1
|
Liu N, Wang B, Zhang G, Shen M, Cheng P, Guo Z, Zuo L, Yang J, Guo M, Wang M, Liu Z, Wu J. Waist-to-hip ratio better reflect beta-cell function and predicts diabetes risk in adult with overweight or obesity. Ann Med 2025; 57:2462447. [PMID: 39921368 PMCID: PMC11809193 DOI: 10.1080/07853890.2025.2462447] [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: 08/31/2024] [Revised: 01/09/2025] [Accepted: 01/24/2025] [Indexed: 02/10/2025] Open
Abstract
OBJECTIVE Controversy remains as to which obesity measures better predict type 2 diabetes (T2D) risk in overweight or obese individuals. The objective of this study is to determine which commonly used obesity measures better reflect beta cell function and predict T2D risk in participants with overweight or obesity and to validate the findings using prospective cohort data. PATIENTS AND METHODS Cross-sectional data from the Obesity Clinic of the Xiangya Hospital of the Central South University and prospective cohort from UK Biobank. BMI, waist circumference (WC), waist-to-hip ratio (WHpR), and waist-to-height ratio (WHtR) were measured. Primary outcomes included beta cell function indices in the cross-sectional study and the occurrence of diabetes obtained from UK Biobank data. RESULTS One thousand four hundred and ninety-seven participants with overweight or obesity (median age 29 years, 41% males) and 322,023 UK Biobank participants without diabetes at baseline (mean age 56.83 years, 50.4% males) were studied. WHpR had a stronger association with beta cell function and central body fat distribution than the other three obesity measures irrespective of glucometabolic states. WHpR associated positively with diabetes risk in participants using the hazard ratio scale (HR per SD increase of WHpR, 2.311, 95% CI 2.250-2.374). CONCLUSIONS WHpR is a superior index in reflecting central body obesity, estimating beta cell function, and predicting T2D risk in people with overweight or obesity compared to BMI, WC, and WHtR.
Collapse
Affiliation(s)
- Na Liu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Obesity and Its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bian Wang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Obesity and Its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guanxiong Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Minxue Shen
- Department of Social Medicine and Health Management, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Peng Cheng
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Obesity and Its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhanjun Guo
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Obesity and Its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Linhui Zuo
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Obesity and Its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Junya Yang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Min Guo
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Min Wang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Obesity and Its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenqi Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jing Wu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Obesity and Its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
2
|
Chen N, Shi S, Zhao S, Shen L, Kong L, Liang Z, Cai S, Chen H, Wang D, Wang Z. Gestational diabetes mellitus and adverse pregnancy outcomes increased on pandemic lockdown: a retrospective analysis. J Matern Fetal Neonatal Med 2025; 38:2460180. [PMID: 40405609 DOI: 10.1080/14767058.2025.2460180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/12/2025] [Accepted: 01/21/2025] [Indexed: 05/24/2025]
Abstract
OBJECTIVES Adverse social contexts, such as lockdowns and disasters during pregnancy, can significantly impact maternal and neonatal outcomes. However, the specific effects on different populations remain unclear. This study aimed to investigate the variations in pregnancy outcomes resulting from the pandemic lockdown and to identify distinct populations in need of targeted intervention. METHODS Women who delivered at our institution spanning from 2017 to 2019 (pre-pandemic) and from 2020 to 2022 (during the pandemic lockdown) were included in this study. A comparison was conducted on maternal and neonatal outcomes across a total of 19,382 singleton pregnancies, with a specific focus on those affected by gestational diabetes mellitus (GDM). RESULTS As a total of 19,382 singleton pregnant women were included, this study found a significant increase in the incidence rates of GDM with an odds ratio of 1.194 (95%CI: 1.109-1.286, p < .001). Additionally, following the pandemic lockdown, there was an increase in rates of premature birth, premature rupture of membranes, and intrahepatic cholestasis of pregnancy. Further analysis of the GDM cohort revealed a notable rise in the risk of Group B Streptococcus infection and neonatal small for gestational age (SGA). Specifically, GDM patients with a body mass index (BMI) less than 18.5 kg/m2 exhibited an increased risk of fetal growth restriction and SGA. CONCLUSIONS The pandemic lockdown adversely affected pregnancy outcomes. Therefore, targeted screening and clinical management for specific populations should be prioritized.
Collapse
Affiliation(s)
- Niankun Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shaole Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shanshan Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lixia Shen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lingyi Kong
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zanxiong Liang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shiqin Cai
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Haitian Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Dongyu Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zilian Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
3
|
Feigerlova E. Novel estrogen receptor-α gene inactivating missense variant in a woman: Therapeutic challenge and long-term follow-up data. Bone 2025; 194:117427. [PMID: 40032016 DOI: 10.1016/j.bone.2025.117427] [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: 11/29/2024] [Revised: 01/26/2025] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Rare mutations of the ESR1 gene, encoding the estrogen receptor alpha (ERα), have been shown to cause estrogen resistance in humans. Phenotypic features include impaired maturation of epiphyseal cartilage, osteoporosis, and infertility. Clinical phenotype is not well described and the available data reflect inconsistency. To date, there are no effective therapeutic options. METHOD This retrospective study provides a detailed description of bone and metabolic phenotype and 8-year follow-up data of a female with a novel p.Met543Thr missense variant in the homozygous state, localized in the ligand-binding domain. RESULTS The patient, first seen at the age of 21.3 years, presented with a tall stature (+2.2 SD), a delayed bone age (13 years). She had no breast development, normal axillary and pubic pilosity and bilateral axillary acanthosis nigricans. The metabolic phenotype included insulin resistance, decreased insulin sensitivity and increased leptinemia. The patient presented a continuous linear growth (height + 3SD at the age of 28.6 years). She had a severe osteoporosis of the lumbar spine (Z-score -3.9) and osteopenia of the femoral neck (Z-score -1.8). Osteoporosis worsened (Z-score -5.6 at the lumbar spine; Z-score -4.4 at the femoral neck) despite successive treatments with ethinyl-estradiol and tamoxifen (selective estrogen modulator). Markers of bone turnover were increased and unresponsive to treatment. Treatment with ethinyl-estradiol improved insulin sensitivity, lowered leptinemia, increased some estrogen-regulated liver proteins and the E2/T ratio. CONCLUSION This report brings new insights to the estrogen resistance syndrome and improves our understanding of the skeletal and tissue specific roles of ERα in humans.
Collapse
Affiliation(s)
- Eva Feigerlova
- Centre de référence des maladies héréditaires du métabolisme, Hôpital d'enfants, Centre Hospitalier universitaire of Nancy and Faculty of Medicine, Midwifery and Health Professions, Université de Lorraine, F- 54000 Nancy, France; Centre Universitaire d'Enseignement par Simulation - CUESim, Virtual Hospital of Lorraine, Faculty of Medicine, Midwifery and Health Professions, Université de Lorraine, Nancy F- 54000, France; Centre de référence des maladies héréditaires du métabolisme, Hôpital d'enfants, rue Morvan, CHRU de Nancy, F- 54000 Nancy, France; Université de Lorraine, Inserm, DCAC, Nancy, France.
| |
Collapse
|
4
|
Vahlhaus J, Peters B, Hornemann S, Ost AC, Kruse M, Busjahn A, Pfeiffer AFH, Pivovarova-Ramich O. Later eating timing in relation to an individual internal clock is associated with lower insulin sensitivity and affected by genetic factors. EBioMedicine 2025; 116:105737. [PMID: 40305967 DOI: 10.1016/j.ebiom.2025.105737] [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: 10/24/2024] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Although the contribution of the circadian clock to metabolic regulation is widely recognized, the role of eating timing in glucose metabolism and diabetes risk remains insufficiently studied. This study aimed (i) to investigate the link between the eating timing pattern relative to individual clock and glucose homoeostasis and (ii) to explore the contribution of genetic and environmental factors to eating timing parameters. METHODS In 92 adult twins (NCT01631123), glycaemic traits were assessed using the oral glucose tolerance test. Parameters of eating timing pattern (eating timing itself, daily calorie distribution, and eating frequency) were extracted from five-day food records. Caloric midpoint defined as the time point at which 50% of daily calories are consumed. Circadian timing of eating was determined as a time interval between the clock time of eating and a corrected midpoint of sleep, a chronotype marker. Heritability of eating timing components was estimated by comparing correlations within monozygotic and dizygotic twin pairs and fitting genetic structural equation models. FINDINGS Among components of eating timing, the most associations were found for the circadian time of caloric midpoint (CCM). Later CCM was significantly associated with poorer insulin sensitivity, i.e. with lower ISI Stumvoll (β = 0.304, p = 5.9 × 10-4) and higher HOMA-IR (β = -0.258, p = 0.011) indices, as well as with higher fasting insulin levels (β = -0.259, p = 0.013), even after the model adjustment for sex, age, daily energy intake, and sleep duration. Later CCM also demonstrated robust associations with higher BMI and waist circumference. All eating timing components showed high or moderate heritability and were strongly related to individual sleep timing. INTERPRETATION Later eating timing in relation to an individual internal clock is associated with lower insulin sensitivity. Shifting the main calorie intake to earlier circadian times may improve glucose metabolism, but genetic factors could influence the feasibility and effectiveness of eating-timing based interventions. The findings should be investigated in a larger cohort. FUNDING This work was supported by the German Research Foundation (DFG RA 3340/4-1 to OP-R, project number 530918029), by the European Association for the Study of Diabetes (Morgagni Prize 2020 to OP-R), and by the German Federal Ministry of Education and Research (BMBF NUGAT 0315424 to AFHP). The DZD is funded by the German Federal Ministry for Education and Research (01GI0925).
Collapse
Affiliation(s)
- Janna Vahlhaus
- Department of Molecular Metabolism and Precision Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; University of Lübeck, Lübeck, Germany
| | - Beeke Peters
- Department of Molecular Metabolism and Precision Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Silke Hornemann
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Anne-Cathrin Ost
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Michael Kruse
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology, Diabetes and Nutrition, Campus Benjamin Franklin, Berlin, Germany
| | | | - Andreas F H Pfeiffer
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology, Diabetes and Nutrition, Campus Benjamin Franklin, Berlin, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolism, Berlin, Germany
| | - Olga Pivovarova-Ramich
- Department of Molecular Metabolism and Precision Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolism, Berlin, Germany.
| |
Collapse
|
5
|
Göbl CS, Linder T, Eppel D, Kotzaeridi G, Weidinger L, Zarotti S, Fischer T, Bernasconi MT, Kunze M, Ochsenbein-Koelble N, Winzeler B, Hoesli I, Huhn EA, Tura A. Early prediction of gestational diabetes mellitus: the role of the pregnancy-specific triglycerides-glucose index and other fasting parameters in combination with dynamic testing. Acta Diabetol 2025:10.1007/s00592-025-02490-7. [PMID: 40167633 DOI: 10.1007/s00592-025-02490-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025]
Abstract
The identification of mothers at risk for gestational diabetes mellitus (GDM) at start of pregnancy may be beneficial to improve perinatal outcomes. This study aims evaluating the predictive performance of fasting and dynamic indices of glucose metabolism at first trimester and their association with later GDM development. A cohort of 198 women received detailed metabolic assessment at median gestational age (13 weeks) including 75-g oral glucose tolerance test (OGTT) with assessment of glucose, insulin and C-peptide, and biochemical markers (including triglycerides) to calculate different indices of insulin sensitivity either at fasting and in the OGTT dynamic conditions. Moreover, parameters of β-cell function were assessed. A second OGTT was performed between 24 and 28 gestational weeks (GW) to identify women with GDM. We found that 28 women developed GDM, and, in univariable analysis, this was fairly predicted by several first trimester indices, both at fasting and in dynamic conditions. However, fasting indices containing maternal triglycerides showed better accuracy as compared to traditional indices (even the dynamic ones). In multivariable analysis, the best predictive model of GDM development included fasting and OGTT glucose values, HbA1c, and an insulin sensitivity marker that includes triglycerides (e.g. the improved triglyceride-glucose index, TyGIS). β-Cell function was not included in such predictive model, but at 24-28 GW it showed remarkable impairment in women with GDM. In conclusion, both fasting and dynamic parameters of glucose homeostasis at early pregnancy showed fair predictive accuracy for later GDM, with TyGIS showing excellent performance. β-Cell dysfunction role needs being further elucidated.
Collapse
Affiliation(s)
- Christian S Göbl
- Division of Obstetrics and Feto-Maternal Medicine, Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria.
| | - Tina Linder
- Division of Obstetrics and Feto-Maternal Medicine, Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| | - Daniel Eppel
- Division of Obstetrics and Feto-Maternal Medicine, Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| | - Grammata Kotzaeridi
- Division of Obstetrics and Feto-Maternal Medicine, Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| | - Laura Weidinger
- Division of Obstetrics and Feto-Maternal Medicine, Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| | - Sophie Zarotti
- Division of Obstetrics and Feto-Maternal Medicine, Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| | - Thorsten Fischer
- Department of Obstetrics and Gynaecology, Paracelsus Medical University, Salzburger Landeskrankenhaus, Salzburg, Austria
| | | | - Mirjam Kunze
- Department of Obstetrics and Gynaecology, University Hospital Freiburg, Freiburg im Breisgau, Germany
| | | | - Bettina Winzeler
- Department of Endocrinology and Diabetology, University Hospital Basel, Basel, Switzerland
| | - Irene Hoesli
- Department of Obstetrics and Gynaecology, University and University Hospital Basel, Basel, Switzerland
| | - Evelyn A Huhn
- Department of Obstetrics and Gynaecology, University and University Hospital Basel, Basel, Switzerland
- Clinic of Obstetrics and Prenatal Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Andrea Tura
- Institute of Neuroscience, CNR, Padua, Italy.
| |
Collapse
|
6
|
Jones DL, Kusinski LC, Gillies C, Meek CL. A critique of measurement of defective insulin secretion and insulin sensitivity as a precision approach to gestational diabetes. Diabetologia 2025; 68:752-765. [PMID: 39621104 PMCID: PMC11950144 DOI: 10.1007/s00125-024-06334-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/09/2024] [Indexed: 03/28/2025]
Abstract
AIMS/HYPOTHESIS Precision medicine approaches to gestational diabetes mellitus (GDM) have categorised patients according to disease pathophysiology (insulin resistance, insulin insufficiency or both), and demonstrated associations with clinical outcomes. We aimed to assess whether using enhanced processing to determine indices of insulin secretion and sensitivity is analytically robust, reproducible in a different population, and useful diagnostically and prognostically in clinical practice. METHODS A total of 1308 pregnant women with one or more risk factors for GDM who underwent a 75 g OGTT at one of nine hospital sites were recruited to this observational study. Specimens were collected for determination of glucose levels using standard and enhanced procedures, HbA1c and insulin analysis. GDM diagnosis and management followed National Institute for Health and Care Excellence guidance. We categorised women into pathophysiological subtypes: insulin-resistant GDM (HOMA2-S < 25th centile of the population with normal glucose tolerance [NGT]), insulin-insufficient GDM (HOMA2-B < 25th centile), both or neither. We assessed associations with pregnancy outcomes using logistic regression. RESULTS Using enhanced specimen handling, 1027/1308 (78.5%) women had NGT, with 281/1308 (21.5%) being classified as having GDM. Of this group, 135/281 (48.0%) had insulin-resistant GDM, 73/281 (26.0%) had insulin-insufficient GDM and 2/281 (0.7%) had both insulin-resistant and insulin-insufficient GDM. Unexpectedly, 71 patients (25.3%) had GDM with both HOMA2-S and HOMA2-B ≥ 25th centile (GDM-neither). This novel subgroup appeared to be relatively insulin-sensitive in the fasting state but developed marked post-load hyperglycaemia and hyperinsulinaemia, suggesting an isolated postprandial defect in insulin sensitivity that was not captured by HOMA2-B or HOMA2-S. Women within most GDM subgroups had comparable pregnancy outcomes to those of normoglycaemic women, and HOMA2-B and HOMA2-S were weak predictors of pregnancy outcomes. Maternal BMI predicted a similar number of outcomes to HOMA2-S, suggesting that there was no additional predictive value in adding HOMA2-S. Similar findings were obtained when using different indices and standard specimen handling techniques. CONCLUSIONS/INTERPRETATION Precision categorisation of GDM using HOMA2-S and HOMA2-B does not provide useful diagnostic or prognostic information, but did distinguish a novel subgroup of patients with GDM, characterised by an isolated postprandial defect in insulin sensitivity.
Collapse
Affiliation(s)
- Danielle L Jones
- Institute of Metabolic Science Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Laura C Kusinski
- Institute of Metabolic Science Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
- Leicester Diabetes Centre, Leicester General Hospital, University of Leicester, Leicester, UK
| | - Clare Gillies
- Leicester Diabetes Centre, Leicester General Hospital, University of Leicester, Leicester, UK
| | - Claire L Meek
- Institute of Metabolic Science Metabolic Research Laboratories, University of Cambridge, Cambridge, UK.
