pancreatic beta cell function and insulin resistance profiles in first-degree relatives of patients with prediabetes and type 2 diabetes. Acta Diabetol 2025; 62:253-261. [PMID: 39150512 DOI: 10.1007/s00592-024-02352-8] [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: 05/13/2024] [Accepted: 07/30/2024] [Indexed: 08/17/2024]
Abstract
AIMS To evaluate insulin secretion and insulin resistance profiles in individuals with family history of prediabetes and type 2 diabetes. METHODS This was a cross-sectional study to evaluate clinical and metabolic profiles between individuals with type 2 diabetes, prediabetes and their relatives. There were 911 subjects divided into five groups: (i) normoglycemic (NG), (ii) type 2 diabetes, (iii) prediabetes, (iv) first-degree relatives of patients with type 2 diabetes (famT2D), and (v) first-degree relatives of patients with prediabetes (famPD); anthropometrical, biochemical and nutritional evaluation, as well as insulin resistance and pancreatic beta cell function measurement was performed by oral glucose tolerance to compare between groups. RESULTS The most prevalent type 2 diabetes risk factors were dyslipidemia (81%), family history of type 2 diabetes (76%), central obesity (73%), male sex (63%), and sedentary lifestyle (60%), and most of them were progressively associated to prediabetes and type 2 diabetes groups. Insulin sensitivity was lower in famT2D groups in comparison to NG group (p < 0.0001). FamPD and famT2D had a 10% lower pancreatic beta cell function (DI) than the NG group (NG group 2.78 ± 1.0, famPD 2.5 ± 0.85, famT2D 2.4 ± 0.75, p˂0.001). CONCLUSIONS FamPD and famT2D patients had lower pancreatic beta cell function than NG patients, highlighting that defects in insulin secretion and insulin sensitivity appear long time before the development of hyperglycemia in patients genetically predisposed.
Collapse
Affiliation(s)
- Anaid Herrerías-García
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, Blvd. Milenio 1001, Predio San Carlos, 37670, León, Guanajuato, Mexico
| | - Emmanuel Jacobo-Tovar
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, Blvd. Milenio 1001, Predio San Carlos, 37670, León, Guanajuato, Mexico
| | - Claudia Mariana Hernández-Robles
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, Blvd. Milenio 1001, Predio San Carlos, 37670, León, Guanajuato, Mexico
| | - Rodolfo Guardado-Mendoza
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, Blvd. Milenio 1001, Predio San Carlos, 37670, León, Guanajuato, Mexico.
| |
Collapse
|
7
|
Tamaroff J, Nguyen S, Wilson NE, Stefanovski D, Xiao R, Scattergood T, Capiola C, Schur GM, Dunn J, Dedio A, Wade K, Shah H, Sharma R, Mootha VK, Kelly A, Lin KY, Lynch DR, Reddy R, Rickels MR, McCormack SE. Insulin Sensitivity and Insulin Secretion in Adults With Friedreich's Ataxia: The Role of Skeletal Muscle. J Clin Endocrinol Metab 2025; 110:317-333. [PMID: 39109797 PMCID: PMC11747682 DOI: 10.1210/clinem/dgae545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/28/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024]
Abstract
INTRODUCTION Friedreich's ataxia (FRDA) is a multisystem disorder caused by frataxin deficiency. FRDA-related diabetes mellitus (DM) is common. Frataxin supports skeletal muscle mitochondrial oxidative phosphorylation (OXPHOS) capacity, a mediator of insulin sensitivity. Our objective was to test the association between skeletal muscle health and insulin sensitivity and secretion in adults with FRDA without DM. METHODS Case-control study (NCT02920671). Glucose and insulin metabolism (stable-isotope oral glucose tolerance tests), body composition (dual-energy x-ray absorptiometry), physical activity (self-report), and skeletal muscle OXPHOS capacity (creatine chemical exchange saturation transfer magnetic resonance imaging) were assessed. RESULTS Participants included 11 individuals with FRDA (4 female), median age 27 years (interquartile range 23, 39), body mass index 26.9 kg/m2 (24.1, 29.4), and 24 controls (11 female), 29 years (26, 39), 24.4 kg/m2 (21.8, 27.0). Fasting glucose was higher in FRDA [91 vs 83 mg/dL (5.0 vs 4.6 mmol/L), P < .05]. Individuals with FRDA had lower insulin sensitivity (whole-body insulin sensitivity index 2.8 vs 5.3, P < .01), higher postprandial insulin secretion (insulin secretory rate incremental area under the curve 30-180 minutes, 24 652 vs 17,858, P < .05), and more suppressed postprandial endogenous glucose production (-.9% vs 26.9% of fasting endogenous glucose production, P < .05). In regression analyses, lower OXPHOS and inactivity explained some of the difference in insulin sensitivity. More visceral fat contributed to lower insulin sensitivity independent of FRDA. Insulin secretion accounting for sensitivity (disposition index) was not different. CONCLUSION Lower mitochondrial OXPHOS capacity, inactivity, and visceral adiposity contribute to lower insulin sensitivity in FRDA. Higher insulin secretion appears compensatory and, when inadequate, could herald DM. Further studies are needed to determine if muscle- or adipose-focused interventions could delay FRDA-related DM.
Collapse
Affiliation(s)
- Jaclyn Tamaroff
- Division of Pediatric Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Pediatric Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sara Nguyen
- Division of Pediatric Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Neil E Wilson
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Darko Stefanovski
- New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, PA 19348, USA
| | - Rui Xiao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Theresa Scattergood
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher Capiola
- Division of Pediatric Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Gayatri Maria Schur
- Division of Pediatric Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Medical Scientist Training Program, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Julia Dunn
- Division of Pediatric Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Anna Dedio
- Division of Pediatric Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kristin Wade
- Division of Pediatric Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Hardik Shah
- Department of Molecular Biology, Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute, Cambridge, MA 02142, USA
- Metabolomics Platform, Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
| | - Rohit Sharma
- Department of Molecular Biology, Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute, Cambridge, MA 02142, USA
| | - Vamsi K Mootha
- Department of Molecular Biology, Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute, Cambridge, MA 02142, USA
| | - Andrea Kelly
- Division of Pediatric Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kimberly Y Lin
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Pediatric Cardiology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - David R Lynch
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ravinder Reddy
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael R Rickels
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shana E McCormack
- Division of Pediatric Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
8
|
Zhang S, Bian W, Wang Y, Shen M, Qian Y, Dai H, Zheng S, Fu Q, Xu K, Yang T, Jiang H. The MTNR1B Rs724030 variant is associated with islet function and women waist-to-hip ratio in healthy subjects. Front Endocrinol (Lausanne) 2025; 15:1398687. [PMID: 39886031 PMCID: PMC11779613 DOI: 10.3389/fendo.2024.1398687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 12/26/2024] [Indexed: 02/01/2025] Open
Abstract
Objective This study aims to investigate the associations between MTNR1B rs724030 A>G variant and prediabetes risk, along with their correlations with clinical features, including plasma glucose and serum insulin levels during oral glucose tolerance test (OGTT), islet function, insulin resistance, and plasma lipid levels. In particular, we investigated whether there are sex dimorphisms in the impact of this variant on islet function/insulin resistance. Methods We included 3415 glucose-tolerant healthy and 1744 prediabetes individuals based on OGTT. Binary logistic regression was performed to evaluate the relationships between rs724030 in MTNR1B and prediabetes under the additive model. Additionally, multiple linear regression was utilized to investigate the associations between this variant and glycemic-related quantitative traits and lipid levels. Results While no association was observed between the rs724030 variant in MTNR1B and prediabetes risk in the overall cohort (P > 0.05), we found the G allele of this variant was associated with higher fasting and 30-minute plasma glucose levels, decreased Insulinogenic Index (IGI), and oral disposition index (DIo) (P = 0.009, 0.001, 0.001, and 0.007, respectively) in the normal glucose tolerance (NGT) individuals with normal BMI levels. Furthermore, we also found significant associations between this variant and IGI, corrected insulin response (CIR), and DIo (All P < 0.001) in female individuals whose waist-to-hip ratio (WHR) is greater than 0.85, with considerable heterogeneity (Phet = 0.009, 0.030, and 0.049, respectively) to male participants in the NGT individuals, but not in the impaired fasting glucose (IFG)/impaired glucose tolerance (IGT) individuals. Additionally, no association was observed between this variant and insulin clearance (All P > 0.05). Conclusions The MTNR1B rs724030 variant contributes to glycemic traits and islet function, and its effects have sex dimorphisms in the NGT individuals after stratifying by WHR. All these findings provide a basis for accurately assessing islet function in healthy populations and offer a new perspective on precision prevention.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Tao Yang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hemin Jiang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
9
|
Yuan F, Fang D, Xu H, Nie Y, Cai W, Gu T. One-hour post-load glucose is associated with biopsy-proven metabolic dysfunction-associated steatotic liver disease in obese individuals. DIABETES & METABOLISM 2025; 51:101588. [PMID: 39645036 DOI: 10.1016/j.diabet.2024.101588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/17/2024] [Accepted: 11/03/2024] [Indexed: 12/09/2024]
Abstract
AIM To investigate the association between one-hour post-load plasma glucose (1h-PG) levels and metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic-associated steatohepatitis (MASH). METHODS Clinical data were collected from 538 obese participants who underwent oral glucose tolerance tests (OGTT) and liver biopsy in the Drum Tower Hospital affiliated with the Nanjing University Medical School. Participants were categorized into normal glucose tolerance (NGT), prediabetes (pre-DM), and type 2 diabetes (T2DM) groups, with further stratification of the NGT group into 1h-PG Low and 1h-PG High. The diagnosis of MASLD and MASH was performed using the NASH Clinical Research Network scoring system. Logistic regression analyses were performed to determine the association between each category and MASLD/ MASH. RESULTS The prevalence of MASLD and MASH progressively increased across NGT 1h-PG Low, NGT 1h-PG High, pre-DM and T2DM groups. Compared to the NGT 1h-PG Low group, the NGT 1h-PG High group had a significantly higher risk of MASLD (OR 3.27 [95 % CI 1.32;8.09]) and MASH (OR 3.08 [1.55;6.11]), which is similar to the pre-DM group. Additionally, elevated 1h-PG levels were associated with hepatic steatosis, lobular inflammation, and fibrosis. Mediation analysis indicated that Matsuda insulin sensitivity index and disposition index played a sequential mediating role between 1h-PG and hepatic steatosis, accounting for 14.23 % of total effect (β 0.014 [95 % CI 0.002;0.035]). CONCLUSION Elevated 1h-PG levels increase the risk of MASLD and MASH in obese individuals. Early screening and management of MASLD are necessary, even in NGT individuals when 1h-PG levels are ≥8.6 mmol/l.
Collapse
Affiliation(s)
- Fangqin Yuan
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine; Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Da Fang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University; Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Hao Xu
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine; Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Yuanyuan Nie
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine; Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Wen Cai
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University; Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Tianwei Gu
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine; Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University; Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China.
| |
Collapse
|
10
|
Centofanti S, Heilbronn LK, Wittert G, Dorrian J, Coates AM, Kennaway D, Gupta C, Stepien JM, Catcheside P, Yates C, Grosser L, Matthews RW, Banks S. Fasting as an intervention to alter the impact of simulated night-shift work on glucose metabolism in healthy adults: a cluster randomised controlled trial. Diabetologia 2025; 68:203-216. [PMID: 39422718 DOI: 10.1007/s00125-024-06279-1] [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: 01/30/2024] [Accepted: 07/05/2024] [Indexed: 10/19/2024]
Abstract
AIMS/HYPOTHESIS Night-shift work causes circadian misalignment and impairs glucose metabolism. We hypothesise that food intake during night shifts may contribute to this phenomenon. METHODS This open-label, multi-arm, single-site, parallel-group controlled trial involved a 6 day stay at the University of South Australia's sleep laboratory (Adelaide, SA, Australia). Healthy, non-shift-working adults without obesity (N=55; age 24.5 ± 4.8 years; BMI 24.8 ± 2.8 kg/m2) were assigned to the next available run date and cluster randomised (1:1:1) to fasting-at-night (N=20), snack-at-night (N=17), or meal-at-night (N=18) conditions. One participant withdrew from each group, prior to starting the study. Due to study design, neither participants nor people collecting their measurements could be blinded. Statistical and laboratory staff were concealed to study allocation. Participants were fed at calculated energy balance, with the macronutrient composition of meals being similar across conditions. The primary outcomes were a linear mixed-effects model of glucose, insulin and NEFA AUC in response to a 75 g OGTT that was conducted prior to and after 4 consecutive nights of shift work plus 1 night of recovery sleep. Insulin sensitivity, insulinogenic and disposition indexes were also calculated. RESULTS Night-shift work impaired insulin sensitivity, as measured by insulin AUC (p=0.035) and the insulin sensitivity index (p=0.016) across all conditions. Insulin secretion, as measured by the insulinogenic index, was increased in the fasting-at-night condition only (p=0.030), resulting in a day×condition interaction in glucose AUC (p<0.001) such that glucose tolerance was impaired in the meal-at night (+2.00 [95% CI 1.45, 2.56], p<0.001) and snack at-night (+0.96 [0.36, 1.56], p=0.022) conditions vs the fasting-at-night (+0.34 [-0.21, 0.89]) condition. A day×condition interaction was also observed in NEFA AUC (p<0.001), being higher in the meal-at-night (+0.07 [0.03, 0.10]. p=0.001) and snack-at-night (0.01 [-0.03, 0.05], p=0.045) conditions vs the fasting-at-night condition (-0.02 [-0.06, 0.01]). No adverse events occurred. CONCLUSIONS/INTERPRETATION The timing of food intake has a critical effect on glucose metabolism during simulated night-shift work, which was readily amendable to a meal re-timing intervention. TRIAL REGISTRATION Australian New Zealand Clinical Trials Registry (ANZCTR) ACTRN12616001556437 FUNDING: This work was funded by the National Health and Medical Research Council (NHMRC), APP1099077.
