1
|
George J, King D, Shojaee-Moradie F, Whyte M, Scott M, Rockall T. Effect of prehabilitation on cardiopulmonary fitness and insulin sensitivity before pancreatic surgery. Clin Nutr ESPEN 2025; 68:274-282. [PMID: 40383256 DOI: 10.1016/j.clnesp.2025.05.021] [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: 02/22/2025] [Revised: 04/28/2025] [Accepted: 05/10/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Pancreatic surgery is associated with significant morbidity. Cardiopulmonary fitness can predict postoperative outcomes. Similarly, poor insulin sensitivity is associated with postoperative complications. Supervised exercise training can improve cardiorespiratory fitness and insulin sensitivity. Immunonutrition may work synergistically with exercise. The study aimed to assess the impact of a 4-week multimodal prehabilitation programme on cardiopulmonary fitness and insulin sensitivity in patients awaiting pancreatic surgery, compared to a control group receiving standard postoperative care. METHODS In this prospective, non-randomised pilot study, we recruited patients with benign or malignant pathology, listed for pancreatic resection. Subjects underwent a baseline cardiopulmonary exercise test. We measured insulin sensitivity using the gold standard technique, the hyperinsulinaemic-euglycaemic clamp. The 4-week intervention comprised: resistance and high intensity interval training (ten sessions) plus daily omega-3 fatty supplements (2g) and extra virgin olive oil (30 ml). Cardiopulmonary exercise tests and clamps were repeated after four weeks. These were compared with contemporaneous non-randomised controls. RESULTS Of 21 recruited patients, 12 out of 12 (prehabilitation) and 6 out of 9 (control) completed the study. Prehabilitation led to an improvement in oxygen uptake at anaerobic threshold (+2.0 (95 per cent CI 0.1 to 4.0) ml kg-1 min-1) and peak exercise (+3.0 (95 per cent CI 0.7 to 5.3) ml kg-1 min-1), compared to controls. Within the prehabilitation group, there were significant improvements in peak power (P = 0.001), oxygen uptake at anaerobic threshold (P = 0.017) and peak exercise (P = 0.002). Cardiopulmonary fitness parameters were unchanged amongst controls. Insulin sensitivity did not change in either group. CONCLUSION Prehabilitation resulted in a significant improvement in cardiopulmonary fitness, before pancreatic surgery. The potential benefits of improved cardiopulmonary fitness must be balanced against the oncological and metabolic consequences of delay.
Collapse
Affiliation(s)
- Jason George
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK; Minimal Access Therapy Training Unit, Royal Surrey County Hospital, Guildford, Surrey, UK.
| | - David King
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK; Institute for Life and Earth Sciences, Heriot-Watt University, Riccarton, Edinburgh, UK
| | | | - Martin Whyte
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK
| | - Michael Scott
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, USA
| | - Timothy Rockall
- Minimal Access Therapy Training Unit, Royal Surrey County Hospital, Guildford, Surrey, UK
| |
Collapse
|
2
|
Aliyu U, Toor SM, Abdalhakam I, Elrayess MA, Abou−Samra AB, Albagha OME. Evaluating indices of insulin resistance and estimating the prevalence of insulin resistance in a large biobank cohort. Front Endocrinol (Lausanne) 2025; 16:1591677. [PMID: 40421243 PMCID: PMC12104043 DOI: 10.3389/fendo.2025.1591677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 04/11/2025] [Indexed: 05/28/2025] Open
Abstract
Introduction Insulin resistance (IR) is involved in the pathogenesis of various metabolic disorders. Several surrogate indices of IR have been proposed. We assessed the performance of seven clinically relevant indirect measures of IR and estimated the prevalence of IR in a large population-based cohort. Methods The study was conducted on fasting individuals from the Qatar biobank (QBB) participants (n = 7,875). Individuals were considered insulin sensitive (IS) if lean, not diagnosed with diabetes, no hypertriglyceridemia, and not on lipid-lowering drugs, while individuals with Type 2 diabetes (T2D) were considered insulin resistant (IR). Cut-offs were determined as the top or lowest quartile values in the IS participants. The performance of IR indices was based on area under the curve (AUC), sensitivity and specificity. Results The cut-off for HOMA-IR was determined at 1.878, HOMA2-IR (insulin); 1.128, HOMA2-IR (C-peptide); 1.307, QUICKI; 0.347, TyG; 8.281, McAi; 7.727 and 1.718 for TG/HDL. All IR indices analyzed yielded AUC values ranging from 0.83 to 0.92. TyG was the most robust measure for IR (AUC = 0.92, Sensitivity = 0.90, Specificity = 0.79). The overall prevalence of IR in Qatar was estimated at ~51 - 65%. Conclusions TyG index was the most robust index for determining IR in the Qatari population. The proposed cut-offs could serve as a reference in Middle Eastern populations for IR screening.
Collapse
Affiliation(s)
- Usama Aliyu
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Salman M. Toor
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | | | - Mohamed A. Elrayess
- Biomedical Research Center, QU Health, Qatar University, Doha, Qatar
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Omar M. E. Albagha
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
3
|
Ku CW, Chan HG, Sia AL, Huang C, Quek J, Cheung YB, Tan KML, Lai JS, Godfrey KM, Chan JKY, Yap F, Loy SL. One-carbon metabolism, insulin resistance, and fecundability in a Singapore prospective preconception cohort study. Am J Clin Nutr 2025:S0002-9165(25)00252-7. [PMID: 40334751 DOI: 10.1016/j.ajcnut.2025.04.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/27/2025] [Accepted: 04/30/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND One-carbon metabolism, which consists of the folate cycle, methionine cycle, and trans-sulphuration pathway, is associated with nucleotide synthesis. However, the association between one-carbon metabolites, metabolic status, and reproductive health remains poorly understood. OBJECTIVES We examined the association between the one-carbon cycle plasma metabolites and fecundability and determined whether it is modified by metabolic health status, as assessed by insulin resistance (IR). METHODS This prospective cohort study utilized data from the Singapore PREconception Study of long-Term maternal and child Outcomes. Fasting blood samples were collected, and one-carbon cycle metabolites were measured. Fecundability was measured by time to pregnancy in menstrual cycles within a year of enrollment. We identified patterns in plasma one-carbon cycle metabolites using principal component (PC) analysis. We estimated fecundability ratios (FRs) and confidence intervals (CIs), with confounder adjustment using discrete-time proportional hazards models. IR was determined using the Homeostatic Model Assessment 2 Insulin Resistance score, classified into lower IR (<0.65) and higher IR (≥0.65). The role of IR was examined through interaction tests and stratification. RESULTS We identified 3 one-carbon cycle PCs. PC1, characterized by higher folate and lower homocysteine concentrations; PC2, characterized by higher concentrations of dimethylglycine, choline, methionine, and betaine; and PC3, characterized by higher concentrations of vitamins B2, B12, and B6. Each z-score increase in PC1 was associated with a 17% increase in fecundability (FR: 1.17; 95% CI: 1.03, 1.33). The association between PC1 and fecundability was more evident in women with lower IR (FR: 1.30; 95% CI: 1.08, 1.57) but was attenuated in those with higher IR (FR: 1.09; 95% CI: 0.92, 1.30), with a P-for-interaction of 0.127. PC2 and PC3 were not associated with fecundability. CONCLUSIONS Our findings suggest that higher folate and lower homocysteine concentrations, which reflected the interlinked folate and methionine cycles, were associated with higher fecundability in preconception women with lower IR but less so in those with higher IR. CLINICAL TRIAL REGISTRATION This trial was registered at ClinicalTrials.gov as NCT03531658 (https://www. CLINICALTRIALS gov/study/NCT03531658).
Collapse
Affiliation(s)
- Chee Wai Ku
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore; Duke-NUS Medical School, Singapore, Singapore
| | - Hiu Gwan Chan
- Endocrinology Service, Department of Pediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Alexandrea Lishan Sia
- Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore, Singapore
| | - Christine Huang
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jessica Quek
- National University Health System, Singapore, Singapore
| | - Yin Bun Cheung
- Program in Health Services & Systems Research, Duke-NUS Medical School, Singapore, Singapore; Tampere Centre for Child, Adolescent and Maternal Health Research, Tampere University, Tampere, Finland
| | - Karen Mei Ling Tan
- Singapore Institute for Clinical Services, Agency for Science, Technology and Research, Singapore; Medical Research Council Lifecourse Epidemiology Centre, University of Southampton, United Kingdom
| | - Jun Shi Lai
- Singapore Institute for Clinical Services, Agency for Science, Technology and Research, Singapore; Medical Research Council Lifecourse Epidemiology Centre, University of Southampton, United Kingdom
| | - Keith M Godfrey
- National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, United Kingdom; National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton, Southampton, United Kingdom; National Health Service Foundation Trust, University Hospital Southampton, Southampton, United Kingdom
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore; Duke-NUS Medical School, Singapore, Singapore
| | - Fabian Yap
- Duke-NUS Medical School, Singapore, Singapore; Endocrinology Service, Department of Pediatrics, KK Women's and Children's Hospital, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - See Ling Loy
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore; Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
4
|
Preston EV, Lytel-Sternberg J, Quinn MR, Williams PL, Seely EW, Brown FM, Hacker MR, McElrath TF, Cantonwine DE, Wylie BJ, Powe CE, James-Todd T. Associations of personal care product use during pregnancy and the postpartum period with markers of postpartum glycemic control - Results from the ERGO Study. Int J Hyg Environ Health 2025; 266:114569. [PMID: 40158509 PMCID: PMC12044551 DOI: 10.1016/j.ijheh.2025.114569] [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: 10/25/2024] [Revised: 02/24/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Personal care products frequently contain endocrine disrupting chemicals (EDCs) including parabens and phthalates, which can alter glucose metabolism. The postpartum period is a time of rapid metabolic change, but whether EDC-associated product use impacts postpartum glucose metabolism is unknown. METHODS We included 270 participants from the Boston, MA-based Environmental Reproductive and Glucose Outcomes (ERGO) pregnancy cohort with data on self-reported personal care product use at ≤4 pregnancy visits (median: 11, 19, 26, 36 weeks of gestation) and 1 postpartum visit (median: 9 weeks). We quantified postpartum hemoglobin A1c (HbA1c), fasting insulin, fasting- and 2-h glucose post-75-g oral glucose tolerance test, and calculated homeostatic model assessment for insulin sensitivity (HOMA2-S) and beta-cell function (HOMA2-B). Using covariate-adjusted linear regression, we estimated visit-specific associations of product use with postpartum glycemic outcomes. RESULTS Associations of product use with postpartum glycemic measures were mixed. Users of certain hair products had lower postpartum insulin sensitivity compared to non-users (e.g., Visit1 hair gel/spray: 22.8% difference [95% CI: 39.2, -1.9] in mean HOMA2-S). Conversely, users of products like deodorant, liquid- and bar soap, had higher insulin sensitivity and lower glucose levels (e.g., postpartum deodorant: 32.1% difference [95% CI: 7.0, 63.1] in mean HOMA2-S; -3.1 mg/dL [95% CI: 6.3, -0.04] mean fasting glucose). Associations with other products were inconsistent across timepoints or null. CONCLUSION Use of certain personal care products during the perinatal period was associated with altered postpartum glucose metabolism. Larger studies are needed to understand the impacts of product use patterns on glycemic outcomes.
Collapse
Affiliation(s)
- Emma V Preston
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, 02115, USA.
| | - Jennie Lytel-Sternberg
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, 02115, USA.
| | - Marlee R Quinn
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, 02115, USA.
| | - Paige L Williams
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA.
| | - Ellen W Seely
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA.
| | - Florence M Brown
- Joslin Diabetes Center, Harvard Medical School, 1 Joslin Place, Boston, MA, 02215, USA.
| | - Michele R Hacker
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA; Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA, 02215, USA.
| | - Thomas F McElrath
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA; Division of Maternal Fetal Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA.
| | - David E Cantonwine
- Division of Maternal Fetal Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA.
| | - Blair J Wylie
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA, 02215, USA; Departments of Obstetrics and Gynecology and Environmental Health Sciences, Columbia University Medical Center, 622 West 168th St, New York, NY, 10032, USA.
| | - Camille E Powe
- Diabetes Unit, Massachusetts General Hospital, Harvard Medical School, 50 Staniford St, Boston, MA, 02114, USA; Department of Medicine, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, Boston, MA, 02114, USA; Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, 32 Fruit St, Boston, MA, 02114, USA.
| | - Tamarra James-Todd
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, 02115, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA.
| |
Collapse
|
5
|
Bai S, Lin C, Cai X, Hu S, Wu J, Chen L, Yang W, Ji L. Developing CGMap: Characterizing Continuous Glucose Monitoring Data in Patients with Type 2 Diabetes. Biomedicines 2025; 13:1080. [PMID: 40426908 PMCID: PMC12109104 DOI: 10.3390/biomedicines13051080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/18/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Objectives: This study will characterize continuous glucose monitoring (CGM) data in patients with type 2 diabetes in China, and assess the relationship between CGM-derived indicators and diabetes-related clinical parameters. Methods: The data for this study were collected from a randomized trial in China (ChiCTR2000039424) from February 2020 to July 2022 in which patients wore a CGM device for 14 days. Glycemia risk index (GRI), coefficient of variation (CV), standard deviation (SD), mean amplitude of glycemic excursions (MAGE), time in range (TIR), time above range (TAR), time below range (TBR), and estimate glycated hemoglobin (eA1c) were analyzed. Ordinary least square linear regression and the Spearman method were used to test the relationship between CGM-derived indicators and diabetes-related clinical parameters. Results: In all, 528 patients with type 2 diabetes from a randomized controlled trial were analyzed. It was shown that CV, SD, and MAGE increased with age and diabetes duration, but decreased with an increase in body mass index. Higher fasting plasma glucose, higher baseline HbA1c, and higher insulin resistance levels were associated with higher GRI, SD, MAGE, TAR, and eA1c, and they were associated with lower TIR. In addition, higher HOMA-2β was associated with higher TIR and TBR, and with lower TAR and eA1c. Hemoglobin had positive correlations to SD, TAR, and eA1c. Conclusions: It was found that glucose variability increased with age and the duration of diabetes. However, glucose variability decreased with increased BMI. Meanwhile, greater glycemic variability was associated with worse islet function, higher baseline glucose level, and higher hemoglobin.