- Leicester Diabetes Centre, Leicester General Hospital, University of Leicester, Leicester, UK.
- University Hospitals Leicester NHS Trust, Leicester General Hospital, Gwendoline Road, Leicester, UK.
| |
Collapse
|
7
|
Zhao J, Massoudian SD, Stray-Gundersen S, Wojan F, Lalande S. Short bouts of hypoxia improve insulin sensitivity in adults with type 2 diabetes. J Appl Physiol (1985) 2025; 138:873-880. [PMID: 40013508 DOI: 10.1152/japplphysiol.00932.2024] [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: 11/27/2024] [Revised: 12/28/2024] [Accepted: 02/20/2025] [Indexed: 02/28/2025] Open
Abstract
Hypoxia stimulates glucose uptake independently from the action of insulin. The purpose of this study was to determine the effect of intermittent hypoxia, consisting of alternating short bouts of breathing hypoxic and room air, on glucose concentration, insulin concentration, and insulin sensitivity during an oral glucose tolerance test in adults with type 2 diabetes and adults with normal glycemic control. Nine adults with type 2 diabetes (2 women, HbA1c: 7.3 ± 1.5%, age: 52 ± 13 yr) and nine adults with normal glycemic control (4 women, HbA1c: 5.4 ± 0.1%, age: 24 ± 4 yr) performed a 2-h oral glucose tolerance test on two separate visits to the laboratory. Following ingestion of the glucose drink, participants were exposed to either an intermittent hypoxia protocol, consisting of eight 4-min hypoxic cycles at a targeted oxygen saturation of 80% interspersed with breathing room air to resaturation, or a sham protocol consisting of eight 4-min normoxic cycles interspersed with breathing room air. Intermittent hypoxia did not attenuate the increase in glucose concentration but attenuated the increase in insulin concentration in response to an oral glucose tolerance test in comparison with the sham protocol in adults with type 2 diabetes. Insulin sensitivity was greater during intermittent hypoxia in comparison with the sham protocol in adults with type 2 diabetes (0.043 ± 0.036 vs. 0.032 ± 0.046 μmol/kg/min/pmol, P = 0.01), but did not change in the control group (0.122 ± 0.015 vs. 0.128 ± 0.008 μmol/kg/min/pmol, P = 0.12). In conclusion, intermittent hypoxia improved insulin sensitivity in adults with type 2 diabetes.NEW & NOTEWORTHY The aim of this study was to determine the effect of short bouts of hypoxia, which stimulates glucose uptake, on glucose concentration, insulin concentration, and insulin sensitivity during an oral glucose tolerance test in adults with type 2 diabetes and adults with normal glycemic control. Intermittent hypoxia acutely improved insulin sensitivity in adults with type 2 diabetes.
Collapse
Affiliation(s)
- Jiahui Zhao
- Department of Kinesiology and Health EducationThe University of Texas at Austin, Austin, Texas, United States
| | - Sahar D Massoudian
- Department of Kinesiology and Health EducationThe University of Texas at Austin, Austin, Texas, United States
| | - Sten Stray-Gundersen
- Department of Kinesiology and Health EducationThe University of Texas at Austin, Austin, Texas, United States
| | - Frank Wojan
- Department of Kinesiology and Health EducationThe University of Texas at Austin, Austin, Texas, United States
| | - Sophie Lalande
- Department of Kinesiology and Health EducationThe University of Texas at Austin, Austin, Texas, United States
| |
Collapse
|
8
|
Biyikoglu H, Robertson MD, Collins AL. Isolating the acute metabolic effects of carbohydrate restriction on postprandial metabolism with or without energy restriction: a crossover study. Eur J Nutr 2025; 64:133. [PMID: 40111529 PMCID: PMC11926029 DOI: 10.1007/s00394-025-03646-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 03/05/2025] [Indexed: 03/22/2025]
Abstract
Low-carbohydrate diets and intermittent energy restriction may offer metabolic advantages in fuel utilisation, that are independent of weight loss. The underlying mechanisms for these effects are unclear but may involve extensions of the catabolic phase and/or attenuation of insulin secretion. To address this gap, we aimed to investigate the independent acute metabolic effect of carbohydrate restriction at varying energy levels. Twelve, (six female) healthy overweight/obese participants (27.3 ± 1.8 years; 25.2 ± 1.6 kg/m2) completed this three-way study. Volunteers followed three diets for one day (36 h, covering the intervention day and overnight fasting), separated by 5-day washout: a normal carbohydrate, energy-balanced diet (nEB, 55% CHO), a low-carbohydrate, energy-balanced diet (LCEB, 50 g/day CHO), and a low-carbohydrate, energy-restricted diet (LC25, 50 g/day CHO with 75% energy restriction). Fasting and serial postprandial (360 min) measurements to a mixed test meal were collected the following morning. Additionally, subjective appetite responses and two-day subsequent ad libitum food intake was assessed. Both low-carbohydrate with and without energy restriction diets induced comparable decrease in triacylglycerol iAUC (p = 0.02, p = 0.04, respectively), and respiratory quotient (both p < 0.01) along with increase in non-esterified fatty acids (both p < 0.01) and 3-hydroxybutyrate (p = 0.001, p = 0.01, respectively) levels. Compared to a non-restricted carbohydrate, energy-balanced diet, postprandial glucose levels significantly increased in the LCEB arm (p = 0.024) and showed a rising trend in the LC25 arm (p = 0.07). Neither insulin responses nor resting, and diet-induced thermogenesis were significantly altered by variations in energy or carbohydrate content. These findings demonstrate that carbohydrate restriction, without altering energy intake, can elicit effects similar to those observed in short-term fasting. As such we propose a strategy of repeated carbohydrate restriction cycles alone may be an emerging alternative approach for the enhancement of cardiometabolic health, warranting further investigation.
Collapse
Affiliation(s)
- Hayriye Biyikoglu
- Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Surrey, GU2 7XH, UK
| | - M Denise Robertson
- Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Surrey, GU2 7XH, UK
- School of Life and Health Sciences, University of Roehampton, London, SW15 5PH, UK
| | - Adam L Collins
- Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Surrey, GU2 7XH, UK.
| |
Collapse
|
9
|
Neville J, Foley K, Lacey S, Tuthill A, Kgosidialwa O, O'Riordan M, O'Halloran F, Costelloe SJ. Impact of changes in gestational diabetes mellitus diagnostic criteria during the COVID-19 pandemic. Ir J Med Sci 2025:10.1007/s11845-025-03926-3. [PMID: 40072817 DOI: 10.1007/s11845-025-03926-3] [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: 08/28/2024] [Accepted: 02/19/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND/AIMS During the COVID-19 pandemic, the Health Service Executive (HSE) and Royal College of Obstetricians and Gynaecologists (RCOG) recommended fasting and random plasma glucose (FPG/RPG) alongside glycated haemoglobin (HbA1c) to replace the oral glucose tolerance test (OGTT) for diagnosing Gestational Diabetes Mellitus (GDM). METHODS The study compared testing patterns and diagnostic rates for GDM before and after implementing the RCOG guidelines (01/05/2020) in pregnancies beginning 01/11/2018 to 31/03/2021. Trends were inspected using Cochrane-Armitage tests. Differences between General Practice (GP) and Secondary Care (SCare) were assessed by chi-square analysis. A significance level of p < 0.05 was used for all analyses. Information on maternal and pregnancy characteristics was accessed where available. RESULTS Data indicated a significant reduction in OGTTs requested by GPs and SCare. Conversely, HbA1c, FPG and RPG test requests increased significantly in both locations. The overall GDM positivity rate increased significantly from 7.4% to 22.0% in GP and 16.9% to 39.0% in SCare following RCOG guideline implementation. CONCLUSIONS The RCOG guidelines appear to have been well adopted by GPs and SCare, with greater adherence in SCare. Using FPG, RPG and HbA1c to a greater extent than the OGTT corresponded with increased GDM diagnostic rates. Given the difficulties with interpreting HbA1c in pregnancy, its routine use in diagnosing GDM requires further careful consideration. Relaying changes in diagnostic protocol during pandemics requires strong communication with all requesting clinicians, including GPs. Comparisons between GP and SCare indicated significant differences in test-requesting practices and GDM positivity rates.
Collapse
Affiliation(s)
- Jessica Neville
- Department of Clinical Biochemistry, Cork University Hospital (CUH), T12 P928, Wilton, Cork, Ireland.
- Department of Biological Science, Munster Technological University, Bishopstown Campus, T12 P928, Cork, Ireland.
| | - Kelly Foley
- Department of Clinical Biochemistry, Cork University Hospital (CUH), T12 P928, Wilton, Cork, Ireland
| | - Seán Lacey
- Research Integrity & Compliance Officer, Munster Technological University, Rossa Ave, Bishopstown, T12 P928, Cork, Ireland
| | | | | | - Mairead O'Riordan
- Department of Obstetrics and Gynaecology, CUMH, T12 P928, Cork, Ireland
| | - Fiona O'Halloran
- Department of Biological Science, Munster Technological University, Bishopstown Campus, T12 P928, Cork, Ireland
| | - Seán J Costelloe
- Department of Clinical Biochemistry, Cork University Hospital (CUH), T12 P928, Wilton, Cork, Ireland
| |
Collapse
|
10
|
Osmulski ME, Yu Y, Kuang A, Josefson JL, Hivert MF, Scholtens DM, Lowe WL. Subtypes of Gestational Diabetes Mellitus Are Differentially Associated With Newborn and Childhood Metabolic Outcomes. Diabetes Care 2025; 48:390-399. [PMID: 39787502 PMCID: PMC11870284 DOI: 10.2337/dc24-1735] [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: 08/16/2024] [Accepted: 11/22/2024] [Indexed: 01/12/2025]
Abstract
OBJECTIVE Subtypes of gestational diabetes mellitus (GDM) based on insulin sensitivity and secretion have been described. We addressed the hypothesis that GDM subtypes are differentially associated with newborn and child anthropometric and glycemic outcomes. RESEARCH DESIGN AND METHODS Newborn and child (age 11-14 years) outcomes were examined in 7,970 and 4,160 mother-offspring dyads, respectively, who participated in the Hyperglycemia and Adverse Pregnancy Outcome (HAPO) study and HAPO Follow-Up Study. GDM was classified as insulin-deficient GDM (insulin secretion <25th percentile with preserved insulin sensitivity), insulin-resistant GDM (insulin sensitivity <25th percentile with preserved insulin secretion), or mixed-defect GDM (both <25th percentile). Regression models for newborn and child outcomes included adjustment for field center, maternal BMI, and other pregnancy covariates. Child models also included adjustment for child age, sex, and family history of diabetes. RESULTS Compared with mothers with normal glucose tolerance, all three GDM subtypes were associated with birth weight and sum of skinfolds >90th percentile. Insulin-resistant and mixed-defect GDM were associated with higher risk of cord C-peptide levels >90th percentile. Insulin-resistant GDM was associated with higher risk of neonatal hypoglycemia. Insulin-resistant GDM was associated with higher risk of neonatal hypoglycemia and childhood obesity (odds ratio [OR] 1.53, 95% CI 1.127-2.08). The risk of childhood impaired glucose tolerance was higher with insulin-resistant GDM (OR 2.21, 95% CI 1.50-3.25) and mixed-defect GDM (OR 3.01, 95% CI 1.47-6.19). CONCLUSIONS GDM subtypes are differentially associated with newborn and childhood outcomes. Better characterizing individuals with GDM could help identify at-risk offspring to offer targeted, preventative interventions early in life.
Collapse
Affiliation(s)
- Meredith E. Osmulski
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Yuanzhi Yu
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Alan Kuang
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Jami L. Josefson
- Division of Endocrinology, Department of Pediatrics, Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago, IL
| | - Marie-France Hivert
- Department of Medicine, Massachusetts General Hospital, Boston, MA
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Denise M. Scholtens
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - William L. Lowe
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
11
|
Thirunavukkarasu S, Ziegler TR, Weber MB, Staimez L, Lobelo F, Millard-Stafford ML, Schmidt MD, Venkatachalam A, Bajpai R, El Fil F, Prokou M, Kumar S, Tapp RJ, Shaw JE, Pasquel FJ, Nocera JR. High-Intensity Interval Training for Individuals With Isolated Impaired Fasting Glucose: Protocol for a Proof-of-Concept Randomized Controlled Trial. JMIR Res Protoc 2025; 14:e59842. [PMID: 39977858 PMCID: PMC11888011 DOI: 10.2196/59842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/16/2024] [Accepted: 10/09/2024] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Standard lifestyle interventions have shown limited efficacy in preventing type 2 diabetes among individuals with isolated impaired fasting glucose (i-IFG). Hence, tailored intervention approaches are necessary for this high-risk group. OBJECTIVE This study aims to (1) assess the feasibility of conducting a high-intensity interval training (HIIT) study and the intervention acceptability among individuals with i-IFG, and (2) investigate the preliminary efficacy of HIIT in reducing fasting plasma glucose levels and addressing the underlying pathophysiology of i-IFG. METHODS This study is a 1:1 proof-of-concept randomized controlled trial involving 34 physically inactive individuals (aged 35-65 years) who are overweight or obese and have i-IFG. Individuals will undergo a 3-step screening procedure to determine their eligibility: step 1 involves obtaining clinical information from electronic health records, step 2 consists of completing questionnaires, and step 3 includes blood tests. All participants will be fitted with continuous glucose monitoring devices for approximately 80 days, including 10 days prior to the intervention, the 8-week intervention period, and 10 days following the intervention. Intervention participants will engage in supervised HIIT sessions using stationary "spin" cycle ergometers in groups of 5 or fewer. The intervention will take place 3 times a week for 8 weeks at the Aerobic Exercise Laboratory in the Rehabilitation Hospital at Emory University. Control participants will be instructed to refrain from engaging in intense physical activities during the study period. All participants will receive instructions to maintain a eucaloric diet throughout the study. Baseline and 8-week assessments will include measurements of weight, blood pressure, body composition, waist and hip circumferences, as well as levels of fasting plasma glucose, 2-hour plasma glucose, and fasting insulin. Primary outcomes include feasibility parameters, intervention acceptability, and participants' experiences, perceptions, and satisfaction with the HIIT intervention, as well as facilitators and barriers to participation. Secondary outcomes comprise between-group differences in changes in clinical measures and continuous glucose monitoring metrics from baseline to 8 weeks. Quantitative data analysis will include descriptive statistics, correlation, and regression analyses. Qualitative data will be analyzed using framework-driven and thematic analyses. RESULTS Recruitment for the study is scheduled to begin in February 2025, with follow-up expected to be completed by the end of September 2025. We plan to publish the study findings by the end of 2025. CONCLUSIONS The study findings are expected to guide the design and execution of an adequately powered randomized controlled trial for evaluating HIIT efficacy in preventing type 2 diabetes among individuals with i-IFG. TRIAL REGISTRATION Clinicaltrials.gov NCT06143345; https://clinicaltrials.gov/study/NCT06143345. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) PRR1-10.2196/59842.
Collapse
Affiliation(s)
- Sathish Thirunavukkarasu
- Department of Family and Preventive Medicine, School of Medicine, Emory University, Atlanta, GA, United States
- Emory Global Diabetes Research Center, Woodruff Health Sciences Center, Emory University, Atlanta, GA, United States
| | - Thomas R Ziegler
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Mary Beth Weber
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Lisa Staimez
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Felipe Lobelo
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Mindy L Millard-Stafford
- Exercise Physiology Laboratory, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Michael D Schmidt
- Department of Kinesiology, University of Georgia, Atlanta, GA, United States
| | | | - Ram Bajpai
- School of Medicine, Keele University, Staffordshire, United Kingdom
| | - Farah El Fil
- University of Nicosia Medical School, Nicosia, Cyprus
| | - Maria Prokou
- University of Nicosia Medical School, Nicosia, Cyprus
| | - Siya Kumar
- College of Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Robyn J Tapp
- Research Institute for Health and Wellbeing, Coventry University, Coventry, United Kingdom
| | | | - Francisco J Pasquel
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Joe R Nocera
- Division of Physical Therapy, Departments of Neurology and Rehabilitation Medicine, School of Medicine, Emory University, Atlanta, GA, United States
- Center for Visual and Neurocognitive Rehabilitation, Atlanta, GA, United States
| |
Collapse
|
12
|
Moffa S, Sorice GP, Di Giuseppe G, Cinti F, Ciccarelli G, Soldovieri L, Brunetti M, Sonnino R, Nista EC, Gasbarrini A, Pontecorvi A, Mezza T, Giaccari A. A single bout of physical exercise improves 1-hour post-load plasma glucose in healthy young adults. J Endocrinol Invest 2025; 48:455-464. [PMID: 39347907 PMCID: PMC11785650 DOI: 10.1007/s40618-024-02438-8] [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: 02/24/2024] [Accepted: 07/29/2024] [Indexed: 10/01/2024]
Abstract
PURPOSE Physical exercise is a key component in the treatment of type 2 diabetes and plays an important role in maintaining a healthy glucose metabolism even in healthy subjects. To date, no studies have investigated the effect of a single bout of aerobic physical exercise on glucose metabolism in young, moderately active, healthy adults. METHODS We performed an OGTT 7 days before and 24 h after a single bout of physical exercise, to evaluate 1-hour post-load plasma glucose and surrogate indexes of insulin sensitivity and insulin secretion. RESULTS Glucose levels were significantly reduced after exercise at baseline and one hour after glucose load; similarly, insulin was significantly lower 1 h after glucose load. We found a significant increase in the Matsuda index, confirmed by OGIS index, QUICKI index, and by significant reduction in HOMA-IR. Conversely, we observed a trend to increase in HOMA-B. CONCLUSION This is the first study to evaluate the effect of a single bout of exercise on 1-hour glucose levels following OGTT. We found a significant reduction in 1-hour glucose levels following OGTT together with an increased insulin sensitivity. A single 30-minute bout of aerobic exercise also seemed to improve the insulin secretion pattern. Modifications in beta cell secretory capacity during exercise are likely secondary to an improvement in insulin action in insulin dependent tissues.