Collapse
Affiliation(s)
- Stephanie Centofanti
- Behaviour-Brain-Body Research Centre, UniSA Justice and Society, University of South Australia, Adelaide, SA, Australia
| | - Leonie K Heilbronn
- Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Gary Wittert
- Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- The Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, SA, Australia
| | - Jillian Dorrian
- Behaviour-Brain-Body Research Centre, UniSA Justice and Society, University of South Australia, Adelaide, SA, Australia
| | - Alison M Coates
- Behaviour-Brain-Body Research Centre, UniSA Justice and Society, University of South Australia, Adelaide, SA, Australia
- Alliance for Research in Exercise, Nutrition and Activity, University of South Australia, Adelaide, SA, Australia
| | - David Kennaway
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Charlotte Gupta
- Appleton Institute, Central Queensland University, Rockhampton, QLD, Australia
| | - Jacqueline M Stepien
- Behaviour-Brain-Body Research Centre, UniSA Justice and Society, University of South Australia, Adelaide, SA, Australia
| | - Peter Catcheside
- Flinders Health and Medical Research Institute: Sleep Health (formerly Adelaide Institute for Sleep Health), Flinders University, Adelaide, SA, Australia
| | - Crystal Yates
- Behaviour-Brain-Body Research Centre, UniSA Justice and Society, University of South Australia, Adelaide, SA, Australia
| | - Linda Grosser
- Behaviour-Brain-Body Research Centre, UniSA Justice and Society, University of South Australia, Adelaide, SA, Australia
| | - Raymond W Matthews
- Appleton Institute, Central Queensland University, Rockhampton, QLD, Australia
| | - Siobhan Banks
- Behaviour-Brain-Body Research Centre, UniSA Justice and Society, University of South Australia, Adelaide, SA, Australia.
| |
Collapse
|
11
|
Baltogianni M, Dermitzaki N, Giapros V, Balomenou F, Kosmeri C, Ladomenou F, Kantza E, Serbis A. Indicators of Glucose Metabolism in Children and Adolescents Characterized as Having "Metabolically Healthy" and "Metabolically Unhealthy" Obesity. CHILDREN (BASEL, SWITZERLAND) 2025; 12:50. [PMID: 39857881 PMCID: PMC11763677 DOI: 10.3390/children12010050] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND/OBJECTIVES Some individuals with obesity may exhibit fewer metabolic disturbances and face a lower long-term risk of complications; however, the existence of this so-called "metabolically healthy obesity" (MHO) compared to "metabolically unhealthy obesity" (MUO) remains controversial. We hypothesized that children with MHO might have a more favorable profile than children with MUO. Markers of glucose metabolism and insulin sensitivity were compared between children and adolescents diagnosed with MHO and MUO. METHODS This study recruited prospectively 104 children and adolescents (aged 6-16 years, 47 boys) with obesity. All participants underwent an oral glucose tolerance test (OGTT), and a comparative analysis was performed on HOMA-IR, QUICKI, insulin sensitivity index (ISI), insulinogenic index (IGI), disposition index (DI), and oral disposition index (oDI). Glucose metabolism indices were compared in these subgroups according to pubertal status. RESULTS Forty-seven children (45.2%) were diagnosed with MHO. The whole-body ISI differed significantly between the MHO and MUO groups (4.02 vs. 2.7, p < 0.01). The IGI was statistically lower in the MHO group compared to MUO (1.26 vs. 1.54, p < 0.01), while neither the DI nor the oDI differed significantly. A higher ISI (4.5 vs. 3.9, p < 0.01) was observed in prepubertal MHO individuals compared to MHO adolescents. CONCLUSIONS Children classified as MHO according to the more recent criteria exhibit a more favorable metabolic profile than those with MUO. However, a completely healthy profile was not demonstrated in the MHO group, as many crucial metabolic profile parameters were comparable to those observed in the MUO group. The findings of this study indicate that all children with obesity, irrespective of whether they are categorized as having MUO or MHO, necessitate close monitoring.
Collapse
Affiliation(s)
- Maria Baltogianni
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (M.B.); (N.D.); (F.B.)
| | - Niki Dermitzaki
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (M.B.); (N.D.); (F.B.)
| | - Vasileios Giapros
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (M.B.); (N.D.); (F.B.)
| | - Foteini Balomenou
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (M.B.); (N.D.); (F.B.)
| | - Chrysoula Kosmeri
- Pediatric Department, School of Medicine, University of Ioannina, 45500 Ioannina, Greece (F.L.); (E.K.); (A.S.)
| | - Fani Ladomenou
- Pediatric Department, School of Medicine, University of Ioannina, 45500 Ioannina, Greece (F.L.); (E.K.); (A.S.)
| | - Evanthia Kantza
- Pediatric Department, School of Medicine, University of Ioannina, 45500 Ioannina, Greece (F.L.); (E.K.); (A.S.)
| | - Anastasios Serbis
- Pediatric Department, School of Medicine, University of Ioannina, 45500 Ioannina, Greece (F.L.); (E.K.); (A.S.)
| |
Collapse
|
12
|
Goedecke JH, Kufe CN, Masemola M, Lichaba M, Seipone ID, Mendham AE, Gibson H, Hawley J, Selva DM, Magodoro I, Kengne AP, Chikowore T, Crowther NJ, Norris SA, Karpe F, Olsson T, Storbeck KH, Micklesfield LK. Sex hormone-binding globulin, testosterone and type 2 diabetes risk in middle-aged African women: exploring the impact of HIV and menopause. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.25.24319619. [PMID: 39763542 PMCID: PMC11703316 DOI: 10.1101/2024.12.25.24319619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Objectives Sex hormone-binding globulin (SHBG) and testosterone are differentially associated with type 2 diabetes (T2D) risk. We investigated whether these associations differ by HIV and menopausal status in Black South African women living with (WLWH) and without HIV (WLWOH). Design Cross-sectional observational. Methods Eighty one premenopausal (57 WLWOH, 24 WLWH) and 280 postmenopausal (236 WLWOH, 44 WLWH) women from the Middle-Aged Soweto Cohort (MASC) completed the following measures: circulating SHBG and sex hormones, body composition (dual energy x-ray absorptiometry), oral glucose tolerance test to estimate insulin sensitivity (Matsuda index), secretion (insulinogenic index, IGI) and clearance, and beta-cell function (disposition index, DI). Dysglycaemia was defined as either impaired fasting or postprandial glucose or T2D. Results SHBG was higher and total and free testosterone were lower in postmenopausal WLWH than WLWOH (all p≤0.023). Irrespective of HIV serostatus, SHBG was positively associated with Matsuda index, insulin clearance and DI and inversely with HOMA-IR (all p<0.011). The association between SHBG and Matsuda index was stronger in premenopausal than postmenopausal women (p=0.043 for interaction). Free testosterone (and not total testosterone) was only negatively associated with basal insulin clearance (p=0.021), and positively associated with HOMA-IR in premenopausal and not post-menopausal women (p=0.015 for interaction). Conclusions We show for the first time that midlife African WLWH have higher SHBG and lower total and free testosterone than WLWOH, which corresponded to their higher beta-cell function, suggesting a putative protective effect of SHBG on T2D risk in WLWH.
Collapse
Affiliation(s)
- Julia H. Goedecke
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa
- South African Medical Research Council/WITS Developmental Pathways for Health Research Unit (DPHRU), Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, Umeå, Sweden
| | - Clement Nyuyki Kufe
- South African Medical Research Council/WITS Developmental Pathways for Health Research Unit (DPHRU), Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Anaesthesiology, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Maphoko Masemola
- South African Medical Research Council/WITS Developmental Pathways for Health Research Unit (DPHRU), Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mamosilo Lichaba
- Department of Exercise Science and Sports Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ikanyeng D. Seipone
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa
| | - Amy E Mendham
- South African Medical Research Council/WITS Developmental Pathways for Health Research Unit (DPHRU), Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Riverland Mallee Coorong Local Health Network, South Australia Health, Berri, South Australia, Australia
- Health through Physical Activity, Lifestyle and Sport Research Centre (HPALS), FIMS International Collaborating Centre of Sports Medicine, Division of Physiological Sciences, Department of Human Biology, Faculty of Health Sciences, University of Cape Town
| | - Hylton Gibson
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - James Hawley
- Department of Clinical Biochemistry, University Hospital of South Manchester, Manchester, United Kingdom
| | - David M. Selva
- Diabetes and Metabolism Department, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Itai Magodoro
- Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Andre Pascal Kengne
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Tinashe Chikowore
- South African Medical Research Council/WITS Developmental Pathways for Health Research Unit (DPHRU), Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nigel J. Crowther
- Department of Chemical Pathology, National Health Laboratory Service and University of the Witwatersrand Faculty of Health Sciences, Johannesburg, South Africa
| | - Shane A Norris
- South African Medical Research Council/WITS Developmental Pathways for Health Research Unit (DPHRU), Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- School of Human Development and Health, University of the Southampton, UK
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK and
- National Institute for Health Research, Oxford Biomedical Research Centre, Oxford Radcliffe Hospitals Trust, OCDEM, Churchill Hospital, Oxford, UK
| | - Tommy Olsson
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, Umeå, Sweden
| | - Karl-Heinz Storbeck
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Lisa K. Micklesfield
- South African Medical Research Council/WITS Developmental Pathways for Health Research Unit (DPHRU), Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
13
|
Adeerjiang Y, Sidike A, Gan XX, Li QT, Jiang S. The Role of Wnt3a/β-Catenin/TCF7L2 Pathway in Diabetes and Cardiorenal Complications. Cardiorenal Med 2024; 15:72-82. [PMID: 39709946 PMCID: PMC11844670 DOI: 10.1159/000543145] [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: 08/22/2024] [Accepted: 12/11/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Diabetes mellitus is a prevalent chronic disease that is becoming increasingly common worldwide and can lead to a number of dangerous complications. The Wnt signaling pathway is important for the onset and progression of diabetes. Wnt3a is a typical Wnt ligand that can increase the stability of β-catenin, control TCF7L2 expression, promote β-cell proliferation, and reduce apoptosis. SUMMARY The involvement of the Wnt3a/β-catenin/TCF7L2 signaling pathway in the development of diabetes and associated problems related to the kidneys is reviewed in this article. KEY MESSAGE We believe that a thorough comprehension of the molecular connections between diabetes and signaling pathways will eventually lead to improved diabetes management.
Collapse
Affiliation(s)
- Yilinuer Adeerjiang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China,
- Department of Endocrinology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,
| | - Abudulimu Sidike
- Department of Endocrinology, The First People's Hospital of Kashgar Region, Kashgar, China
| | - Xiao-Xue Gan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Qin-Tian Li
- First Clinical Medical College of Xinjiang Medical University, Urumqi, China
| | - Sheng Jiang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
14
|
Bai MR, Abirami K, Gayathri R, Vedantham S, Shobana S, Nagarajan LP, Gunasekaran G, Nagamuthu G, Malini HM, Gokulakrishnan K, Sandhya N, Ganesh Jeevan R, Anjana RM, Unnikrishnan R, Krishnaswamy K, Sudha V, Mohan V. Effect of low vs high dietary-advanced glycation end products on insulin-sensitivity and inflammatory- markers among overweight/obese Asian-Indian adults-A randomised controlled trial. Int J Food Sci Nutr 2024; 75:835-845. [PMID: 39360559 DOI: 10.1080/09637486.2024.2405121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 08/31/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024]
Abstract
The present study investigated the effect of low vs high-dietary-Advanced Glycation End products-based diets on oral disposition index-(DIo)-a marker of islet β-cell function and cardiometabolic risks factors in 38-overweight and obese Asian Indian-adults (aged 25-45 years with body-mass-index (BMI) ≥23kg/m2) through 12-week isocaloric crossover feeding trial. Biochemical-measures included-glucose tolerance test (GTT), Insulin assay (0,30 and 120 min), lipid-profile, serum-adiponectin, serum-AGE and serum-Thiobarbituric acid reactive substances-(TBARS) assessed both at baseline and end of each intervention. Generalised linear models showed that low-dAGE diet significantly improved in oral disposition index [Least Square Mean (SE), +0.3 (0.1); p = 0.03] compared to high-dAGE diet. The low-dAGE diet also showed a significant reduction in 30-minutes plasmapost-glucose-challenge-value:(-8.1[3.8] (mg/dl) vs 3.8 [3.8] (mg/dl); p = 0.01), serum-AGEs-(-3.2 [0.2] (μg/ml) vs -0.8 [0.2] (μg/ml); p = <0.0001) compared to high-dAGE diet. In summary, low-dAGE diets exhibited improvement in the insulin-sensitivity and reduction in the inflammatory levels compared to high-dAGE diets. Hence, study first time in India revealed that low dAGE diets could be a potential strategy to reduce diabetes risk.
Collapse
Affiliation(s)
- Mookambika Ramya Bai
- Department of Foods, Nutrition & Dietetics Research, Madras Diabetes Research Foundation, Chennai, India
- Department of Biotechnology, Sastra University, Thanjavur, India
| | - Kuzhandaivelu Abirami
- Department of Foods, Nutrition & Dietetics Research, Madras Diabetes Research Foundation, Chennai, India
| | - Rajagopal Gayathri
- Department of Foods, Nutrition & Dietetics Research, Madras Diabetes Research Foundation, Chennai, India
| | | | - Shanmugam Shobana
- Department of Foods, Nutrition & Dietetics Research, Madras Diabetes Research Foundation, Chennai, India
- Department of Diabetes Food Technology, Madras Diabetes Research Foundation, Chennai, India
| | - Lakshmi Priya Nagarajan
- Department of Foods, Nutrition & Dietetics Research, Madras Diabetes Research Foundation, Chennai, India
| | - Geetha Gunasekaran
- Department of Foods, Nutrition & Dietetics Research, Madras Diabetes Research Foundation, Chennai, India
| | - Gayathri Nagamuthu
- Department of Foods, Nutrition & Dietetics Research, Madras Diabetes Research Foundation, Chennai, India
| | - Hudgekar Madhav Malini
- Department of Foods, Nutrition & Dietetics Research, Madras Diabetes Research Foundation, Chennai, India
| | - Kuppan Gokulakrishnan
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - Narasimhan Sandhya
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - Ramajeevan Ganesh Jeevan
- Department of Foods, Nutrition & Dietetics Research, Madras Diabetes Research Foundation, Chennai, India
| | - Ranjit Mohan Anjana
- Department of Diabetology, Madras Diabetes Research Foundation, ICMR Centre for Advanced Research in Diabetes, Dr. Mohan's Diabetes Specialties Centre, IDF Centre Excellence in Diabetes, Chennai, India
| | - Ranjit Unnikrishnan
- Department of Diabetology, Madras Diabetes Research Foundation, ICMR Centre for Advanced Research in Diabetes, Dr. Mohan's Diabetes Specialties Centre, IDF Centre Excellence in Diabetes, Chennai, India
| | - Kamala Krishnaswamy
- Department of Foods, Nutrition & Dietetics Research, Madras Diabetes Research Foundation, Chennai, India
| | - Vasudevan Sudha
- Department of Foods, Nutrition & Dietetics Research, Madras Diabetes Research Foundation, Chennai, India
| | - Viswanathan Mohan
- Department of Diabetology, Madras Diabetes Research Foundation, ICMR Centre for Advanced Research in Diabetes, Dr. Mohan's Diabetes Specialties Centre, IDF Centre Excellence in Diabetes, Chennai, India
| |
Collapse
|
15
|
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
|
16
|
Martino M, Galderisi A, Evans-Molina C, Dayan C. Revisiting the Pattern of Loss of β-Cell Function in Preclinical Type 1 Diabetes. Diabetes 2024; 73:1769-1779. [PMID: 39106185 DOI: 10.2337/db24-0163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/25/2024] [Indexed: 08/09/2024]
Abstract
Type 1 diabetes (T1D) results from β-cell destruction due to autoimmunity. It has been proposed that β-cell loss is relatively quiescent in the early years after seroconversion to islet antibody positivity (stage 1), with accelerated β-cell loss only developing around 6-18 months prior to clinical diagnosis. This construct implies that immunointervention in this early stage will be of little benefit, since there is little disease activity to modulate. Here, we argue that the apparent lack of progression in early-stage disease may be an artifact of the modality of assessment used. When substantial β-cell function remains, the standard assessment, the oral glucose tolerance test, represents a submaximal stimulus and underestimates the residual function. In contrast, around the time of diagnosis, glucotoxicity exerts a deleterious effect on insulin secretion, giving the impression of disease acceleration. Once glucotoxicity is relieved by insulin therapy, β-cell function partially recovers (the honeymoon effect). However, evidence from recent trials suggests that glucose control has little effect on the underlying disease process. We therefore hypothesize that the autoimmune destruction of β-cells actually progresses at a more or less constant rate through all phases of T1D and that early-stage immunointervention will be both beneficial and desirable. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Mariangela Martino
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K
- PhD Program in Immunology, Molecular Medicine, and Applied Biotechnologies, University of Rome "Tor Vergata," Rome, Italy
| | | | - Carmella Evans-Molina
- Indiana University School of Medicine, Indianapolis, IN
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
- Richard L. Roudebush VA Medical Center, Indianapolis, IN
| | - Colin Dayan
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K
| |
Collapse
|
17
|
Kim JY, Lee J, Kim SG, Kim NH. Recent Glycemia Is a Major Determinant of β-Cell Function in Type 2 Diabetes Mellitus. Diabetes Metab J 2024; 48:1135-1146. [PMID: 38889769 PMCID: PMC11621653 DOI: 10.4093/dmj.2023.0359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/26/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGRUOUND Progressive deterioration of β-cell function is a characteristic of type 2 diabetes mellitus (T2DM). We aimed to investigate the relative contributions of clinical factors to β-cell function in T2DM. METHODS In a T2DM cohort of 470 adults (disease duration 0 to 41 years), β-cell function was estimated using insulinogenic index (IGI), disposition index (DI), oral disposition index (DIO), and homeostasis model assessment of β-cell function (HOMA-B) derived from a 75 g oral glucose tolerance test (OGTT). The relative contributions of age, sex, disease duration, body mass index, glycosylated hemoglobin (HbA1c) levels (at the time of the OGTT), area under the curve of HbA1c over time (HbA1c AUC), coefficient of variation in HbA1c (HbA1c CV), and antidiabetic agents use were compared by standardized regression coefficients. Longitudinal analyses of these indices were also performed. RESULTS IGI, DI, DIO, and HOMA-B declined over time (P<0.001 for all). Notably, HbA1c was the most significant factor affecting IGI, DI, DIO, and HOMA-B in the multivariable regression analysis. Compared with HbA1c ≥9%, DI was 1.9-, 2.5-, 3.7-, and 5.5-fold higher in HbA1c of 8%-<9%, 7%-<8%, 6%-<7%, and <6%, respectively, after adjusting for confounding factors (P<0.001). Conversely, β-cell function was not affected by the type or duration of antidiabetic agents, HbA1c AUC, or HbA1c CV. The trajectories of the IGI, DI, DIO, and HOMA-B mirrored those of HbA1c. CONCLUSION β-Cell function declines over time; however, it is flexible, being largely affected by recent glycemia in T2DM.