Collapse
Affiliation(s)
| | | | - Xiaoling Cai
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing 100044, China; (S.B.); (C.L.); (S.H.); (J.W.); (L.C.); (W.Y.)
| | | | | | | | | | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing 100044, China; (S.B.); (C.L.); (S.H.); (J.W.); (L.C.); (W.Y.)
| |
Collapse
|
6
|
Chen YE, Ku CW, Chong MF, Yap F, Chan JKY, Loy SL, Chen LW. Associations of >1-h compared with 1-h meal timing variability (eating jetlag) with plasma glycemic parameters and continuous glucose monitoring measures among pregnant females: a prospective cohort study. Am J Clin Nutr 2025:S0002-9165(25)00243-6. [PMID: 40294750 DOI: 10.1016/j.ajcnut.2025.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 04/01/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Eating jetlag (EJL), the difference in eating times between weekdays and weekends, disrupts circadian alignment and may affect metabolic health. However, its influence on glucose tolerance and continuous glucose monitoring (CGM) during pregnancy remains unknown. OBJECTIVES We aimed to investigate the associations between EJL and glycemic parameters during pregnancy. METHODS This secondary analysis was conducted on a cohort of 248 healthy pregnant females from Singapore. EJL, derived from 4-d food diaries at 20-wk of gestation, was the absolute difference in average meal times between weekdays and weekends for the first (EJLfirst) and last (EJLlast) meals and categorized as ≤1-h (reference) or >1-h. Primary outcomes at 25-wk of gestation included results from the 75-g oral glucose tolerance test, fasting insulin, homeostasis model assessment of insulin resistance (HOMA2-IR), and β-cell function (HOMA2-%B). Secondary outcomes at 20-wk of gestation included glycemic control and variability measured over 10-d using CGM. Skewed glycemic variables were log-transformed for normality, and associations between EJL and glycemic outcomes were analyzed using multivariable regressions. RESULTS After adjusting for baseline sociodemographic, lifestyle, and dietary factors, EJLlast >1-h was associated with higher fasting insulin [geometric mean ratio (95% confidence intervals): 1.21 (1.05, 1.39)], HOMA2-IR [1.21 (1.05, 1.39)], HOMA2-%B [1.11 (1.01, 1.22)], and CGM-based measures, including mean glucose [1.05 (1.00, 1.09)], J-index [1.11 (1.01, 1.22)], and glucose management indicator [1.03 (1.00, 1.06)]. EJLfirst >1-h was associated with higher CGM-based mean amplitude of glycemic excursions (MAGE) [1.09 (1.01, 1.19)]. For CGM-based glycemic variability outcomes (standard deviation, coefficient of variation [CV], MAGE), there were interactions between EJLfirst and 1) diet quality [adherence to Dietary Approaches to Stop Hypertension (DASH)] (P-interactions = 0.06-0.09), and 2) prepregnancy body mass index (BMI) (P-interaction=0.07 for CV). In females with a prepregnancy BMI ≥23 kg/m2 and low diet quality (DASH score ≤median), EJLfirst >1 h was associated with higher CGM-based glycemic variability. CONCLUSIONS EJL was associated with unfavorable glycemic parameters during pregnancy. Dietary interventions could promote consistent meal timing, especially in higher risk groups with suboptimal nutritional status. This trial was registered at clinicaltrials.gov as NCT03803345.
Collapse
Affiliation(s)
- Yu-En Chen
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Chee Wai Ku
- Duke-NUS Medical School, Singapore, Singapore; Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Mary Ff Chong
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Fabian Yap
- Duke-NUS Medical School, Singapore, Singapore; Endocrinology Service, KK Women's and Children's Hospital, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jerry Kok Yen Chan
- Duke-NUS Medical School, Singapore, Singapore; Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - See Ling Loy
- Duke-NUS Medical School, Singapore, Singapore; Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Ling-Wei Chen
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan; Master of Public Health Program, College of Public Health, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
7
|
Kang F, Zhang Z, Fu H, Sun J, Zhang J, Wang Q. β-Cell Dedifferentiation in HOMA-βlow and HOMA-βhigh Subjects. J Clin Endocrinol Metab 2025; 110:e1430-e1438. [PMID: 39133811 PMCID: PMC12012814 DOI: 10.1210/clinem/dgae538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 09/12/2024]
Abstract
CONTEXT β-Cell dedifferentiation ratio is increased in type 2 diabetes; but its direct link to in vivo β-cell function in human remains unclear. OBJECTIVE The present study was designed to investigate whether β-cell dedifferentiation in situ was closely associated with β-cell function in vivo and to identify targets crucial for β-cell dedifferentiation/function in human. METHODS We acquired homeostasis model assessment of β-cell function (HOMA-β) values, calculated the number of hormone-negative endocrine cells, and evaluated important markers and novel candidates for β-cell dedifferentiation/function on paraneoplastic pancreatic tissues from 13 patients with benign pancreatic cystic neoplasm or intrapancreatic accessory spleen. RESULTS Both the β-cell dedifferentiation ratio and the dedifferentiation marker (Aldh1a3) were inversely related to in vivo β-cell function (HOMA-β) and in situ β-cell functional markers Glut2 and Ucn3 in humans. Moreover, the islets from HOMA-βlow subjects were manifested as (1) increased β-cell dedifferentiation ratio, (2) enriched dedifferentiation maker Aldh1a3, and (3) lower expression of Glut2 and Ucn3 compared with those from HOMA-βhigh subjects. We found that basic leucine zipper transcription factor 2 (Bach2) expression was significantly induced in islets from HOMA-βlow patients and was positively correlated with the ratio of β-cell dedifferentiation in humans. CONCLUSION Our findings emphasize the contribution of β-cell dedifferentiation to β-cell dysfunction in humans. Bach2 induction in β-cells with higher frequency of dedifferentiation observed in HOMA-βlow subjects reinforces its distinctive role as a pharmaceutical target of β-cell dedifferentiation for the treatment of people with diabetes.
Collapse
Affiliation(s)
- Fuyun Kang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhuo Zhang
- Department of Surgery, Shanghai United Family Hospital, Shanghai 200021, China
| | - Hui Fu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiajun Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jun Zhang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qidi Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
8
|
Pather R, Khathi A, Ngubane P. The effects of obesity on thyroid function in a metabolically healthy high-fat, high-carbohydrate diet-induced obese rat model. Front Endocrinol (Lausanne) 2025; 16:1538627. [PMID: 40331142 PMCID: PMC12052566 DOI: 10.3389/fendo.2025.1538627] [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: 12/03/2024] [Accepted: 03/28/2025] [Indexed: 05/08/2025] Open
Abstract
Introduction Obesity is a recognized exacerbator of thyroid dysfunction due to its detrimental effects on energy homeostasis, appetite regulation, basal metabolic rate, thermogenesis, and metabolism. However, almost all the reported findings on obesity-related thyroid dysfunction are based on models of metabolically unhealthy obesity (MUO) in the presence of insulin resistance. There are currently no reported studies using a metabolically healthy obesity (MHO) model characterized by the absence of insulin resistance to investigate thyroid dysfunction. Hence, this study aimed to investigate the association between thyroid dysfunction and obesity in a metabolically healthy high-fat high-carbohydrate diet-induced obese rat model. Materials and methods Male Sprague Dawley rats were randomly divided into either the control diet or the high-fat high-carbohydrate diet group (HFHC) (n=9, per group). During the 5-month induction period, the control group did not develop obesity while consuming a standard diet with water. The HFHC diet group consumed the HFHC diet and water for the same duration and was diagnosed with obesity. Post-obesity confirmation, the animals continued with the respective diets for a further 7 months to maintain the obese state. Caloric intake, fasting blood glucose (FBG) and BMI were measured once a month for the duration of the experiment. Glucose homeostasis and thyroid functional parameters were assessed terminally, accompanied by satiety and pro-inflammatory markers. Results The HFHC diet group presented with higher BMI, caloric intake and FBG, and elevated insulin, HOMA-IR, Hb1Ac, leptin and IL-6 levels compared to the control diet group. The HFHC diet group presented with significantly elevated levels of TSH, fT3 and fT4. These observations suggest that thyroid homeostasis is disturbed in the obese state. However, the reported elevated glycemic status indicators and IL-6 concentrations in the HFHC diet group did not satisfy the minimum criteria to be characterized as MUO. Conclusion The HFHC diet has induced MHO in male Sprague Dawley rats. This warrants using this model to investigate the homeostatic changes that occur during the metabolically healthy obese state. This can open new avenues for developing preventative measures to avoid progressing to MUO.
Collapse
Affiliation(s)
- Reveshni Pather
- Department of Human Physiology, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | | | | |
Collapse
|
9
|
Dunne F, Newman C, Alvarez-Iglesias A, O'Shea P, Devane D, Gillespie P, Egan A, O'Donnell M, Smyth A. Postnatal Cardiometabolic Health After Metformin Use in Gestational Diabetes: A Secondary Analysis of the EMERGE Trial. J Clin Endocrinol Metab 2025; 110:e1566-e1572. [PMID: 39056256 DOI: 10.1210/clinem/dgae522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/08/2024] [Accepted: 07/24/2024] [Indexed: 07/28/2024]
Abstract
AIM Women with gestational diabetes mellitus (GDM) display adverse lifetime cardiometabolic health. We examined whether early metformin in GDM could impact cardiometabolic risk factors postpartum. METHODS EMERGE, a double-blind, placebo-controlled trial, randomized pregnancies 1:1 to placebo or metformin at GDM diagnosis and followed participants from randomization until 12 ± 4 weeks postpartum. In total, 478 pregnancies were available for postpartum maternal assessment, 237 and 241 assigned to metformin and placebo respectively. Weight (kg), body mass index (BMI) (kg/m2), waist circumference (cm), and blood pressure (mmHg) were measured, infant feeding method documented, and blood specimens drawn for a 75-gram oral glucose tolerance test, fasting insulin, C-peptide, and lipid analysis. RESULTS Despite similar weight and BMI at trial randomization, participants receiving metformin had significantly lower weight (79.5 ± 15.9 vs 82.6 ± 16.9 kg; P = .04) and BMI (29.3 [5.6] vs 30.5 [5.4]; P = .018) at the postpartum visit. However, no difference in weight change from randomization to 12 weeks postpartum was observed between metformin and placebo groups. Overall, 29% (n = 139) of the cohort met criteria for prediabetes or diabetes, with no positive impact with metformin. There were also no differences in measurements of insulin resistance, blood pressure, or lipids between groups. CONCLUSION Early metformin use in GDM did not impact important cardiometabolic parameters in the early postpartum period despite significant benefits in weight gain and insulin use in pregnancy.
Collapse
Affiliation(s)
- Fidelma Dunne
- Institute for Clinical Trials, College of Medicine, Nursing and Health Sciences, University of Galway, Galway H91 TK33, Ireland
- HRB Clinical Research Facility Galway, University of Galway, Galway H91 TK33, Ireland
- Galway University Hospital, Galway H91 YR71, Ireland
| | - Christine Newman
- Institute for Clinical Trials, College of Medicine, Nursing and Health Sciences, University of Galway, Galway H91 TK33, Ireland
- HRB Clinical Research Facility Galway, University of Galway, Galway H91 TK33, Ireland
- Galway University Hospital, Galway H91 YR71, Ireland
| | | | - Paula O'Shea
- HRB Clinical Research Facility Galway, University of Galway, Galway H91 TK33, Ireland
| | - Declan Devane
- Institute for Clinical Trials, College of Medicine, Nursing and Health Sciences, University of Galway, Galway H91 TK33, Ireland
- HRB Clinical Research Facility Galway, University of Galway, Galway H91 TK33, Ireland
| | - Paddy Gillespie
- School of Business and Economics, University of Galway, Galway H91 TK33, Ireland
| | - Aoife Egan
- Institute for Clinical Trials, College of Medicine, Nursing and Health Sciences, University of Galway, Galway H91 TK33, Ireland
- Department of Endocrinology, Mayo Clinic, Rochester, MN AZ85054, USA
| | - Martin O'Donnell
- Institute for Clinical Trials, College of Medicine, Nursing and Health Sciences, University of Galway, Galway H91 TK33, Ireland
- HRB Clinical Research Facility Galway, University of Galway, Galway H91 TK33, Ireland
- Galway University Hospital, Galway H91 YR71, Ireland
| | - Andrew Smyth
- Institute for Clinical Trials, College of Medicine, Nursing and Health Sciences, University of Galway, Galway H91 TK33, Ireland
- HRB Clinical Research Facility Galway, University of Galway, Galway H91 TK33, Ireland
- Galway University Hospital, Galway H91 YR71, Ireland
| |
Collapse
|
10
|
Dansinger ML, Gleason JA, Maddalena J, Asztalos BF, Diffenderfer MR. Lifestyle Modification in Prediabetes and Diabetes: A Large Population Analysis. Nutrients 2025; 17:1333. [PMID: 40284198 PMCID: PMC12030603 DOI: 10.3390/nu17081333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Aims: Diabetes mellitus is a major cause of atherosclerotic cardiovascular disease (ASCVD). We examined a large population and tested the efficacy of a voluntary lifestyle program in prediabetic and diabetic subjects. Methods: Of 133,764 subjects, 56.3% were healthy, 36.2% were prediabetic, and 7.5% were diabetic. Fasting serum measurements of glucose, insulin, adiponectin, glycosylated hemoglobin (HbA1c), high-sensitivity C-reactive protein (hs-CRP), glycated serum protein (GSP), fibrinogen, myeloperoxidase (MPO), lipoprotein-associated phospholipase A2 (LpPLA2), as well as standard lipids, direct low-density lipoprotein cholesterol (LDL-C), and small dense LDL-C (sdLDL-C) were performed using standard automated assays. Follow-up sampling at 6-12 months occurred in 20.1% of the prediabetic and 22.2% of the diabetic subjects; of these, 12.2% of the prediabetic and 9.7% of the diabetic subjects participated in a voluntary, real-world, digital dietitian-directed lifestyle-modification program with a 10-year diabetes risk being calculated using a biochemical model (Framingham). Results: Prediabetic and diabetic subjects had significantly elevated triglycerides, sdLDL-C, and hs-CRP and decreased HDL-C. They were insulin resistant as compared to healthy subjects, but only diabetics had significant reductions in insulin production. Lifestyle modification significantly reduced diabetes risk by 45.6% in prediabetics and significantly increased (2.4-fold) the percentage of diabetics that were in remission at follow-up (8.2% versus 3.4%) with increased weight loss (6.5 versus 2.0 pounds). Lifestyle intervention resulted in significant favorable effects on many metabolic markers. Conclusions: The measurement of fasting glucose and insulin is essential for the detection of decreased insulin production in diabetics. A digital lifestyle program can have favorable effects on ASCVD risk factors and diabetic status.