Collapse
Affiliation(s)
- Simona Moffa
- Centro per le Malattie Endocrine e Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Gian Pio Sorice
- Sezione di Medicina Interna, Endocrinologia, Andrologia e Malattie Metaboliche, Dipartimento di Medicina di Precisione e Rigenerativa e Area Jonica - (DiMePre-J), Università Degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Gianfranco Di Giuseppe
- Centro per le Malattie Endocrine e Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Francesca Cinti
- Centro per le Malattie Endocrine e Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Gea Ciccarelli
- Centro per le Malattie Endocrine e Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Laura Soldovieri
- Centro per le Malattie Endocrine e Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Michela Brunetti
- Centro per le Malattie Endocrine e Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Rebecca Sonnino
- Centro per le Malattie Endocrine e Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Enrico C Nista
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
- Pancreas Unit, Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell'Apparato Digerente, Fondazione Policlinico Universitario Gemelli IRCCS, Roma, Italy
| | - Antonio Gasbarrini
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
- Pancreas Unit, Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell'Apparato Digerente, Fondazione Policlinico Universitario Gemelli IRCCS, Roma, Italy
| | - Alfredo Pontecorvi
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Teresa Mezza
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy.
- Pancreas Unit, Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell'Apparato Digerente, Fondazione Policlinico Universitario Gemelli IRCCS, Roma, Italy.
| | - Andrea Giaccari
- Centro per le Malattie Endocrine e Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
13
|
Xu F, Wu S, Gao S, Li X, Huang C, Chen Y, Zhu P, Liu G. Causal association between insulin sensitivity index and Alzheimer's disease. J Neurochem 2025; 169:e16254. [PMID: 39479764 DOI: 10.1111/jnc.16254] [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: 04/13/2024] [Revised: 10/06/2024] [Accepted: 10/07/2024] [Indexed: 02/11/2025]
Abstract
Evidence from observational and Mendelian randomization (MR) studies suggested that insulin resistance (IR) was associated with Alzheimer's disease (AD). However, the causal effects of different indicators of IR on AD remain inconsistent. Here, we aim to assess the causal association between the insulin sensitivity index (ISI), a measure of post-prandial IR, and the risk of AD. We first conducted primary and secondary univariable MR analyses. We selected 8 independent genome-wide significant (p < 5E-08, primary analyses) and 61 suggestive (p < 1E-05, secondary analyses) ISI genetic variants from large-scale genome-wide association studies (GWAS; N = 53 657), respectively, and extracted their corresponding GWAS summary statistics from AD GWAS, including IGAP2019 (N = 63 926) and FinnGen_G6_AD_WIDE (N = 412 181). We selected five univariable MR methods and used heterogeneity, horizontal pleiotropy test, and leave-one-out sensitivity analysis to confirm the stability of MR estimates. Finally, we conducted a meta-analysis to combine MR estimates from two non-overlapping AD GWAS datasets. We further performed multivariable MR (MVMR) to assess the potential mediating role of type 2 diabetes (T2D) on the association between ISI and AD using two MVMR methods. In univariable MR, utilizing 8 genetic variants in primary analyses, we found a significant causal association of genetically increased ISI with decreased risk of AD (OR = 0.79, 95% CI: 0.68-0.92, p = 0.003). Utilizing 61 genetic variants in secondary analyses, we found consistent findings of a causal effect of genetically increased ISI on the decreased risk of AD (OR = 0.89, 95% CI: 0.82-0.96, p = 0.003). Heterogeneity, horizontal pleiotropy test, and leave-one-out sensitivity analysis ensured the reliability of the MR estimates. In MVMR, we found no causal relationship between ISI and AD after adjusting for T2D (p > 0.05). We provide genetic evidence that increased ISI is significantly and causally associated with reduced risk of AD, which is mediated by T2D. These findings may inform prevention strategies directed toward IR-associated T2D and AD.
Collapse
Affiliation(s)
- Fang Xu
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, China
| | - Shiyang Wu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Shan Gao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Xuan Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Chen Huang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Yan Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, Wuhu, Anhui, China
| | - Ping Zhu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Guiyou Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, Wuhu, Anhui, China
- Brain Hospital, Shengli Oilfield Central Hospital, Dongying, China
- Beijing Key Laboratory of Hypoxia Translational Medicine, National Engineering Laboratory of Internet Medical Diagnosis and Treatment Technology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Wang J, Yan D, Wang S, Zhao A, Hou X, Zheng X, Guo J, Shen L, Bao Y, Jia W, Yu X, Hu C, Zhang Z. Osteopenia Metabolomic Biomarkers for Early Warning of Osteoporosis. Metabolites 2025; 15:66. [PMID: 39852408 PMCID: PMC11767427 DOI: 10.3390/metabo15010066] [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: 12/13/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
Introduction: This study aimed to capture the early metabolic changes before osteoporosis occurs and identify metabolomic biomarkers at the osteopenia stage for the early prevention of osteoporosis. Materials and Methods: Metabolomic data were generated from normal, osteopenia, and osteoporosis groups with 320 participants recruited from the Nicheng community in Shanghai. We conducted individual edge network analysis (iENA) combined with a random forest to detect metabolomic biomarkers for the early warning of osteoporosis. Weighted Gene Co-Expression Network Analysis (WGCNA) and mediation analysis were used to explore the clinical impacts of metabolomic biomarkers. Results: Visual separations of the metabolic profiles were observed between three bone mineral density (BMD) groups in both genders. According to the iENA approach, several metabolites had significant abundance and association changes in osteopenia participants, confirming that osteopenia is a critical stage in the development of osteoporosis. Metabolites were further selected to identify osteopenia (nine metabolites in females; eight metabolites in males), and their ability to discriminate osteopenia was improved significantly compared to traditional bone turnover markers (BTMs) (female AUC = 0.717, 95% CI 0.547-0.882, versus BTMs: p = 0.036; male AUC = 0.801, 95% CI 0.636-0.966, versus BTMs: p = 0.007). The roles of the identified key metabolites were involved in the association between total fat-free mass (TFFM) and osteopenia in females. Conclusion: Osteopenia was identified as a tipping point during the development of osteoporosis with metabolomic characteristics. A few metabolites were identified as candidate early-warning biomarkers by machine learning analysis, which could indicate bone loss and provide new prevention guidance for osteoporosis.
Collapse
Affiliation(s)
- Jie Wang
- Department of Osteoporosis, Metabolic Bone Disease and Genetic Research Unit, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Dandan Yan
- Department of Endocrinology and Metabolism, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Diabetes Institute, Shanghai Clinical Center of Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200233, China
| | - Suna Wang
- Clinical Research Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Aihua Zhao
- Center for Translational Medicine, and Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xuhong Hou
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xiaojiao Zheng
- Center for Translational Medicine, and Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jingyi Guo
- Clinical Research Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Li Shen
- Clinical Research Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yuqian Bao
- Department of Endocrinology and Metabolism, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Diabetes Institute, Shanghai Clinical Center of Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200233, China
| | - Wei Jia
- Hong Kong Phenome Research Centre, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China
| | - Xiangtian Yu
- Clinical Research Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Zhenlin Zhang
- Department of Osteoporosis, Metabolic Bone Disease and Genetic Research Unit, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| |
Collapse
|
15
|
Ramírez-Vélez R, Correa-Rodríguez M, Calderón-González JC, Dávila-Grisales A, González-Ruíz K, Correa-Bautista JE, Izquierdo M. The association between insulin resistance and cytokines in adolescents with excess of adiposity. Curr Probl Cardiol 2025; 50:102925. [PMID: 39510396 DOI: 10.1016/j.cpcardiol.2024.102925] [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: 11/04/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND AND AIMS Increased adiposity has been directly associated with insulin resistance (IR), and cytokines released by adipose tissue seem to link adiposity to IR in youth. We used an antibody-based array to investigate the differential levels of serum cytokines according to insulin status in a cohort of overweight/obese and inactive adolescents and evaluated their potential associations with clinical and metabolic characteristics. METHODS AND RESULTS We performed a cross-sectional data analysis from 122 adolescents (11-17 years of age). We assessed body composition, cardiometabolic risk factors, biochemical variables, and physical fitness. The concentration of 55 cytokines was quantified in blood samples. The homeostasis model assessment insulin resistance (HOMA-IR) and AST/ALT and TG/HDL ratios were calculated. IR adolescents as defined as HOMA-IR >2.5. The number of adolescents with IR in the study was 91 (66 % girls). In the IS group, after controlling for confounders, higher IL-15 levels were significantly associated with higher alanine aminotransferase levels and lower AST/ALT ratio, respectively (Ps<0.05). In the same line, there were significantly higher alanine aminotransferase levels and lower AST/ALT ratio, respectively, with FGF-9 (Ps<0.05). Likewise, higher alanine aminotransferase levels were significantly associated positively with HGF (p=0.045). Additionally, leptin levels are associated with six adiposity indexes (i.e., fat mass/height index, body fat, body mass index, android fat mass and gynoid fat mass) in overweight/obese adolescents with IR (Ps<0.05). CONCLUSIONS These data may provide novel insights into the pathogenic mechanisms underlying IR in youth, offering new targets for prevention.
Collapse
Affiliation(s)
- Robinson Ramírez-Vélez
- Navarrabiomed, Hospital Universitario de Navarra (HUN)-Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain; CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain; Facultad de Ciencias de la Educación, Unidad Central del Valle del Cauca (UCEVA), Túlua, Colombia.
| | - María Correa-Rodríguez
- Faculty of Health Science, Department of Nursing, University of Granada, Av. Ilustración, 60 18016 Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | | | - Albeiro Dávila-Grisales
- Facultad de Ciencias de la Educación, Unidad Central del Valle del Cauca (UCEVA), Túlua, Colombia
| | - Katherine González-Ruíz
- Grupo de Investigación Salud y Movimiento. Programa de Fisioterapia. Facultad de Salud, Universidad Santiago de Cali 760035, Colombia
| | | | - Mikel Izquierdo
- Navarrabiomed, Hospital Universitario de Navarra (HUN)-Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain; CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
16
|
Low JL, Marcotte-Chénard A, Tremblay R, Islam H, Falkenhain K, Mampuya WM, Mari A, McManus AM, Riesco E, Little JP. An acute bout of 4 × 4-min or 10 × 1-min HIIT improves β cell glucose sensitivity in postmenopausal females with type 2 diabetes: a secondary analysis. J Appl Physiol (1985) 2025; 138:311-317. [PMID: 39694495 DOI: 10.1152/japplphysiol.00777.2024] [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: 10/08/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Improvements in glycemic control following acute exercise are typically attributed to improved postexercise insulin sensitivity (IS) with comparatively little known about how acute exercise impacts β cell function, especially in postmenopausal females. We determined how two high-intensity interval training (HIIT) protocols, matched for total estimated energy expenditure, impact β cell function in postmenopausal females with type 2 diabetes. Thirteen postmenopausal females (70 ± 5 yr; 12 ± 7 yr since diagnosis, 80.9 ± 13.8 kg, 32.4 ± 5.6 kg·m2; HbA1c-49.8 ± 10.3 mmol/mol [6.7 ± 1.0]) living with type 2 diabetes were included in this semirandomized crossover trial. The trial involved an initial resting control condition followed by two HIIT conditions [4 × 4-min HIIT (HIIT4) and 10 × 1-min HIIT (HIIT10)] completed in a randomized order 2-4 days apart. β cell function (glucose sensitivity) and insulin sensitivity were determined from a 2-h mixed-meal tolerance test performed 2 h after rest or HIIT. Both HIIT4 and HIIT10 significantly improved β cell glucose sensitivity compared with control (15 pmol/min/m2/[mmol/L], [95% confidence interval (CI) 6, 23]; P = 0.002 and 16 pmol/min/m2/[mmol/L], [95% CI 7, 25]; P = 0.002, respectively), with no difference between HIIT protocols (1 [-8, 10], P = 0.79). There were no significant differences in IS metrics (Matsuda index, OGIS, Stumvoli, and QUICKI) between the conditions. An acute bout of 4 × 4-min or 10 × 1-min HIIT improves β cell glucose sensitivity in postmenopausal females living with type 2 diabetes. ClinicalTrials.gov: NCT04986345.NEW & NOTEWORTHY This is the first study to explore the effects of acute high-intensity interval training (HIIT) on β cell function in postmenopausal women with type 2 diabetes. Our crossover trial compares two HIIT protocols, matched for total estimated energy expenditure, examining their impacts on β cell function and insulin sensitivity. Despite the absence of an insulin-sensitizing effect, we show robust effects of HIIT on β-cell function, including an improvement in β-cell glucose sensitivity.
Collapse
Affiliation(s)
- J L Low
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, British Columbia, Canada
| | - A Marcotte-Chénard
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, British Columbia, Canada
- Research Centre on Aging, CIUSSS de l'Estrie-CHUS, Sherbrooke, Quebec, Canada
- Faculty of Physical Activity Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - R Tremblay
- Research Centre on Aging, CIUSSS de l'Estrie-CHUS, Sherbrooke, Quebec, Canada
- Faculty of Physical Activity Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - H Islam
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, British Columbia, Canada
| | - K Falkenhain
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, British Columbia, Canada
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, United States
| | - W M Mampuya
- CHUS Research Centre, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - A Mari
- Institute of Neuroscience, National Research Council, Padova, Italy
| | - A M McManus
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, British Columbia, Canada
| | - E Riesco
- Research Centre on Aging, CIUSSS de l'Estrie-CHUS, Sherbrooke, Quebec, Canada
- Faculty of Physical Activity Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - J P Little
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, British Columbia, Canada
| |
Collapse
|
17
|
Metwally AA, Perelman D, Park H, Wu Y, Jha A, Sharp S, Celli A, Ayhan E, Abbasi F, Gloyn AL, McLaughlin T, Snyder MP. Prediction of metabolic subphenotypes of type 2 diabetes via continuous glucose monitoring and machine learning. Nat Biomed Eng 2024:10.1038/s41551-024-01311-6. [PMID: 39715896 DOI: 10.1038/s41551-024-01311-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 11/01/2024] [Indexed: 12/25/2024]
Abstract
The classification of type 2 diabetes and prediabetes does not consider heterogeneity in the pathophysiology of glucose dysregulation. Here we show that prediabetes is characterized by metabolic heterogeneity, and that metabolic subphenotypes can be predicted by the shape of the glucose curve measured via a continuous glucose monitor (CGM) during standardized oral glucose-tolerance tests (OGTTs) performed in at-home settings. Gold-standard metabolic tests in 32 individuals with early glucose dysregulation revealed dominant or co-dominant subphenotypes (muscle or hepatic insulin-resistance phenotypes in 34% of the individuals, and β-cell-dysfunction or impaired-incretin-action phenotypes in 40% of them). Machine-learning models trained with glucose time series from OGTTs from the 32 individuals predicted the subphenotypes with areas under the curve (AUCs) of 95% for muscle insulin resistance, 89% for β-cell deficiency and 88% for impaired incretin action. With CGM-generated glucose curves obtained during at-home OGTTs, the models predicted the muscle-insulin-resistance and β-cell-deficiency subphenotypes of 29 individuals with AUCs of 88% and 84%, respectively. At-home identification of metabolic subphenotypes via a CGM may aid the risk stratification of individuals with early glucose dysregulation.