Collapse
Affiliation(s)
- Ji Yoon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jiyoon Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Sin Gon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Nam Hoon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Kim MK, Kwon HS, Baek KH, Song KH. Bile acids, fibroblast growth factor-19, and glucagon-like peptide-1 levels in the long term after bariatric surgery. Asian J Surg 2024:S1015-9584(24)02313-3. [PMID: 39424505 DOI: 10.1016/j.asjsur.2024.10.004] [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: 01/19/2024] [Revised: 03/28/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024] Open
Abstract
OBJECTIVES Glucagon-like peptide-1(GLP-1) is a hormone often measured in the short-term following Roux-en-Y gastric bypass (RYGB) due to its elevation and association with improvement of glucose metabolism. We examined the durability of this effect in patients with type 2 diabetes mellitus (DM) in the long term after RYGB. METHODS Obese patients with type 2 DM who had received RYGB 10 years ago (n = 10) were enrolled and a meal tolerance test (MTT) was performed. A matched control group with type 2 DM (n = 5) underwent MTT. RESULTS Glucose levels during the MTT did not differ between patients with RYGB and the nonsurgical group. Insulin, C-peptide and GLP-1 levels during MTT were significantly higher in patients with RYGB compared with the nonsurgical group (Area under the curve [AUC] of insulin; 57.4 ± 22.9 vs. 27.7 ± 11.1 mIU/L•hr, P = 0.008; AUC of total GLP-1; 189.4 ± 74.72 vs. 52.13 ± 10.23 pM •hr, P = 0.002), and in particular, peak insulin, C-peptide and GLP-1 levels observed 30-45 min after eating were markedly different from those in the nonsurgical group. Bile acids (BAs) and fibroblast growth factor 19 (FGF-19) levels during MTT were higher in patients with RYGB compared with the nonsurgical group. Peak BAs and FGF-19 levels tended to be higher in the RYGB. CONCLUSIONS An enhanced GLP-1 response was noted 10 years after RYGB, strongly suggesting a durability of this effect. BAs and FGF-19 were increased in the RYGB group, but not as much as the pronounced increase in GLP-1 secretion.
Collapse
Affiliation(s)
- Mee Kyoung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 07345, South Korea
| | - Hyuk-Sang Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 07345, South Korea
| | - Ki-Hyun Baek
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 07345, South Korea
| | - Ki-Ho Song
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 07345, South Korea.
| |
Collapse
|
19
|
Yuzbashian E, Berg E, de Campos Zani SC, Chan CB. Cow's Milk Bioactive Molecules in the Regulation of Glucose Homeostasis in Human and Animal Studies. Foods 2024; 13:2837. [PMID: 39272602 PMCID: PMC11395457 DOI: 10.3390/foods13172837] [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: 07/03/2024] [Revised: 08/26/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Obesity disrupts glucose metabolism, leading to insulin resistance (IR) and cardiometabolic diseases. Consumption of cow's milk and other dairy products may influence glucose metabolism. Within the complex matrix of cow's milk, various carbohydrates, lipids, and peptides act as bioactive molecules to alter human metabolism. Here, we summarize data from human studies and rodent experiments illustrating how these bioactive molecules regulate insulin and glucose homeostasis, supplemented with in vitro studies of the mechanisms behind their effects. Bioactive carbohydrates, including lactose, galactose, and oligosaccharides, generally reduce hyperglycemia, possibly by preventing gut microbiota dysbiosis. Milk-derived lipids of the milk fat globular membrane improve activation of insulin signaling pathways in animal trials but seem to have little impact on glycemia in human studies. However, other lipids produced by ruminants, including polar lipids, odd-chain, trans-, and branched-chain fatty acids, produce neutral or contradictory effects on glucose metabolism. Bioactive peptides derived from whey and casein may exert their effects both directly through their insulinotropic effects or renin-angiotensin-aldosterone system inhibition and indirectly by the regulation of incretin hormones. Overall, the results bolster many observational studies in humans and suggest that cow's milk intake reduces the risk of, and can perhaps be used in treating, metabolic disorders. However, the mechanisms of action for most bioactive compounds in milk are still largely undiscovered.
Collapse
Affiliation(s)
- Emad Yuzbashian
- Department of Agriculture, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Emily Berg
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | | | - Catherine B Chan
- Department of Agriculture, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
20
|
Minato-Inokawa S, Tsuboi-Kaji A, Honda M, Takeuchi M, Kitaoka K, Kurata M, Wu B, Kazumi T, Fukuo K. Low muscle mass is associated with low insulin sensitivity, impaired pancreatic β cell function, and high glucose excursion in nondiabetic nonobese Japanese women. Metabol Open 2024; 23:100306. [PMID: 39188637 PMCID: PMC11347059 DOI: 10.1016/j.metop.2024.100306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/28/2024] Open
Abstract
Aim We tested whether skeletal muscle mass is associated with insulin sensitivity, pancreatic β-cell function, and postglucose glycemia. Methods Appendicular skeletal muscle mass (ASM) (relative to body size, %ASM) by DXA, surrogate measures of insulin sensitivity, insulin secretion and the disposition index (insulin sensitivity adjusted insulin secretion: a product of the insulinogenic index and Matsuda insulin sensitivity index) inferred from serum insulin kinetics during a 75 g oral glucose tolerance test (OGTT) were evaluated in 168 young and 65 middle-aged women, whose BMI averaged <23.0 kg/m2 and HbA1c ≦ 5.5 %. Results In two groups of women, %ASM was associated negatively with homeostasis model assessment insulin resistance (HOMA-IR) and 2-h insulin (both p < 0.01 or less). In middle-aged women not in young women, %ASM was associated inversely with the Matsuda index (p < 0.001). In middle-aged women only, it also showed a positive association with the disposition index (p = 0.02) and inverse associations with 1-h and 2-h glucose (both p < 0.01) and area under the glucose concentration curve during OGTT (p = 0.006). On multivariate linear regression analyses, 2-h insulin emerged as a determinant of %ASM independently of HOMA-IR in young women (standardized β: 0.287, p < 0.001, R2 = 0.077). In middle-aged women, the Matsuda index emerged as a determinant of %ASM (standardized β: 0.476, p < 0.001) independently of HOMA-IR, log ODI and AUCg and explained 21.3 % of %ASM variability. Post-glucose glycemia and AUCg were higher and log ODI was lower in middle-aged women with low compared with high %ASM. Conclusion Low skeletal muscle mass (relative to body size) was associated with low insulin sensitivity in young and middle-aged Japanese women who were neither obese nor diabetic. Middle-aged women with low muscle mass had low disposition index, an early marker of inadequate pancreatic β-cell compensation, and hence high glucose excursion. Low skeletal muscle mass may be associated with the development of type 2 diabetes at a much lower BMI in Japanese people.
Collapse
Affiliation(s)
- Satomi Minato-Inokawa
- Research Institute for Nutrition Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Laboratory of Community Health and Nutrition, Department of Bioscience, Graduate School of Agriculture, Ehime University, Matsuyama, Ehime, Japan
| | - Ayaka Tsuboi-Kaji
- Research Institute for Nutrition Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Nutrition, Osaka City Juso Hospital, Osaka, Japan
| | - Mari Honda
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Health, Sports, and Nutrition, Faculty of Health and Welfare, Kobe Women's University, Kobe, Hyogo, Japan
| | - Mika Takeuchi
- Research Institute for Nutrition Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Kaori Kitaoka
- Research Institute for Nutrition Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Advanced Epidemiology, Noncommunicable Disease (NCD) Epidemiology Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Miki Kurata
- Research Institute for Nutrition Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Food Sciences and Nutrition, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Bin Wu
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Endocrinology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Tsutomu Kazumi
- Research Institute for Nutrition Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Medicine, Hakuhoukai Kakogawa Hospital, Kakogawa, Hyogo, Japan
| | - Keisuke Fukuo
- Research Institute for Nutrition Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Food Sciences and Nutrition, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| |
Collapse
|
21
|
Abrams EJ, Jao J, Madlala HP, Zerbe A, Catalano P, Gerschenson M, Goedecke JH, Gomba Y, Josefson J, Kurland IJ, Legbedze J, McComsey GA, Matyesini S, Mukonda E, Robinson D, Myer L. An observational cohort study to investigate the impact of dolutegravir in pregnancy and its obesogenic effects on the metabolic health of women living with HIV and their children: Study protocol. PLoS One 2024; 19:e0307296. [PMID: 39159183 PMCID: PMC11332920 DOI: 10.1371/journal.pone.0307296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 08/21/2024] Open
Abstract
INTRODUCTION Dolutegravir (DTG)-based antiretroviral therapy is the World Health Organization's preferred first-line regimen for all persons with HIV, including pregnant women. While DTG has been implicated as an obesogen associated with greater weight gain compared to other antiretrovirals, there is a paucity of data in pregnant women and their children. The Obesogenic oRigins of maternal and Child metabolic health Involving Dolutegravir (ORCHID) study is investigating associations between DTG, weight gain, and metabolic outcomes in the context of HIV. MATERIALS & METHODS ORCHID is a prospective observational study taking place in Cape Town, South Africa (NCT04991402). A total of 1920 pregnant women with and without HIV infection are being followed from ≤18 weeks gestational age to 24 months postpartum with their children. Participants attend eleven study visits: 3 antenatal, delivery, and 7 postnatal visits. Several embedded sub-studies address specific scientific aims. Primary outcome measurements in mothers include anthropometry, blood pressure, body composition, dysglycemia, insulin resistance (IR), and dyslipidemia. Other maternal measures include demographics, resting energy expenditure, viral load, physical activity, dietary intake, hepatic steatosis, and repository specimens. Sub-study measurements include markers of adipose inflammation, gut integrity, and satiety/hunger, subcutaneous adipose tissue morphology and mitochondrial function, and metabolomics. Primary outcome measurements in children include anthropometry, adipose tissue mass, dysglycemia, IR, and dyslipidemia. Other variables include fetal growth, birth outcomes, medical/breastfeeding history, caloric intake, neurodevelopment, and repository specimens. Sub-study measurements include metabolites/lipid subspecies in umbilical cord blood, as well as breast milk composition and DTG exposure. DISCUSSION ORCHID will play a pivotal role in defining obesogenic mechanisms and clinical consequences of DTG use in pregnancy in women with HIV and their children. It will provide insights into metabolic disease risk reduction in the context of HIV/DTG, identify intervention targets, and inform public health approaches to diminish chronic metabolic co-morbidities for women and children.
Collapse
Affiliation(s)
- Elaine J. Abrams
- ICAP at Columbia University, Mailman School of Public Health, Columbia University, New York, NY, United States of America
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States of America
- Department of Pediatrics, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, United States of America
| | - Jennifer Jao
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Hlengiwe P. Madlala
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Western Cape, Cape Town, South Africa
| | - Allison Zerbe
- ICAP at Columbia University, Mailman School of Public Health, Columbia University, New York, NY, United States of America
| | - Patrick Catalano
- Maternal Infant Research Institute, Obstetrics and Gynecology Research, Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University School of Medicine, Tufts University, Boston, MA, United States of America
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States of America
| | - Julia H. Goedecke
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa
- Health through Physical Activity, Lifestyle and Sport Research Centre (HPALS), Division of Physiological Sciences, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Yolanda Gomba
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Western Cape, Cape Town, South Africa
| | - Jami Josefson
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Irwin J. Kurland
- Department of Medicine, Stable Isotope and Metabolomics Core Facility, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Justine Legbedze
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Grace A. McComsey
- University Hospitals Health System, Cleveland, OH, United States of America
- Department of Medicine and Pediatrics of Case Western Reserve University, Cleveland, OH, United States of America
| | - Sandisiwe Matyesini
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Western Cape, Cape Town, South Africa
| | - Elton Mukonda
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Western Cape, Cape Town, South Africa
| | - Daniel Robinson
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Landon Myer
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Western Cape, Cape Town, South Africa
| |
Collapse
|
22
|
Bar Ziv O, Cahn A, Jansen T, Istomin V, Kedem E, Olshtain-Pops K, Israel S, Oster Y, Orenbuch-Harroch E, Korem M, Strahilevitz J, Levy I, Valdés-Mas R, Ivanova V, Elinav E, Shahar E, Elinav H. Diagnosis and Risk Factors of Prediabetes and Diabetes in People Living With Human Immunodeficiency Virus: Evaluation of Clinical and Microbiome Parameters. J Infect Dis 2024; 230:411-420. [PMID: 38557867 DOI: 10.1093/infdis/jiae167] [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: 06/07/2023] [Revised: 03/08/2024] [Accepted: 03/28/2024] [Indexed: 04/04/2024] Open
Abstract
Diabetes mellitus (DM) is more common among people living with human immunodeficiency virus (PLWH) compared with healthy individuals. In a prospective multicenter study (N = 248), we identified normoglycemic (48.7%), prediabetic (44.4%), and diabetic (6.9%) PLWH. Glycosylated hemoglobin (HbA1c) and fasting blood glucose (FBG) sensitivity in defining dysglycemia was 96.8%, while addition of oral glucose tolerance test led to reclassification of only 4 patients. Inclusion of 93 additional PLWH with known DM enabled identification of multiple independent predictors of dysglycemia or diabetes: older age, higher body mass index, Ethiopian origin, HIV duration, lower integrase inhibitor exposure, and advanced disease at diagnosis. Shotgun metagenomic microbiome analysis revealed 4 species that were significantly expanded with hyperglycemia/hyperinsulinemia, and 2 species that were differentially more prevalent in prediabetic/diabetic PLWH. Collectively, we uncover multiple potential host and microbiome predictors of altered glycemic status in PLWH, while demonstrating that FBG and HbA1c likely suffice for diabetes screening. These potential diabetic predictors merit future prospective validation.