Collapse
Affiliation(s)
- Michael L. Dansinger
- Boston Heart Diagnostics Corporation, 200 Crossing Boulevard, Framingham, MA 01702, USA; (J.A.G.); (J.M.); (B.F.A.)
| | - Joi A. Gleason
- Boston Heart Diagnostics Corporation, 200 Crossing Boulevard, Framingham, MA 01702, USA; (J.A.G.); (J.M.); (B.F.A.)
| | - Julia Maddalena
- Boston Heart Diagnostics Corporation, 200 Crossing Boulevard, Framingham, MA 01702, USA; (J.A.G.); (J.M.); (B.F.A.)
| | - Bela F. Asztalos
- Boston Heart Diagnostics Corporation, 200 Crossing Boulevard, Framingham, MA 01702, USA; (J.A.G.); (J.M.); (B.F.A.)
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
| | - Margaret R. Diffenderfer
- Boston Heart Diagnostics Corporation, 200 Crossing Boulevard, Framingham, MA 01702, USA; (J.A.G.); (J.M.); (B.F.A.)
| |
Collapse
|
11
|
Hamamoto Y, Oura T, Hirase T. Insulin Sensitivity and Beta-Cell Function Following Tirzepatide in Japanese Patients with Type 2 Diabetes: A SURPASS J-mono Analysis. Diabetes Ther 2025; 16:717-729. [PMID: 39951042 PMCID: PMC11926299 DOI: 10.1007/s13300-025-01704-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/03/2025] [Indexed: 03/21/2025] Open
Abstract
INTRODUCTION The objective of this work was to assess the association of tirzepatide with changes in insulin sensitivity and beta-cell function in Japanese patients with type 2 diabetes (T2D). METHODS This was a prespecified analysis of SURPASS J-mono, a multicenter, randomized, double-blind, active-controlled phase 3 study in Japanese participants with T2D. Participants were randomly assigned to receive tirzepatide 5, 10, 15 mg, or dulaglutide 0.75 mg once weekly for 52 weeks. Changes from baseline in homeostatic model assessment (HOMA2) parameters were assessed by a mixed model for repeated measures. Post hoc subgroup analyses were conducted based on high and low baseline C-peptide categories (cutoff: median 1.86 µg/l). RESULTS This analysis included 636 participants (tirzepatide 5 mg: n = 159; 10 mg: n = 158; 15 mg: n = 160; dulaglutide 0.75 mg: n = 159). Fasting insulin and C-peptide levels were significantly reduced from baseline at week 52 at all three tirzepatide doses compared with dulaglutide 0.75 mg (estimated treatment differences [ETDs] for tirzepatide 5, 10, and 15 mg, respectively: insulin, - 22.6%, - 29.8%, and - 31.0%; C-peptide, - 14.2%, - 21.5%, and - 20.7%; p < 0.001 all comparisons). Insulin sensitivity and beta-cell function indices significantly increased in all tirzepatide groups compared with the dulaglutide 0.75-mg group (ETDs for tirzepatide 5, 10, and 15 mg, respectively: HOMA2-%S [insulin], 31.8%, 43.2%, and 49.9%; HOMA2-%S [C-peptide], 24.8%, 33.9%, and 38.2%; HOMA2-%B [insulin], 27.3%, 34.9%, and 40.6%; HOMA2-%B [C-peptide], 31.6%, 40.1%, and 46.7%; p < 0.001 all comparisons). Regardless of baseline C-peptide level, the tirzepatide groups showed significant improvement in glycated hemoglobin at week 52 compared with the dulaglutide group (C-peptide < 1.86 µg/l: - 2.31% to - 2.75% vs. - 1.45%; ≥ 1.86 µg/l: - 2.43% to - 2.89% vs. - 1.12%; p < 0.001). CONCLUSIONS In these prespecified and post hoc analyses, tirzepatide was associated with improved insulin sensitivity and insulin secretion in Japanese participants with T2D, which may result in reduced fasting insulin levels. CLINICALTRIALS GOV: NCT03861052.
Collapse
Affiliation(s)
- Yoshiyuki Hamamoto
- Center for Diabetes, Endocrinology and Metabolism, Kansai Electric Power Hospital, Osaka, Japan
| | - Tomonori Oura
- Japan Drug Development and Medical Affairs, Eli Lilly Japan K.K., Kobe, 651-0086, Japan
| | - Tetsuaki Hirase
- Japan Drug Development and Medical Affairs, Eli Lilly Japan K.K., Kobe, 651-0086, Japan.
| |
Collapse
|
12
|
Askeland A, Rasmussen RW, Gjela M, Frøkjær JB, Højlund K, Mellergaard M, Handberg A. Non-invasive liver fibrosis markers are increased in obese individuals with non-alcoholic fatty liver disease and the metabolic syndrome. Sci Rep 2025; 15:10652. [PMID: 40148373 PMCID: PMC11950363 DOI: 10.1038/s41598-025-85508-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/03/2025] [Indexed: 03/29/2025] Open
Abstract
The need for early non-invasive diagnostic tools for chronic liver fibrosis is growing, particularly in individuals with obesity, non-alcoholic fatty liver disease (NAFLD), and the metabolic syndrome (MetS) since prevalence of these conditions is increasing. This case-control study compared non-invasive liver fibrosis markers in obesity with NAFLD and MetS (NAFLD-MetS, n = 33), in obese (n = 28) and lean (n = 27) control groups. We used MRI (T1 relaxation times (T1) and liver stiffness), circulating biomarkers (CK18, PIIINP, and TIMP1), and algorithms (FIB-4 index, Forns score, FNI, and MACK3 score) to assess their potential in predicting liver fibrosis risk. We found that T1 (892 ± 81 ms vs. 818 ± 64 ms, p < 0.001), FNI (15 ± 12% vs. 9 ± 7%, p = 0.018), CK18 (166 ± 110 U/L vs. 113 ± 41 U/L, p = 0.019), and MACK3 (0.18 ± 0.15 vs. 0.05 ± 0.04, p < 0.001) were higher in the NAFLD-MetS group compared with the obese control group. Moreover, correlations were found between CK18 and FNI (r = 0.69, p < 0.001), CK18 and T1 (r = 0.41, p < 0.001), FNI and T1 (r = 0.33, p = 0.006), MACK3 and FNI (r = 0.79, p < 0.001), and MACK3 and T1 (r = 0.50, p < 0.001). We show that liver fibrosis markers are increased in obese individuals with NAFLD and MetS without clinical signs of liver fibrosis. More studies are needed to validate the use of these non-invasive biomarkers for early identification of liver fibrosis risk.
Collapse
Affiliation(s)
- Anders Askeland
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | | - Mimoza Gjela
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Radiology, Aalborg University Hospital, Aalborg, Denmark
| | - Jens Brøndum Frøkjær
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Radiology, Aalborg University Hospital, Aalborg, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Maiken Mellergaard
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark.
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
13
|
Ai M, Otokozawa S, Liu CT, Asztalos BF, Maddalena J, Diffenderfer MR, Russo G, Thongtang N, Dansinger ML. Diabetes Mellitus Risk Prediction in the Framingham Offspring Study and Large Population Analysis. Nutrients 2025; 17:1117. [PMID: 40218874 PMCID: PMC11990307 DOI: 10.3390/nu17071117] [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: 02/03/2025] [Revised: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Diabetes mellitus is a major cause of death and a significant risk factor for cardiovascular disease, kidney failure, neuropathy, and retinopathy. Our objectives were to develop a diabetes risk model and apply it to a large population. Methods: Non-diabetic adults in the Framingham Offspring Study (n = 2416) were followed for 10 years for new diabetes. At baseline, the fasting serum glucose, adiponectin, insulin, glycated albumin, total cholesterol, triglycerides (TG), and high-density lipoprotein cholesterol (HDL-C) were measured using standardized automated assays. Standard health information was collected. Diabetes risk prediction models were developed using logistic regression analysis and applied to a large population (n = 133,764). Results: In this prospective study, 166 subjects (6.9%) developed new-onset diabetes. Glucose, body mass index (BMI), log adiponectin, % log glycated albumin, parental diabetes, TG, and the use of cholesterol-lowering medications entered the model (C statistic: 0.924; 0.898, biochemical variables: 0.898, and fasting glucose: only 0.876). In the population in non-diabetic subjects (56.3) and prediabetic subjects (36.2%), the predicted 10-year diabetes risk rates were 0.4% and 5.5% with the biochemical model, respectively. Prediabetic and diabetic subjects were insulin-resistant compared to non-diabetic subjects, but only those with diabetes had significant reductions in their insulin production. Conclusions: The 10-year risk of diabetes can be accurately predicted and applied to large populations. Fasting glucose alone is diagnostic for diabetes and is an excellent predictor of future diabetes, with having prediabetes increasing the risk 6-fold. Insulin and C-peptide measurements are useful in diabetic subjects to detect decreased insulin production and the need for insulin therapy.
Collapse
Affiliation(s)
- Masumi Ai
- Cardiovascular Nutrition Laboratory, Human Nutrition Research Center on Aging at Tufts University, Tufts University School of Medicine, Boston, MA 02111, USA; (S.O.); (B.F.A.); (M.R.D.); (G.R.); (N.T.); (M.L.D.)
- Department of Insured Medical Care Management, Institute of Science Tokyo, Tokyo 113-8519, Japan
| | - Seiko Otokozawa
- Cardiovascular Nutrition Laboratory, Human Nutrition Research Center on Aging at Tufts University, Tufts University School of Medicine, Boston, MA 02111, USA; (S.O.); (B.F.A.); (M.R.D.); (G.R.); (N.T.); (M.L.D.)
- Department of Public Health, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA;
- Framingham Heart Study, Framingham, MA 01702, USA
| | - Bela F. Asztalos
- Cardiovascular Nutrition Laboratory, Human Nutrition Research Center on Aging at Tufts University, Tufts University School of Medicine, Boston, MA 02111, USA; (S.O.); (B.F.A.); (M.R.D.); (G.R.); (N.T.); (M.L.D.)
- Boston Heart Diagnostics, Framingham, MA 01702, USA;
| | - Julia Maddalena
- Boston Heart Diagnostics, Framingham, MA 01702, USA;
- Perennial Inc., Boulder, CO 80301, USA
| | - Margaret R. Diffenderfer
- Cardiovascular Nutrition Laboratory, Human Nutrition Research Center on Aging at Tufts University, Tufts University School of Medicine, Boston, MA 02111, USA; (S.O.); (B.F.A.); (M.R.D.); (G.R.); (N.T.); (M.L.D.)
- Boston Heart Diagnostics, Framingham, MA 01702, USA;
| | - Giuseppina Russo
- Cardiovascular Nutrition Laboratory, Human Nutrition Research Center on Aging at Tufts University, Tufts University School of Medicine, Boston, MA 02111, USA; (S.O.); (B.F.A.); (M.R.D.); (G.R.); (N.T.); (M.L.D.)
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Nuntakorn Thongtang
- Cardiovascular Nutrition Laboratory, Human Nutrition Research Center on Aging at Tufts University, Tufts University School of Medicine, Boston, MA 02111, USA; (S.O.); (B.F.A.); (M.R.D.); (G.R.); (N.T.); (M.L.D.)
- Division of Endocrinology and Metabolism, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Michael L. Dansinger
- Cardiovascular Nutrition Laboratory, Human Nutrition Research Center on Aging at Tufts University, Tufts University School of Medicine, Boston, MA 02111, USA; (S.O.); (B.F.A.); (M.R.D.); (G.R.); (N.T.); (M.L.D.)
- Boston Heart Diagnostics, Framingham, MA 01702, USA;
| |
Collapse
|
14
|
Gill H, Badulescu S, Di Vincenzo JD, Tabassum A, McKenzie A, Shah H, Amin M, Llach CD, Rosenblat JD, McIntyre RS, Mansur RB. Metabolic factors modulate effort-based decision-making in major depressive disorder. J Affect Disord 2025; 373:88-93. [PMID: 39732399 DOI: 10.1016/j.jad.2024.12.090] [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: 06/20/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND Abnormalities in effort-based decision-making have been consistently reported in major depressive disorder (MDD). Evidence indicates that metabolic factors, such as insulin resistance and dyslipidemia, which are highly prevalent in MDD, are independently associated with reward disturbances. Herein, we investigate the moderating effect of metabolic factors on effort-based decision-making in individuals with MDD. METHODS Forty-nine adults with MDD completed the Effort Expenditure for Rewards Task (EEfRT). Anthropometric and laboratorial parameters were assessed in all participants. We conducted a factor analysis to identify combinations of correlated metabolic variables, and reduce the number of comparisons. RESULTS Proxy markers of elevated insulin resistance (OR: 0.816, p < 0.001) and hyperglycemia (OR: 0.898, p = 0.021) were associated with a lower willingness to exert physical effort for rewards in the EEfRT. In contrast, elevated HDL (OR: 1.165, p = 0.004), and elevated non-HDL cholesterol and triglycerides (OR: 1.184, p < 0.001) were associated with increased frequency of hard task choices. These associations were independent of age, sex, depressive symptoms severity and medication use. Computational modeling revealed that the insulin resistance (β = 0.275, p = 0.035) and cholesterol factors (β = 0.565, p < 0.001) were independently associated with increased effort discounting. LIMITATIONS Post-hoc analysis using a relatively small sample of convenience. CONCLUSIONS Metabolic factors significantly and independently modulated effort-based decision-making in patients with MDD. These results have implications for our understanding of reward disturbances in MDD, and offer insights for further mechanistic investigations.