Collapse
Affiliation(s)
- Ahmed A Metwally
- Department of Genetics, Stanford University, Stanford, CA, USA
- Systems and Biomedical Engineering Department, Cairo University, Giza, Egypt
- Google LLC, Mountain View, CA, USA
| | - Dalia Perelman
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - Heyjun Park
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Yue Wu
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Alokkumar Jha
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Seth Sharp
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | | | - Ekrem Ayhan
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - Fahim Abbasi
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - Anna L Gloyn
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Centre, Stanford University, Stanford, CA, USA
| | | | | |
Collapse
|
18
|
Tang Y, Li L, Li J. Correlations of the triglyceride-glucose index and modified indices with arterial stiffness in overweight or obese adults. Front Endocrinol (Lausanne) 2024; 15:1499120. [PMID: 39741881 PMCID: PMC11685072 DOI: 10.3389/fendo.2024.1499120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
Background Insulin resistance (IR) contributes substantially to the development of cardiovascular disease (CVD) and metabolic disorders, particularly obesity. The homeostatic model assessment of IR is a prevalent IR indicator, but insulin measurement is quite impractical for widely use. Given its convenience and accessibility, the triglyceride-glucose (TyG) index, along with modified indices such as the triglyceride-glucose-waist circumference (TyG-WC) and triglyceride-glucose-waist-height ratio (TyG-WHtR), are gaining recognition as practical tools for assessing IR. This study aimed to investigate the specific correlation between the TyG index and its modified indices with arterial stiffness in an overweight or obese population and to explore novel, self-defined modified TyG indices for identifying individuals at elevated risk for such conditions. Methods This retrospective study included 1,143 overweight or obese individuals from 2021 to 2023. Medical data, including brachial-ankle pulse wave velocity (baPWV), were collected. Two novel modified TyG indices, TyG-1h and TyG-2h, were defined by substituting the fasting glucose level in the TyG formula with 1-hour and 2-hour post-load plasma glucose levels, respectively. Multivariate logistic regression analyses were conducted to identify parameters that demonstrated a statistically significant correlation with arterial stiffness, defined as a baPWV threshold of ≥ 1400 cm/s. Additionally, restricted cubic spline (RCS) modelling was employed to further explore these relationships in a visually interpretable manner. To evaluate and compare the diagnostic accuracy of the conventional TyG index and its novel modified versions, receiver operating characteristic (ROC) curve analyses were performed. Results Our findings revealed that individuals with arterial stiffness presented significantly elevated TyG index and all its modified versions (P< 0.05). By utilizing a binary logistic regression model and adjusting for potential confounders, we determined that all TyG-related parameters independently correlated with an increased risk of developing arterial stiffness. Moreover, TyG-WHtR displayed the best correlation (OR 3.071, 95% CI 1.496-6.303) when stratified by quartiles, followed by TyG-1h (OR 2.298, 95% CI 1.248-4.234) and TyG-2h (OR 2.115, 95% CI 1.175-3.807). ROC curves suggested that TyG-1h and TyG-2h demonstrated superior diagnostic performance compared to TyG, with AUCs of 0.685, 0.679 and 0.673, respectively. Conclusions The modified TyG indices exhibited strong effectiveness in identifying arterial stiffness in Chinese overweight or obese individuals.
Collapse
Affiliation(s)
| | | | - Jialin Li
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Ningbo
University, Ningbo, China
| |
Collapse
|
19
|
Mather KJ, Mari A, Heise T, DeVries JH, Hua M, Urva S, Coskun T, Haupt A, Heine RJ, Pratt E, Thomas MK, Milicevic Z. Effects of Tirzepatide vs Semaglutide on β-Cell Function, Insulin Sensitivity, and Glucose Control During a Meal Test. J Clin Endocrinol Metab 2024; 109:3046-3054. [PMID: 38795393 DOI: 10.1210/clinem/dgae319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/16/2024] [Accepted: 05/06/2024] [Indexed: 05/27/2024]
Abstract
CONTEXT In a clinical study, tirzepatide, a glucose-dependent insulinotropic polypeptide/glucagon-like peptide-1 receptor agonist (GIP/GLP-1RA), provided superior glycemic control vs the GLP-1RA semaglutide. The physiologic mechanisms are incompletely understood. OBJECTIVE This work aimed to evaluate treatment effects by model-based analyses of mixed-meal tolerance test (MMTT) data. METHODS A 28-week double-blind, randomized, placebo-controlled trial of patients with type 2 diabetes treated with metformin was conducted at 2 clinical research centers in Germany. Interventions included tirzepatide 15 mg, semaglutide 1 mg, and placebo. Main outcome measures included glycemic control, model-derived β-cell function indices including insulin secretion rate (ISR) at 7.2-mmol/L glucose (ISR7.2), β-cell glucose sensitivity (β-CGS), insulin sensitivity, and estimated hepatic insulin-to-glucagon ratio. RESULTS Tirzepatide significantly reduced fasting glucose and MMTT total glucose area under the curve (AUC) vs semaglutide (P < .01). Incremental glucose AUC did not differ significantly between treatments; therefore, greater total glucose AUC reduction with tirzepatide was mainly attributable to greater suppression of fasting glucose. A greater reduction in total ISR AUC was achieved with tirzepatide vs semaglutide (P < .01), in the context of greater improvement in insulin sensitivity with tirzepatide (P < .01). ISR7.2 was significantly increased with tirzepatide vs semaglutide (P < .05), showing improved β-CGS. MMTT-derived β-CGS was increased but not significantly different between treatments. Both treatments reduced fasting glucagon and total glucagon AUC, with glucagon AUC significantly reduced with tirzepatide vs semaglutide (P < .01). The estimated hepatic insulin-to-glucagon ratio did not change substantially with either treatment. CONCLUSION These results suggest that the greater glycemic control observed for tirzepatide manifests as improved fasting glucose and glucose excursion control, due to improvements in ISR, insulin sensitivity, and glucagon suppression.
Collapse
Affiliation(s)
- Kieren J Mather
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, 2-35122 Padua, Italy
| | - Tim Heise
- Profil Institute for Metabolic Research, 41460 Neuss, Germany
| | - J Hans DeVries
- Profil Institute for Metabolic Research, 41460 Neuss, Germany
| | - Ming Hua
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Shweta Urva
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Tamer Coskun
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Axel Haupt
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Robert J Heine
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Edward Pratt
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Melissa K Thomas
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Zvonko Milicevic
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| |
Collapse
|
20
|
Liu J, An Y, Yang N, Xu Y, Wang G. Longitudinal associations of dietary fiber and its source with 48-week weight loss maintenance, cardiometabolic risk factors and glycemic status under metformin or acarbose treatment: a secondary analysis of the March randomized trial. Nutr Diabetes 2024; 14:81. [PMID: 39358341 PMCID: PMC11447090 DOI: 10.1038/s41387-024-00340-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
AIMS To examine longitudinal and dose-d ependent associations between dietary fiber intake and various clinical outcomes over 48 weeks of pharmacological treatment in T2DM patients. METHODS In this secondary analysis, we used data from the MARCH trial, which was designed to compare the efficacy of acarbose or metformin monotherapy as the initial therapy in Chinese patients newly diagnosed with T2DM. Dietary data were obtained using a 24-h dietary recall method to evaluate the intakes of dietary fiber from different sources as well as the carbohydrate-to-fiber ratio. RESULTS A total of 551 newly-diagnosed patients with T2DM complete dietary records (286 in the acarbose group and 265 in the metformin group) were included. Higher intake of total fiber and whole grain fiber was positively associated with better β-cell function, insulin sensitivity and postprandial glycemic control under acarbose treatment. Higher intake of legume fiber was associated with better glycemic control under both acarbose and metformin treatment but with better weight loss only under metformin treatment. A high-carbohydrate-low-fiber diet was associated with worse glycemic control and lower HDL-C under acarbose treatment but with higher insulin sensitivity and better weight loss under metformin treatment. CONCLUSIONS The notable effects of various dietary fibers when combined with different oral glucose-lowering medications should be considered to maximize therapeutic benefit.
Collapse
Affiliation(s)
- Jia Liu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yu An
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ning Yang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yuan Xu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
21
|
Madsen AL, Bonàs-Guarch S, Gheibi S, Prasad R, Vangipurapu J, Ahuja V, Cataldo LR, Dwivedi O, Hatem G, Atla G, Guindo-Martínez M, Jørgensen AM, Jonsson AE, Miguel-Escalada I, Hassan S, Linneberg A, Ahluwalia TS, Drivsholm T, Pedersen O, Sørensen TIA, Astrup A, Witte D, Damm P, Clausen TD, Mathiesen E, Pers TH, Loos RJF, Hakaste L, Fex M, Grarup N, Tuomi T, Laakso M, Mulder H, Ferrer J, Hansen T. Genetic architecture of oral glucose-stimulated insulin release provides biological insights into type 2 diabetes aetiology. Nat Metab 2024; 6:1897-1912. [PMID: 39420167 PMCID: PMC11496110 DOI: 10.1038/s42255-024-01140-6] [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: 07/30/2023] [Accepted: 09/02/2024] [Indexed: 10/19/2024]
Abstract
The genetics of β-cell function (BCF) offer valuable insights into the aetiology of type 2 diabetes (T2D)1,2. Previous studies have expanded the catalogue of BCF genetic associations through candidate gene studies3-7, large-scale genome-wide association studies (GWAS) of fasting BCF8,9 or functional islet studies on T2D risk variants10-14. Nonetheless, GWAS focused on BCF traits derived from oral glucose tolerance test (OGTT) data have been limited in sample size15,16 and have often overlooked the potential for related traits to capture distinct genetic features of insulin-producing β-cells17,18. We reasoned that investigating the genetic basis of multiple BCF estimates could provide a broader understanding of β-cell physiology. Here, we aggregate GWAS data of eight OGTT-based BCF traits from ~26,000 individuals of European descent, identifying 55 independent genetic associations at 44 loci. By examining the effects of BCF genetic signals on related phenotypes, we uncover diverse disease mechanisms whereby genetic regulation of BCF may influence T2D risk. Integrating BCF-GWAS data with pancreatic islet transcriptomic and epigenomic datasets reveals 92 candidate effector genes. Gene silencing in β-cell models highlights ACSL1 and FAM46C as key regulators of insulin secretion. Overall, our findings yield insights into the biology of insulin release and the molecular processes linking BCF to T2D risk, shedding light on the heterogeneity of T2D pathophysiology.
Collapse
Affiliation(s)
- A L Madsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - S Bonàs-Guarch
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - S Gheibi
- Department of Clinical Sciences, Unit of Molecular Metabolism, Lund University, Malmö, Sweden
| | - R Prasad
- Department of Clinical Sciences, Unit of Genomics, Diabetes and Endocrinology, Lund University, Malmö, Sweden
| | - J Vangipurapu
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - V Ahuja
- Institute for Molecular Medicine Finland and Research Program of Clinical and Molecular Medicine, University of Helsinki, Helsinki, Finland
| | - L R Cataldo
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
- Department of Clinical Sciences, Unit of Molecular Metabolism, Lund University, Malmö, Sweden
| | - O Dwivedi
- Institute for Molecular Medicine Finland and Research Program of Clinical and Molecular Medicine, University of Helsinki, Helsinki, Finland
- Folkhalsan Research Centre, Helsinki, Finland
| | - G Hatem
- Department of Clinical Sciences, Unit of Genomics, Diabetes and Endocrinology, Lund University, Malmö, Sweden
| | - G Atla
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - M Guindo-Martínez
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - A M Jørgensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - A E Jonsson
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - I Miguel-Escalada
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - S Hassan
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - A Linneberg
- Center for Clinical Research and Prevention, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health Sciences, UCPH, Copenhagen, Denmark
| | - Tarunveer S Ahluwalia
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- The Bioinformatics Center, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - T Drivsholm
- Center for Clinical Research and Prevention, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Section of General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - O Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - T I A Sørensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
- Department of Public Health Sciences (Section of Epidemiology), University of Copenhagen, Copenhagen, Denmark
| | - A Astrup
- Novo Nordisk Fonden, Hellerup, Denmark
| | - D Witte
- Institut for Folkesundhed-Epidemiologi, Aarhus University, Aarhus, Denmark
| | - P Damm
- Center for Pregnant Women with Diabetes and Department of Gynecology, Fertility, and Obstetrics and Department of Clinical Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - T D Clausen
- Center for Pregnant Women with Diabetes and Department of Gynecology, Fertility, and Obstetrics and Department of Clinical Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - E Mathiesen
- Center for Pregnant Women with Diabetes, Department of Nephrology and Endocrinology and Department of Clinical Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - T H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - R J F Loos
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - L Hakaste
- Institute for Molecular Medicine Finland and Research Program of Clinical and Molecular Medicine, University of Helsinki, Helsinki, Finland
- Folkhalsan Research Centre, Helsinki, Finland
| | - M Fex
- Department of Clinical Sciences, Unit of Molecular Metabolism, Lund University, Malmö, Sweden
| | - N Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - T Tuomi
- Department of Clinical Sciences, Unit of Genomics, Diabetes and Endocrinology, Lund University, Malmö, Sweden
- Institute for Molecular Medicine Finland and Research Program of Clinical and Molecular Medicine, University of Helsinki, Helsinki, Finland
- Folkhalsan Research Centre, Helsinki, Finland
- Helsinki University Hospital, Abdominal Centre / Endocrinology, Helsinki, Finland
| | - M Laakso
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - H Mulder
- Department of Clinical Sciences, Unit of Molecular Metabolism, Lund University, Malmö, Sweden
| | - J Ferrer
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - T Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen (UCPH), Copenhagen, Denmark.
| |
Collapse
|
22
|
Scifres CM, Davis EM, Orris S, Costacou T, Lalama C, Abebe KZ, Catalano P. Metabolic factors and perinatal outcomes among pregnant individuals with mild glucose intolerance. Diabetes Res Clin Pract 2024; 216:111830. [PMID: 39159865 DOI: 10.1016/j.diabres.2024.111830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/11/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
AIMS Metabolic characteristics and outcomes were compared among pregnant individuals with varying levels of glucose intolerance. METHODS 827 participants from a randomized clinical trial comparing the IADPSG and Carpenter Coustan Criteria were grouped as follows: normal glucose tolerance, mild glucose intolerance (100 g OGTT with one abnormal value) and treated GDM (diagnosed by Carpenter Coustan or IADPSG criteria). Differences in metabolic characteristics and perinatal outcomes were assessed using inverse probability of treatment weighting. RESULTS Mild glucose intolerance had lower insulin sensitivity and beta cell response than normal glucose tolerance, and similar findings to treated GDM. Small for gestational age (SGA) (OR 0.13, 95% CI 0.08-0.24) and neonatal composite morbidity were lower (OR 0.53, 95% CI 0.38-0.74), and maternal composite morbidity higher (OR 2.03, 95% CI 1.57-2.62) when comparing mild intolerance to normal glucose tolerance. Large for gestational age (OR 3.42 95% CI 1.39-8.41) was higher while SGA (OR 0.21, 95% CI 0.05-0.81) and neonatal composite morbidity (OR 0.31, 95% CI 0.17-0.57) were lower with mild glucose intolerance compared to treated GDM. CONCLUSIONS Mild glucose intolerance has a similar metabolic profile to treated GDM, and outcome differences are likely related to knowledge of diagnosis and treatment. CLINICAL TRIALS REGISTRY NCT02309138.
Collapse
Affiliation(s)
| | - Esa M Davis
- University of Maryland School of Medicine, Baltimore, MD, United States.
| | - Steve Orris
- University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.
| | - Tina Costacou
- University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.
| | - Christna Lalama
- University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.
| | - Kaleab Z Abebe
- University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.
| | | |
Collapse
|
23
|
Bonnefond A, Froguel P. Genetics brings new insight to β-cell function. Nat Metab 2024; 6:1848-1849. [PMID: 39420166 DOI: 10.1038/s42255-024-01131-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Affiliation(s)
- Amélie Bonnefond
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France.
- Department of Metabolism, Imperial College London, London, UK.
| | - Philippe Froguel
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France.