Collapse
Affiliation(s)
- Omer Bar Ziv
- Department of Military Medicine and "Zameret," Faculty of Medicine, Hebrew University, and Israel and Medical Corps, Israel Defense Forces
| | - Avivit Cahn
- Diabetes Unit, Department of Endocrinology and Metabolism, Hadassah Medical Center
- Faculty of Medicine, Hebrew University, Jerusalem
| | - Tallulah Jansen
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot
| | | | - Eynat Kedem
- Allergy, Immunology and AIDS Unit, Rambam Medical Center, Haifa
| | - Karen Olshtain-Pops
- Faculty of Medicine, Hebrew University, Jerusalem
- Hadassah AIDS Center, Department of Clinical Microbiology and Infectious Diseases, Hadassah Medical Center, Jerusalem
| | - Sarah Israel
- Faculty of Medicine, Hebrew University, Jerusalem
- Hadassah AIDS Center, Department of Clinical Microbiology and Infectious Diseases, Hadassah Medical Center, Jerusalem
| | - Yonatan Oster
- Faculty of Medicine, Hebrew University, Jerusalem
- Hadassah AIDS Center, Department of Clinical Microbiology and Infectious Diseases, Hadassah Medical Center, Jerusalem
| | - Efrat Orenbuch-Harroch
- Faculty of Medicine, Hebrew University, Jerusalem
- Hadassah AIDS Center, Department of Clinical Microbiology and Infectious Diseases, Hadassah Medical Center, Jerusalem
| | - Maya Korem
- Faculty of Medicine, Hebrew University, Jerusalem
- Hadassah AIDS Center, Department of Clinical Microbiology and Infectious Diseases, Hadassah Medical Center, Jerusalem
| | - Jacob Strahilevitz
- Faculty of Medicine, Hebrew University, Jerusalem
- Hadassah AIDS Center, Department of Clinical Microbiology and Infectious Diseases, Hadassah Medical Center, Jerusalem
| | - Itzchak Levy
- Infectious Diseases Unit, Sheba Medical Center, Tel Hashomer, Israel
| | - Rafael Valdés-Mas
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot
| | - Valeria Ivanova
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot
- Division of Microbiome and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Eduardo Shahar
- Allergy, Immunology and AIDS Unit, Rambam Medical Center, Haifa
| | - Hila Elinav
- Faculty of Medicine, Hebrew University, Jerusalem
- Hadassah AIDS Center, Department of Clinical Microbiology and Infectious Diseases, Hadassah Medical Center, Jerusalem
| |
Collapse
|
23
|
Minato-Inokawa S, Honda M, Tsuboi-Kaji A, Takeuchi M, Kitaoka K, Kurata M, Wu B, Kazumi T, Fukuo K. Associations of adipose insulin resistance index with pancreatic β cell function (inverse) and glucose excursion (positive) in young Japanese women. Sci Rep 2024; 14:18590. [PMID: 39127728 PMCID: PMC11316777 DOI: 10.1038/s41598-024-69181-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
The relationship of adipose tissue insulin resistance (AT-IR, a product of fasting insulin and free fatty acids) and homeostasis-model assessment-insulin resistance (HOMA-IR) to β-cell function was studied cross-sectionally in the setting of subtle glucose dysregulation. Associations of AT-IR and HOMA-IR with fasting and post-glucose glycemia and β-cell function inferred from serum insulin kinetics during a 75 g oral glucose tolerance test were studied in 168 young female Japanese students. β-cell function was evaluated by disposition index calculated as a product of the insulinogenic index (IGI) and Matsuda index. AT-IR, not HOMA-IR, showed positive associations with post-glucose glycemia and area under the glucose response curve although both indices were associated with fasting glycemia. HOMA-IR, not AT-IR, was associated positively with log IGI whereas both indices were inversely associated with Matsuda index. AT-IR, not HOMA-IR, showed inverse associations with log disposition index. Associations of adipose tissue insulin resistance with β-cell function (inverse) and glucose excursion in young Japanese women may suggest that lipotoxicity to pancreatic β-cells for decades may be associated with β cell dysfunction found in Japanese patients with type 2 diabetes. Positive association of HOMA-IR with insulinogenic index may be associated with compensatory increased insulin secretion.
Collapse
Affiliation(s)
- Satomi Minato-Inokawa
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-Cho, Nishinomiya, Hyogo, 663-8558, Japan
- Laboratory of Community Health and Nutrition, Department of Bioscience, Graduate School of Agriculture, Ehime University, Matsuyama, Ehime, Japan
| | - Mari Honda
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Health, Sports, and Nutrition, Faculty of Health and Welfare, Kobe Women's University, Kobe, Hyogo, Japan
| | - Ayaka Tsuboi-Kaji
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-Cho, Nishinomiya, Hyogo, 663-8558, Japan
- Department of Nutrition, Osaka City Juso Hospital, Osaka, Japan
| | - Mika Takeuchi
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-Cho, Nishinomiya, Hyogo, 663-8558, Japan
| | - Kaori Kitaoka
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-Cho, Nishinomiya, Hyogo, 663-8558, Japan
- Department of Advanced Epidemiology, Noncommunicable Disease (NCD) Epidemiology Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Miki Kurata
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-Cho, Nishinomiya, Hyogo, 663-8558, Japan
- Department of Food Sciences and Nutrition, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Bin Wu
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Endocrinology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Tsutomu Kazumi
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-Cho, Nishinomiya, Hyogo, 663-8558, Japan.
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan.
- Department of Medicine, Kohan Kakogawa Hospital, Kakogawa, Hyogo, Japan.
| | - Keisuke Fukuo
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-Cho, Nishinomiya, Hyogo, 663-8558, Japan
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Food Sciences and Nutrition, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| |
Collapse
|
24
|
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
|
25
|
Sawicki CM, Ren Y, Kanaya AM, Kandula N, Gadgil M, Liang L, Haslam DE, Bhupathiraju SN. Metabolite Profiles of Plant-Based Diets and Cardiometabolic Risk in the Mediators of Atherosclerosis in South Asians Living in America Study. J Nutr 2024; 154:2501-2513. [PMID: 38901635 PMCID: PMC11375466 DOI: 10.1016/j.tjnut.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/21/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Healthy plant-based diets have been associated with lower risk of type 2 diabetes (T2D). Metabolomics can be leveraged to identify potential pathways through which diet influences disease risk. OBJECTIVES This study aimed to identify profiles of serum metabolites reflective of plant-based diets of varying quality and examine associations with cardiometabolic risk and T2D. METHODS We included data from 687 participants of the Mediators of Atherosclerosis in South Asians Living in America (MASALA) cohort. An overall plant-based diet index (PDI), healthy PDI (hPDI), and unhealthy PDI (uPDI) were estimated from food frequency questionnaires. Serum metabolites were assayed using ultraperformance liquid chromatography mass spectrometry. Elastic net regression was used to identify sets of metabolites predictive of each diet index, and metabolite profile scores were calculated as the weighted sum of the selected metabolites. Cross-sectional associations between metabolite profile scores and cardiometabolic measures and prospective associations with incident T2D were evaluated with multivariable-adjusted linear and logistic regressions. RESULTS Metabolite profiles for PDI, hPDI, and uPDI consisted of n = 51, 55, and 45 metabolites, respectively. Metabolites strongly positively correlated with diet indices included phosphatidylcholine (16:0/18:3) for PDI, phosphatidylethanolamine (20:1/20:4) and pantothenate for hPDI, and lysophosphatidylglycerol (18:2/0:0), proline, and lauric acid for uPDI. Higher metabolite profile scores for PDI and hPDI were associated with lower glycemia and lipids measures, whereas a higher uPDI metabolite score was associated with higher triglycerides and lower low density lipoprotein cholesterol and high density lipoprotein cholesterol. A higher metabolite score for hPDI was additionally associated with lower adiposity measures, higher liver fat attenuation, higher adiponectin, lower odds of overweight (odds ratio [OR]: 0.64; 95% confidence interval [CI]: 0.51, 0.81) and obesity (OR: 0.59; 95% CI: 0.48, 0.74), and lower odds of incident T2D (OR: 0.66; 95% CI: 0.45, 0.97). CONCLUSIONS Metabolite profiles of different plant-based diets were identified. Metabolite profiles of overall and healthy plant-based diets were associated with favorable cardiometabolic risk profiles.
Collapse
Affiliation(s)
- Caleigh M Sawicki
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Yin Ren
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Alka M Kanaya
- Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Namratha Kandula
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Meghana Gadgil
- Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Liming Liang
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Danielle E Haslam
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Shilpa N Bhupathiraju
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States.
| |
Collapse
|
26
|
Mitchell CM, Stinson EJ, Chang DC, Krakoff J. A mixed meal tolerance test predicts onset of type 2 diabetes in Southwestern Indigenous adults. Nutr Diabetes 2024; 14:50. [PMID: 38987291 PMCID: PMC11237083 DOI: 10.1038/s41387-024-00269-3] [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: 05/05/2023] [Revised: 01/05/2024] [Accepted: 02/23/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND/OBJECTIVE To identify predictors of incident type 2 diabetes using a mixed meal tolerance test (MMTT). METHODS Adult Indigenous Americans without diabetes (n = 501) from a longitudinal cohort underwent at baseline a 4-h MMTT, measures of body composition, an oral glucose tolerance test, an intravenous glucose tolerance test for acute insulin response (AIR), and a hyperinsulinemic-euglycemic clamp for insulin action (M). Plasma glucose responses from the MMTT were quantified by the total and incremental area under the curve (AUC/iAUC). RESULTS At follow-up (median time 9.6 [inter-quartile range: 5.6-13.5] years), 169 participants were diagnosed with diabetes. Unadjusted Cox proportional hazards models, glucose AUC180-min (HR: 1.98, 95% CI: 1.67, 2.34, p < 0.0001), AUC240-min (HR: 1.93, 95% CI: 1.62, 2.31, p < 0.0001), and iAUC180-min (HR: 1.43, 95% CI: 1.20, 1.71, p < 0.0001) were associated with an increased risk of diabetes. After adjustment for covariates (age, sex, body fat percentage, M, AIR, Indigenous American heritage) in three subsequent models, AUC180-min (HR: 1.44, 95% CI: 1.10, 1.88, p = 0.007) and AUC240-min (HR: 1.41, 95% CI: 1.09, 1.84, p < 0.01) remained associated with increased risk of diabetes. CONCLUSIONS Glucose responses to a mixed meal predicted the development of type 2 diabetes. This indicates that a mixed nutritional challenge provides important information on disease risk. CLINICAL TRIAL REGISTRY ClinicalTrials.gov identifier : NCT00340132, NCT00339482.
Collapse
Affiliation(s)
- Cassie M Mitchell
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA.
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, USA.
| | - Emma J Stinson
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Douglas C Chang
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Jonathan Krakoff
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| |
Collapse
|
27
|
Healey E, Kohane I. Model-Based Insulin Sensitivity and Beta-Cell Function Estimation from Daily Continuous Glucose Monitoring. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-4. [PMID: 40038964 DOI: 10.1109/embc53108.2024.10781685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Type 2 diabetes (T2D) is a prevalent chronic illness with many different options for treatment management. Continuous glucose monitors (CGM) offer physiological data that clinicians can access when making treatment decisions. However, the utility of CGM in management of T2D remains an active area of research. In our work, we demonstrate the feasibility of exploiting raw daily CGM data to estimate the physiological parameters of insulin sensitivity and beta-cell function that correlate with estimates derived from laboratory findings. We use a peak extraction algorithm to extract peaks from daily CGM data and implement a model-based approach to infer physiological parameters. We demonstrate that the inferred parameter estimates of insulin sensitivity and beta-cell function correlate to the ground truth measurements as determined by an oral glucose tolerance test (OGTT).
Collapse
|
28
|
van den Burg EL, Schoonakker MP, van Peet PG, van den Akker-van Marle EM, Lamb HJ, Longo VD, Numans ME, Pijl H. Integration of a fasting-mimicking diet programme in primary care for type 2 diabetes reduces the need for medication and improves glycaemic control: a 12-month randomised controlled trial. Diabetologia 2024; 67:1245-1259. [PMID: 38546821 PMCID: PMC11153305 DOI: 10.1007/s00125-024-06137-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 02/22/2024] [Indexed: 06/06/2024]
Abstract
AIMS/HYPOTHESIS The aim of this study was to evaluate the impact on metabolic control of periodic use of a 5-day fasting-mimicking diet (FMD) programme as an adjunct to usual care in people with type 2 diabetes under regular primary care surveillance. METHODS In this randomised, controlled, assessor-blinded trial, people with type 2 diabetes using metformin as the only glucose-lowering drug and/or diet for glycaemic control were randomised to receive 5-day cycles of an FMD monthly as an adjunct to regular care by their general practitioner or to receive regular care only. The primary outcomes were changes in glucose-lowering medication (as reflected by the medication effect score) and HbA1c levels after 12 months. Moreover, changes in use of glucose-lowering medication and/or HbA1c levels in individual participants were combined to yield a clinically relevant outcome measure ('glycaemic management'), which was categorised as improved, stable or deteriorated after 1 year of follow-up. Several secondary outcome measures were also examined, including changes in body weight. RESULTS One hundred individuals with type 2 diabetes, age 18-75 years, BMI ≥27 kg/m2, were randomised to the FMD group (n=51) or the control group (n=49). Eight FMD participants and ten control participants were lost to follow-up. Intention-to-treat analyses, using linear mixed models, revealed adjusted estimated treatment effects for the medication effect score (-0.3; 95% CI -0.4, -0.2; p<0.001), HbA1c (-3.2 mmol/mol; 95% CI -6.2, -0.2 and -0.3%; 95% CI -0.6, -0.0; p=0.04) and body weight (-3.6 kg; 95% CI -5.2, -2.1; p<0.001) at 12 months. Glycaemic management improved in 53% of participants using FMD vs 8% of control participants, remained stable in 23% vs 33%, and deteriorated in 23% vs 59% (p<0.001). CONCLUSIONS/INTERPRETATION Integration of a monthly FMD programme in regular primary care for people with type 2 diabetes who use metformin as the only glucose-lowering drug and/or diet for glycaemic control reduces the need for glucose-lowering medication, improves HbA1c despite the reduction in medication use, and appears to be safe in routine clinical practice. TRIAL REGISTRATION ClinicalTrials.gov NCT03811587 FUNDING: The project was co-funded by Health~Holland, Top Sector Life Sciences & Health, the Dutch Diabetes Foundation and L-Nutra.