Collapse
Affiliation(s)
- Hartej Gill
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Sebastian Badulescu
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Joshua D Di Vincenzo
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Aniqa Tabassum
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Andrea McKenzie
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Hiya Shah
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Mahrus Amin
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Cristian-Daniel Llach
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Joshua D Rosenblat
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Rodrigo B Mansur
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
15
|
Kwon Y, Yoon H, Ha J, Lee HS, Pahk K, Kwon H, Kim S, Park S. Changes in pancreatic levodopa uptake in patients with obesity and new-onset type 2 diabetes: an 18F-FDOPA PET-CT study. Front Endocrinol (Lausanne) 2025; 16:1460253. [PMID: 40099262 PMCID: PMC11911206 DOI: 10.3389/fendo.2025.1460253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 01/23/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Levodopa (L-3,4-dihydroxyphenylalanine)g, a dopamine precursor that circulates in the peripheral region, is involved in pancreatic glycemic control. Although previous animal studies have shown that peripheral levodopa is correlated with insulin secretion in pancreatic beta cells, the mechanism by which the pancreas uses levodopa differently in humans with obesity and type 2 diabetes remains unknown. Our study aimed to observe how the pancreas uptakes and utilizes levodopa differently under obese and diabetic conditions. Materials and method 18F-fluoro-L-dopa positron emission tomography-computed tomography (18F-FDOPA PET-CT) was used to visualize how the human body uses levodopa under obese and diabetic conditions and presented its clinical implications. 10 patients were divided into 3 groups: 1) Group A, normal weight without type 2 diabetes; 2) Group B, obese without type 2 diabetes; and 3) Group C, obese with new-onset type 2 diabetes. All patients' lifestyle modification was conducted prior to 18F-FDOPA PET-CT, and plasma samples were collected to confirm changes in amino acid metabolites. Results Pancreatic levodopa uptake increased in obese patients with insulin resistance, whereas it decreased in obese patients with new-onset type 2 diabetes [standardized uptake value (SUV) mean in participants with normal weight, 2.6 ± 0.7; SUVmean in patients with obesity, 3.6 ± 0.1; SUVmean in patients with obesity and new-onset type 2 diabetes, 2.6 ± 0.1, P = 0.02]. Conclusions This suggested that the alterations in the functional capacity of pancreatic beta cells to take up circulating levodopa are potentially linked to the insulin resistance and the pathogenesis of type 2 diabetes. The differences in the uptake values between the groups implied that pancreatic levodopa uptake could be an early indicator of type 2 diabetes.
Collapse
Affiliation(s)
- Yeongkeun Kwon
- Center for Obesity and Metabolic Diseases, Korea University Anam Hospital, Seoul, Republic of Korea
- Gut & Metabolism Laboratory, Korea University College of Medicine, Seoul, Republic of Korea
- Division of Foregut Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hanseok Yoon
- Division of Biotechnology, Korea University, Seoul, Republic of Korea
| | - Jane Ha
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, United States
| | - Hyeon-seong Lee
- Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung, Republic of Korea
| | - Kisoo Pahk
- Department of Nuclear Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyunwoo Kwon
- Department of Nuclear Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sungeun Kim
- Department of Nuclear Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sungsoo Park
- Center for Obesity and Metabolic Diseases, Korea University Anam Hospital, Seoul, Republic of Korea
- Gut & Metabolism Laboratory, Korea University College of Medicine, Seoul, Republic of Korea
- Division of Foregut Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
16
|
Vliebergh J, Gesquiere I, Foulon V, Augustijns P, Lannoo M, Deleus E, Meulemans A, Mathieu C, Mertens A, Matthys C, Van der Schueren B, Vangoitsenhoven R. Change in carbohydrate intake one year after Roux-en-Y gastric bypass: A prospective study. Nutr Health 2025; 31:209-216. [PMID: 37006189 DOI: 10.1177/02601060231166821] [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: 04/04/2023]
Abstract
Background and objectives: To investigate the effect of carbohydrate intake before laparoscopic Roux-en-Y gastric bypass (LRYGB) on body weight, body composition and glycaemic status after surgery. Methods: In a tertiary centre cohort study, dietary habits, body composition and glycaemic status were evaluated before and 3, 6 and 12 months after LRYGB. Detailed dietary food records were processed by specialized dietitians on the basis of a standard protocol. The study population was subdivided according to relative carbohydrate intake before surgery. Results: Before surgery, 30 patients had a moderate relative carbohydrate intake (26%-45%, M-CHO), a mean body mass index (BMI) of 40.4 ± 3.9 kg/m² and a mean glycated haemoglobin A1c (A1C) of 6.5 ± 1.2% compared to 20 patients with a high relative carbohydrate intake (> 45%, H-CHO), mean BMI of 40.9 ± 3.7 kg/m² (non-significant, NS) and a mean A1C of 6.2% (NS). One year after surgery, body weight, body composition and glycaemic status were similar in the M-CHO (n = 25) and H-CHO groups (n = 16), despite less caloric intake in the H-CHO group (1317 ± 285 g vs. 1646 ± 345 g in M-CHO, p < 0.01). Their relative carbohydrate intake converged to 46% in both groups, but the H-CHO group reduced the absolute total carbohydrate consumption more than the M-CHO group (190 ± 50 g in M-CHO vs. 153 ± 39 g in H-CHO, p < 0.05), and this was especially pronounced for the mono- and disaccharides (86 ± 30 g in M-CHO vs. 65 ± 27 g in H-CHO, p < 0.05). Conclusion: A high relative carbohydrate intake before LRYGB, did not influence the change in body composition or diabetes status after surgery, despite a significantly lower total energy intake and less mono- and disaccharide consumption after surgery.
Collapse
Affiliation(s)
- Joke Vliebergh
- Department of Endocrinology, University Hospitals Leuven, Belgium
| | - Ina Gesquiere
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Belgium
| | - Veerle Foulon
- Department of Pharmaceutical and Pharmacological Sciences, Clinical Pharmacology and Pharmacotherapy, KU Leuven, Belgium
| | - Patrick Augustijns
- Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery and Disposition, KU Leuven, Belgium
| | - Matthias Lannoo
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Belgium
- Department of Abdominal Surgery, University Hospitals Leuven, Belgium
| | - Ellen Deleus
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Belgium
- Department of Abdominal Surgery, University Hospitals Leuven, Belgium
| | - Ann Meulemans
- Department of Endocrinology, University Hospitals Leuven, Belgium
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Belgium
| | - Chantal Mathieu
- Department of Endocrinology, University Hospitals Leuven, Belgium
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Belgium
| | - Ann Mertens
- Department of Endocrinology, University Hospitals Leuven, Belgium
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Belgium
| | - Christophe Matthys
- Department of Endocrinology, University Hospitals Leuven, Belgium
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Belgium
| | - Bart Van der Schueren
- Department of Endocrinology, University Hospitals Leuven, Belgium
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Belgium
| | - Roman Vangoitsenhoven
- Department of Endocrinology, University Hospitals Leuven, Belgium
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Belgium
| |
Collapse
|
17
|
Dwibedi C, Axelsson AS, Abrahamsson B, Fahey JW, Asplund O, Hansson O, Ahlqvist E, Tremaroli V, Bäckhed F, Rosengren AH. Effect of broccoli sprout extract and baseline gut microbiota on fasting blood glucose in prediabetes: a randomized, placebo-controlled trial. Nat Microbiol 2025; 10:681-693. [PMID: 39929977 PMCID: PMC11879859 DOI: 10.1038/s41564-025-01932-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/13/2025] [Indexed: 03/06/2025]
Abstract
More effective treatments are needed for impaired fasting glucose or glucose intolerance, known as prediabetes. Sulforaphane is an isothiocyanate that reduces hepatic gluconeogenesis in individuals with type 2 diabetes and is well tolerated when provided as a broccoli sprout extract (BSE). Here we report a randomized, double-blind, placebo-controlled trial in which drug-naive individuals with prediabetes were treated with BSE (n = 35) or placebo (n = 39) once daily for 12 weeks. The primary outcome was a 0.3 mmol l-1 reduction in fasting blood glucose compared with placebo from baseline to week 12. Gastro-intestinal side effects but no severe adverse events were observed in response to treatment. BSE did not meet the prespecified primary outcome, and the overall effect in individuals with prediabetes was a 0.2 mmol l-1 reduction in fasting blood glucose (95% confidence interval -0.44 to -0.01; P = 0.04). Exploratory analyses to identify subgroups revealed that individuals with mild obesity, low insulin resistance and reduced insulin secretion had a pronounced response (0.4 mmol l-1 reduction) and were consequently referred to as responders. Gut microbiota analysis further revealed an association between baseline gut microbiota and pathophysiology and that responders had a different gut microbiota composition. Genomic analyses confirmed that responders had a higher abundance of a Bacteroides-encoded transcriptional regulator required for the conversion of the inactive precursor to bioactive sulforaphane. The abundance of this gene operon correlated with sulforaphane serum concentration. These findings suggest a combined influence of host pathophysiology and gut microbiota on metabolic treatment response, and exploratory analyses need to be confirmed in future trials. ClinicalTrials.gov registration: NCT03763240 .
Collapse
Affiliation(s)
- Chinmay Dwibedi
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology and Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Annika S Axelsson
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Birgitta Abrahamsson
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jed W Fahey
- Departments of Medicine, Pharmacology and Molecular Sciences, and Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Olof Asplund
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - Ola Hansson
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
- Institute for Molecular Medicine Finland, Helsinki University, Helsinki, Finland
| | - Emma Ahlqvist
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - Valentina Tremaroli
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anders H Rosengren
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
18
|
Yu G, Tam CHT, Lim CKP, Shi M, Lau ESH, Ozaki R, Lee HM, Ng ACW, Hou Y, Fan B, Huang C, Wu H, Yang A, Cheung HM, Lee KF, Siu SC, Hui G, Tsang CC, Lau KP, Leung JYY, Cheung EYN, Tsang MW, Kam G, Lau IT, Li JKY, Yeung VTF, Lau E, Lo S, Fung S, Cheng YL, Szeto CC, Hong Kong Diabetes Biobank Study Group, Chow E, Kong APS, Tam WH, Luk AOY, Weedon MN, So WY, Chan JCN, Oram RA, Ma RCW, TRANSCEND Consortium. Type 2 diabetes pathway-specific polygenic risk scores elucidate heterogeneity in clinical presentation, disease progression and diabetic complications in 18,217 Chinese individuals with type 2 diabetes. Diabetologia 2025; 68:602-614. [PMID: 39531041 PMCID: PMC11832604 DOI: 10.1007/s00125-024-06309-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/09/2024] [Indexed: 11/16/2024]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes is a complex and heterogeneous disease and the aetiological components underlying the heterogeneity remain unclear in the Chinese and East Asian population. Therefore, we aimed to investigate whether specific pathophysiological pathways drive the clinical heterogeneity in type 2 diabetes. METHODS We employed newly developed type 2 diabetes hard-clustering and soft-clustering pathway-specific polygenic risk scores (psPRSs) to characterise individual genetic susceptibility to pathophysiological pathways implicated in type 2 diabetes in 18,217 Chinese patients from Hong Kong. The 'total' type 2 diabetes polygenic risk score (PRS) was summed by genome-wide significant type 2 diabetes signals (n=1289). We examined the associations between psPRSs and cardiometabolic profile, age of onset, two glycaemic deterioration outcomes (clinical requirement of insulin treatment, defined by two consecutive HbA1c values ≥69 mmol/mol [8.5%] more than 3 months apart during treatment with two or more oral glucose-lowering drugs, and insulin initiation), three renal (albuminuria, end-stage renal disease and chronic kidney disease) outcomes and five cardiovascular outcomes. RESULTS Although most psPRSs and total type 2 diabetes PRS were associated with an earlier and younger onset of type 2 diabetes, the psPRSs showed distinct associations with clinical outcomes. In particular, individuals with normal weight showed higher psPRSs for beta cell dysfunction and lipodystrophy than those who were overweight. The psPRSs for obesity were associated with faster progression to clinical requirement of insulin treatment (adjusted HR [95% CI] 1.09 [1.05, 1.13], p<0.0001), end-stage renal disease (1.10 [1.04, 1.16], p=0.0007) and CVD (1.10 [1.05, 1.16], p<0.0001) while the psPRSs for beta cell dysfunction were associated with reduced incident end-stage renal disease (0.90 [0.85, 0.95], p=0.0001) and heart failure (0.83 [0.73, 0.93], p=0.0011). Major findings remained significant after adjusting for a set of clinical variables. CONCLUSIONS/INTERPRETATION Beta cell dysfunction and lipodystrophy could be the driving pathological pathways in type 2 diabetes in individuals with normal weight. Genetic risks of beta cell dysfunction and obesity represent two major genetic drivers of type 2 diabetes heterogeneity in disease progression and diabetic complications, which are shared across ancestry groups. Type 2 diabetes psPRSs may help inform patient stratification according to aetiology and guide precision diabetes care.