- Department of Metabolism, Imperial College London, London, UK.
| |
Collapse
|
24
|
Lin Y, Zhang Y, Shen X, Weng Z, Huang L, Zhao F, Yan S. Body Composition Changes Impact Islet β-Cell Function in Patients With Type 2 Diabetes Mellitus. J Lipids 2024; 2024:4986998. [PMID: 39376578 PMCID: PMC11458290 DOI: 10.1155/2024/4986998] [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: 06/25/2024] [Accepted: 09/18/2024] [Indexed: 10/09/2024] Open
Abstract
Background: Identifying β-cells dysregulation in type 2 diabetes mellitus (T2DM) is crucial. Weight fluctuations are frequently observed during diabetes treatment. However, the relationship between body composition changes and islet β-cell function in individuals with T2DM remains insufficiently investigated. Methods: This retrospective longitudinal study encompassed a cohort of 775 T2DM patients, who underwent body composition measuring using dual-energy X-ray absorptiometry (DEXA) and followed up for a median of 2.29 years. Key metrics included body mass index (BMI), fat mass index (FMI), trunk fat mass index (TFMI), muscle mass index (MMI), appendicular skeletal muscle mass index (ASMI), muscle/fat mass ratio (M/F), and the appendicular skeletal muscle mass/trunk fat mass ratio (A/T) were then categorized and grouped. Insulin, C-peptide, and glucose levels were assessed concurrently following a glucose load. β-cell function included insulin resistance (HOMA-IR), insulin sensitivity (Matsuda index (MI)), and insulin secretion evaluated by HOMA-β and C-peptidogenic index (CGI). Results: Although no significant changes in BMI were observed, patients with T2DM at readmission exhibited higher FMI, TFMI, and ASMI, as well as elevated levels of HOMA-IR, MI, and CGI compared to baseline measurements. And lower MI, higher levels of CGI, and HOMA-IR were observed in BMI increased group. Univariate correlation analysis revealed a negative association between changes in BMI (ΔBMI) and ΔMI, while positive associations were observed in both ΔHOMA-IR and ΔCGI. Among body composition indexes, ΔFMI exhibited the strongest correlation with ΔHOMA-IR (r = 0.255, p < 0.001), and ΔASMI was positively associated with ΔMI and ΔCGI (r = 0.131 and 0.194, respectively). Moreover, increased levels of BMI and FMI were associated with a greater risk of progressive insulin resistance compared to the decreased, whereas the trend was converse in ASMI and A/T. Conclusions: Increased FMI may partially contribute to the deterioration of insulin resistance, while increased ASMI is associated with improved insulin sensitivity and secretion. Maintaining an appropriate BMI and muscle/fat ratio is conductive to prevent the progression of insulin resistance in patients with T2DM.
Collapse
Affiliation(s)
- Yuxi Lin
- Department of Endocrinologythe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Department of EndocrinologyNational Regional Medical CenterBinhai Campus of the First Affiliated HospitalFujian Medical University, Fuzhou 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Provincethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolismthe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Diabetes Research Institute of Fujian Provincethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Metabolic Diseases Research Institutethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
| | - Yongze Zhang
- Department of Endocrinologythe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Department of EndocrinologyNational Regional Medical CenterBinhai Campus of the First Affiliated HospitalFujian Medical University, Fuzhou 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Provincethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolismthe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Diabetes Research Institute of Fujian Provincethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Metabolic Diseases Research Institutethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
| | - Ximei Shen
- Department of Endocrinologythe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Department of EndocrinologyNational Regional Medical CenterBinhai Campus of the First Affiliated HospitalFujian Medical University, Fuzhou 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Provincethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolismthe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Diabetes Research Institute of Fujian Provincethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Metabolic Diseases Research Institutethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
| | - Zhiyan Weng
- Department of Endocrinologythe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Department of EndocrinologyNational Regional Medical CenterBinhai Campus of the First Affiliated HospitalFujian Medical University, Fuzhou 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Provincethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolismthe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Diabetes Research Institute of Fujian Provincethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Metabolic Diseases Research Institutethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
| | - Lingning Huang
- Department of Endocrinologythe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Department of EndocrinologyNational Regional Medical CenterBinhai Campus of the First Affiliated HospitalFujian Medical University, Fuzhou 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Provincethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolismthe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Diabetes Research Institute of Fujian Provincethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Metabolic Diseases Research Institutethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
| | - Fengying Zhao
- Department of Endocrinologythe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Department of EndocrinologyNational Regional Medical CenterBinhai Campus of the First Affiliated HospitalFujian Medical University, Fuzhou 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Provincethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolismthe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Diabetes Research Institute of Fujian Provincethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Metabolic Diseases Research Institutethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
| | - Sunjie Yan
- Department of Endocrinologythe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Department of EndocrinologyNational Regional Medical CenterBinhai Campus of the First Affiliated HospitalFujian Medical University, Fuzhou 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Provincethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolismthe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Diabetes Research Institute of Fujian Provincethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
- Metabolic Diseases Research Institutethe First Affiliated HospitalFujian Medical University, Fuzhou 350005, China
| |
Collapse
|
25
|
Metwally AA, Perelman D, Park H, Wu Y, Jha A, Sharp S, Celli A, Ayhan E, Abbasi F, Gloyn AL, McLaughlin T, Snyder M. Predicting Type 2 Diabetes Metabolic Phenotypes Using Continuous Glucose Monitoring and a Machine Learning Framework. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.20.24310737. [PMID: 39108516 PMCID: PMC11302614 DOI: 10.1101/2024.07.20.24310737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/11/2024]
Abstract
Type 2 diabetes (T2D) and prediabetes are classically defined by the level of fasting glucose or surrogates such as hemoglobin HbA1c. This classification does not take into account the heterogeneity in the pathophysiology of glucose dysregulation, the identification of which could inform targeted approaches to diabetes treatment and prevention and/or predict clinical outcomes. We performed gold-standard metabolic tests in a cohort of individuals with early glucose dysregulation and quantified four distinct metabolic subphenotypes known to contribute to glucose dysregulation and T2D: muscle insulin resistance, β-cell dysfunction, impaired incretin action, and hepatic insulin resistance. We revealed substantial inter-individual heterogeneity, with 34% of individuals exhibiting dominance or co-dominance in muscle and/or liver IR, and 40% exhibiting dominance or co-dominance in β-cell and/or incretin deficiency. Further, with a frequently-sampled oral glucose tolerance test (OGTT), we developed a novel machine learning framework to predict metabolic subphenotypes using features from the dynamic patterns of the glucose time-series ("shape of the glucose curve"). The glucose time-series features identified insulin resistance, β-cell deficiency, and incretin defect with auROCs of 95%, 89%, and 88%, respectively. These figures are superior to currently-used estimates. The prediction of muscle insulin resistance and β-cell deficiency were validated using an independent cohort. We then tested the ability of glucose curves generated by a continuous glucose monitor (CGM) worn during at-home OGTTs to predict insulin resistance and β-cell deficiency, yielding auROC of 88% and 84%, respectively. We thus demonstrate that the prediabetic state is characterized by metabolic heterogeneity, which can be defined by the shape of the glucose curve during standardized OGTT, performed in a clinical research unit or at-home setting using CGM. The use of at-home CGM to identify muscle insulin resistance and β-cell deficiency constitutes a practical and scalable method by which to risk stratify individuals with early glucose dysregulation and inform targeted treatment to prevent T2D.
Collapse
Affiliation(s)
- Ahmed A. Metwally
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Dalia Perelman
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Heyjun Park
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Yue Wu
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Alokkumar Jha
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Seth Sharp
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Alessandra Celli
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Ekrem Ayhan
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Fahim Abbasi
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Anna L Gloyn
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
- Stanford Diabetes Research Centre, Stanford University, Stanford, CA 94305, USA
| | - Tracey McLaughlin
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
- These authors contributed equally
| | - Michael Snyder
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- These authors contributed equally
| |
Collapse
|
26
|
Fellinger E, Brandt T, Creutzburg J, Rommerskirchen T, Schmidt A. Analytical Performance of the FreeStyle Libre 2 Glucose Sensor in Healthy Male Adults. SENSORS (BASEL, SWITZERLAND) 2024; 24:5769. [PMID: 39275680 PMCID: PMC11397946 DOI: 10.3390/s24175769] [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: 08/05/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/16/2024]
Abstract
Continuous Glucose Monitoring (CGM) not only can be used for glycemic control in chronic diseases (e.g., diabetes), but is increasingly being utilized by individuals and athletes to monitor fluctuations in training and everyday life. However, it is not clear how accurately CGM reflects plasma glucose concentration in a healthy population in the absence of chronic diseases. In an oral glucose tolerance test (OGTT) with forty-four healthy male subjects (25.5 ± 4.5 years), the interstitial fluid glucose (ISFG) concentration obtained by a CGM sensor was compared against finger-prick capillary plasma glucose (CPG) concentration at fasting baseline (T0) and 30 (T30), 60 (T60), 90 (T90), and 120 (T120) min post OGTT to investigate differences in measurement accuracy. The overall mean absolute relative difference (MARD) was 12.9% (95%-CI: 11.8-14.0%). Approximately 100% of the ISFG values were within zones A and B in the Consensus Error Grid, indicating clinical accuracy. A paired t-test revealed statistically significant differences between CPG and ISFG at all time points (T0: 97.3 mg/dL vs. 89.7 mg/dL, T30: 159.9 mg/dL vs. 144.3 mg/dL, T60: 134.8 mg/dL vs. 126.2 mg/dL, T90: 113.7 mg/dL vs. 99.3 mg/dL, and T120: 91.8 mg/dL vs. 82.6 mg/dL; p < 0.001) with medium to large effect sizes (d = 0.57-1.02) and with ISFG systematically under-reporting the reference system CPG. CGM sensors provide a convenient and reliable method for monitoring blood glucose in the everyday lives of healthy adults. Nonetheless, their use in clinical settings wherein implications are drawn from CGM readings should be handled carefully.
Collapse
Affiliation(s)
- Eva Fellinger
- NextGenerationEU, dtec.bw Project Smart Health Lab, University of the Bundeswehr Munich, 85579 Neubiberg, Germany
| | - Tom Brandt
- Institute of Sport Sciences, University of the Bundeswehr Munich, 85579 Neubiberg, Germany
| | - Justin Creutzburg
- Institute of Sport Sciences, University of the Bundeswehr Munich, 85579 Neubiberg, Germany
| | - Tessa Rommerskirchen
- Institute of Sport Sciences, University of the Bundeswehr Munich, 85579 Neubiberg, Germany
| | - Annette Schmidt
- NextGenerationEU, dtec.bw Project Smart Health Lab, University of the Bundeswehr Munich, 85579 Neubiberg, Germany
- Institute of Sport Sciences, University of the Bundeswehr Munich, 85579 Neubiberg, Germany
- Research Center Smart Digital Health, University of the Bundeswehr Munich, 85579 Neubiberg, Germany
| |
Collapse
|
27
|
Wrench E, Subar DA, Bampouras TM, Lauder RM, Gaffney CJ. Myths and methodologies: Assessing glycaemic control and associated regulatory mechanisms in human physiology research. Exp Physiol 2024; 109:1461-1477. [PMID: 39014995 PMCID: PMC11363129 DOI: 10.1113/ep091433] [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: 12/15/2023] [Accepted: 06/17/2024] [Indexed: 07/18/2024]
Abstract
Accurate measurements of glycaemic control and the underpinning regulatory mechanisms are vital in human physiology research. Glycaemic control is the maintenance of blood glucose concentrations within optimal levels and is governed by physiological variables including insulin sensitivity, glucose tolerance and β-cell function. These can be measured with a plethora of methods, all with their own benefits and limitations. Deciding on the best method to use is challenging and depends on the specific research question(s). This review therefore discusses the theory and procedure, validity and reliability and any special considerations of a range common methods used to measure glycaemic control, insulin sensitivity, glucose tolerance and β-cell function. Methods reviewed include glycosylated haemoglobin, continuous glucose monitors, the oral glucose tolerance test, mixed meal tolerance test, hyperinsulinaemic euglycaemic clamp, hyperglycaemic clamp, intravenous glucose tolerance test and indices derived from both fasting concentrations and the oral glucose tolerance test. This review aims to help direct understanding, assessment and decisions regarding which method to use based on specific physiology-related research questions.
Collapse
Affiliation(s)
- Elizabeth Wrench
- Lancaster Medical School, Health Innovation One, Sir John Fisher DriveLancaster UniversityLancasterUK
| | - Daren A. Subar
- Royal Blackburn HospitalEast Lancashire Hospitals NHS TrustBlackburnUK
| | | | - Robert M. Lauder
- Lancaster Medical School, Health Innovation One, Sir John Fisher DriveLancaster UniversityLancasterUK
| | - Christopher J. Gaffney
- Lancaster Medical School, Health Innovation One, Sir John Fisher DriveLancaster UniversityLancasterUK
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Ham JY, Jang YK, Jeon BY, Shon YH. Magnesium from Deep Seawater as a Potentially Effective Natural Product against Insulin Resistance: A Randomized Trial. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1265. [PMID: 39202546 PMCID: PMC11355969 DOI: 10.3390/medicina60081265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 09/03/2024]
Abstract
Background and Objectives: Deep seawater has been shown to restore pancreatic function in obese diabetic mice and considerably improve the homeostatic model assessment for insulin resistance, total cholesterol, and low-density lipoprotein cholesterol concentrations in patients with impaired fasting glucose or glucose tolerance. In this study, the effect of 12-week daily consumption of magnesium (Mg2+)-containing deep seawater mineral extracts on blood glucose concentration and insulin metabolism-associated indicators was investigated in patients with impaired glucose tolerance. Materials and methods: In this 12-week randomized, double-blind trial, patients (n = 37) with impaired glucose tolerance consumed deep seawater mineral extracts. Changes in blood glucose concentration and related indicators were compared between the treatment group and placebo group (n = 38). Results: The fasting insulin, C-peptide, homeostatic model assessment for insulin resistance, quantitative insulin sensitivity check index, homeostatic model assessment of beta-cell function, and Stumvoll insulin sensitivity index values in the deep seawater mineral extract group showed improvements compared with the placebo group. However, no significant differences between groups were observed in fasting blood glucose, postprandial blood glucose, glycated hemoglobin, or incremental area under the curve values. Conclusions: Oral supplementation with deep seawater mineral extracts enriched in Mg2+ markedly improves insulin sensitivity in patients with pre-diabetes. This study illustrates the potential clinical application of natural Mg2+ from deep seawater to alleviate insulin resistance in patients with pre-diabetes. Trial registration: This trial was retrospectively registered with Clinical Research information Service (CRIS), No. KCT0008695, on 8 August 2023.
Collapse
Affiliation(s)
- Ji Yeon Ham
- Department of Laboratory Medicine, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, 807 Hogukro Buk-gu, Daegu 41404, Republic of Korea
| | - You Kyung Jang
- QBM Research Institute, QBM Co., Ltd., 7-25 Gangnam-daero 27-gil, Seocho-gu, Seoul 06752, Republic of Korea
| | - Byong Yeob Jeon
- QBM Research Institute, QBM Co., Ltd., 7-25 Gangnam-daero 27-gil, Seocho-gu, Seoul 06752, Republic of Korea
| | - Yun Hee Shon
- Bio-Medical Research Institute, Kyungpook National University Hospital, 135 Dongdukro Jung-gu, Daegu 41940, Republic of Korea
| |
Collapse
|
31
|
Cui R, Wei Y, Liu J, Wang G. Hepatic insulin resistance affects the efficacy of metformin in patients with newly diagnosed type 2 diabetes: A Sub-Analysis of the MARCH trial. Diabetes Res Clin Pract 2024; 214:111788. [PMID: 39032659 DOI: 10.1016/j.diabres.2024.111788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
AIM Our study aimed to analyze how hepatic insulin resistance (IR) influences the efficacy of 48 weeks of metformin treatment in newly diagnosed type 2 diabetes patients. METHODS We chose 291 participants who were allocated to a 48-week metformin treatment in the "Metformin and Acarbose in Chinese as initial Hypoglycemic treatment" (MARCH) trial and calculated their hepatic insulin resistance indexes (HIRI). We equally grouped the subjects into tertiles: low, medium, and high HIRI groups based on baseline HIRI; Low, medium, and high ΔHIRI groups based on the decreasing extent of HIRI after a 48-week metformin treatment. RESULTS Multiple linear regression showed that baseline HIRI was positively associated with the rising degree of Matsuda index and the falling range of fasting blood glucose, fasting insulin, homeostasis model assessment of insulin resistance (HOMA-IR), and HIRI. Furthermore, baseline fasting insulin, homeostatic model assessment of β cell function (HOMA-β), HOMA-IR, and HIRI were positively associated with the decreasing extent of HIRI, while baseline Matsuda index had a negative association with the falling extent of HIRI. CONCLUSIONS Patients with higher levels of hepatic IR obtained better curative effects from metformin in terms of glycemic control, insulin saving, insulin sensitivity enhancement, and IR improvement. Higher fasting blood glucose, fasting insulin, HOMA-β, IR, and lower Matsuda index were indicators of better hepatic IR improvement.
Collapse
Affiliation(s)
- Ruixiang Cui
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing 100020, China
| | - Ying Wei
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing 100020, China
| | - Jia Liu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing 100020, China.