Collapse
Affiliation(s)
- Elske L van den Burg
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, the Netherlands.
| | - Marjolein P Schoonakker
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - Petra G van Peet
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - Elske M van den Akker-van Marle
- Department of Biomedical Data Sciences, Medical Decision Making, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - Hildo J Lamb
- Department of Radiology, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - Valter D Longo
- Longevity Institute, Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- FIRC Institute of Molecular Oncology, Milan, Italy
| | - Mattijs E Numans
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - Hanno Pijl
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
- Department of Internal Medicine, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| |
Collapse
|
29
|
Sabatini S, Nolan JJ, O'Donoghue G, Kennedy A, Petrie J, Walker M, O'Gorman DJ, Gastaldelli A. Baseline phenotypes with preserved β-cell function and high insulin concentrations have the best improvements in glucose tolerance after weight loss: results from the prospective DEXLIFE and EGIR-RISC studies. Metabolism 2024; 155:155910. [PMID: 38599278 DOI: 10.1016/j.metabol.2024.155910] [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: 11/27/2023] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND Weight loss and lifestyle intervention improve glucose tolerance delaying the onset of type 2 diabetes (T2D), but individual responses are highly variable. Determining the predictive factors linked to the beneficial effects of weight loss on glucose tolerance could provide tools for individualized prevention plans. Thus, the aim was to investigate the relationship between pre-intervention values of insulin sensitivity and secretion and the improvement in glucose metabolism after weight loss. METHODS In the DEXLIFE cohort (373 individuals at high risk of T2D, assigned 3:1 to a 12-week lifestyle intervention or a control arm, Trial Registration: ISRCTN66987085), K-means clustering and logistic regression analysis were performed based on pre-intervention indices of insulin sensitivity, insulin secretion (AUC-I), and glucose-stimulated insulin response (ratio of incremental areas of insulin and glucose, iAUC I/G). The response to the intervention was evaluated in terms of reduction of OGTT-glucose concentration. Clusters' validation was done in the prospective EGIR-RISC cohort (n = 1538). RESULTS Four replicable clusters with different glycemic and metabolomic profiles were identified. Individuals had similar weight loss, but improvement in glycemic profile and β-cell function was different among clusters, highly depending on pre-intervention insulin response to OGTT. Pre-intervention high insulin response was associated with the best improvement in AUC-G, while clusters with low AUC-I and iAUC I/G showed no beneficial effect of weight loss on glucose control, as also confirmed by the logistic regression model. CONCLUSIONS Individuals with preserved β-cell function and high insulin concentrations at baseline have the best improvement in glucose tolerance after weight loss.
Collapse
Affiliation(s)
- Silvia Sabatini
- Institute of Clinical Physiology, National Research Council, CNR, Pisa, Italy
| | - John J Nolan
- Department of Clinical Medicine, Trinity College Dublin, Ireland
| | - Grainne O'Donoghue
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland
| | - Aileen Kennedy
- School of Biological, Health and Sports Sciences, Technological University Dublin, Dublin, Ireland
| | - John Petrie
- School of Health and Wellbeing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Mark Walker
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Donal J O'Gorman
- School of Health and Human Performance, Dublin City University, Glasnevin, Dublin, Ireland
| | - Amalia Gastaldelli
- Institute of Clinical Physiology, National Research Council, CNR, Pisa, Italy.; Diabetes Division, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
30
|
Gupta Y, Goyal A, Ambekar S, Kalaivani M, Bhatla N, Tandon N. Postpartum glycemic and cardiometabolic profile of women testing positive for gestational diabetes mellitus by International Association of Diabetes and Pregnancy Study Groups (IADPSG) but negative by alternate criteria: Insights from CHIP-F study. Diabetes Metab Syndr 2024; 18:103064. [PMID: 38959545 DOI: 10.1016/j.dsx.2024.103064] [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: 10/24/2023] [Revised: 06/16/2024] [Accepted: 06/19/2024] [Indexed: 07/05/2024]
Abstract
OBJECTIVE To evaluate burden of postpartum diabetes and other cardiometabolic risk factors among women who test positive for gestational diabetes mellitus (GDM) by International Association of Diabetes and Pregnancy Study Groups (IADPSG) criteria, but negative by alternate criteria. METHODS This prospective cross-sectional study was conducted from 2019 to 2022 and is a sub-study of the CHIP-F cohort (Cohort Study of Indian Women with Hyperglycemia in Pregnancy and their Families). RESULTS Study participants (n = 826; 183 with normoglycemia and 643 with GDM using IADPSG criteria) were evaluated at a median (IQR) postpartum interval of 31 (21-45) months. Using the United Kingdom National Institute of Health and Care Excellence (UK NICE), Canadian Diabetes Association (CDA), and Diabetes in Pregnancy Study Group India (DIPSI) criteria, 251 (39.0 %), 148 (23.0 %) and 384 (59.7 %) women who tested positive for GDM by IADPSG criteria, would have tested negative. The incidence of postpartum diabetes among such women was 30.4, 34.3, and 48.2 per 1000 women-years, respectively, which was significantly higher than those testing negative by both IADPSG and UK NICE (5.0 per 1000 women-years), IADPSG and CDA (9.2/1000 women-years) and IADPSG and DIPSI criteria (5.0/1000 women-years). The burden of obesity and metabolic syndrome was also significantly higher in such women. CONCLUSIONS We found a significant burden of postpartum diabetes and cardiometabolic risk factors among women who tested positive for GDM by IADPSG, but negative by alternate criteria. There are potential clinical implications of a "failed" diagnosis for future cardiometabolic diseases that need to be carefully examined.
Collapse
Affiliation(s)
- Yashdeep Gupta
- Department of Endocrinology & Metabolism, All India Institute of Medical Sciences, New Delhi, India.
| | - Alpesh Goyal
- Department of Endocrinology & Metabolism, All India Institute of Medical Sciences, New Delhi, India
| | - Samita Ambekar
- Department of Endocrinology & Metabolism, All India Institute of Medical Sciences, New Delhi, India
| | - Mani Kalaivani
- Department of Statistics, All India Institute of Medical Sciences, New Delhi, India
| | - Neerja Bhatla
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, New Delhi, India
| | - Nikhil Tandon
- Department of Endocrinology & Metabolism, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
31
|
Stinson SE, Fernández de Retana Alzola I, Brünner Hovendal ED, Lund MAV, Fonvig CE, Holm LA, Jonsson AE, Frithioff-Bøjsøe C, Christiansen M, Pedersen O, Ängquist L, Sørensen TIA, Holst JJ, Hartmann B, Holm JC, Hansen T. Altered Glucagon and GLP-1 Responses to Oral Glucose in Children and Adolescents With Obesity and Insulin Resistance. J Clin Endocrinol Metab 2024; 109:1590-1600. [PMID: 38087928 PMCID: PMC11099488 DOI: 10.1210/clinem/dgad728] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Indexed: 05/18/2024]
Abstract
CONTEXT Pediatric obesity is characterized by insulin resistance, yet it remains unclear whether insulin resistance contributes to abnormalities in glucagon and incretin secretion. OBJECTIVE To examine whether fasting and stimulated glucagon, glucagon-like peptide-1 (GLP-1), and glucose-dependent insulinotropic polypeptide (GIP) concentrations differ between children and adolescents with obesity and insulin resistance (OIR), obesity and normal insulin sensitivity (OIS), and controls with normal weight (NW). METHODS 80 (34 boys) children and adolescents, aged 7-17 years with OIR (n = 22), OIS (n = 22), and NW (n = 36) underwent an oral glucose tolerance test with measurements of serum insulin, plasma glucose, glucagon, total GLP-1, and total GIP. Homeostatic model assessment of insulin resistance (HOMA-IR), single point insulin sensitivity estimator (SPISE), Matsuda index, insulinogenic index (IGI), and oral disposition index (ODI) were calculated. RESULTS Fasting concentrations of glucagon and GLP-1 were higher in the OIR group, with no significant differences for GIP. The OIR group had higher glucagon total area under the curve (tAUC0-120) and lower GLP-1 incremental AUC (iAUC0-120), with no significant differences in GIP iAUC0-120. Higher fasting glucagon was associated with higher HOMA-IR, lower Matsuda index, lower SPISE, higher IGI, and higher plasma alanine transaminase, whereas higher fasting GLP-1 was associated with higher HOMA-IR, lower Matsuda index, and lower ODI. Higher glucagon tAUC0-120 was associated lower SPISE and lower Matsuda index, whereas lower GLP-1 iAUC0-120 was associated with a higher HOMA-IR, lower Matsuda index, and lower ODI. CONCLUSION Children and adolescents with OIR have elevated fasting concentrations of glucagon and GLP-1, higher glucagon and lower GLP-1 responses during an OGTT compared to those with OIS and NW. In contrast, individuals with OIS have similar hormone responses to those with NW.
Collapse
Affiliation(s)
- Sara Elizabeth Stinson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ierai Fernández de Retana Alzola
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Emilie Damgaard Brünner Hovendal
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Holbæk Hospital, 4300 Holbæk, Denmark
| | - Morten Asp Vonsild Lund
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Holbæk Hospital, 4300 Holbæk, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Cilius Esmann Fonvig
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Holbæk Hospital, 4300 Holbæk, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Louise Aas Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Holbæk Hospital, 4300 Holbæk, Denmark
| | - Anna Elisabet Jonsson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christine Frithioff-Bøjsøe
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Holbæk Hospital, 4300 Holbæk, Denmark
| | - Michael Christiansen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department for Congenital Disorders, Statens Serum Institute, 2300 Copenhagen, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Center for Clinical Metabolic Research, Herlev-Gentofte University Hospital, 2900 Copenhagen, Denmark
| | - Lars Ängquist
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Thorkild I A Sørensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, 1353 Copenhagen, Denmark
| | - Jens Juul Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Bolette Hartmann
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jens-Christian Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Holbæk Hospital, 4300 Holbæk, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
32
|
Hani NS, Vajravelu ME, Meijer JL, McCaffery H, Sturza J, Dhadphale E, Lee JM. The Reproducibility and Reliability of Insulin Sensitivity and Secretion Indices in Children and Adolescents. Pediatr Diabetes 2024; 2024:2136173. [PMID: 39726936 PMCID: PMC11671114 DOI: 10.1155/2024/2136173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2024] Open
Abstract
Context Insulin sensitivity and secretion indices can be useful tools in understanding insulin homeostasis in children at risk for diabetes. There have been few studies examining the reproducibility of these measures in pediatrics. Objective To determine whether fasting or oral glucose tolerance test (OGTT)-derived insulin measures would be more reproducible and whether there would be differences based on weight, sex, race, and pubertal status. Design Observational study. Setting Clinical research unit. Patients or Other Participants Two hundred fifty-seven overweight/obese (BMI ≥ 85th%, n = 186) and normal weight (BMI < 85th%, n = 71) children without diabetes between ages of 8 and 17 were included in the study. Methods OGTT tests performed in study participants at two separate visits within a 3-week period. We performed two formal oral glucose tolerance tests within a 3-week period. The reproducibility of fasting measures was compared with OGTT-derived measures by weight categories and compared by weight, sex, race, and pubertal status. Comparisons were made between the correlation coefficients of fasting vs. OGTT-derived measures and between normal weight vs. obese/overweight participants, male vs. female, White vs. Black, and pre- vs. post-midpubertal. Intraclass correlation coefficients were calculated for each comparison as well. Results For insulin sensitivity, the OGTT-derived measure was more reproducible than the fasting measures. There were no significant differences in reproducibility in the overweight/obese population compared to the normal weight population nor by sex, race, or pubertal status. Conclusions Nonfasting insulin sensitivity measures are more reproducible than fasting insulin sensitivity measures, regardless of weight category. Insulin secretion measures have poor reproducibility overall. Weight status, sex, race, and midpubertal stage do not impact the reproducibility of insulin sensitivity and secretion measures.
Collapse
Affiliation(s)
- Nellie Said Hani
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Michigan, Ann Arbor, USA
| | - Mary Ellen Vajravelu
- Division of Pediatric Endocrinology, Diabetes and Metabolism, UPMC—Children’s Hospital of Pittsburgh, Pittsburgh, USA
| | - Jennifer L Meijer
- Geisel School of Medicine, Department of Pediatrics, Dartmouth College, Hanover, USA
| | - Harlan McCaffery
- Susan B. Meister Child Health Evaluation and Research Center, Division of Pediatric Endocrinology, University of Michigan, Ann Arbor, USA
| | - Julie Sturza
- Susan B. Meister Child Health Evaluation and Research Center, Division of Pediatric Endocrinology, University of Michigan, Ann Arbor, USA
| | - Emily Dhadphale
- Susan B. Meister Child Health Evaluation and Research Center, Division of Pediatric Endocrinology, University of Michigan, Ann Arbor, USA
| | - Joyce M Lee
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Michigan, Ann Arbor, USA
| |
Collapse
|
33
|
Staimez LR, Dutta A, Beyh YS, Gupta R, Noule HK, Sapna V, Deepa K, Stein AD, Narayan KV, Prabhakaran D, Kalpana B, Prabhakaran P. Pancreatic Beta Cell Function in Infants Varies by Maternal Weight. Metabolites 2024; 14:208. [PMID: 38668336 PMCID: PMC11052198 DOI: 10.3390/metabo14040208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/19/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
The Asian Indian Beta Cell function (ABCs) in Infants Study examined the associations of maternal weight on infant pancreatic beta cell function across 7 months postpartum. Pregnant women aged 18-35 years were recruited in Hyderabad, India. Women were classified by first trimester weight as underweight (UW), BMI < 18.5 kg/m2; normal weight (NW), BMI 18.5-22.9 kg/m2; or overweight (OW), BMI 23.0 through <28.5 kg/m2. At age > 7 months, infants had an oral glucose tolerance test (OGTT, 1.75 g glucose/kg bodyweight) following a 3 h fast. Infant blood samples were assayed for C-peptide and glucose. Infant beta cell function (HOMA2-B; disposition index, DI) and insulin resistance (HOMA2-IR) were compared across maternal weight groups. Mothers (UW n = 63; NW n = 43; OW n = 29) had similar age at delivery and second trimester 50 g glucose challenge test results. Cord HOMA2-B values were 51% greater for IUW (83.5, SD 55.2) and 44% greater for IOW (79.9, SD 60.8) vs. INW (55.4, SD 51.5), forming a U-shaped relationship between maternal weight and HOMA2-B. No qualitative differences in HOMA2-IR were found at birth. However, at 7 months postpartum, HOMA2-IR changed most within IUW (-64% median reduction) and changed the least in IOW (-7% median reduction). At seven months postpartum, DI was higher in IUW vs. the other groups (geometric mean IUW 1.9 SD 2.5; INW 1.3 SD 2.6 or vs. IOW mean 1.2 SD 3.7), reflecting a +49% difference in DI. Evidence from this study illustrates adaptations in the pancreatic functional response of infants associated with the maternal nutritional environment.
Collapse
Affiliation(s)
- Lisa R. Staimez
- Emory Global Diabetes Research Center, Woodruff Health Sciences Center, Emory University, Atlanta, GA 30322, USA; (Y.S.B.)
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, USA
| | - Anubrati Dutta
- Centre for Chronic Disease Control, New Delhi 110016, Delhi, India; (A.D.); (R.G.); (D.P.); (P.P.)
| | - Yara S. Beyh
- Emory Global Diabetes Research Center, Woodruff Health Sciences Center, Emory University, Atlanta, GA 30322, USA; (Y.S.B.)
| | - Ruby Gupta
- Centre for Chronic Disease Control, New Delhi 110016, Delhi, India; (A.D.); (R.G.); (D.P.); (P.P.)
| | - Hari Krishna Noule
- Centre for Chronic Disease Control, New Delhi 110016, Delhi, India; (A.D.); (R.G.); (D.P.); (P.P.)
- Share India, MediCiti Institute for Medical Sciences, Medchal 501401, Telangana, India; (V.S.); (B.K.)
- Administrative Staff College of India, Hyderabad 500034, Telanaga, India
| | - Vyakaranam Sapna
- Share India, MediCiti Institute for Medical Sciences, Medchal 501401, Telangana, India; (V.S.); (B.K.)
| | - Kothapally Deepa
- Share India, MediCiti Institute for Medical Sciences, Medchal 501401, Telangana, India; (V.S.); (B.K.)
| | - Aryeh D. Stein
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, USA
| | - K.M. Venkat Narayan
- Emory Global Diabetes Research Center, Woodruff Health Sciences Center, Emory University, Atlanta, GA 30322, USA; (Y.S.B.)