Collapse
Affiliation(s)
- Gechang Yu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-SJTU Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Claudia H T Tam
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-SJTU Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Cadmon K P Lim
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-SJTU Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Mai Shi
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-SJTU Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Eric S H Lau
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Risa Ozaki
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Heung-Man Lee
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-SJTU Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Alex C W Ng
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-SJTU Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yong Hou
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-SJTU Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Baoqi Fan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-SJTU Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Chuiguo Huang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-SJTU Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hongjiang Wu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Aimin Yang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hoi Man Cheung
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ka Fai Lee
- Department of Medicine and Geriatrics, Kwong Wah Hospital, Hong Kong, China
| | - Shing Chung Siu
- Diabetes Centre, Tung Wah Eastern Hospital, Hong Kong, China
| | - Grace Hui
- Diabetes Centre, Tung Wah Eastern Hospital, Hong Kong, China
| | - Chiu Chi Tsang
- Diabetes and Education Centre, Alice Ho Miu Ling Nethersole Hospital, Hong Kong, China
| | | | - Jenny Y Y Leung
- Department of Medicine and Geriatrics, Ruttonjee Hospital, Hong Kong, China
| | - Elaine Y N Cheung
- Department of Medicine and Geriatrics, United Christian Hospital, Hong Kong, China
| | - Man Wo Tsang
- Department of Medicine and Geriatrics, United Christian Hospital, Hong Kong, China
| | - Grace Kam
- Department of Medicine and Geriatrics, United Christian Hospital, Hong Kong, China
| | - Ip Tim Lau
- Tseung Kwan O Hospital, Hong Kong, China
| | - June K Y Li
- Department of Medicine, Yan Chai Hospital, Hong Kong, China
| | - Vincent T F Yeung
- Centre for Diabetes Education and Management, Our Lady of Maryknoll Hospital, Hong Kong, China
| | - Emmy Lau
- Department of Medicine, Pamela Youde Nethersole Eastern Hospital, Hong Kong, China
| | - Stanley Lo
- Department of Medicine, Pamela Youde Nethersole Eastern Hospital, Hong Kong, China
| | - Samuel Fung
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong, China
| | - Yuk Lun Cheng
- Department of Medicine, Alice Ho Miu Ling Nethersole Hospital, Hong Kong, China
| | - Cheuk Chun Szeto
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Elaine Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Alice P S Kong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Wing Hung Tam
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
- CUHK Medical Centre, Hong Kong, China
| | - Andrea O Y Luk
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-SJTU Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Wing-Yee So
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-SJTU Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-SJTU Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Ronald C W Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China.
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China.
- CUHK-SJTU Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China.
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
- Diabetes Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| | | |
Collapse
|
19
|
Liu J, Fu Y, Liang P, Song Z, Li Y, Wu H. The association between novel metabolic parameters and all-cause/cardiovascular mortality in patients with metabolic syndrome is modified by age. Cardiovasc Diabetol 2025; 24:96. [PMID: 40022121 PMCID: PMC11871838 DOI: 10.1186/s12933-025-02587-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/07/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Triglyceride glucose index (TyG) serves as an effective parameter for assessing metabolic status. However, it remains uncertain whether TyG and other metabolic parameters can predict clinical outcomes in people with metabolic syndrome (MetS). We investigated the association of TyG, triglyceride glucose-waist to height ratio (TyG-WHtR), and metabolic score for insulin resistance (METS-IR) with all-cause and cardiovascular mortality in the MetS cohort and determined whether this association changes with age. METHOD Participants enrolled in the National Health and Nutrition Examination Survey (NHANES) between 2001 and 2018 were selected and categorized into two groups: younger individuals (age < 65 years) and older individuals (age ≥ 65 years). Three new metabolic indices of TyG, TyG-WHtR, and METS-IR were constructed. The weighted Cox proportional hazards model and restricted cubic spline (RCS) models were employed to evaluate the relation between three indices and mortality outcomes. The time-dependent receiver operating characteristic (ROC) curve assessed the ability of different indices to predict mortality. Sensitivity analysis was conducted to evaluate the robustness and reliability of the findings. RESULTS The study comprised a total of 8271 participants, including 5456 younger participants and 2815 older participants, and 1407 deaths were observed over a median follow-up period of 8.3 years. Compared with the first quartile (Q1), the fourth quartile's (Q4) TyG, TyG-WHtR, and METS-IR were linked to an increased risk of all-cause mortality (HR 1.63, 95% CI 1.12-2.39; HR 2.78, 95% CI 1.68-4.61; HR 1.36, 95% CI 1.12-2.02, respectively) and cardiovascular mortality (HR 2.04, 95% CI 1.15-4.90; HR 4.99, 95% CI 1.76-14.11; HR 2.69, 95% CI 1.89-8.15, respectively) in the younger group but not in the older group. The RCS results showed no significant non-linear associations between TyG, TyG-WHtR, METS-IR, and all-cause (P = 0.082; P = 0.712; P = 0.062, respectively) or cardiovascular mortality (P = 0.176; P = 0.793; P = 0.482, respectively) in the older age group. TyG-WHtR demonstrated the highest area under the curve for predicting 3-year mortality in the younger age group, with values of 0.653 for all-cause mortality and 0.688 for cardiovascular mortality. CONCLUSION Our results highlight the predictive value of TyG, TyG-WHtR, and METS-IR in the MetS population, providing new evidence for medical practice and public health.
Collapse
Affiliation(s)
- Jiajun Liu
- Shanghai University of Traditional Chinese Medicine, Shenzhen Hospital, Shenzhen, China
- Luohu District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Yihui Fu
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pengpeng Liang
- Shanghai University of Traditional Chinese Medicine, Shenzhen Hospital, Shenzhen, China
- Luohu District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Zhangxiao Song
- Shanghai University of Traditional Chinese Medicine, Shenzhen Hospital, Shenzhen, China
- Luohu District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Yue Li
- Shanghai University of Traditional Chinese Medicine, Shenzhen Hospital, Shenzhen, China.
- Luohu District Hospital of Traditional Chinese Medicine, Shenzhen, China.
| | - Hongyan Wu
- Shanghai University of Traditional Chinese Medicine, Shenzhen Hospital, Shenzhen, China.
- Luohu District Hospital of Traditional Chinese Medicine, Shenzhen, China.
| |
Collapse
|
20
|
de Carvalho GB, Brandão-Lima PN, Santos RKF, de Sousa Rocha V, Oliveira AS, da Cruz Santos B, Santos CB, Reis AR, Maia CSC, de Oliveira E Silva AM, Pires LV. Relationship Between the Single Nucleotide Polymorphism A35C in the Cu/Zn-Superoxide Dismutase-1 Gene and Glycemic Control in Individuals with Type 2 Diabetes Mellitus. Biol Trace Elem Res 2025:10.1007/s12011-025-04555-8. [PMID: 39994114 DOI: 10.1007/s12011-025-04555-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 02/15/2025] [Indexed: 02/26/2025]
Abstract
Hyperglycemia in type 2 diabetes mellitus (T2DM) increases oxidative stress. Furthermore, the presence of the single nucleotide polymorphism A35C (SNP A35C) in Cu/Zn-superoxide dismutase1 (SOD1) gene is closely related to this increase in oxidative stress and the development and progression of T2DM and its complications. This study aimed to evaluate the association between SNP A35C (rs2234694) genotypes and glycemic control in T2DM individuals. A total of 110 individuals were evaluated for anthropometric parameters, body composition, glycemic metabolism markers (fasting serum glucose, %HbA1c, insulin, C-peptide, and HOMA-IR, -%B, -%S), and SOD activity. Individuals were grouped according to SNP A35C genotypes. Variables of interest were assessed according to groups. The T-test for independent samples or the Mann-Whitney U test was used to analyze the differences in continuous variables between groups, and the chi-square test was performed for categorical variables. A binary logistic regression model was constructed, with p < 0.05 considered significant. Overweight was found in 81.8% of individuals with T2DM. Individuals with the AC genotype for SNP A35C had higher levels of fasting serum glucose (p = 0.018) and lower values of HOMA-%B (p = 0.044). The presence of the variant allele was positively associated with higher values of fasting serum glucose (OR: 11.340; 95%IC 1.173-109.649; p = 0.036) and HOMA-IR (OR: 9.987; 95%IC 1.127-88.506; p = 0.039). Individuals with the AC genotype of SNP A35C had poorer glycemic control than individuals with the AA genotype, and the presence of the variant allele was associated with poor glycemic control in T2DM individuals.
Collapse
Affiliation(s)
- Gabrielli Barbosa de Carvalho
- Department of Nutrition, Nutrition Sciences Post-Graduate Program, Federal University of Sergipe, Marcelo Deda Chagas Avenue, S/n - Jardim Rosa Elze, São Cristóvão, Sergipe, 49107-230, Brazil
- Nutritional Biochemistry Laboratory, Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Paula Nascimento Brandão-Lima
- Nutritional Biochemistry Laboratory, Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
- Post-Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Ramara Kadija Fonseca Santos
- Department of Nutrition, Nutrition Sciences Post-Graduate Program, Federal University of Sergipe, Marcelo Deda Chagas Avenue, S/n - Jardim Rosa Elze, São Cristóvão, Sergipe, 49107-230, Brazil
- Nutritional Biochemistry Laboratory, Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | | | - Alan Santos Oliveira
- Post-Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Beatriz da Cruz Santos
- Department of Nutrition, Nutrition Sciences Post-Graduate Program, Federal University of Sergipe, Marcelo Deda Chagas Avenue, S/n - Jardim Rosa Elze, São Cristóvão, Sergipe, 49107-230, Brazil
- Nutritional Biochemistry Laboratory, Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Cynthia Batista Santos
- Department of Nutrition, Nutrition Sciences Post-Graduate Program, Federal University of Sergipe, Marcelo Deda Chagas Avenue, S/n - Jardim Rosa Elze, São Cristóvão, Sergipe, 49107-230, Brazil
- Nutritional Biochemistry Laboratory, Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Aline Rocha Reis
- Department of Nutrition, Nutrition Sciences Post-Graduate Program, Federal University of Sergipe, Marcelo Deda Chagas Avenue, S/n - Jardim Rosa Elze, São Cristóvão, Sergipe, 49107-230, Brazil
- Nutritional Biochemistry Laboratory, Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | | | - Ana Mara de Oliveira E Silva
- Department of Nutrition, Nutrition Sciences Post-Graduate Program, Federal University of Sergipe, Marcelo Deda Chagas Avenue, S/n - Jardim Rosa Elze, São Cristóvão, Sergipe, 49107-230, Brazil
- Post-Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Liliane Viana Pires
- Department of Nutrition, Nutrition Sciences Post-Graduate Program, Federal University of Sergipe, Marcelo Deda Chagas Avenue, S/n - Jardim Rosa Elze, São Cristóvão, Sergipe, 49107-230, Brazil.
- Nutritional Biochemistry Laboratory, Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil.
| |
Collapse
|
21
|
Athira AS, Abhijith B, Sruthi PK, Ragavamenon AC, Lankalapalli RS, Reshma MV. Lyophilized ash gourd ( Benincasa hispida (Thunb.) Cogn.) juice alleviates diet-induced prediabetes in a rat model. Food Funct 2025; 16:1534-1549. [PMID: 39903217 DOI: 10.1039/d4fo05327c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Prediabetes is characterized by elevated blood sugar levels, indicating an increased risk of developing diabetes. This study evaluated the effects of ash gourd (AG), a tropical fruit from the Cucurbitaceae family, on prediabetes, as well as its phytochemical composition. A prediabetic rat model was developed in Sprague-Dawley (SD) rats by administering a high fat diet (HFD) for 16 weeks. This model exhibited reduced pancreatic function, heightened insulin resistance, and decreased insulin sensitivity compared to a standard diet group, leading to hyperglycemia and dyslipidemia, hallmarks of prediabetic conditions. Histological analysis of hepatic tissue revealed macro- and microvesicular fat accumulation and inflammatory changes, supporting these findings. This study highlights the utility of HFD-induced SD rats as a model for prediabetic conditions. Following this, lyophilized ash gourd juice (LAGJ) powder was administered to the prediabetic rat model to assess its potential for reversing prediabetic conditions. LAGJ administration resulted in a significant reduction in fasting blood sugar (FBS) levels, glucose intolerance, and insulin resistance. Additionally, LAGJ significantly mitigated fatty liver changes compared to the prediabetic untreated control (PUC) group. Histological examination of liver tissue in the LAGJ treated group showed a typical architecture similar to that of the normal control group. These findings indicate that LAGJ could be a promising intervention for individuals with prediabetes who are at risk of developing type 2 diabetes and fatty liver disease. Phytochemical analysis of AG pulp revealed the presence of stigmasterol, stigmasteryl β-glucoside, and 6'-O-palmitoyl stigmasteryl β-glucoside.