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing 100020, China.
| |
Collapse
|
32
|
Tremblay EJ, Peyrel P, Karelis AD, Rabasa-Lhoret R, Tchernof A, Joanisse DR, Mauriège P. Resistance training and cardiometabolic risk in women with metabolically healthy and unhealthy obesity. Appl Physiol Nutr Metab 2024; 49:1068-1082. [PMID: 38648673 DOI: 10.1139/apnm-2023-0279] [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] [Indexed: 04/25/2024]
Abstract
Despite some reported benefits, there is a low quality of evidence for resistance training (RT) improving metabolic health of individuals with overweight or obesity. We evaluated the impact of RT on body composition, cardiorespiratory fitness (CRF) and physical performance, lipid-lipoprotein profile, inflammation, and glucose-insulin homeostasis in 51 postmenopausal women versus 29 controls matched for age, obesity, and physical activity. Exercised women were further subdivided for comparison of RT effects into those presenting metabolically healthy obesity (MHO) and those with metabolically unhealthy obesity (MUHO) classified according to Karelis and Rabasa-Lhoret or an approach based on adipose tissue secretory dysfunction using the plasma adiponectin(A)/leptin (L) ratio. Participants followed a 4-month weekly RT program targeting major muscle groups (3 × 10 repetitions at 80% one repetition maximum (1-RM)). Percent fat marginally decreased and lean body mass increased (0.01 < p < 0.05) while CRF and muscular strength improved in all women, after RT (effect size (ES): 0.11-1.21 (trivial to large effects), p ˂ 0.01). Fasting plasma triacylglycerol and high-density lipoprotein-cholesterol levels slightly increased and decreased, respectively, in participants with MHO using the A/L ratio approach (ES: -0.47 to 1.07 (small to large effects), p ˂ 0.05). Circulating interleukin-6 soluble receptor decreased in both groups and soluble tumor necrosis factor receptor-1/soluble tumor necrosis factor receptor-2 in women with MUHO only, irrespective of definition (ES: -0.42 to -0.84 (small to large effects), p ˂ 0.05). Glucose-insulin homeostasis was unchanged regardless of group or definition. RT improved physical performance and body composition but had a lesser impact on cardiometabolic risk in women with obesity, irrespective of their metabolic phenotype.
Collapse
Affiliation(s)
- Eve-Julie Tremblay
- École de nutrition, Université Laval, Québec, QC, Canada
- Centre de recherche de l'institut de cardiologie et pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Paul Peyrel
- Centre de recherche de l'institut de cardiologie et pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Département de kinésiologie, Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Antony D Karelis
- Département des sciences de l'activité physique, Université du Québec à Montréal, Montréal, QC, Canada
| | - Rémi Rabasa-Lhoret
- Département de nutrition, Université de Montréal, Montréal, QC, Canada
- Institut de recherches cliniques de Montréal, Montréal, QC, Canada
| | - André Tchernof
- École de nutrition, Université Laval, Québec, QC, Canada
- Centre de recherche de l'institut de cardiologie et pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Denis R Joanisse
- Centre de recherche de l'institut de cardiologie et pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Département de kinésiologie, Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Pascale Mauriège
- Centre de recherche de l'institut de cardiologie et pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Département de kinésiologie, Faculté de médecine, Université Laval, Québec, QC, Canada
| |
Collapse
|
33
|
Ginoudis A, Ioannidou S, Tsakiroglou G, Kazeli K, Vagdatli E, Lymperaki E. Correlation of Albumin, Red Cell Distribution Width and Other Biochemical and Hematological Parameters with Glycated Hemoglobin in Diabetic, Prediabetic and Non-Diabetic Patients. Int J Mol Sci 2024; 25:8037. [PMID: 39125606 PMCID: PMC11311434 DOI: 10.3390/ijms25158037] [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: 07/03/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Diabetes mellitus is a chronic metabolic disease that affects more than 10.5% of the world's adult population. Biochemical and hematological parameters, such as albumin (ALB) and red cell distribution width (RDW), have been shown to be altered in diabetic patients. This study aimed to correlate hematological and biochemical parameters with glycated hemoglobin (HbA1c). A total of 777 adults (372 women and 405 men, aged 19-85 years) were divided into three groups: 218 participants with HbA1c < 5.7% (group A: non-diabetic), 226 with HbA1c ≥ 5.7% and <6.5% (group B: prediabetic) and 333 with HbA1c ≥ 6.5% (group C: diabetic). Biochemical and hematological parameters were compared among the three groups. An analysis of variance was performed to determine the correlations of the parameters among the groups. The ALB and sodium (Na) levels were significantly lower in group C than in groups A (ALB: 3.8 g/dL vs. 4.1 g/dL, p < 0.0001, Na: 138.4 mmol/L vs. 139.3 mmol/L, p < 0.001) and B (ALB: 3.8 g/dL vs. 4.0 g/dL, p < 0.0001, Na: 138.4 mmol/L vs. 139.6 mmol/L, p < 0.0001), whereas the RDW-standard deviation (RDW-SD) and urea were increased in group C as compared to group A (RDW: 45.8 vs. 43.9 fL, p < 0.0001, urea: 55.6 mg/dL vs. 38.5 mg/dL, p < 0.0001). The mean platelet volume (MPV) was increased in group C as compared to group A (9.3 fL vs. 9.1 fL, p < 0.05, respectively). Τhe increase in RDW-SD in group A as compared to B and C demonstrates the impact of hyperglycemia on red blood cells. Albumin and RDW might improve risk assessment for the development of diabetes. These results highlight the potential role of these parameters as an indication for prediabetes that would alert for measurement of HbA1c.
Collapse
Affiliation(s)
- Argyrios Ginoudis
- School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Stavroula Ioannidou
- Department of Biomedical Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (S.I.); (G.T.); (K.K.); (E.V.)
| | - Georgia Tsakiroglou
- Department of Biomedical Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (S.I.); (G.T.); (K.K.); (E.V.)
| | - Konstantina Kazeli
- Department of Biomedical Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (S.I.); (G.T.); (K.K.); (E.V.)
| | - Eleni Vagdatli
- Department of Biomedical Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (S.I.); (G.T.); (K.K.); (E.V.)
- Hippokration General Hospital of Thessaloniki, 54642 Thessaloniki, Greece
| | - Evgenia Lymperaki
- Department of Biomedical Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (S.I.); (G.T.); (K.K.); (E.V.)
| |
Collapse
|
34
|
Reik A, Schauberger G, Wiechert M, Hauner H, Holzapfel C. Association Between the Postprandial Response to an Oral Glucose Tolerance Test and Anthropometric Changes After an 8-Week Low-Calorie Formula Diet - Results From the Lifestyle Intervention (LION) Study. Mol Nutr Food Res 2024; 68:e2400106. [PMID: 38850172 DOI: 10.1002/mnfr.202400106] [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: 02/09/2024] [Revised: 05/07/2024] [Indexed: 06/10/2024]
Abstract
SCOPE Interindividual variations in postprandial metabolism and weight loss outcomes have been reported. The literature suggests links between postprandial metabolism and weight regulation. Therefore, the study aims to evaluate if postprandial glucose metabolism after a glucose load predicts anthropometric outcomes of a weight loss intervention. METHODS AND RESULTS Anthropometric data from adults with obesity (18-65 years, body mass index [BMI] 30.0-39.9 kg m-2) are collected pre- and post an 8-week formula-based weight loss intervention. An oral glucose tolerance test (OGTT) is performed at baseline, from which postprandial parameters are derived from glucose and insulin concentrations. Linear regression models explored associations between these parameters and anthropometric changes (∆) postintervention. A random forest model is applied to identify predictive parameters for anthropometric outcomes after intervention. Postprandial parameters after an OGTT of 158 participants (63.3% women, age 45 ± 12, BMI 34.9 ± 2.9 kg m-2) reveal nonsignificant associations with changes in anthropometric parameters after weight loss (p > 0.05). Baseline fat-free mass (FFM) and sex are primary predictors for ∆ FFM [kg]. CONCLUSION Postprandial glucose metabolism after a glucose load does not predict anthropometric outcomes after short-term weight loss via a formula-based low-calorie diet in adults with obesity.
Collapse
Affiliation(s)
- Anna Reik
- Institute for Nutritional Medicine, School of Medicine and Health, Technical University of Munich, 80992, Munich, Germany
| | - Gunther Schauberger
- Chair of Epidemiology, School of Medicine and Health, Technical University of Munich, 80992, Munich, Germany
| | - Meike Wiechert
- Institute for Nutritional Medicine, School of Medicine and Health, Technical University of Munich, 80992, Munich, Germany
| | - Hans Hauner
- Institute for Nutritional Medicine, School of Medicine and Health, Technical University of Munich, 80992, Munich, Germany
- Else Kroener-Fresenius-Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
| | - Christina Holzapfel
- Institute for Nutritional Medicine, School of Medicine and Health, Technical University of Munich, 80992, Munich, Germany
- Department of Nutritional, Food and Consumer Sciences, Fulda University of Applied Sciences, 36037, Fulda, Germany
| |
Collapse
|
35
|
Chan SY, Zhang H, Wong JT, Chang HF, Chen LW, Barton SJ, Nield H, El-Heis S, Kenealy T, Lavalle L, Ramos-Nieves JM, Godin JP, Silva-Zolezzi I, Cutfield WS, Godfrey KM. Higher early pregnancy plasma myo-inositol associates with increased postprandial glycaemia later in pregnancy: Secondary analyses of the NiPPeR randomized controlled trial. Diabetes Obes Metab 2024; 26:1658-1669. [PMID: 38312016 DOI: 10.1111/dom.15468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 02/06/2024]
Abstract
AIM Myo-inositol supplementation from ~13 weeks' gestation reportedly improves glycaemia regulation in metabolically at-risk women, with speculation that earlier supplementation might bring further improvement. However, the NiPPeR trial of a myo-inositol-containing supplement starting preconception did not lower gestational glycaemia in generally healthy women. We postulated that the earlier timing of supplementation influences the maternal metabolic adaptation for gestational glycaemia regulation. METHODS In total, 585 women were recruited from Singapore, UK and New Zealand for the NiPPeR study. We examined associations of plasma myo-inositol concentrations at 7 and 28 weeks' gestation with 28 weeks plasma glucose (PG; fasting, and 1 h and 2 h in 75 g oral glucose tolerance test) and insulin indices using linear regression adjusting for covariates. RESULTS Higher 7-week myo-inositol, but not 28-week myo-inositol, associated with higher 1 h PG [βadj (95% confidence intervals) 0.05 (0.01, 0.09) loge mmol/L per loge μmol/L, p = .022] and 2 h PG [0.08 (0.03, 0.12), p = .001]; equivalent to 0.39 mmol/L increase in 2 h PG for an average 7-week myo-inositol increase of 23.4 μmol/L with myo-inositol supplementation. Higher 7-week myo-inositol associated with a lower 28-week Stumvoll index (first phase), an approximation of insulin secretion [-0.08 (-0.15, -0.01), p = .020] but not with 28-week Matsuda insulin sensitivity index. However, the clinical significance of a 7-week myo-inositol-related increase in glycaemia was limited as there was no association with gestational diabetes risk, birthweight and cord C-peptide levels. In-silico modelling found higher 28-week myo-inositol was associated with lower gestational glycaemia in White, but not Asian, women after controlling for 7-week myo-inositol effects. CONCLUSION To our knowledge, our study provides the first evidence that increasing first trimester plasma myo-inositol may slightly exacerbate later pregnancy post-challenge glycaemia, indicating that the optimal timing for starting prenatal myo-inositol supplementation needs further investigation.
Collapse
Affiliation(s)
- Shiao-Yng Chan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology, National University Hospital, Singapore, Singapore
| | - Han Zhang
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jui-Tsung Wong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Hsin F Chang
- Department of Obstetrics and Gynaecology, National University Hospital, Singapore, Singapore
| | - Ling-Wei Chen
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Sheila J Barton
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Heidi Nield
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Sarah El-Heis
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Timothy Kenealy
- Liggins Institute and A Better Start - National Science Challenge, The University of Auckland, Auckland, New Zealand
| | - Luca Lavalle
- Nestlé Research, Société des Produits Nestlé SA, Lausanne, Switzerland
| | | | | | | | - Wayne S Cutfield
- Liggins Institute and A Better Start - National Science Challenge, The University of Auckland, Auckland, New Zealand
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
36
|
Lee K, Kuang A, Bain JR, Hayes MG, Muehlbauer MJ, Ilkayeva OR, Newgard CB, Powe CE, Hivert MF, Scholtens DM, Lowe WL. Metabolomic and genetic architecture of gestational diabetes subtypes. Diabetologia 2024; 67:895-907. [PMID: 38367033 DOI: 10.1007/s00125-024-06110-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 01/12/2024] [Indexed: 02/19/2024]
Abstract
AIMS/HYPOTHESIS Physiological gestational diabetes mellitus (GDM) subtypes that may confer different risks for adverse pregnancy outcomes have been defined. The aim of this study was to characterise the metabolome and genetic architecture of GDM subtypes to address the hypothesis that they differ between GDM subtypes. METHODS This was a cross-sectional study of participants in the Hyperglycemia and Adverse Pregnancy Outcome (HAPO) study who underwent an OGTT at approximately 28 weeks' gestation. GDM was defined retrospectively using International Association of Diabetes and Pregnancy Study Groups/WHO criteria, and classified as insulin-deficient GDM (insulin secretion <25th percentile with preserved insulin sensitivity) or insulin-resistant GDM (insulin sensitivity <25th percentile with preserved insulin secretion). Metabolomic analyses were performed on fasting and 1 h serum samples in 3463 individuals (576 with GDM). Genome-wide genotype data were obtained for 8067 individuals (1323 with GDM). RESULTS Regression analyses demonstrated striking differences between the metabolomes for insulin-deficient or insulin-resistant GDM compared to those with normal glucose tolerance. After adjustment for covariates, 33 fasting metabolites, including 22 medium- and long-chain acylcarnitines, were uniquely associated with insulin-deficient GDM; 23 metabolites, including the branched-chain amino acids and their metabolites, were uniquely associated with insulin-resistant GDM; two metabolites (glycerol and 2-hydroxybutyrate) were associated with the same direction of association with both subtypes. Subtype differences were also observed 1 h after a glucose load. In genome-wide association studies, variants within MTNR1B (rs10830963, p=3.43×10-18, OR 1.55) and GCKR (rs1260326, p=5.17×10-13, OR 1.43) were associated with GDM. Variants in GCKR (rs1260326, p=1.36×10-13, OR 1.60) and MTNR1B (rs10830963, p=1.22×10-9, OR 1.49) demonstrated genome-wide significant association with insulin-resistant GDM; there were no significant associations with insulin-deficient GDM. The lead SNP in GCKR, rs1260326, was associated with the levels of eight of the 25 fasting metabolites that were associated with insulin-resistant GDM and ten of 41 1 h metabolites that were associated with insulin-resistant GDM. CONCLUSIONS/INTERPRETATION This study demonstrates that physiological GDM subtypes differ in their metabolome and genetic architecture. These findings require replication in additional cohorts, but suggest that these differences may contribute to subtype-related adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Kristen Lee
- Department of Medicine, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Alan Kuang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - James R Bain
- Duke Molecular Physiology Institute, Durham, NC, USA
- Department of Medicine, Duke University School of Medicine, Duke University, Durham, NC, USA
| | - M Geoffrey Hayes
- Department of Medicine, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Olga R Ilkayeva
- Duke Molecular Physiology Institute, Durham, NC, USA
- Department of Medicine, Duke University School of Medicine, Duke University, Durham, NC, USA
| | - Christopher B Newgard
- Duke Molecular Physiology Institute, Durham, NC, USA
- Department of Medicine, Duke University School of Medicine, Duke University, Durham, NC, USA
| | - Camille E Powe
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Marie-France Hivert
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Denise M Scholtens
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - William L Lowe
- Department of Medicine, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
37
|
Stone SI, Balasubramanyam A, Posey JE. Atypical Diabetes: What Have We Learned and What Does the Future Hold? Diabetes Care 2024; 47:770-781. [PMID: 38329838 PMCID: PMC11043229 DOI: 10.2337/dci23-0038] [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: 11/21/2023] [Indexed: 02/10/2024]
Abstract
As our understanding of the pathophysiology of diabetes evolves, we increasingly recognize that many patients may have a form of diabetes that does not neatly fit with a diagnosis of either type 1 or type 2 diabetes. The discovery and description of these forms of "atypical diabetes" have led to major contributions to our collective understanding of the basic biology that drives insulin secretion, insulin resistance, and islet autoimmunity. These discoveries now pave the way to a better classification of diabetes based on distinct endotypes. In this review, we highlight the key biological and clinical insights that can be gained from studying known forms of atypical diabetes. Additionally, we provide a framework for identification of patients with atypical diabetes based on their clinical, metabolic, and molecular features. Helpful clinical and genetic resources for evaluating patients suspected of having atypical diabetes are provided. Therefore, appreciating the various endotypes associated with atypical diabetes will enhance diagnostic accuracy and facilitate targeted treatment decisions.