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, USA
| | - Dorairaj Prabhakaran
- Centre for Chronic Disease Control, New Delhi 110016, Delhi, India; (A.D.); (R.G.); (D.P.); (P.P.)
- Public Health Foundation of India, Gurgaon 110030, Haryana, India
| | - Basany Kalpana
- Share India, MediCiti Institute for Medical Sciences, Medchal 501401, Telangana, India; (V.S.); (B.K.)
| | - Poornima Prabhakaran
- Centre for Chronic Disease Control, New Delhi 110016, Delhi, India; (A.D.); (R.G.); (D.P.); (P.P.)
| |
Collapse
|
34
|
Lee MJ, Bae JH, Khang AR, Yi D, Yun MS, Kang YH. Triglyceride-glucose index predicts type 2 diabetes mellitus more effectively than oral glucose tolerance test-derived insulin sensitivity and secretion markers. Diabetes Res Clin Pract 2024; 210:111640. [PMID: 38548110 DOI: 10.1016/j.diabres.2024.111640] [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: 01/19/2024] [Revised: 03/01/2024] [Accepted: 03/25/2024] [Indexed: 04/01/2024]
Abstract
AIMS We explored the role of the triglyceride-glucose (TyG) index as an early and superior predictor of type 2 diabetes mellitus (T2DM) in individuals with normal glucose tolerance (NGT) using a community-based Korean cohort over 18 years. METHODS We retrospectively examined 6,072 adults with NGT from the Korean Genome and Epidemiology Study. Cox proportional hazard regression models were employed to evaluate the risk of incidence of T2DM and receiver operating characteristic analysis was used to calculate the area under the curve (AUC). RESULTS At baseline, the TyG index correlated with the homeostasis model assessment of insulin resistance (HOMA-IR) and the composite insulin sensitivity index (ISI) (β: 0.045, p < 0.001; β: -0.105, p < 0.001, respectively). Over the 18-year follow-up period, 999 individuals developed T2DM. An increase in the TyG quartile independently predicted the incidence of T2DM [hazard ratio, 2.36 (1.9-2.93) for Q4]. The AUC value of the TyG index was 0.642, the highest value among HOMA-IR and OGTT-derived insulin sensitivity and secretion markers. CONCLUSIONS The TyG index is associated with HOMA-IR and composite ISI even with NGT. The TyG index demonstrated independent predictability for T2DM incidence in individuals with NGT, better than OGTT-derived insulin sensitivity and secretion markers.
Collapse
Affiliation(s)
- Min Jin Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea; Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Ji Hyun Bae
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea; Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Ah Reum Khang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea; Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Dongwon Yi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea; Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Mi Sook Yun
- Division of Biostatistics, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Yang Ho Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea; Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea.
| |
Collapse
|
35
|
Bae JC, Wander PL, Lemaitre RN, Fretts AM, Sitlani CM, Bui HH, Thomas MK, Leonetti D, Fujimoto WY, Boyko EJ, Utzschneider KM. Associations of plasma sphingolipids with measures of insulin sensitivity, β-cell function, and incident diabetes in Japanese Americans. Nutr Metab Cardiovasc Dis 2024; 34:633-641. [PMID: 38161124 PMCID: PMC10922320 DOI: 10.1016/j.numecd.2023.10.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND AND AIMS To prospectively investigate associations of plasma sphingolipids with insulin sensitivity, β-cell function, and incident diabetes in the Japanese American Community Diabetes Study. METHODS AND RESULTS Baseline plasma samples from adults without diabetes (n = 349; mean age 56.7 years, 51 % men) were assayed for circulating ceramide and sphingomyelin species. Adjusted regression models examined cross-sectional and longitudinal associations with insulin sensitivity (HOMA2-%S), β-cell function (oral disposition index: DIo) and with incident diabetes over 5 years follow-up. Concentrations of four species (Ceramide C16:0, C18:0, C20:0, and C22:0) were inversely associated with HOMA2-%S at baseline (all P values < 0.05, Q values < 0.05) and change in HOMA2-%S over 5 years (all P values < 0.05, Q values < 0.05). No sphingolipids were associated with baseline or change in DIo. Of the four species associated with HOMA2-%S, only Ceramide C18:0 was significantly and positively associated with incident diabetes (RR/1SD 1.44, 95 % CI 1.10-1.80, P = 0.006, Q = 0.024). The association of plasma Ceramide C18:0 with the risk of diabetes was partially mediated by change in HOMA2-%S between baseline and 5 years (mediation proportion: 61.5 %, 95 % CI 21.1%-212.5 %). CONCLUSION Plasma Ceramide C18:0 was associated with higher risk of incident diabetes which was partially mediated through a decrease in insulin sensitivity between baseline and five years. Circulating Ceramide C18:0 could be a potential biomarker for identifying those at risk of developing diabetes.
Collapse
Affiliation(s)
- Ji Cheol Bae
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, South Korea
| | - Pandora L Wander
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Rozenn N Lemaitre
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Amanda M Fretts
- Department of Epidemiology, University of Washington, Seattle, WA, United States
| | - Colleen M Sitlani
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Hai H Bui
- Eli Lilly and Company, Indianapolis, IN, United States
| | | | - Donna Leonetti
- Department of Anthropology, University of Washington, Seattle, WA, United States
| | - Wilfred Y Fujimoto
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Edward J Boyko
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States; Department of Medicine, University of Washington, Seattle, WA, United States
| | - Kristina M Utzschneider
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States; Department of Medicine, University of Washington, Seattle, WA, United States.
| |
Collapse
|
36
|
Cao C, Koh HCE, Reeds DN, Patterson BW, Klein S, Mittendorfer B. Critical Evaluation of Indices Used to Assess β-Cell Function. Diabetes 2024; 73:391-400. [PMID: 38015795 PMCID: PMC10882145 DOI: 10.2337/db23-0613] [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: 08/02/2023] [Accepted: 11/18/2023] [Indexed: 11/30/2023]
Abstract
The assessment of β-cell function, defined as the relationship between insulin secretion rate (ISR) and plasma glucose, is not standardized and often involves any of a number of β-cell function indices. We compared β-cell function by using popular indices obtained during basal conditions and after glucose ingestion, including the HOMA-B index, the basal ISR (or plasma insulin)-to-plasma glucose concentration ratio, the insulinogenic and ISRogenic indices, the ISR (or plasma insulin)-to-plasma glucose concentration areas (or incremental areas) under the curve ratio, and the disposition index, which integrates a specific β-cell function index value with an estimate of insulin sensitivity, between lean people with normal fasting glucose (NFG) and normal glucose tolerance (NGT) (n = 50) and four groups of people with obesity (n = 188) with 1) NFG-NGT, 2) NFG and impaired glucose tolerance (IGT), 3) impaired fasting glucose (IFG) and IGT, and 4) type 2 diabetes. We also plotted the ISR-plasma glucose relationship before and after glucose ingestion and used a statistical mixed-effects model to evaluate group differences in this relationship (i.e., β-cell function). Index-based group differences in β-cell function produced contradicting results and did not reflect the group differences of the actual observed ISR-glucose relationship or, in the case of the disposition index, group differences in glycemic status. The discrepancy in results is likely due to incorrect mathematical assumptions that are involved in computing indices, which can be overcome by evaluating the relationship between ISR and plasma glucose with an appropriate statistical model. Data obtained with common β-cell function indices should be interpreted cautiously. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Chao Cao
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Han-Chow E. Koh
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Dominic N. Reeds
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Bruce W. Patterson
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Samuel Klein
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
- Sansum Diabetes Research Institute, Santa Barbara, CA
| | - Bettina Mittendorfer
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
- Departments of Medicine and Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
| |
Collapse
|
37
|
Aslamy A, Wood AC, Jensen ET, Bertoni AG, Sheridan PA, Wong KE, Ramesh G, Rotter JI, Chen YDI, Goodarzi MO. Increased Plasma Branched Short-Chain Fatty Acids and Improved Glucose Homeostasis: The Microbiome and Insulin Longitudinal Evaluation Study (MILES). Diabetes 2024; 73:385-390. [PMID: 37992186 PMCID: PMC10882143 DOI: 10.2337/db23-0401] [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: 05/19/2023] [Accepted: 11/18/2023] [Indexed: 11/24/2023]
Abstract
Short-chain fatty acids (SCFAs) have been extensively studied for potential beneficial roles in glucose homeostasis and risk of diabetes; however, most of this research has focused on butyrate, acetate, and propionate. The effect on metabolism of branched SCFAs (BSCFAs; isobutyrate, isovalerate, and methylbutyrate) is largely unknown. In a cohort of 219 non-Hispanic White participants and 126 African American participants, we examined the association of BSCFA with dysglycemia (prediabetes and diabetes) and oral glucose tolerance test-based measures of glucose and insulin homeostasis, as well as with demographic, anthropometric, lifestyle, and lipid traits, and other SCFAs. We observed a bimodal distribution of BSCFAs, with 25 individuals having high levels (H-BSCFA group) and 320 individuals having lower levels (L-BSCFA group). The prevalence of dysglycemia was lower in the H-BSCFA group compared with the L-BSCFA group (16% vs. 49%; P = 0.0014). This association remained significant after adjustment for age, sex, race, BMI, and levels of other SCFAs. Consistent with the lower rate of dysglycemia, fasting and postprandial glucose levels were lower and the disposition index was higher in the H-BSCFA group. Additional findings in H-BSCFA versus L-BSCFA included lower fasting and postprandial C-peptide levels and lower insulin clearance without differences in insulin levels, insulin sensitivity, insulin secretion, or other variables examined, including diet and physical activity. As one of the first human studies associating higher BSCFA levels with lower odds of dysglycemia and improved glucose homeostasis, this study sets the stage for further investigation of BSCFA as a novel target for prevention or treatment of diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Arianne Aslamy
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Alexis C. Wood
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX
| | - Elizabeth T. Jensen
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC
| | - Alain G. Bertoni
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC
| | | | | | - Gautam Ramesh
- School of Medicine, University of California San Diego, La Jolla, CA
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation and Department of Pediatrics, UCLA Medical Center, Harbor-Torrance, CA
| | - Yii-Der I. Chen
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation and Department of Pediatrics, UCLA Medical Center, Harbor-Torrance, CA
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
38
|
Ren S, Wu D, Li P. Evaluation of insulin secretion and insulin sensitivity in pregnant women: Application value of simple indices. Clin Chim Acta 2024; 554:117753. [PMID: 38185282 DOI: 10.1016/j.cca.2023.117753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 12/29/2023] [Accepted: 12/29/2023] [Indexed: 01/09/2024]
Abstract
The prevalence of gestational diabetes mellitus (GDM) is increasing annually, which poses substantial harm to the health of both mothers and children. Therefore, selection of clinically applicable and easily detectable indicators in the assessment of maternal insulin secretory function and insulin sensitivity in pregnant women undoubtedly holds great importance in evaluating the risk of GDM, guiding the choice of GDM therapy modalities, and improving the ability to provide early warning of adverse pregnancy outcomes. Compared with the classic clamp technique, many simple indices are more suited for use among pregnant women due to the low frequency of blood sampling and simple administration involved. While indices derived from fasting blood glucose and fasting insulin levels are most readily available, they are unable to provide information on the ability of insulin to manage the glucose load during pregnancy. Although the indices derived from the insulin and glucose values at each time point of the oral glucose tolerance test can provide a more comprehensive picture of the insulin sensitivity and insulin secretory function of the body, their application is constrained by the complexity of the procedure and associated high costs. Concomitantly, the findings from different studies are influenced by a variety of confounding factors, such as the gestational age during testing, race, and detection method. Furthermore, insulin secretory function and insulin sensitivity in pregnant women differ from those in non-pregnant women in that they change significantly with prolonged pregnancy; hence, there is an urgent need to develop a pregnancy-specific reference range. This article reviews the progress in the application of simple indices to help clinicians better understand their potential application in detecting GDM.
Collapse
Affiliation(s)
- Shuying Ren
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, People's Republic of China
| | - Dan Wu
- Department of Endocrinology, 242 Hospital Affilliated to Shenyang Medical College, Shenyang, Liaoning Province, People's Republic of China
| | - Ping Li
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, People's Republic of China.
| |
Collapse
|
39
|
Hampe CS, Shojaie A, Brooks-Worrell B, Dibay S, Utzschneider K, Kahn SE, Larkin ME, Johnson ML, Younes N, Rasouli N, Desouza C, Cohen RM, Park JY, Florez HJ, Valencia WM, Palmer JP, Balasubramanyam A. GAD65Abs Are Not Associated With Beta-Cell Dysfunction in Patients With T2D in the GRADE Study. J Endocr Soc 2024; 8:bvad179. [PMID: 38333889 PMCID: PMC10853002 DOI: 10.1210/jendso/bvad179] [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: 09/27/2023] [Indexed: 02/10/2024] Open
Abstract
Context Autoantibodies directed against the 65-kilodalton isoform of glutamic acid decarboxylase (GAD65Abs) are markers of autoimmune type 1 diabetes (T1D) but are also present in patients with Latent Autoimmune Diabetes of Adults and autoimmune neuromuscular diseases, and also in healthy individuals. Phenotypic differences between these conditions are reflected in epitope-specific GAD65Abs and anti-idiotypic antibodies (anti-Id) against GAD65Abs. We previously reported that 7.8% of T2D patients in the GRADE study have GAD65Abs but found that GAD65Ab positivity was not correlated with beta-cell function, glycated hemoglobin (HbA1c), or fasting glucose levels. Context In this study, we aimed to better characterize islet autoantibodies in this T2D cohort. This is an ancillary study to NCT01794143. Methods We stringently defined GAD65Ab positivity with a competition assay, analyzed GAD65Ab-specific epitopes, and measured GAD65Ab-specific anti-Id in serum. Results Competition assays confirmed that 5.9% of the patients were GAD65Ab positive, but beta-cell function was not associated with GAD65Ab positivity, GAD65Ab epitope specificity or GAD65Ab-specific anti-Id. GAD65-related autoantibody responses in GRADE T2D patients resemble profiles in healthy individuals (low GAD65Ab titers, presence of a single autoantibody, lack of a distinct epitope pattern, and presence of anti-Id to diabetes-associated GAD65Ab). In this T2D cohort, GAD65Ab positivity is likely unrelated to the pathogenesis of beta-cell dysfunction. Conclusion Evidence for islet autoimmunity in the pathophysiology of T2D beta-cell dysfunction is growing, but T1D-associated autoantibodies may not accurately reflect the nature of their autoimmune process.