Collapse
Affiliation(s)
- A S Athira
- Agro-processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, Kerala, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Balan Abhijith
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
| | - P K Sruthi
- Department of Biochemistry, Amala Cancer Research Centre, Amala Nagar, Thrissur-680555, Kerala, India
| | - Achuthan C Ragavamenon
- Department of Biochemistry, Amala Cancer Research Centre, Amala Nagar, Thrissur-680555, Kerala, India
| | - Ravi S Lankalapalli
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
| | - M V Reshma
- Agro-processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, Kerala, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
22
|
Weber KS, Schlesinger S, Goletzke J, Straßburger K, Zaharia OP, Trenkamp S, Wagner R, Lieb W, Buyken AE, Roden M, Herder C. Associations of carbohydrate quality and cardiovascular risk factors vary among diabetes subtypes. Cardiovasc Diabetol 2025; 24:53. [PMID: 39915806 PMCID: PMC11804081 DOI: 10.1186/s12933-025-02580-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/04/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Assess the intake of carbohydrate quality and their association with cardiovascular risk factors among diabetes subtypes. METHODS Participants of the German Diabetes Study (GDS) (recent-onset diabetes (n = 487) and 5-years thereafter (n = 209)) were allocated into severe autoimmune diabetes (SAID, 35%), severe insulin-deficient diabetes (SIDD, 3%), severe insulin-resistant diabetes (SIRD, 5%), mild obesity-related diabetes (MOD, 28%), and mild age-related diabetes (MARD, 29%). Dietary glycemic index (GI), glycemic load (GL), and intake of higher- (≥ 55) and low-GI (< 55) foods, dietary fiber, and total sugar were derived from a validated food frequency questionnaire and cross-sectionally associated with cardiovascular risk factors (blood lipids, subclinical inflammation, blood pressure, fatty liver index) using multivariable linear regression analysis for subtypes with prevalences ≥ 10%. RESULTS Intake of carbohydrate quality parameters was broadly comparable between the subtypes. Among SAID higher total sugar intake was associated with lower HDL-cholesterol (ß (95% CI) relative change per 1 SD increment: - 3.4% (- 6.7; - 0.1)). No clear associations were seen among MOD. Among MARD, a higher dietary GL and higher-GI carbohydrate intake were associated with higher serum triglycerides (10.9% (2.4; 20.1), 12.4% (3.9; 21.5)) and fatty liver index (absolute change: 0.18 (0.06; 0.31), 0.17 (0.05; 0.28)) and lower HDL-cholesterol (- 4.1% (- 7.6; - 0.4), - 4.4% (- 7.8; - 0.8)), whilst higher intake of low-GI carbohydrates and dietary fiber were associated with lower high-sensitivity C-reactive protein (- 16.0% (- 25.7; - 5.1), - 13.9% (- 24.2; - 2.2)). CONCLUSIONS Associations of carbohydrate quality parameters with blood lipids, subclinical inflammation, and fatty liver index differed between diabetes subtypes. However, evidence is too preliminary to derive subtype-specific recommendations. TRIAL REGISTRATION Clinicaltrials.gov: NCT01055093.
Collapse
Affiliation(s)
- Katharina S Weber
- Institute of Epidemiology, Kiel University, Niemannsweg 11, 24105, Kiel, Germany.
| | - Sabrina Schlesinger
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Munich-Neuherberg, Germany
| | - Janina Goletzke
- Faculty of Natural Sciences, Institute of Nutrition, Consumption and Health, Paderborn University, Paderborn, Germany
| | - Klaus Straßburger
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Munich-Neuherberg, Germany
| | - Oana-Patricia Zaharia
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Munich-Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sandra Trenkamp
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Munich-Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Robert Wagner
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Munich-Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Wolfgang Lieb
- Institute of Epidemiology, Kiel University, Niemannsweg 11, 24105, Kiel, Germany
| | - Anette E Buyken
- Faculty of Natural Sciences, Institute of Nutrition, Consumption and Health, Paderborn University, Paderborn, Germany
| | - Michael Roden
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Munich-Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christian Herder
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Munich-Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
23
|
Tokgöz S, Boss M, Jansen TJ, Meijer R, Frielink C, van Bon AC, Tack CJ, de Galan BE, Gotthardt M. Activation of the HPA Axis Does Not Explain Nonresponsiveness to GLP-1R Agonist Treatment in Individuals With Type 2 Diabetes. Diabetes 2025; 74:212-222. [PMID: 39561332 PMCID: PMC11755685 DOI: 10.2337/db24-0463] [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: 06/04/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
ARTICLE HIGHLIGHTS It is unclear why some individuals with type 2 diabetes are unresponsive to treatment with glucagon-like peptide 1 receptor (GLP-1R) agonists, but hypothalamic-pituitary-adrenal (HPA) axis activation could play a role. We used [68Ga]Ga-NODAGA-exendin-4 positron emission tomography/computed tomography to compare pituitary GLP-1R expression between responders and nonresponders to treatment with GLP-1R agonists. Pituitary GLP-1R expression and HPA axis activation did not differ between responders and nonresponders to GLP-1R agonist treatment. In addition, pituitary radiolabeled exendin uptake was markedly higher in men than in women. Further study is required to explain treatment differences and understand sex differences in pituitary radiolabeled exendin uptake.
Collapse
Affiliation(s)
- Sevilay Tokgöz
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marti Boss
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
| | - Theodorus J.P. Jansen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rick Meijer
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cathelijne Frielink
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Arianne C. van Bon
- Department of Internal Medicine, Rijnstate Hospital, Arnhem, the Netherlands
| | - Cees J. Tack
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Bastiaan E. de Galan
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| | - Martin Gotthardt
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
24
|
Stobbe AY, de Klerk ES, van Wilpe R, Kievit AJ, Choi KF, Preckel B, Hollmann MW, Hermanides J, van Stijn MFM, Hulst AH. Study protocol of the PRINCESS trial-PReoperative INtermittent fasting versus CarbohydratE loading to reduce inSulin resiStance versus standard of care in orthopaedic patients: a randomised controlled trial. BMJ Open 2025; 15:e087260. [PMID: 39842917 PMCID: PMC11956279 DOI: 10.1136/bmjopen-2024-087260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
INTRODUCTION Surgical trauma induces a metabolic stress response, resulting in reduced insulin sensitivity and hyperglycaemia. Postoperative insulin resistance (IR) is associated with postoperative complications, and extended preoperative fasting may further aggravate the postoperative metabolic stress response. Nutritional strategies, such as carbohydrate loading (CHL), have been successfully used to attenuate postoperative IR. Recent evidence suggests that time-restricted feeding (TRF), a form of intermittent fasting, improves IR in the general population, even after a short period of TRF. We hypothesise that TRF, as well as CHL, improve postoperative IR. METHODS AND ANALYSIS This open-label, single-centre, randomised controlled trial will compare the effect of short-term preoperative TRF, CHL and standard preoperative fasting on perioperative IR. A total of 75 orthopaedic patients presenting for elective intermediate to major surgery at a Dutch academic hospital will be randomly assigned to a control group (standard preoperative fasting), a TRF group or a CHL group. The primary outcome is postoperative IR, based on the updated homeostasis model assessment of IR, on the first day after surgery. Statistical analyses are performed using Student's t-tests or Mann-Whitney U tests. ETHICS AND DISSEMINATION The local medical ethics committee of the Amsterdam UMC, the Netherlands, approved the trial protocol in January 2023 (NL81556.018.22). No publication restrictions apply, and the results of the study will be disseminated through a peer-reviewed journal. TRIAL REGISTRATION NUMBER NCT05760339.
Collapse
Affiliation(s)
- Ayla Y Stobbe
- Department of Anaesthesiology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Department of Endocrinology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Quality of Care, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam, The Netherlands
| | - Eline S de Klerk
- Department of Anaesthesiology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Quality of Care, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Robert van Wilpe
- Department of Anaesthesiology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Arthur J Kievit
- Department of Orthopaedic Surgery and Sports Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Musculoskeletal Health, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Kee Fong Choi
- Department of Anaesthesiology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Benedikt Preckel
- Department of Anaesthesiology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Quality of Care, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Markus W Hollmann
- Department of Anaesthesiology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Quality of Care, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Jeroen Hermanides
- Department of Anaesthesiology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Quality of Care, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam, The Netherlands
| | - Mireille F M van Stijn
- Department of Anaesthesiology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Quality of Care, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam, The Netherlands
| | - Abraham H Hulst
- Department of Anaesthesiology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Quality of Care, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Lu B, Li P, Crouse AB, Grimes T, Might M, Ovalle F, Shalev A. Data-driven Cluster Analysis Reveals Increased Risk for Severe Insulin-deficient Diabetes in Black/African Americans. J Clin Endocrinol Metab 2025; 110:387-395. [PMID: 39078946 PMCID: PMC11747757 DOI: 10.1210/clinem/dgae516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/27/2024] [Accepted: 07/23/2024] [Indexed: 10/05/2024]
Abstract
CONTEXT Diabetes is a heterogenic disease and distinct clusters have emerged, but the implications for diverse populations have remained understudied. OBJECTIVE Apply cluster analysis to a diverse diabetes cohort in the US Deep South. DESIGN Retrospective hierarchical cluster analysis of electronic health records from 89 875 patients diagnosed with diabetes between January 1, 2010, and December 31, 2019, at the Kirklin Clinic of the University of Alabama at Birmingham, an ambulatory referral center. PATIENTS Adult patients with International Classification of Diseases diabetes codes were selected based on available data for 6 established clustering parameters (glutamic acid decarboxylase autoantibody; hemoglobin A1c; body mass index; diagnosis age; HOMA2-B; HOMA2-IR); ∼42% were Black/African American. MAIN OUTCOME MEASURE(S) Diabetes subtypes and their associated characteristics in a diverse adult population based on clustering analysis. We hypothesized that racial background would affect the distribution of subtypes. Outcome and hypothesis were formulated prior to data collection. RESULTS Diabetes cluster distribution was significantly different in Black/African Americans compared to Whites (P < .001). Black/African Americans were more likely to have severe insulin-deficient diabetes (OR, 1.83; 95% CI, 1.36-2.45; P < .001), associated with more serious metabolic perturbations and a higher risk for complications (OR, 1.42; 95% CI, 1.06-1.90; P = .020). Surprisingly, Black/African Americans specifically had more severe impairment of β-cell function (homoeostatic model assessment 2 estimates of β-cell function, C-peptide) (P < .001) but not being more obese or insulin resistant. CONCLUSION Racial background greatly influences diabetes cluster distribution and Black/African Americans are more frequently and more severely affected by severe insulin-deficient diabetes. This may further help explain the disparity in outcomes and have implications for treatment choice.
Collapse
Affiliation(s)
- Brian Lu
- Comprehensive Diabetes Center, Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Peng Li
- School of Nursing, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Andrew B Crouse
- Hugh Kaul Precision Medicine Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Tiffany Grimes
- Comprehensive Diabetes Center, Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Matthew Might
- Hugh Kaul Precision Medicine Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Fernando Ovalle
- Comprehensive Diabetes Center, Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Anath Shalev
- Comprehensive Diabetes Center, Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
26
|
Mertens J, Roosens L, Braspenning R, Vandebeeck J, Francque S, De Block C. The 13C Glucose Breath Test Accurately Identifies Insulin Resistance in People With Type 1 Diabetes. J Clin Endocrinol Metab 2025; 110:e432-e442. [PMID: 38487831 DOI: 10.1210/clinem/dgae175] [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: 10/06/2023] [Indexed: 01/22/2025]
Abstract
OBJECTIVE This study investigated whether the delta-over-baseline of exhaled 13CO2 (Δ13CO2), generated from a 13C glucose breath test (13C-GBT), measured insulin resistance (IR) in people with type 1 diabetes, using the hyperinsulinemic-euglycemic clamp (HEC) as a reference method. The secondary objective was to compare the 13C-GBT with the estimated glucose disposal rate (eGDR). METHODS A 40 mU/m2/min HEC and 2 separate 13C-GBTs (euglycemic with insulin bolus and hyperglycemic without bolus) were consecutively performed in 44 adults with type 1 diabetes with varying body compositions. eGDR was calculated based on hemoglobin A1c (HbA1c), presence of hypertension, and waist circumference. RESULTS The mean glucose disposal rate (M-value) was 5.9 ± 3.1 mg/kg/min and mean euglycemic Δ13CO2 was 6.4 ± 2.1 δ‰, while median eGDR was 5.9 [4.3-9.8] mg/kg/min. The hyperglycemic Δ13CO2 did not correlate with the M-value, while the euglycemic Δ13CO2 and the M-value correlated strongly (r = 0.74, P < .001). The correlation between M-value and eGDR was more moderate (Spearman's rho = 0.63, P < .001). Linear regression showed an association between Δ13CO2 and M-value, adjusted for age, sex, and HbA1c ]adjusted R² = 0.52, B = 1.16, 95% confidence interval (CI) .80-1.52, P < .001]. The area under the receiver-operator characteristics curve for Δ13CO2 to identify subjects with IR (M-value < 4.9 mg/kg/min) was 0.81 (95% CI .68-.94, P < .001). The optimal cut-off for Δ13CO2 to identify subjects with IR was ≤ 5.8 δ‰. CONCLUSION Under euglycemic conditions, the 13C-GBT accurately identified individuals with type 1 diabetes and concurrent IR, suggesting its potential as a valuable noninvasive index. Clinical Trial Identifier: NCT04623320.
Collapse
Affiliation(s)
- Jonathan Mertens
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, 2650 Edegem, Belgium
- Department of Gastroenterology & Hepatology, Antwerp University Hospital, 2650 Edegem, Belgium
- Laboratory of Experimental Medicine and Paediatrics, University of Antwerp, 2610 Wilrijk, Belgium
| | - Laurence Roosens
- Laboratory of Clinical Chemistry, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Rie Braspenning
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Joeri Vandebeeck
- Laboratory of Clinical Chemistry, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Sven Francque
- Department of Gastroenterology & Hepatology, Antwerp University Hospital, 2650 Edegem, Belgium
- Laboratory of Experimental Medicine and Paediatrics, University of Antwerp, 2610 Wilrijk, Belgium
| | - Christophe De Block
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, 2650 Edegem, Belgium
- Laboratory of Experimental Medicine and Paediatrics, University of Antwerp, 2610 Wilrijk, Belgium
| |
Collapse
|
27
|
Kim Y, Rimmer JH, Lai B, Oster R, Cowan R, Young HJ, Fisher G, Kim Y, Giannone J, Wilroy JD. Cardiometabolic Health Intervention Using Music and Exercise (CHIME) Delivered via Telehealth to Wheelchair Users: Protocol for a Randomized Controlled Trial. JMIR Res Protoc 2025; 14:e57423. [PMID: 39814364 PMCID: PMC11780300 DOI: 10.2196/57423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/11/2024] [Accepted: 12/17/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Wheelchair users live predominantly sedentary lifestyles and have a substantially higher risk for cardiometabolic disease and mortality compared to people without disabilities. Exercise training has been found to be effective in improving cardiometabolic health (CMH) outcomes among people without disabilities, but research on wheelchair users is limited and of poor quality. OBJECTIVE The primary aim of this study is to examine the immediate and sustained effects of a 24-week, telehealth, movement-to-music cardiovascular (M2M-C) exercise program on core indicators of CMH among adult wheelchair users compared to an active control group. The secondary aim is to explore the beneficial effects of M2M-C exercises on cardiovascular capacity, physical activity, and quality of life. Intervention components include tailored exercises and remote performance monitoring, delivered via live videoconference training by a telecoach and asynchronous videos. METHODS This study's design is a parallel-arm randomized controlled trial enrolling 132 physically inactive adult wheelchair users with poor cardiometabolic profiles. The M2M-C intervention group involves 24 weeks of virtual live and monitored home exercise training (3×/wk, 15-40 min/session), followed by a 12-week maintenance period where participants have access to an online media library of exercise videos. The control group involves 36 weeks of self-guided exercise through access to a media library of exercise videos, including videos for range of motion, muscle strength, and balance. The primary outcomes are cardiometabolic indicators of health, and assessors are blinded. RESULTS Recruitment procedures started in January 2024 with the first participant enrolled on March 18, 2024. All data are anticipated to be collected by November 2027, and the main results of the trial are anticipated to be published by February 2028. Secondary analyses of data will be subsequently published. A total of 16 participants have been recruited as of paper submission. CONCLUSIONS The knowledge obtained from this trial will provide evidence to inform exercise prescriptions aimed at improving CMH among adult wheelchair users. TRIAL REGISTRATION ClinicalTrials.gov NCT05606432; https://clinicaltrials.gov/study/NCT05606432. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/57423.