Collapse
Affiliation(s)
- Stephen I. Stone
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Ashok Balasubramanyam
- Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX
| | - Jennifer E. Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| |
Collapse
|
38
|
Knudsen LL, Knorr S, Prange SK, Wolff C, Nørgaard H, Torp AM, Madsen LR, Mortensen L, Thomsen HH, Sørensen LP, Ovesen PG, Fuglsang J, Kampmann U. Clinical and Metabolic Characterization of Women With Gestational Diabetes Mellitus Within the First Year Postpartum. J Endocr Soc 2024; 8:bvae044. [PMID: 38601785 PMCID: PMC11004785 DOI: 10.1210/jendso/bvae044] [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: 09/06/2023] [Indexed: 04/12/2024] Open
Abstract
Context Women with gestational diabetes mellitus (GDM) have an increased risk of long-term complications, including impaired glucose metabolism, type 2 diabetes (T2DM), cardiovascular disease, and obesity. In current clinical practice, a 1 size fits all approach to GDM is applied, although heterogeneity among women with GDM has been recognized. Objective To give the most adequate preventive care and postpartum (PP) guidance, we aimed to make a metabolic characterization and identify subgroups of women with previous GDM within the first year PP. Methods In this prospective cohort study, we collected data in gestational week 34-38, at 3 months, and 1 year PP on women with GDM who participated in a PP follow-up program in Central Region Denmark from April 2019 to December 2022. Results In total, 1270 women were included in the program in late pregnancy. Of the 768 women participating in either the oral glucose tolerance test 3 months PP (n = 545) or the 1-year follow-up (n = 493) or both (n = 261), 608 (79.2%) were normoglycemic, 137 (17.8%) had prediabetes, 20 (2.6%) had T2DM, and 3 (.4%) had developed T1DM. More than 40% of the women gained weight in the first year PP compared with their pregestational weight. Conclusion Our study shows that 20.8% of women with GDM who volunteered to participate in a clinical follow-up program developed prediabetes or diabetes (T1DM and T2DM) within the first year PP. The GDM diagnosis encompasses a heterogenetic group of women and a deeper characterization may provide an opportunity for a more personalized risk assessment to prevent the progression to T2DM.
Collapse
Affiliation(s)
| | - Sine Knorr
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Charlotte Wolff
- Department of Gynecology and Obstetrics, Aarhus University Hospital, Aarhus, Denmark
| | - Helle Nørgaard
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Anne Mette Torp
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Lene Ring Madsen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Internal Medicine, Gødstrup Hospital, Herning, Denmark
- Danish Diabetes Academy, Odense University Hospital, Odense, Denmark
| | - Lene Mortensen
- Department of Internal Medicine, Horsens Regional Hospital, Horsens, Denmark
| | - Henrik Holm Thomsen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Internal Medicine, Viborg Regional Hospital, Viborg, Denmark
| | - Lars Peter Sørensen
- Department of Internal Medicine, Randers Regional Hospital, Randers, Denmark
| | - Per Glud Ovesen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Gynecology and Obstetrics, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Fuglsang
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Gynecology and Obstetrics, Aarhus University Hospital, Aarhus, Denmark
| | - Ulla Kampmann
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
39
|
Ha J, Chung ST, Springer M, Kim JY, Chen P, Chhabra A, Cree MG, Diniz Behn C, Sumner AE, Arslanian SA, Sherman AS. Estimating insulin sensitivity and β-cell function from the oral glucose tolerance test: validation of a new insulin sensitivity and secretion (ISS) model. Am J Physiol Endocrinol Metab 2024; 326:E454-E471. [PMID: 38054972 PMCID: PMC11639675 DOI: 10.1152/ajpendo.00189.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 11/27/2023] [Accepted: 11/27/2023] [Indexed: 12/07/2023]
Abstract
Efficient and accurate methods to estimate insulin sensitivity (SI) and β-cell function (BCF) are of great importance for studying the pathogenesis and treatment effectiveness of type 2 diabetes (T2D). Existing methods range in sensitivity, input data, and technical requirements. Oral glucose tolerance tests (OGTTs) are preferred because they are simpler and more physiological than intravenous methods. However, current analytical methods for OGTT-derived SI and BCF also range in complexity; the oral minimal models require mathematical expertise for deconvolution and fitting differential equations, and simple algebraic surrogate indices (e.g., Matsuda index, insulinogenic index) may produce unphysiological values. We developed a new insulin secretion and sensitivity (ISS) model for clinical research that provides precise and accurate estimates of SI and BCF from a standard OGTT, focusing on effectiveness, ease of implementation, and pragmatism. This model was developed by fitting a pair of differential equations to glucose and insulin without need of deconvolution or C-peptide data. This model is derived from a published model for longitudinal simulation of T2D progression that represents glucose-insulin homeostasis, including postchallenge suppression of hepatic glucose production and first- and second-phase insulin secretion. The ISS model was evaluated in three diverse cohorts across the lifespan. The new model had a strong correlation with gold-standard estimates from intravenous glucose tolerance tests and insulin clamps. The ISS model has broad applicability among diverse populations because it balances performance, fidelity, and complexity to provide a reliable phenotype of T2D risk.NEW & NOTEWORTHY The pathogenesis of type 2 diabetes (T2D) is determined by a balance between insulin sensitivity (SI) and β-cell function (BCF), which can be determined by gold standard direct measurements or estimated by fitting differential equation models to oral glucose tolerance tests (OGTTs). We propose and validate a new differential equation model that is simpler to use than current models and requires less data while maintaining good correlation and agreement with gold standards. Matlab and Python code is freely available.
Collapse
Affiliation(s)
- Joon Ha
- Department of Mathematics, Howard University, Washington, District of Columbia, United States
| | - Stephanie T Chung
- Section on Pediatric Diabetes, Obesity, and Metabolism, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Max Springer
- Department of Mathematics, University of Maryland, College Park, Maryland, United States
| | - Joon Young Kim
- Department of Exercise Science, David B. Falk College of Sport and Human Dynamics, Syracuse University, Syracuse, New York, United States
| | | | - Aaryan Chhabra
- Department of Biology, Indian Institute of Science Education and Research, Pune, India
| | - Melanie G Cree
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Cecilia Diniz Behn
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Applied Mathematics and Statistics, Colorado School of Mines, Golden, Colorado, United States
| | - Anne E Sumner
- Intramural Research Program, National Institute on Minority Health and Health Disparities (NIMHD), National Institutes of Health, Bethesda, Maryland, United States
- Section on Ethnicity and Health, Diabetes Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, Maryland, United States
- Hypertension in Africa Research Team, North-West University, Potchefstroom, South Africa
| | - Silva A Arslanian
- Division of Pediatric Endocrinology, Metabolism and Diabetes Mellitus, Center for Pediatric Research in Obesity and Metabolism, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Arthur S Sherman
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
40
|
Yuan L, Zhao J, Liu Y, Zhao J, Olnood CG, Xu YJ, Liu Y. Multiomics analysis revealed the mechanism of the anti-diabetic effect of Salecan. Carbohydr Polym 2024; 327:121694. [PMID: 38171651 DOI: 10.1016/j.carbpol.2023.121694] [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: 09/13/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024]
Abstract
Salecan, a natural β-glucan compromising nine residues connected by β-(1 → 3)/α-(1 → 3) glycosidic bonds, is one of the newly approved food ingredients. Salecan has multiple health-improving effects, yet its mechanism against Type 2 diabetes mellitus (T2DM) remains poorly understood. In this study, the hypoglycemic effect and underlying mechanism of Salecan intervention on STZ-induced diabetic model mice were investigated. After 8 weeks of gavage, Salecan attenuated insulin resistance and repaired pancreatic β cells in a dose-dependent manner. In addition, Salecan supplement remodel the structure of the gut microbiota and altered the level of intestinal metabolites. Serum metabolites, especially unsaturated fatty acids, were also affected significantly. In addition, tight junction proteins in the colon and autophagy-related proteins in the pancreas were upregulated. Multiomics analysis indicated that Lactobacillus johnsonii, Muribaculaceae, and Lachnoclostridium were highly associated with fatty acid esters of hydroxy fatty acids (FAHFA) levels in the colon, accordingly enhancing arachidonic acid and linoleic acid in serum, and promoting GLP-1 release in the intestine and insulin secretion in the pancreas, thus relieving insulin resistance and exhibiting hypoglycemic effects. These findings provide a novel understanding of the anti-diabetic effect of Salecan in mice from a molecular perspective, paving the way for the wide use of Salecan.
Collapse
Affiliation(s)
- Liyang Yuan
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, China
| | - Juan Zhao
- Sichuan Synlight Biotech Ltd, 88 Keyuan South Road, Chengdu 610000, Sichuan, China
| | - Yanjun Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, China
| | - Jialiang Zhao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, China
| | - Chen Guang Olnood
- Sichuan Synlight Biotech Ltd, 88 Keyuan South Road, Chengdu 610000, Sichuan, China
| | - Yong-Jiang Xu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, China.
| | - Yuanfa Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, China.
| |
Collapse
|
41
|
Zhang Z, Li M, Ji G, Zhang L. Causal relationship between sleep apnea and non-alcoholic fatty liver disease: A Mendelian randomization study. Eur J Clin Invest 2024; 54:e14116. [PMID: 37916519 DOI: 10.1111/eci.14116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Observational studies indicate that sleep apnea is associated with non-alcoholic fatty liver disease (NAFLD) and its related metabolic features, independent of confounding factors including obesity. However, the causal relationships remain to be determined. METHODS Univariable and multivariable Mendelian randomization (MR) analyses were performed to investigate the causal relationship between sleep apnea and NAFLD, along with its typical features including liver function, glycemic traits and lipid profiles. Summary-level data for sleep apnea were obtained from the Finngen consortium (33,423 cases and 307,648 controls). Summary-level data for NAFLD were available from a GWAS meta-analysis (8434 cases and 770,180 controls), and data for 12 NAFLD-related features from corresponding published GWASs. The inverse variance weighted (IVW) analysis was employed as the primary statistical method. Bidirectional MR and CAUSE analysis were conducted to avoid reverse causality and false positive findings. RESULTS In univariable MR analyses, we found evidence to support a causal effect of genetically predicted sleep apnea on NAFLD (OR = 1.50, 95% CI = 1.18-1.91) and HDL-C (β = -0.045, 95% CI = -0.090 to -0.001). In reverse MR, genetically predicted serum TG was associated with an increased risk of sleep apnea (OR = 1.07, 95% CI = 1.02-1.12), while genetically predicted HDL-C was associated with a decreased risk of sleep apnea (OR = 0.93, 95% CI = 0.89-0.98). After adjusting body mass index or educational attainment, none of these causal associations were retained. However, CAUSE method and MR analyses focusing on lipoprotein subfractions supported a causal effect of sleep apnea on HDL-C and HDL subfractions. CONCLUSION This MR study indicated that sleep apnea has no direct causal association with NAFLD, elevated liver enzymes and insulin resistance. Our results showed suggestive inverse associations of genetically predicted sleep apnea on HDL-C and HDL subfractions, indicating that both HDL-C levels and HDL function may be causally implicated in sleep apnea.
Collapse
Affiliation(s)
- Ziqi Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meng Li
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
42
|
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
|
43
|
Guo M, Li M, Cui F, Wang H, Ding X, Gao W, Fang X, Chen L, Niu P, Ma J. Mediation effect of serum zinc on insulin secretion inhibited by methyl tert-butyl ether in gas station workers. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:8952-8962. [PMID: 38183540 DOI: 10.1007/s11356-023-31772-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 12/26/2023] [Indexed: 01/08/2024]
Abstract
Methyl tert-butyl ether (MTBE), a type of gasoline additive, has been found to affect insulin function and glucose homeostasis in animal experiments, but there is still no epidemiological evidence. Zinc (Zn) is a key regulatory element of insulin secretion and function, and Zn homeostasis can be disrupted by MTBE exposure through inducing oxidative stress. Therefore, we suspected that Zn might be involved and play an important role in the process of insulin secretion inhibited by MTBE exposure. In this study, we recruited 201 male subjects including occupational and non-occupational MTBE exposure from Anhui Province, China in 2019. Serum insulin and functional analog fibroblast growth factor 1 (FGF1) and blood MTBE were detected by Elisa and headspace solid-phase microextraction and gas chromatography-high-resolution mass spectrometry. According to MTBE internal exposure level, the workers were divided into low- and high-exposed groups and found that the serum insulin level in the high-exposed group was significantly lower than that in the low-exposed group (p = 0.003) while fasting plasma glucose (FPG) level increased obviously in the high-exposed group compared to the low-exposed group (p = 0.001). Further analysis showed that MTBE exposure level was positively correlated with FPG level, but negatively correlated with serum insulin level, which suggested that the FPG level increase might be related to the decrease of serum insulin level induced by MTBE exposure. The results of further mediation effect analysis showed that changes in serum zinc levels played a major intermediary role in the process of insulin secretion inhibition and blood glucose elevation caused by MTBE exposure. In addition, a significant negative correlation was found between MTBE exposure and serum Zn level, which might play a strong mediating effect on the inhibition of insulin secretion induced by MTBE exposure. In conclusion, our study provided evidence that MTBE could inhibit insulin secretion and interfere with Zn metabolism in gas station workers for the first time, and found that Zn might play an important mediation effect during the process of inhibiting insulin secretion and interfering with glucose metabolism induced by MTBE exposure.
Collapse
Affiliation(s)
- Mingxiao Guo
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Mengdi Li
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Fengtao Cui
- Occupational Disease Prevention and Control Hospital of Huaibei Mining Co., Ltd., Huaibei, 235000, Anhui Province, China
| | - Hanyun Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Xinping Ding
- Occupational Disease Prevention and Control Hospital of Huaibei Mining Co., Ltd., Huaibei, 235000, Anhui Province, China
| | - Wei Gao
- Occupational Disease Prevention and Control Hospital of Huaibei Mining Co., Ltd., Huaibei, 235000, Anhui Province, China
| | - Xingqiang Fang
- Occupational Disease Prevention and Control Hospital of Huaibei Mining Co., Ltd., Huaibei, 235000, Anhui Province, China
| | - Li Chen
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Piye Niu
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Junxiang Ma
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China.
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
44
|
Chen C, Gao K, Chen Z, Zhang Q, Ke X, Mao B, Fan Q, Li Y, Chen S. The supplementation of the multi-strain probiotics WHHPRO™ alleviates high-fat diet-induced metabolic symptoms in rats via gut-liver axis. Front Nutr 2024; 10:1324691. [PMID: 38274203 PMCID: PMC10808617 DOI: 10.3389/fnut.2023.1324691] [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: 10/20/2023] [Accepted: 12/27/2023] [Indexed: 01/27/2024] Open
Abstract
Metabolic syndrome (MS) has emerged as one of the major global health concerns, accompanied by a series of related complications, such as obesity and type-2 diabetes. The gut-liver axis (GLA) is a bidirectional communication between the gut and the liver. The GLA alterations have been revealed to be closely associated with the development of MS. Probiotics within Lactobacillus and Bifidobacterium confer beneficial effects on improving MS symptoms. WHHPRO™ is a mixture of four probiotic strains, with potential MS-improving abilities. This study aimed to investigate the effects of WHHPRO™ on MS symptoms using a high-fat diet (HFD) rat model. Oral administration of WHHPRO™ for 12 weeks improved glucose tolerance, blood lipid, body weight, and liver index in HFD rats. WHHPRO™ shaped the gut microbiome composition by increasing the abundance of Lactobacillus and Akkermansia and normalized the reduced SCFA levels in HFD rats. Besides, WHHPRO™ modulated the fecal bile acids (BAs) profile, with decreased levels of T-b-MCA and 12-KDCA and increased levels of LCA and ILCA. Meanwhile, WHHPRO™ increased total unconjugated BAs in feces and liver and reduced the accumulation of total hepatic BA pool size in HFD rats. Moreover, WHHPRO™ reversed the expression of genes associated with impaired BA metabolism signaling in the ileum and liver. Our findings suggest that WHHPRO™ exerted beneficial effects on improving MS symptoms, involving the modulation of the gut microbiome composition, SCFAs, and the FXR-FGF15 signaling along the GLA. Supplementation of WHHPRO™ may serve as a novel strategy for improving MS symptoms.