Collapse
Affiliation(s)
| | - Ali Shojaie
- Department of Biostatistics, Department of Medicine, University of Washington, Seattle, WA 98185, USA
| | - Barbara Brooks-Worrell
- Department of Biostatistics, Department of Medicine, University of Washington, Seattle, WA 98185, USA
- Department of Medicine, VA Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Sepideh Dibay
- Department of Biostatistics, Department of Medicine, University of Washington, Seattle, WA 98185, USA
| | - Kristina Utzschneider
- Department of Biostatistics, Department of Medicine, University of Washington, Seattle, WA 98185, USA
- Department of Medicine, VA Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Steven E Kahn
- Department of Biostatistics, Department of Medicine, University of Washington, Seattle, WA 98185, USA
- Department of Medicine, VA Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Mary E Larkin
- Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston, MA 02114, USA
| | - Mary L Johnson
- International Diabetes Center, Minneapolis, MN 55416, USA
| | - Naji Younes
- The Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Rockville, MD 20852, USA
| | - Neda Rasouli
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Cyrus Desouza
- Division of Diabetes, Endocrinology and Metabolism, University of Nebraska and Omaha VA Medical Center, Omaha, NE 68198, USA
| | - Robert M Cohen
- Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati and Cincinnati VA Medical Center, Cincinnati, OH 45221, USA
| | | | - Hermes J Florez
- Department of Medicine, University of Miami, Miami, FL 33135, USA
- Division of Endocrinology, Diabetes and Metabolic Diseases, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Willy Marcos Valencia
- Division of Endocrinology, Diabetes and Metabolic Diseases, Medical University of South Carolina, Charleston, SC 29425, USA
- Geriatric Research, Education and Clinical Center, Bruce W. Carter Veterans Affairs Medical Center, Miami, FL 33125, USA
- Robert Stempel Department of Public Health, College of Health and Urban Affairs, Florida International University, Miami, FL 33181, USA
| | - Jerry P Palmer
- Department of Biostatistics, Department of Medicine, University of Washington, Seattle, WA 98185, USA
- Department of Medicine, VA Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Ashok Balasubramanyam
- Department of Medicine: Endocrinology, Diabetes and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
40
|
Naito H, Kaga H, Someya Y, Tabata H, Kakehi S, Tajima T, Ito N, Yamasaki N, Sato M, Kadowaki S, Sugimoto D, Nishida Y, Kawamori R, Watada H, Tamura Y. Fat Accumulation and Elevated Free Fatty Acid Are Associated With Age-Related Glucose Intolerance: Bunkyo Health Study. J Endocr Soc 2024; 8:bvad164. [PMID: 38188453 PMCID: PMC10768880 DOI: 10.1210/jendso/bvad164] [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: 11/02/2023] [Indexed: 01/09/2024] Open
Abstract
Context Older adults have a high prevalence of new-onset diabetes, often attributed to age-related decreases in insulin sensitivity and secretion. It remains unclear whether both insulin sensitivity and secretion continue to deteriorate after age 65. Objective To investigate the effects of aging on glucose metabolism after age 65 and to identify its determinants. Methods This cross-sectional study involved 1438 Japanese older adults without diabetes. All participants underwent a 75-g oral glucose tolerance test (OGTT). Body composition and fat distribution were measured with dual-energy X-ray absorptiometry and magnetic resonance imaging. Participants were divided into 4 groups by age (65-69, 70-74, 75-79, and 80-84 years) to compare differences in metabolic parameters. Results Mean age and body mass index were 73.0 ± 5.4 years and 22.7 ± 3.0 kg/m2. The prevalence of newly diagnosed diabetes increased with age. Fasting glucose, fasting insulin, the area under the curve (AUC)-insulin/AUC-glucose and insulinogenic index were comparable between groups. AUC-glucose and AUC-insulin during OGTT were significantly higher and Matsuda index and disposition index (Matsuda index · AUC-insulin/AUC-glucose) were significantly lower in the age 80-84 group than in the age 65-69 group. Age-related fat accumulation, particularly increased visceral fat area (VFA), and elevated free fatty acid (FFA) levels were observed. Multiple regression revealed strong correlations of both Matsuda index and disposition index with VFA and FFA. Conclusion Glucose tolerance declined with age in Japanese older adults, possibly due to age-related insulin resistance and β-cell deterioration associated with fat accumulation and elevated FFA levels.
Collapse
Affiliation(s)
- Hitoshi Naito
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Hideyoshi Kaga
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Yuki Someya
- Sportology Center, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Hiroki Tabata
- Sportology Center, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Saori Kakehi
- Sportology Center, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Tsubasa Tajima
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Naoaki Ito
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Nozomu Yamasaki
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Motonori Sato
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Satoshi Kadowaki
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Daisuke Sugimoto
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Yuya Nishida
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Ryuzo Kawamori
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
- Sportology Center, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Hirotaka Watada
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
- Sportology Center, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Yoshifumi Tamura
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
- Sportology Center, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| |
Collapse
|
41
|
Shen T, Zheng Q, Zhong L, Zeng X, Yuan X, Mo F, Zhu S, Yang W, Chen Q. Insufficient compensatory pancreatic β-cells function might be closely associated with hyperuricemia in U.S. adults: evidence from the National Health and Nutrition Examination Survey. BMC Public Health 2024; 24:85. [PMID: 38172728 PMCID: PMC10765924 DOI: 10.1186/s12889-023-17471-0] [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: 08/08/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The prevalence of hyperuricemia (HUA) is gradually increasing worldwide. HUA is closely related to diabetes, but the relationship between HUA and pancreatic β-cells function in the population is unclear. The purpose of this article is to investigate the association between pancreatic β-cells and HUA. METHODS This cross-sectional study examined the association between pancreatic β-cells and HUA in 1999-2004 using data from the National Health and Nutrition Examination Survey (NHANES). Subjects were divided into two groups: HUA and non-HUA. Pancreatic β-cells function levels were assessed using homeostasis model assessment version 2-%S (HOMA2-%S), homeostasis model assessment version 2-%B (HOMA2-%B) and disposition index (DI). Multivariate logistic regression models and restricted cubic spline models were fitted to assess the association of pancreatic β-cells function with HUA. RESULTS The final analysis included 5496 subjects with a mean age of 46.3 years (standard error (SE), 0.4). The weighted means of HOMA2-%B, HOMA2-%S and DI were 118.1 (SE, 1.0), 69.9(SE, 1.1) and 73.9 (SE, 0.7), respectively. After adjustment for major confounders, participants in the highest quartile of HOMA2-%B had a higher risk of HUA (OR = 2.55, 95% CI: 1.89-3.43) compared to participants in the lowest quartile. In contrast, participants in the lowest quartile of HOMA2-%S were significantly more likely to have HUA than that in the highest quartile (OR = 3.87, 95% CI: 2.74-5.45), and similar results were observed in DI (OR = 1.98, 95% CI: 1.32-2.97). Multivariate adjusted restricted cubic spline analysis found evidence of non-linear associations between HOMA2-%B, HOAM2-%S, DI and the prevalence of HUA. CONCLUSION Our finding illustrated the indicators of inadequate β-cells compensation might be a new predictor for the presence of HUA in U.S. adults, highlighting a critical role of pancreatic β-cells function on HUA.
Collapse
Affiliation(s)
- Tianran Shen
- Department of Nutrition and Food Hygiene, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, 510006, China
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, 510006, China
| | - Qiutong Zheng
- Department of Nutrition and Food Hygiene, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, 510006, China
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, 510006, China
| | - Liling Zhong
- Department of Nutrition and Food Hygiene, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, 510006, China
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, 510006, China
| | - Xia Zeng
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, 510006, China
- Department of Child and Adolescent Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, 510006, China
| | - Xiaojing Yuan
- Department of Nutrition and Food Hygiene, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, 510006, China
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, 510006, China
| | - Fengxin Mo
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, 510006, China
- Department of Occupational Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangzhou Province, 510006, China
| | - Shiheng Zhu
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, 510006, China
- Department of Occupational Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangzhou Province, 510006, China
| | - Wenhan Yang
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, 510006, China.
- Department of Child and Adolescent Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, 510006, China.
| | - Qingsong Chen
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, 510006, China.
- Department of Occupational Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangzhou Province, 510006, China.
| |
Collapse
|
42
|
Narayanan N, Sahoo J, Kamalanathan S, Sagili H, Zachariah B, Naik D, Roy A, Merugu C. Insulin Sensitivity, Islet Cell Function, and Incretin Axis in Pregnant Women With and Without Gestational Diabetes Mellitus. Indian J Endocrinol Metab 2024; 28:71-79. [PMID: 38533283 PMCID: PMC10962776 DOI: 10.4103/ijem.ijem_7_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 05/29/2023] [Accepted: 09/04/2023] [Indexed: 03/28/2024] Open
Abstract
INTRODUCTION The aim of this study was to compare insulin sensitivity, islet cell function, and incretin axes in pregnant subjects with GDM and normal healthy controls. METHODS Pregnant women at 24 to 28 weeks of gestation were subjected to a 75 g oral glucose tolerance test (OGTT). Samples for glucose, insulin, glucagon, glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) were collected at 0, 30, 60, and 120 min during the OGTT. The Matsuda index (MI) and insulin secretion and sensitivity index-2 (ISSI-2) were assessed. The glucagon suppression index (GSI) was calculated along with the area under the curve (AUC) for glucose, insulin, glucagon, GLP-1, and GIP. RESULTS A total of 48 pregnant women (25 GDM and 23 controls) were finally analysed. The MI and ISSI-2 were low in the GDM group [4.31 vs. 5.42; P = 0.04], [1.99 vs. 3.18, P ≤ 0.01] respectively). Total AUCglucagon was higher in the GDM group (7411.7 vs. 6320.1, P = 0.02). GSI30 was significantly lower in the GDM group (-62.6 vs. -24.7, P = 0.03). Fasting GLP-1 levels were low in GDM women (17.3 vs. 22.2, P = 0.04). The total AUCGLP-1 positively correlated with total GSI in the GDM group. CONCLUSION Asian-Indian GDM women have high insulin insensitivity, islet cell dysfunction, and low fasting GLP-1. Incretin axis dysfunction plays a potential role in their islet cell dysfunction.
Collapse
Affiliation(s)
- Niya Narayanan
- Department of Endocrinology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry, India
| | - Jayaprakash Sahoo
- Department of Endocrinology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry, India
| | - Sadishkumar Kamalanathan
- Department of Endocrinology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry, India
| | - Haritha Sagili
- Department of Obstetrics and Gynaecology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry, India
| | - Bobby Zachariah
- Department of Biochemistry, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry, India
| | - Dukhabandhu Naik
- Department of Endocrinology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry, India
| | - Ayan Roy
- Department of Endocrinology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry, India
| | - Chandhana Merugu
- Department of Endocrinology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry, India
| |
Collapse
|
43
|
Hoffman RP, Yu CY. Hematologic and biochemical inflammatory markers increase with body mass and positively correlate in adolescents. Pediatr Res 2024; 95:223-226. [PMID: 37573380 DOI: 10.1038/s41390-023-02769-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/29/2023] [Accepted: 07/09/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease that has its origins in childhood. The goal of this study was to explore the relationships of hematologic inflammatory markers to body mass, biochemical inflammatory markers and cardiometabolic risk factors. METHODS Healthy, white, non-Hispanic identifying adolescents (n = 75, age 12 to 18 years) were enrolled. Measures studied included body mass index percentile (BMI%), neutrophil and platelet to lymphocyte ratio (NLR, PLR), pan immune inflammation value (PIV), lipids, augmentation index, reactive hyperemia, inflammatory markers (interleukin 6: IL6, c-reactive protein: CRP), complement (C3, C3a, C4, C4a, C5a) insulin secretion and insulin sensitivity (oral glucose tolerance test: Matusda index, and disposition index (DI)). RESULTS NLR (rS = 0.31, p < 0.01), PLR (rS = 0.32, p < 0.01), PIV (rS = 0.32, p < 0.01) and CRP (rS = 0.51, p < 0.001) all positively correlated with BMI% but IL-6 did not. NLR, PLR and PIV all positively correlated with each other. NLR correlated with the reactive hyperemia response (rS = 0.29, p < 0.02) but this relationship was lost when BMI% was included. NLR positively correlated with C3a, C4, CRP and IL6 even when BMI% was included. CONCLUSION In healthy adolescents hematologic markers of inflammation increase with increasing body mass and neutrocyte to lymphocyte ratio is associated with increased complement and inflammatory markers independent of obesity. IMPACT STATEMENT Hematologic and biochemical markers of inflammation increase with increased body mass in healthy adolescents. Hematologic and biochemical markers of inflammation are positively related independent of body mass in healthy adolescents. Hematologic inflammatory markers are not related to markers of cardiometabolic risk in healthy adolescents.
Collapse
Affiliation(s)
- Robert P Hoffman
- Division of Endocrinology, Nationwide Children's Hospital, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| | - Chack-Yung Yu
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
44
|
De Sanctis V, Soliman AT, Daar S, Tzoulis P, Kattamis C. Can we Predict Incipient Diabetes Mellitus in Patients with Transfusion Dependent β-Thalassemia (β-TDT) Referred with a History of Prediabetes? Mediterr J Hematol Infect Dis 2024; 16:e2024005. [PMID: 38223478 PMCID: PMC10786125 DOI: 10.4084/mjhid.2024.005] [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: 09/11/2023] [Accepted: 12/12/2023] [Indexed: 01/16/2024] Open
Abstract
Background Prediabetes and diabetes mellitus (DM) are complications in adult patients with transfusion-dependent β-thalassemia (β-TDT), with their incidence increasing with age. Objective This retrospective observational study describes the glycemic trajectories and evaluates predictive indices of β-cell function and insulin sensitivity/resistance in β-TDT patients with prediabetes, both in a steady state and during 3-h oral glucose tolerance test (OGTT), in order to identify patients at high risk for incipient diabetes. Setting The study was mainly conducted at the Pediatric and Adolescent Outpatient Clinic, Quisisana Hospital, Ferrara (Italy), in collaboration with thalassemia referring centers across Italy. Patients The study included 11 β-TDT (aged 15.11-31.10 years) with prediabetes. Methods: The ADA criteria for the diagnosis of glucose dysregulation were adopted. Investigations included evaluating plasma glucose levels and insulin secretion, analyzing glycemic trajectories and indices of β-cell function, and insulin sensitivity/resistance assessed in steady state and during OGTT. Results The duration of progression from prediabetes to DM, expressed in years, showed a positive direct correlation with corrected insulin response (CIR-30 = r: 0.7606, P: 0.0065), insulinogenic index (IGI 0-120 = r: 0.6121, P:0.045), oral disposition index (oDI = r: 0.7119, P:0.013), insulin growth factor-1 (IGF-1= r: 0.6246, P: 0.039) and an inverse linear correlation with serum ferritin (SF = r: -0.7197, P: 0.012). The number of patients with 1-hour post-load PG value ≥ 155 mg/dL ( ≥ 8.6 mmol/L) was at -4 years: 4/9 (44.4%); -3 years: 8/9 (88.8%); - 2 years: 7/10 (70 %) and at -1 year: 11/11 (100%) (PG range:162-217 mg/dL). Conclusions A progressive increase in 1-hour PG in response to OGTT is associated with progressive β-cell failure, peripheral resistance to insulin action, and reduced oDI and may be considered a relevant marker for incipient DM in β-TDT patients with prediabetes.