Collapse
Affiliation(s)
- Yumi Kim
- Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham, Birmingham, AL, United States
| | - James H Rimmer
- Department of Occupational Therapy, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Byron Lai
- Division of Pediatric Rehabilitation, Children's Hospital of Alabama, Birmingham, AL, United States
| | - Robert Oster
- Department of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rachel Cowan
- Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Hui-Ju Young
- Department of Occupational Therapy, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Gordon Fisher
- Department of Human Studies, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Younguk Kim
- Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham, Birmingham, AL, United States
| | - John Giannone
- Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jereme D Wilroy
- Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
28
|
Si Y, Zhang H, Han X, Liu W, Tu Y, Ma X, Yu H, Bao Y. Nomogram for Predicting Suboptimal Weight Loss at Three Years after Roux-en-Y Gastric Bypass Surgery in Chinese Patients with Obesity and Type 2 Diabetes. Obes Facts 2025; 18:157-168. [PMID: 39813999 PMCID: PMC12017750 DOI: 10.1159/000542923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/29/2024] [Indexed: 01/18/2025] Open
Abstract
INTRODUCTION Strategies to address suboptimal weight loss after Roux-en-Y gastric bypass (RYGB) can be developed if at-risk patients are identified in advance. This study aimed to build a pre-surgery prediction nomogram for early prediction of insufficient weight loss (IWL) or weight regain (WR) after bariatric surgery in Chinese patients. METHODS In this retrospective study, 187 patients with obesity and type 2 diabetes who underwent laparoscopic RYGB were followed yearly for 3 years. Suboptimal weight loss included IWL and WR. IWL was defined as a total weight loss percentage of <25% at 1 year postoperatively, and WR was defined as a maximum weight loss percentage of >20% at 3 years postoperatively. Multivariate logistic regression was performed to identify independent predictors and to establish a nomogram to predict the occurrence of suboptimal weight loss. RESULTS Multivariate logistic regression revealed that male sex (OR 4.268, 95% CI: 1.413-12.890), body mass index (OR 0.816, 95% CI: 0.705-0.946), and glycated hemoglobin (OR 1.493, 95% CI: 1.049-2.126) were independent predictors of IWL/WR. The AUC value of the nomogram constructed from the above three factors was 0.781. The Hosmer-Lemeshow test showed that the model had a good fit (p = 0.143). The calibration curve of the nomogram is close to an ideal diagonal line. Furthermore, the decision curve analysis demonstrated the good net benefits of the model. CONCLUSIONS A nomogram based on pre-surgery factors was developed to predict postoperative IWL/WR. This provides a convenient and useful tool for predicting suboptimal weight loss before surgery.
Collapse
Affiliation(s)
- Yiming Si
- 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, China
| | - Hongwei Zhang
- Department of General Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaodong Han
- Department of General Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weijie Liu
- Department of General Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinfang Tu
- 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, China
| | - Xiaojing Ma
- 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, China
| | - Haoyong Yu
- 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, 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, China
| |
Collapse
|
29
|
McIntyre CC, Lyday RG, Su Y, Nicklas B, Simpson SL, Deep G, Macauley SL, Hugenschmidt CE. Insulin resistance, cognition, and functional brain network topology in older adults with obesity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.26.625552. [PMID: 39829939 PMCID: PMC11741235 DOI: 10.1101/2024.11.26.625552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Objective Cross-sectional data from a sample of older adults with obesity was used to determine how peripheral and neuronal insulin resistance (IR) relate to executive function and functional brain network topology. Methods Older adults (n=71) with obesity but without type 2 diabetes were included. Peripheral IR was quantified by HOMA2-IR. Neuronal IR was quantified according to a proposed neuron-derived exosome-based method (NDE-IR). An executive function composite score, summed scores to the Auditory Verbal Learning Test (AVLT) trials 1-5, and functional brain networks generated from resting-state functional magnetic resonance imaging were outcomes in analyses. We used general linear models and a novel regression framework for brain network analysis to identify relationships between IR measures and brain-related outcomes. Results HOMA2-IR, but not NDE-IR, was negatively associated with executive function. Neither IR measure was associated with AVLT score. Peripheral IR was also related to hippocampal network topology in participants who had undergone functional neuroimaging. Neither peripheral nor neuronal IR were significantly related to network topology of the central executive network. Conclusions Cognitive and functional imaging effects were observed from HOMA2-IR, but not NDE-IR. The hippocampus may be particularly vulnerable to effects of peripheral IR.
Collapse
Affiliation(s)
- Clayton C. McIntyre
- Neuroscience Graduate Program, Wake Forest Graduate School of Arts and Sciences, Winston-Salem, NC, USA
| | - Robert G. Lyday
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Yixin Su
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Barbara Nicklas
- Department of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Sean L. Simpson
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Gagan Deep
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Shannon L. Macauley
- Department of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Physiology, University of Kentucky, Lexington, KY USA
| | - Christina E. Hugenschmidt
- Department of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
30
|
Wang K, Qian Q, Bian C, Sheng P, Zhu L, Teng S, An X. Risk Evaluation of Progression of Proteinuria and Renal Decline Based on a Novel Subgroup Classification in Chinese Patients with Type 2 Diabetes. Diabetes Ther 2025; 16:89-102. [PMID: 39556310 PMCID: PMC11759728 DOI: 10.1007/s13300-024-01667-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/24/2024] [Indexed: 11/19/2024] Open
Abstract
INTRODUCTION Type 2 diabetes mellitus (T2DM) is a highly heterogeneous disease with a varying risk of complications. The recent novel subgroup classification using cluster analysis contributed to the risk evaluation of diabetic complications. However, whether the subgroup classification strategy could be adopted to predict the risk of onset and progression of diabetic kidney disease (DKD) in Chinese individuals with T2DM remains to be elucidated. METHODS In this retrospective study, 612 Chinese patients with T2DM were enrolled, and the median follow-up time was 3.5 years. The T2DM subgroups were categorized by a two-step cluster analysis based on five parameters, including age at onset of diabetes, body mass index (BMI), glycosylated hemoglobin (HbA1c), homeostasis model assessment 2 of insulin resistance (HOMA2-IR), and homeostasis model assessment 2 of β-cell function (HOMA2-β). Clinical characteristics across subgroups were compared using t-tests and chi-square tests. Furthermore, multivariate logistic regression models were adopted to assess the risk of albuminuria progression and renal function decline among different subgroups. RESULTS The cohort was categorized into four groups: severe insulin-deficient diabetes (SIDD), with 146 patients (23.9%); mild insulin resistance (MIRD), with 81 patients (13.2%); moderate glycemic control diabetes (MGCD), with 211 patients (34.5%); and moderate weight insulin deficiency diabetes (MWIDD), with 174 patients (28.4%). The MIRD group exhibited an increased risk of progression from non-albuminuria to albuminuria as compared with the MWIDD group, with an adjusted odds ratio (OR) and 95% confidence interval (CI) of 2.92 (1.06, 8.04). The SIDD group had a higher risk of progression from micro-albuminuria to macro-albuminuria as compared with the MGCD group, with an adjusted OR and 95% CI of 3.39 (1.01, 11.41). There was no significant difference in the glomerular filtration rate (GFR) decline among all groups. CONCLUSION The present study offered the first evidence for risk evaluation of the development of DKD in the novel cluster-based T2DM Chinese subgroups. It suggested that the MIRD subgroup had a higher risk of DKD onset than the MWIDD subgroup. Meanwhile, the SIDD subgroup showed a higher risk of progression of albuminuria than the MGCD subgroup. This novel classification system could be effective in predicting the risk of DKD in Chinese patients with T2DM, which could facilitate the implementation of personalized therapeutic strategies. TRIAL REGISTRATION This study was registered in the Chinese Clinical Trial Registry (ChiCTR2300077183).
Collapse
Affiliation(s)
- Kai Wang
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Han-Zhong Road, Nanjing, 210029, China
| | - Qi Qian
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Han-Zhong Road, Nanjing, 210029, China
| | - Chencheng Bian
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Han-Zhong Road, Nanjing, 210029, China
| | - Pei Sheng
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Han-Zhong Road, Nanjing, 210029, China
| | - Lin Zhu
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Han-Zhong Road, Nanjing, 210029, China.
- Department of Physical Examination Center, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Han-Zhong Road, Nanjing, 210029, China.
| | - Shichao Teng
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Han-Zhong Road, Nanjing, 210029, China.
- Department of Geriatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Han-Zhong Road, Nanjing, 210029, China.
| | - Xiaofei An
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Han-Zhong Road, Nanjing, 210029, China.
| |
Collapse
|
31
|
Menezes-Santos M, Santos BDC, Santos RKF, da Costa SSL, Dos Santos SH, E Silva AMDO, Rocha VDS, Pires LV. Copper Deficiency Associated with Glycemic Control in Individuals with Type 2 Diabetes Mellitus. Biol Trace Elem Res 2025; 203:119-126. [PMID: 38639883 DOI: 10.1007/s12011-024-04185-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/11/2024] [Indexed: 04/20/2024]
Abstract
Adequate copper (Cu) status has been associated with improved glycemic control, partly because of its role in reducing oxidative stress through superoxide dismutase (SOD) activity. Thus, the aim was to investigate the relationship between plasma Cu concentration and markers associated with glycemic control in individuals with type 2 diabetes mellitus (T2DM). This observational and cross-sectional study was conducted in individuals with T2DM of both sexes, aged between 19 and 59 years. Plasma Cu levels were analyzed using inductively coupled plasma optical emission spectrometry (ICP-OES). Fasting glucose and insulin concentrations, C-peptide levels, SOD activity, and glycated hemoglobin (%HbA1c) were measured. Homeostatic model assessments (HOMA%B, HOMA%S, and HOMA-IR) were also performed. Additionally, %body fat and waist circumference were measured, and body mass index was calculated. Participants were categorized based on their plasma Cu concentrations (< 70 µg/dL and ≥ 70 µg/dL). The associations between variables were analyzed using chi-squared or Fisher's test and binary logistic regression models. Statistical significance was set at P < 0.05. Of the 97 participants (74.2% women), 85.5% had Cu deficiency. Cu-deficient individuals showed elevated C-peptide concentrations and HOMA%B values compared to those with adequate Cu levels (2.8 ng/mL vs. 1.8 ng/mL, P = 0.011; and 71.4 vs. 31.0, P = 0.003), respectively. Cu deficiency was associated with insulin resistance (P = 0.044) and decreased likelihood of exceeding the target serum glucose level (OR = 0.147, P = 0.013). However, no significant association was found between SOD activity and plasma Cu concentration. Consequently, Cu deficiency was linked to improved glycemic control, although it was not associated with the other markers.
Collapse
Affiliation(s)
- Matheus Menezes-Santos
- Nutritional Biochemistry Laboratory, Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Beatriz da Cruz Santos
- Nutritional Biochemistry Laboratory, Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
- Nutrition Sciences Post-Graduation Program, Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, Marcelo Deda Avenue, S/n - Jardim Rosa Elze, São Cristóvão, Sergipe, ZIP: 49107-230, Brazil
| | - Ramara Kadija Fonseca Santos
- Nutritional Biochemistry Laboratory, Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
- Health Sciences Post-Graduation Program, Department of Medicine, Center for Biological and Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | | | - Samir Hipólito Dos Santos
- Chemistry Post-Graduation Program, Chemistry Institute, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Ana Mara de Oliveira E Silva
- Nutrition Sciences Post-Graduation Program, Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, Marcelo Deda Avenue, S/n - Jardim Rosa Elze, São Cristóvão, Sergipe, ZIP: 49107-230, Brazil
- Health Sciences Post-Graduation Program, Department of Medicine, Center for Biological and Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | | | - Liliane Viana Pires
- Nutritional Biochemistry Laboratory, Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil.