Collapse
Affiliation(s)
- Cailing Chen
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- Research and Development Department, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Hangzhou Wahaha Technology Co., Ltd., Hangzhou, China
| | - Kan Gao
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- Research and Development Department, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Hangzhou Wahaha Technology Co., Ltd., Hangzhou, China
| | - Zuoguo Chen
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- Research and Development Department, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Hangzhou Wahaha Technology Co., Ltd., Hangzhou, China
| | - Qiwen Zhang
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- Research and Development Department, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Hangzhou Wahaha Technology Co., Ltd., Hangzhou, China
| | - Xueqin Ke
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- Research and Development Department, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Hangzhou Wahaha Technology Co., Ltd., Hangzhou, China
| | - Bingyong Mao
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qiuling Fan
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- Research and Development Department, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Hangzhou Wahaha Technology Co., Ltd., Hangzhou, China
| | - Yanjun Li
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- Research and Development Department, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Hangzhou Wahaha Technology Co., Ltd., Hangzhou, China
| | - Su Chen
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- Research and Development Department, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Hangzhou Wahaha Technology Co., Ltd., Hangzhou, China
| |
Collapse
|
45
|
Hieronimus B, Medici V, Lee V, Nunez MV, Sigala DM, Bremer AA, Cox CL, Keim NL, Schwarz JM, Pacini G, Tura A, Havel PJ, Stanhope KL. Effects of Consuming Beverages Sweetened with Fructose, Glucose, High-Fructose Corn Syrup, Sucrose, or Aspartame on OGTT-Derived Indices of Insulin Sensitivity in Young Adults. Nutrients 2024; 16:151. [PMID: 38201980 PMCID: PMC10780640 DOI: 10.3390/nu16010151] [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: 11/11/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
(1) Background: Clinical results on the effects of excess sugar consumption on insulin sensitivity are conflicting, possibly due to differences in sugar type and the insulin sensitivity index (ISI) assessed. Therefore, we compared the effects of consuming four different sugars on insulin sensitivity indices derived from oral glucose tolerance tests (OGTT). (2) Methods: Young adults consumed fructose-, glucose-, high-fructose corn syrup (HFCS)-, sucrose-, or aspartame-sweetened beverages (SB) for 2 weeks. Participants underwent OGTT before and at the end of the intervention. Fasting glucose and insulin, Homeostatic Model Assessment-Insulin Resistance (HOMA-IR), glucose and insulin area under the curve, Surrogate Hepatic Insulin Resistance Index, Matsuda ISI, Predicted M ISI, and Stumvoll Index were assessed. Outcomes were analyzed to determine: (1) effects of the five SB; (2) effects of the proportions of fructose and glucose in all SB. (3) Results: Fructose-SB and the fructose component in mixed sugars negatively affected outcomes that assess hepatic insulin sensitivity, while glucose did not. The effects of glucose-SB and the glucose component in mixed sugar on muscle insulin sensitivity were more negative than those of fructose. (4) Conclusion: the effects of consuming sugar-SB on insulin sensitivity varied depending on type of sugar and ISI index because outcomes assessing hepatic insulin sensitivity were negatively affected by fructose, and outcomes assessing muscle insulin sensitivity were more negatively affected by glucose.
Collapse
Affiliation(s)
- Bettina Hieronimus
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (B.H.)
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany
| | - Valentina Medici
- Division of Gastroenterology and Hepatology, University of California, Davis, CA 95616, USA
| | - Vivien Lee
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (B.H.)
| | | | - Desiree M. Sigala
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (B.H.)
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany
| | - Andrew A. Bremer
- Department of Pediatrics, School of Medicine, University of California, Davis, CA 95616, USA
| | - Chad L. Cox
- Department of Chemistry and Department of Family and Consumer Sciences, California State University, Sacramento, CA 95819, USA
| | - Nancy L. Keim
- United States Department of Agriculture, Western Human Nutrition Research Center, Davis, CA 95819, USA
| | - Jean-Marc Schwarz
- Department of Basic Sciences, College of Osteopathic Medicine, Touro University California, Vallejo, CA 94592, USA
- Department of Medicine, Division of Endocrinology, Zuckerberg San Francisco General Hospital, University of California San Francisco, San Francisco, CA 94110, USA
| | - Giovanni Pacini
- Department of Medicine, Division of Endocrinology, Zuckerberg San Francisco General Hospital, University of California San Francisco, San Francisco, CA 94110, USA
- Consiglio Nazionale delle Ricerche, Institute of Neuroscience, I-35121 Padova, Italy
| | - Andrea Tura
- Department of Medicine, Division of Endocrinology, Zuckerberg San Francisco General Hospital, University of California San Francisco, San Francisco, CA 94110, USA
- Consiglio Nazionale delle Ricerche, Institute of Neuroscience, I-35121 Padova, Italy
| | - Peter J. Havel
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (B.H.)
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany
| | - Kimber L. Stanhope
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (B.H.)
| |
Collapse
|
46
|
Cohrs CM, Chen C, Atkinson MA, Drotar DM, Speier S. Bridging the Gap: Pancreas Tissue Slices From Organ and Tissue Donors for the Study of Diabetes Pathogenesis. Diabetes 2024; 73:11-22. [PMID: 38117999 PMCID: PMC10784654 DOI: 10.2337/dbi20-0018] [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: 05/04/2021] [Accepted: 10/14/2023] [Indexed: 12/22/2023]
Abstract
Over the last two decades, increased availability of human pancreatic tissues has allowed for major expansions in our understanding of islet biology in health and disease. Indeed, studies of fixed and frozen pancreatic tissues, as well as efforts using viable isolated islets obtained from organ donors, have provided significant insights toward our understanding of diabetes. However, the procedures associated with islet isolation result in distressed cells that have been removed from any surrounding influence. The pancreas tissue slice technology was developed as an in situ approach to overcome certain limitations associated with studies on isolated islets or fixed tissue. In this Perspective, we discuss the value of this novel platform and review how pancreas tissue slices, within a short time, have been integrated in numerous studies of rodent and human islet research. We show that pancreas tissue slices allow for investigations in a less perturbed organ tissue environment, ranging from cellular processes, over peri-islet modulations, to tissue interactions. Finally, we discuss the considerations and limitations of this technology in its future applications. We believe the pancreas tissue slices will help bridge the gap between studies on isolated islets and cells to the systemic conditions by providing new insight into physiological and pathophysiological processes at the organ level. ARTICLE HIGHLIGHTS Human pancreas tissue slices represent a novel platform to study human islet biology in close to physiological conditions. Complementary to established technologies, such as isolated islets, single cells, and histological sections, pancreas tissue slices help bridge our understanding of islet physiology and pathophysiology from single cell to intact organ. Diverse sources of viable human pancreas tissue, each with distinct characteristics to be considered, are available to use in tissue slices for the study of diabetes pathogenesis.
Collapse
Affiliation(s)
- Christian M. Cohrs
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Munich at the University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Chunguang Chen
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Munich at the University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Mark A. Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL
| | - Denise M. Drotar
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL
| | - Stephan Speier
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Munich at the University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
47
|
Jiménez-Sánchez C, Sinturel F, Mezza T, Loizides-Mangold U, Montoya JP, Li L, Di Giuseppe G, Quero G, Guessous I, Jornayvaz F, Schrauwen P, Stenvers DJ, Alfieri S, Giaccari A, Berishvili E, Compagnon P, Bosco D, Riezman H, Dibner C, Maechler P. Lysophosphatidylinositols Are Upregulated After Human β-Cell Loss and Potentiate Insulin Release. Diabetes 2024; 73:93-107. [PMID: 37862465 DOI: 10.2337/db23-0205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
In this study, we identified new lipid species associated with the loss of pancreatic β-cells triggering diabetes. We performed lipidomics measurements on serum from prediabetic mice lacking β-cell prohibitin-2 (a model of monogenic diabetes) patients without previous history of diabetes but scheduled for pancreaticoduodenectomy resulting in the acute reduction of their β-cell mass (∼50%), and patients with type 2 diabetes (T2D). We found lysophosphatidylinositols (lysoPIs) were the main circulating lipid species altered in prediabetic mice. The changes were confirmed in the patients with acute reduction of their β-cell mass and in those with T2D. Increased lysoPIs significantly correlated with HbA1c (reflecting glycemic control), fasting glycemia, and disposition index, and did not correlate with insulin resistance or obesity in human patients with T2D. INS-1E β-cells as well as pancreatic islets isolated from nondiabetic mice and human donors exposed to exogenous lysoPIs showed potentiated glucose-stimulated and basal insulin secretion. Finally, addition of exogenous lysoPIs partially rescued impaired glucose-stimulated insulin secretion in islets from mice and humans in the diabetic state. Overall, lysoPIs appear to be lipid species upregulated in the prediabetic stage associated with the loss of β-cells and that support the secretory function of the remaining β-cells. ARTICLE HIGHLIGHTS Circulating lysophosphatidylinositols (lysoPIs) are increased in situations associated with β-cell loss in mice and humans such as (pre-)diabetes, and hemipancreatectomy. Pancreatic islets isolated from nondiabetic mice and human donors, as well as INS-1E β-cells, exposed to exogenous lysoPIs exhibited potentiated glucose-stimulated and basal insulin secretion. Addition of exogenous lysoPIs partially rescued impaired glucose-stimulated insulin secretion in islets from mice and humans in the diabetic state. LysoPIs appear as lipid species being upregulated already in the prediabetic stage associated with the loss of β-cells and supporting the function of the remaining β-cells.
Collapse
Affiliation(s)
- Cecilia Jiménez-Sánchez
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Flore Sinturel
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Teresa Mezza
- Pancreas Unit, Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli, Institute of Hospitalization and Scientific Care (IRCCS), Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ursula Loizides-Mangold
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Jonathan Paz Montoya
- Proteomics Core Facility, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Lingzi Li
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Gianfranco Di Giuseppe
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Giuseppe Quero
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
- Chirurgia Digestiva, Fondazione Policlinico Universitario Gemelli IRCSS Università Cattolica del Sacro Cuore, Rome, Italy
| | - Idris Guessous
- Department of Primary Care Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - François Jornayvaz
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Division of Endocrinology, Diabetes, Nutrition and Patient Education, Department of Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Dirk Jan Stenvers
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, the Netherlands
| | - Sergio Alfieri
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Chirurgia Digestiva, Fondazione Policlinico Universitario Gemelli IRCSS Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Giaccari
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Ekaterine Berishvili
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
- Cell isolation and Transplantation Center, Geneva University Hospitals, Geneva, Switzerland
| | - Philippe Compagnon
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
- Cell isolation and Transplantation Center, Geneva University Hospitals, Geneva, Switzerland
| | - Domenico Bosco
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
- Cell isolation and Transplantation Center, Geneva University Hospitals, Geneva, Switzerland
| | - Howard Riezman
- Department of Biochemistry, Faculty of Science, National Centre of Competence in Research Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Charna Dibner
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| |
Collapse
|
48
|
Daanje M, Siebelink E, Vrieling F, van den Belt M, van der Haar S, Gerdessen JC, Kersten S, Esser D, Afman LA. Are postprandial glucose responses sufficiently person-specific to use in personalized dietary advice? Design of the RepEAT study: a fully controlled dietary intervention to determine the variation in glucose responses. Front Nutr 2023; 10:1281978. [PMID: 38152465 PMCID: PMC10751339 DOI: 10.3389/fnut.2023.1281978] [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: 08/23/2023] [Accepted: 11/27/2023] [Indexed: 12/29/2023] Open
Abstract
Introduction An elevated postprandial glucose response is associated with an increased risk of cardiometabolic diseases. Existing research suggests large heterogeneity in the postprandial glucose responses to identical meals and food products between individuals, but the effect of other consumed meals during the day and the order of meals during the day on the heterogeneity in postprandial glucose responses still needs to be investigated. In addition, the robustness of the glucose responses to meals or foods is still unknown. Objectives The overall aim of the project is to assess whether the glucose response to a meal is sufficiently person-specific to use in personalized dietary advice. We aim to answer the question: "How replicable are glucose responses to meals within individuals and how consistent is the variation in glucose responses between individuals?" Methods The question will be assessed under standardized conditions of a 9-week fully controlled dietary intervention in which all meals are the same between individuals and consumed in a fixed order at a fixed time. 63 apparently healthy men and women with a BMI of 25-40 kg/m2 and aged 45-75 years were enrolled in the RepEAT study (NCT05456815), of whom 53 participants completed the study. The RepEAT study comprised a fully controlled dietary intervention of nine weeks, consisting of three repetitive periods of three weeks. Within each three-week period, a variety of meals and food products were offered during breakfast, lunch, dinner and in between meal snacks. Throughout the dietary intervention, glucose was continuously monitored using Freestyle Libre Pro IQ monitors. Physical activity was monitored using the ActiGraph and ActivPAL. To measure the association between glucose responses and an individual's phenotype, various measurements were performed before the start of the dietary intervention including an oral glucose tolerance test, a high-fat mixed meal challenge, assessment of body fat distribution including liver fat (MRI/MRS), and cardiometabolic markers. Discussion The repetitive and fully controlled nature of the dietary study allows detailed assessment of the replicability of the glucose responses to meals and food products within individuals. Furthermore, the consistency of the variation between individuals independent of insulin resistance will be determined.
Collapse
Affiliation(s)
- Monique Daanje
- Division of Human Nutrition, Wageningen University & Research, Wageningen, Netherlands
| | - Els Siebelink
- Division of Human Nutrition, Wageningen University & Research, Wageningen, Netherlands
| | - Frank Vrieling
- Division of Human Nutrition, Wageningen University & Research, Wageningen, Netherlands
| | - Maartje van den Belt
- Food and Biobased Research, Wageningen University & Research, Wageningen, Netherlands
| | - Sandra van der Haar
- Food and Biobased Research, Wageningen University & Research, Wageningen, Netherlands
| | - Johanna C. Gerdessen
- Department of Social Sciences, Wageningen University & Research, Wageningen, Netherlands
| | - Sander Kersten
- Division of Human Nutrition, Wageningen University & Research, Wageningen, Netherlands
| | - Diederik Esser
- Food and Biobased Research, Wageningen University & Research, Wageningen, Netherlands
| | - Lydia A. Afman
- Division of Human Nutrition, Wageningen University & Research, Wageningen, Netherlands
| |
Collapse
|
49
|
Jones DL, Petry CJ, Burling K, Barker P, Turner EH, Kusinski LC, Meek CL. Pregnancy glucagon-like peptide 1 predicts insulin but not glucose concentrations. Acta Diabetol 2023; 60:1635-1642. [PMID: 37439859 PMCID: PMC10587288 DOI: 10.1007/s00592-023-02142-8] [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: 03/30/2023] [Accepted: 06/16/2023] [Indexed: 07/14/2023]
Abstract
AIMS Incretin hormones glucagon-like peptide 1 (GLP-1) and gastric inhibitory peptide (GIP) cause increased insulin secretion in non-pregnant adults, but their role in pregnancy, where there are additional metabolically-active hormones from the placenta, is less clear. The aim of the present study was to assess if fasting and post-load incretin concentrations were predictive of pregnancy insulin and glucose concentrations. METHODS Pregnant women (n = 394) with one or more risk factors for gestational diabetes were recruited at 28 weeks for a 75 g oral glucose tolerance test (OGTT). Glucose, insulin, GLP-1 and GIP were measured in the fasting state and 120 min after glucose ingestion. RESULTS Fasting plasma GLP-1 concentrations were associated with plasma insulin (standardised β' 0.393 (0.289-0.498), p = 1.3 × 10-12; n = 306), but not with glucose concentrations (p = 0.3). The association with insulin was still evident when adjusting for BMI (β' 0.271 (0.180-0.362), p = 1.1 × 10-8; n = 297). Likewise, at 120 min the OGTT GLP-1 concentrations were associated with plasma insulin concentrations (β' 0.216 (0.100-0.331), p = 2.7 × 10-4; n = 306) even after adjusting for BMI (β' 0.178 (0.061-0.294), p = 2.9 × 10-3; n = 296), but not with glucose (p = 0.9). GIP concentrations were not associated with insulin or glucose concentrations at either time point (all p > 0.2). In pregnancy plasma GLP-1, but not GIP, concentrations appear to be predictive of circulating insulin concentrations, independently of associations with BMIs. CONCLUSIONS These results suggest that the relationship between insulin and incretins is preserved in pregnancy, but that other factors, such as placental hormones or counter-regulatory hormones, may be more important determinants of glycaemia and gestational diabetes aetiology.
Collapse
Affiliation(s)
- Danielle L Jones
- Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Clive J Petry
- Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Keith Burling
- NIHR Biomedical Research Centre Core Biochemistry Assay Lab, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Peter Barker
- NIHR Biomedical Research Centre Core Biochemistry Assay Lab, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Elizabeth H Turner
- Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Laura C Kusinski
- Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Claire L Meek
- Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK.
- Department of Clinical Biochemistry, Addenbrooke's Hospital, Cambridge, UK.
- Wolfson Diabetes and Endocrinology Clinic, Cambridge University Hospitals, Addenbrooke's Hospital, Cambridge, UK.
| |
Collapse
|
50
|
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
|