Collapse
Affiliation(s)
- Vincenzo De Sanctis
- Coordinator of ICET-A Network (International Network of Clinicians for Endocrinopathies in Thalassemia and Adolescent Medicine) and Pediatric and Adolescent Outpatient Clinic, Quisisana Hospital, Ferrara, Italy
| | - Ashraf T Soliman
- Department of Pediatrics, Division of Endocrinology, Hamad General Hospital, Doha, Qatar
| | - Shahina Daar
- Department of Haematology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Sultanate of Oman
| | - Ploutarchos Tzoulis
- Department of Diabetes and Endocrinology, Whittington Hospital, University College London, London, UK
| | - Christos Kattamis
- Thalassemia Unit, First Department of Paediatrics, National Kapodistrian University of Athens 11527, Greece
| |
Collapse
|
45
|
Gregory JM, Kraft G, Dalla Man C, Slaughter JC, Scott MF, Hastings JR, Edgerton DS, Moore MC, Cherrington AD. A high-fat and fructose diet in dogs mirrors insulin resistance and β-cell dysfunction characteristic of impaired glucose tolerance in humans. PLoS One 2023; 18:e0296400. [PMID: 38134122 PMCID: PMC10745172 DOI: 10.1371/journal.pone.0296400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
This study examined the impact of a hypercaloric high-fat high-fructose diet (HFFD) in dogs as a potential model for human impaired glucose tolerance (IGT) and type 2 diabetes mellitus (T2DM). The HFFD not only led to weight gain but also triggered metabolic alterations akin to the precursors of human T2DM, notably insulin resistance and β-cell dysfunction. Following the HFFD intervention, the dogs exhibited a 50% decrease in insulin sensitivity within the first four weeks, paralleling observations in the progression from normal to IGT in humans. Calculations of the insulinogenic index using both insulin and C-peptide measurements during oral glucose tolerance tests revealed a significant and sustained decrease in early-phase insulin release, with partial compensation in the later phase, predominantly stemming from reduced hepatic insulin clearance. In addition, the Disposition Index, representing the β-cell's capacity to compensate for diminished insulin sensitivity, fell dramatically. These results confirm that a HFFD can instigate metabolic changes in dogs akin to the early stages of progression to T2DM in humans. The study underscores the potential of using dogs subjected to a HFFD as a model organism for studying human IGT and T2DM.
Collapse
Affiliation(s)
- Justin M Gregory
- Ian Burr Division of Pediatric Endocrinology and Diabetes, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Guillaume Kraft
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN United States of America
| | - Chiara Dalla Man
- Department of Information Engineering, University of Padova, Padova, Italy
| | - James C Slaughter
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Melanie F Scott
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN United States of America
| | - Jon R Hastings
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN United States of America
| | - Dale S Edgerton
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN United States of America
| | - Mary C Moore
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN United States of America
| | - Alan D Cherrington
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN United States of America
| |
Collapse
|
46
|
De Sanctis V, Soliman AT, Daar S, Tzoulis P, Karimi M, Saki F, Di Maio S, Kattamis C. A prospective guide for clinical implementation of selected OGTT- derived surrogate indices for the evaluation of β- cell function and insulin sensitivity in patients with transfusion-dependent β- thalassaemia. ACTA BIO-MEDICA : ATENEI PARMENSIS 2023; 94:e2023221. [PMID: 38054665 PMCID: PMC10734222 DOI: 10.23750/abm.v94i6.15329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 12/07/2023]
Abstract
The gold standard for the measurement of insulin secretion is the hyperglycemic clamp and for insulin sensitivity the hyperinsulinemic euglycemic clamp, respectively. A number of surrogate indices, derived from plasma glucose and insulin levels at a fasting state or after oral glucose load, have been proposed to estimate β-cell response, and the ability of β-cells to compensate for changes of insulin sensitivity by modulating insulin secretion (disposition index). Starting from the current recommendations for the annual screening of glucose dysregulation in patients with transfusion dependent β-thalassemia (β-TDT), this article summarizes the most frequently used indirect indices of insulin secretion and resistance derived from the oral glucose tolerance test (OGTT) and discusses the strengths and weaknesses of selected indices and the basic concepts underlying each method for the appropriate evaluation of glucose regulation. Basal indices for β-cell function and insulin sensitivity, albeit simple and cheap, have limited usefulness due to a high coefficient variation and the lack of data about response to glucose load. Therefore, measurement of indices during an OGTT, despite being costly and time-consuming, is suggested since it can detect, even subtle, dynamic changes in insulin secretion and glucose handling. In patients with β-TDT, the indices derived from OGTT may offer an additional factor to evaluate the efficiency of iron chelation therapy and detect patients who may need intensification of iron chelation therapy and/or pharmacological intervention.
Collapse
Affiliation(s)
| | - Ashraf T Soliman
- Department of Pediatrics, Division of Endocrinology, Hamad General Hospital, Doha, Qatar .
| | - Shahina Daar
- Department of Haematology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Sultanate of Oman.
| | - Ploutarchos Tzoulis
- Department of Diabetes and Endocrinology, Whittington Hospital, University College London, London, UK.
| | - Mehran Karimi
- Hematology- Oncology Department, American Hospital Dubai, Dubai, UAE .
| | - Forough Saki
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Salvatore Di Maio
- Emeritus Director in Pediatrics, Children's Hospital "Santobono-Pausilipon", Naples, Italy.
| | - Christos Kattamis
- Τhalassemia Unit, First Department of Paediatrics, National Kapodistrian University of Athens 11527, Greece.
| |
Collapse
|
47
|
De Sanctis V, Daar S, Soliman A, Tzoulis P, Yassin M, Kattamis C. A retrospective study of glucose homeostasis, insulin secretion, sensitivity/resistance in non- transfusion-dependent β-thalassemia patients (NTD- β Thal): reduced β-cell secretion rather than insulin resistance seems to be the dominant defect for glucose dysregulation (GD). ACTA BIO-MEDICA : ATENEI PARMENSIS 2023; 94:e2023262. [PMID: 38054678 PMCID: PMC10734240 DOI: 10.23750/abm.v94i6.15001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/04/2023] [Indexed: 12/07/2023]
Abstract
AIMS Non-transfusion - dependent β-thalassemias (NTD-βThal) can cause iron overload and serious iron-related organ complications as endocrine dysfunction, including glucose dysregulation (GD). PATIENTS AND METHODS We retrieved data of all NTD- β Thal patients referred consecutively to a single Outpatient Italian Clinic from October 2010 to April 2023. All patients underwent a standard 3-h oral glucose tolerance test (OGTT) for analysis of glucose homeostasis, insulin secretion and sensitivity/resistance (IR), using conventional surrogate indices derived from the OGTT. The collected data in NTD- β Thal patients were compared to 20 healthy subjects. RESULTS Seventeen of 26 (65.3 %) NTD- β Thal patients (aged: 7.8 -35.1 years) had normal glucose tolerance, 1/26 (3.8 %) had impaired fasting glucose (IFG), 5/26 (19.2 %) impaired glucose tolerance (IGT), 1/26 (3.8%) IFG plus IGT and 2/26 (7.6%) plasma glucose (PG) level ≥155 mg/dL 1-h after glucose load. GD was observed exclusively in young adult patients; none of them had diabetes mellitus (DM). These findings were associated with a low insulinogenic index (IGI) and oral disposition index. HOMA-IR and QUICKI were not significantly different compared to controls. Interestingly, in young adult patients, ISI-Matsuda index was statistically higher compared to the control group, suggesting an increased insulin sensitivity. CONCLUSIONS This study reported a high prevalence of GD in young adults with NTD- β Thal. The documented reduction of IGI rather than the presence of IR, indicates reduced insulin secretory capacity as the pathophysiological basis of dysglycemia that may represent a novel investigational path for future studies on the mechanism(s) responsible for GD in NTD- β Thal patients.
Collapse
Affiliation(s)
| | - Shahina Daar
- Department of Haematology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Sultanate of Oman.
| | - Ashraf Soliman
- Department of Pediatrics, Division of Endocrinology, Hamad General Hospital, Doha, Qatar .
| | - Ploutarchos Tzoulis
- Department of Diabetes and Endocrinology, Whittington Hospital, University College London, London, UK.
| | - Mohamed Yassin
- Hematology Section, Medical Oncology, National Center for Cancer Care and Research (NCCCR), Hamad Medical Corporation (HMC), Doha, Qatar.
| | - Christos Kattamis
- Τhalassemia Unit, First Department of Paediatrics, National Kapodistrian University of Athens 11527, Greece.
| |
Collapse
|
48
|
Deischinger C, Bastian M, Leitner K, Bancher-Todesca D, Kiss H, Baumgartner-Parzer S, Kautzky-Willer A, Harreiter J. Gremlin-1 in pregnancy and postpartum: relation to the fatty liver index, markers of bone health, glucose metabolism and gestational diabetes mellitus status. Acta Diabetol 2023; 60:1699-1707. [PMID: 37518503 PMCID: PMC10587257 DOI: 10.1007/s00592-023-02151-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023]
Abstract
INTRODUCTION Gremlin-1 is a peptide that functions as an antagonist to bone morphogenic proteins and is overexpressed in obesity and type 2 diabetes mellitus. Gremlin-1 has not yet been investigated in pregnancy, pregnancy-related insulin resistance or gestational diabetes mellitus (GDM). PATIENTS AND METHODS Gremlin-1 levels were measured throughout the pregnancy of 58 women at high risk for GDM at the Medical University of Vienna. Furthermore, an oral glucose tolerance test, fasting insulin, fasting glucose, sex hormones, blood lipids, liver and renal parameters, and markers of bone development were evaluated at two points during pregnancy (< 20 weeks of gestation (GW), GW 24-28) and 12-14 weeks postpartum. RESULTS Gremlin-1 levels decreased from < 20 GW (mean = 9.2 pg/ml, SD = 8.4 pg/ml) to GW 24-28 (mean = 6.7 pg/ml, SD = 5.7 pg/ml, p = 0.033) and increased again postpartum, albeit not significantly (mean = 10.7 pg/ml, SD = 13.1 pg/ml, p = 0.339). During pregnancy, Gremlin-1 levels correlated negatively with osteocalcin and procollagen type I aminoterminal propeptide (P1NP), markers of bone health. Concerning glucose metabolism, Gremlin-1 levels were inversely related to the Insulinogenic Index at GW < 20. However, Gremlin-1 levels were not significantly different between women with normal glucose tolerance and GDM during pregnancy. Postpartum, Gremlin-1 was associated with the fatty liver index, osteocalcin levels, diastolic blood pressure and weight. CONCLUSION Gremlin-1 levels decreased significantly during pregnancy. The biomarker is not related to GDM status, but correlates negatively with the Insulinogenic Index, an index related to beta cell function. Trial Registry Number ACTRN12616000924459.
Collapse
Affiliation(s)
- Carola Deischinger
- Gender Medicine Unit, Clinical Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Magdalena Bastian
- Gender Medicine Unit, Clinical Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Karoline Leitner
- Gender Medicine Unit, Clinical Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Dagmar Bancher-Todesca
- Division of Fetomaternal Medicine, Department of Obstetrics and Gynaecology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Herbert Kiss
- Division of Fetomaternal Medicine, Department of Obstetrics and Gynaecology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Sabina Baumgartner-Parzer
- Gender Medicine Unit, Clinical Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Alexandra Kautzky-Willer
- Gender Medicine Unit, Clinical Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Jürgen Harreiter
- Gender Medicine Unit, Clinical Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
49
|
Thaweethai T, Soetan Z, James K, Florez JC, Powe CE. Distinct Insulin Physiology Trajectories in Euglycemic Pregnancy and Gestational Diabetes Mellitus. Diabetes Care 2023; 46:2137-2146. [PMID: 37126832 PMCID: PMC10698215 DOI: 10.2337/dc22-2226] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/26/2023] [Indexed: 05/03/2023]
Abstract
OBJECTIVE To evaluate changes in insulin physiology in euglycemic pregnancy and gestational diabetes mellitus (GDM). RESEARCH DESIGN AND METHODS Participants underwent oral glucose tolerance tests at ≤15 weeks' gestation (early pregnancy), 24-32 weeks' gestation (mid-late pregnancy), and 6-24 weeks postpartum. We evaluated longitudinal changes in insulin secretory response (log Stumvoll first-phase estimate) and insulin sensitivity (log Matsuda index) using linear mixed models. We then evaluated participants who met GDM criteria in early pregnancy (early GDM) and mid-late pregnancy (classic GDM) separately from those without GDM. We derived the pregnancy insulin physiology (PIP) index to quantify β-cell compensation for insulin resistance. RESULTS Among 166 participants, 21 had early GDM and 24 developed classic GDM. Insulin sensitivity was reduced slightly in early pregnancy (β = -0.20, P < 0.001) and substantially in mid-late pregnancy (β = -0.47, P < 0.001) compared with postpartum. Insulin secretory response (adjusted for insulin sensitivity) was augmented in early pregnancy (β = 0.16, P < 0.001) and mid-late pregnancy (β = 0.16, P = 0.001) compared with postpartum. Compared with postpartum, the PIP index was augmented in early pregnancy (β = 215, P = 0.04) but not mid-late pregnancy (β = 55, P = 0.64). Early GDM was distinguished by a substantial reduction in early pregnancy insulin sensitivity (β = -0.59, P < 0.001) compared with postpartum. Both early and classic GDM lacked evidence of early pregnancy augmentation of insulin secretory response (adjusted for insulin sensitivity) and the PIP index (P > 0.1 vs. postpartum). Early pregnancy PIP index predicted GDM independent of participant characteristics (area under the curve without PIP index 0.70 [95% CI 0.61-0.79], area under the curve with PIP index 0.87 [95% CI 0.80-0.93]). CONCLUSIONS β-Cell function is enhanced in early pregnancy. Deficient first-trimester β-cell function predicts GDM.
Collapse
Affiliation(s)
- Tanayott Thaweethai
- Biostatistics, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Zainab Soetan
- Biostatistics, Massachusetts General Hospital, Boston, MA
| | - Kaitlyn James
- Harvard Medical School, Boston, MA
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA
| | - Jose C. Florez
- Harvard Medical School, Boston, MA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Broad Institute of MIT and Harvard, Boston, MA
| | - Camille E. Powe
- Harvard Medical School, Boston, MA
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Broad Institute of MIT and Harvard, Boston, MA
| |
Collapse
|
50
|
Fuller KN, Bohne EM, Mey JT, Blackburn BK, Miranda VR, Varady KA, Danielson KK, Haus JM. Plasma undercarboxylated osteocalcin dynamics with glycemic stress reflects insulin sensitivity and beta-cell function in humans with and without T2DM. Metabol Open 2023; 20:100264. [PMID: 38115864 PMCID: PMC10728569 DOI: 10.1016/j.metop.2023.100264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/15/2023] [Accepted: 11/22/2023] [Indexed: 12/21/2023] Open
Abstract
This study aimed to better understand the relationship between bone-related biomarkers and nutrient stress in the context of metabolic health. We investigated plasma osteocalcin (OC) during an oral glucose challenge and experimental hyperinsulinemia in Type 2 diabetes (T2DM) and lean healthy controls (LHC). Older individuals with obesity and T2DM (n = 9) and young LHCs (n = 9) underwent a 75g oral glucose tolerance test (OGTT) and a 40 mU/m2/min hyperinsulinemic-euglycemic clamp. Plasma undercarboxylated OC (ucOC) and total OC were measured at baseline, 60mins, and 120mins of the OGTT and clamp via ELISA. In addition, plasma alkaline phosphatase (ALP), leptin, adiponectin, Vitamin D and insulin were measured and indices of insulin sensitivity and β-cell function were derived. The T2DM group had lower (p<0.05) ucOC and ucOC:total OC ratio than LHC during both the OGTT and clamp. Further, baseline ucOC was positively correlated to indices of β-cell function and negatively correlated to indices of insulin resistance when both groups were combined (all p<0.05). Suppression of OC observed in T2DM may be related to glucose intolerance and insulin resistance. Similarly, our data suggest that the observed phenotypic differences between groups are likely a product of long-term glucose dysregulation rather than acute flux in glucose or insulin.
Collapse
Affiliation(s)
- Kelly N.Z. Fuller
- Department of Pediatrics, Section of Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Erin M. Bohne
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, IL, USA
| | - Jacob T. Mey
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Brian K. Blackburn
- Applied Health Sciences and Kinesiology, Humboldt State University, Arcata, CA, USA
| | | | - Krista A. Varady
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, IL, USA
| | - Kirstie K. Danielson
- Division of Endocrinology and Metabolism, University of Illinois at Chicago, IL, USA
| | - Jacob M. Haus
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|