- Nutrition Sciences Post-Graduation Program, Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, Marcelo Deda Avenue, S/n - Jardim Rosa Elze, São Cristóvão, Sergipe, ZIP: 49107-230, Brazil.
| |
Collapse
|
32
|
Liu K, Li T, Zhong P, Zhu Z, Guo X, Liu R, Xiong R, Huang W, Wang W. Retinal and Choroidal Phenotypes Across Novel Subtypes of Type 2 Diabetes Mellitus. Am J Ophthalmol 2025; 269:205-215. [PMID: 39237050 DOI: 10.1016/j.ajo.2024.08.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024]
Abstract
PURPOSE To investigate longitudinal changes in choroidal thickness (CT) and ganglion cell-inner plexiform layer thickness (GC-IPLT) across distinct phenotypes of type 2 diabetes mellitus (T2DM) patients. DESIGN Prospective cohort study. METHODS T2DM patients were categorized into 5 groups (SAID, SIDD, SIRD, MOD, and MARD) using K-means clustering based on β-cell function and insulin resistance. Swept-source optical coherence tomography measured baseline and 4-year follow-up CT and GC-IPLT. Linear mixed-effects models assessed absolute and relative changes in CT and GC-IPLT across subtypes. RESULTS Over a median 4.11-year follow-up, CT and GC-IPLT decreased significantly across all groups. Choroidal thinning rates were most pronounced in SIDD (-6.5 ± 0.53 µm/year and -3.5 ± 0.24%/year) and SAID (-6.27 ± 0.8 µm/year and -3.19 ± 0.37%/year), while MARD showed the slowest thinning rates (-3.63 ± 0.34 µm/year and -1.98 ± 0.25%/year). SIRD exhibited the greatest GC-IPLT loss (-0.66 ± 0.05 µm/year and -0.91 ± 0.07%/year), with the least in SIDD (-0.36 ± 0.05 µm/year and -0.49 ± 0.07%/year), all statistically significant (all P < 0.001). Adjusted for confounding variables, SIDD and SAID groups showed faster CT thinning than MARD [-2.57 µm/year (95% CI: -4.16 to -0.97; P = 0.002) and -2.89 µm/year (95% CI: -4.12 to -1.66; P < 0.001), respectively]. GC-IPLT thinning was notably accelerated in SIRD versus MARD, but slowed in SIDD relative to MARD [differences of -0.16 µm/year (95% CI: -0.3 to -0.03; P = 0.015) and 0.15 µm/year (95% CI: 0.03 to 0.27; P = 0.015), respectively]. CONCLUSIONS Microvascular damage in the choroid is associated with SIDD patients, whereas early signs of retinal neurodegeneration are evident in SIRD patients. All these changes may precede the onset of DR.
Collapse
Affiliation(s)
- Kaiqun Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Guangzhou, China
| | - Ting Li
- Department of Rheumatology and Immunology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Study Center for Obstetrics and Gynecology, The Third Affiliated Hospital (T.L.), Guangzhou Medical University, Guangzhou, China
| | - Pingting Zhong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Guangzhou, China
| | - Ziyu Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Guangzhou, China
| | - Xiao Guo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Guangzhou, China
| | - Riqian Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Guangzhou, China
| | - Ruilin Xiong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Guangzhou, China
| | - Wenyong Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Guangzhou, China.
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Guangzhou, China; Hainan Eye Hospital and Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Haikou, China.
| |
Collapse
|
33
|
Teng L, Luo L, Sun Y, Wang W, Dong Z, Cao X, Ye J, Zhong B. Comparisons of Post-Load Glucose at Different Time Points for Identifying High Risks of MASLD Progression. Nutrients 2024; 17:152. [PMID: 39796589 PMCID: PMC11723153 DOI: 10.3390/nu17010152] [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: 11/19/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Background: The 1-h post-load plasma glucose was proposed to replace the current OGTT criteria for diagnosing prediabetes/diabetes. However, it remains unclear whether it is superior in identifying progressive metabolic dysfunction-associated steatotic liver disease (MASLD), and thus we aimed to clarify this issue. Methods: Consecutive Asian participants (non-MASLD, n = 1049; MASLD, n = 1165) were retrospectively enrolled between June 2012 and June 2024. CT was used to quantify liver steatosis, while the serum liver fibrotic marker was used to evaluate liver fibrosis. Results: Compared with those with normal levels of both 1-h post-glucose (1hPG) and 2-h post-glucose (2hPG), patients with MASLD showed a significant positive association between elevated 1hPG levels and moderate to severe liver steatosis (odds ratio [OR] = 2.19, 95% confidence interval [CI]: 1.13-4.25, p = 0.02]. Elevated levels of both 1hPG and 2hPG were associated with an increased risk of liver injury (OR = 2.03, 95% CI: 1.44-2.86, p < 0.001). Elevated 2hPG levels with or without elevated 1hPG levels were associated with liver fibrosis (OR = 1.99, 95% CI: 1.15-3.45, p < 0.001; OR = 2.72, 95% CI: 1.79-4.11, p < 0.001, respectively). Additionally, either 1hPG or 2hPG levels were associated with atherosclerosis, revealing significant dose-dependent associations between glucose status and atherosclerosis risk (OR = 2.77, 95% CI: 1.55-4.96, p < 0.001 for elevated 1hPG; OR = 2.98, 95% CI = 1.54-5.78, p = 0.001 for elevated 2hPG; OR = 2.41, 95% CI = 1.38-4.21, p = 0.001 for elevated levels of both 1hPG and 2hPG). The areas under the ROC for predicting steatosis, liver injury, liver fibrosis, and atherosclerosis were 0.64, 0.58, 0.58, and 0.64 for elevated 1hPG (all p < 0.05) and 0.50, 0.60, 0.56, and 0.62 for elevated 2hPG (all p < 0.05), respectively. Conclusions: These findings underscore the necessity for clinicians to acknowledge that the screening and management of MALSD requires the monitoring of 1hPG levels.
Collapse
Affiliation(s)
- Long Teng
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou 510080, China; (L.T.); (L.L.)
| | - Ling Luo
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou 510080, China; (L.T.); (L.L.)
| | - Yanhong Sun
- Department of Clinical Laboratory, The First Affiliated Hospital, Sun Yat-sen University, No. 183 Huangpu East Road, Huangpu District, Guangzhou 510080, China;
| | - Wei Wang
- Department of Ultrasound, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou 510080, China;
| | - Zhi Dong
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou 510080, China;
| | - Xiaopei Cao
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou 510080, China;
| | - Junzhao Ye
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou 510080, China; (L.T.); (L.L.)
| | - Bihui Zhong
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou 510080, China; (L.T.); (L.L.)
| |
Collapse
|
34
|
Maanaoui M, Debillon F, Lenain R, Defrance F, Vantyghem MC, Van Triempont M, Provôt F, Chetboun M, Kerr-Conte J, Hamroun A, Frimat M, Raverdy V, Pattou F, Hazzan M, Glowacki F. Association between HOMA2 based beta-cell function or insulin resistance and long-term outcomes in kidney transplant recipients with type 2 diabetes. Sci Rep 2024; 14:32081. [PMID: 39738741 PMCID: PMC11685385 DOI: 10.1038/s41598-024-83817-2] [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: 01/23/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025] Open
Abstract
Type 2 diabetes (T2D) is a common comorbidity in kidney transplant recipients, representing a significant proportion of the candidate pool. Post-kidney transplantation management of T2D remains challenging, leading to inferior long-term outcomes compared to non-diabetic recipients. This study aimed to assess the association between Homeostatic Model Assessment 2 (HOMA2) derived insulin resistance and beta-cell function on kidney graft outcomes in T2D kidney transplant recipients. We conducted a monocentric retrospective study at the University Hospital of Lille, including all consecutive adult T2D patients who underwent kidney transplantation between January 2007 and December 2018. HOMA2 indexes (HOMA2 IR for insulin resistance and HOMA2 B for beta-cell function) were calculated at one-year post-transplantation. Primary endpoint was graft survival and secondary endpoints were death-censored graft survival, patient survival and post-transplant metabolic control. Among 1620 kidney transplant recipients, 138 patients with T2D were included, with a median follow-up of 1837 days [1283-2726]. HOMA2 IR was significantly associated with an increased risk of kidney graft failure or death (HR per unit = 1.11 (1.02-1.21). Beta-cell function was not associated with graft prognosis, but decreased beta-cell function was associated with poorer metabolic control over time. This study highlights the significance of insulin resistance as a potential determinant of long-term outcomes in T2D kidney transplant recipients.
Collapse
Affiliation(s)
- Mehdi Maanaoui
- Department of Nephrology, CHU Lille, University of Lille, Lille, 59000, France.
- U1190 Translational Research for Diabetes, INSERM, Institut Pasteur de Lille, Université Lille, Lille, France.
- Service de Nephrologie, Hôpital Huriez, CHRU de Lille, Lille, 59037, France.
| | - Florence Debillon
- Department of Nephrology, CHU Lille, University of Lille, Lille, 59000, France
| | - Rémi Lenain
- Department of Nephrology, CHU Lille, University of Lille, Lille, 59000, France
| | - Frédérique Defrance
- U1190 Translational Research for Diabetes, INSERM, Institut Pasteur de Lille, Université Lille, Lille, France
- Department of Endocrinology, Diabetology, and Metabolism, CHU Lille, University of Lille, Lille, 59000, France
| | - Marie-Christine Vantyghem
- U1190 Translational Research for Diabetes, INSERM, Institut Pasteur de Lille, Université Lille, Lille, France
- Department of Endocrinology, Diabetology, and Metabolism, CHU Lille, University of Lille, Lille, 59000, France
| | | | - François Provôt
- Department of Nephrology, CHU Lille, University of Lille, Lille, 59000, France
| | - Mikael Chetboun
- U1190 Translational Research for Diabetes, INSERM, Institut Pasteur de Lille, Université Lille, Lille, France
- Department of General and Endocrine Surgery, CHU Lille, University of Lille, Lille, 59000, France
| | - Julie Kerr-Conte
- U1190 Translational Research for Diabetes, INSERM, Institut Pasteur de Lille, Université Lille, Lille, France
| | - Aghiles Hamroun
- Santé Publique, Epidémiologie - UMR 1167 Ridage, Institut Pasteur de Lille, Univ Lille, Chu Lille, Lille, France
| | - Marie Frimat
- Department of Nephrology, CHU Lille, University of Lille, Lille, 59000, France
- University of Lille, INSERM, UMR 1167, RID-AGE, Lille, France
| | - Violeta Raverdy
- U1190 Translational Research for Diabetes, INSERM, Institut Pasteur de Lille, Université Lille, Lille, France
| | - François Pattou
- U1190 Translational Research for Diabetes, INSERM, Institut Pasteur de Lille, Université Lille, Lille, France
- Department of General and Endocrine Surgery, CHU Lille, University of Lille, Lille, 59000, France
| | - Marc Hazzan
- Department of Nephrology, CHU Lille, University of Lille, Lille, 59000, France
| | - François Glowacki
- Department of Nephrology, CHU Lille, University of Lille, Lille, 59000, France
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, 59000, France
| |
Collapse
|
35
|
Rosendo-Silva D, Lopes E, Monteiro-Alfredo T, Falcão-Pires I, Eickhoff H, Viana S, Reis F, Pires AS, Abrantes AM, Botelho MF, Seiça R, Matafome P. The adipose tissue melanocortin 3 receptor is targeted by ghrelin and leptin and may be a therapeutic target in obesity. Mol Cell Endocrinol 2024; 594:112367. [PMID: 39293775 DOI: 10.1016/j.mce.2024.112367] [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: 07/24/2024] [Revised: 09/03/2024] [Accepted: 09/07/2024] [Indexed: 09/20/2024]
Abstract
OBJECTIVE Obesity is linked to perturbations in energy balance mechanisms, including ghrelin and leptin actions at the hypothalamic circuitry of neuropeptide Y (NPY) and melanocortin. However, information about the regulation of this system in the periphery is still scarce. Our objective was to study the regulation of the NPY/melanocortin system in the adipose tissue (AT) and evaluate its therapeutic potential for obesity and type 2 diabetes. METHODS The expression of the NPY/melanocortin receptors' levels was assessed in the visceral AT of individuals with obesity and altered metabolism. Protein levels of these receptors were evaluated in cultured adipocytes incubated with ghrelin (30 and 100 ng/mL) and leptin (1 and 10 nM) and in the AT of an animal model with a mutation in the leptin receptor (ZSF1 rat), to understand their regulation by leptin and ghrelin. The vertical sleeve gastrectomy animal model was used to evaluate the putative therapeutic potential of the NPY/melanocortin system. RESULTS In this study, we unravelled that leptin (1 nM and 10 nM) selectively reduced the levels of NPY5R and MC3R but no other NPYR/MCRs in cultured adipocytes. In turn, acylated ghrelin (100 ng/mL) significantly increased NPY1R, but the inhibition of its receptor also abrogates MC3R levels. However, in the Lepr-deficient ZSF1 rat, both NPY5R and MC3R levels were reduced, along with other NPYRs and MCRs, suggesting that leptin resistance negatively affects NPY and melanocortin signalling. In human adipose tissue, we found a downregulation of genes encoding the NPY and melanocortin receptors in the visceral AT of individuals with obesity and insulin resistance, being correlated with genes regulating metabolic activity. Additionally, diabetic obese rats submitted to vertical sleeve gastrectomy showed increased levels of NPY, melanocortin, ghrelin, and leptin receptors in the AT, including MC3R, suggesting it may constitute a therapeutic target in obesity. CONCLUSIONS Our results suggest that the AT NPY/melanocortin system, particularly the MC3R, may be involved in the neuroendocrine regulation of adipocyte metabolism. Altogether, our work shows MC3R is under the control of the ghrelin/leptin duo, is reduced in patients with obesity and prediabetes, and may constitute a therapeutic target in obesity.
Collapse
Affiliation(s)
- Daniela Rosendo-Silva
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.
| | - Eduardo Lopes
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Tamaeh Monteiro-Alfredo
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Inês Falcão-Pires
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Hans Eickhoff
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal
| | - Sofia Viana
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; University of Coimbra, iCBR and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, Portugal; Polytechnic University of Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal
| | - Flávio Reis
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; University of Coimbra, iCBR and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, Portugal
| | - Ana Salomé Pires
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; University of Coimbra, iCBR Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, Faculty of Medicine, Coimbra, Portugal
| | - Ana Margarida Abrantes
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; University of Coimbra, iCBR Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, Faculty of Medicine, Coimbra, Portugal
| | - Maria Filomena Botelho
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; University of Coimbra, iCBR Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, Faculty of Medicine, Coimbra, Portugal
| | - Raquel Seiça
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal
| | - Paulo Matafome
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; Polytechnic University of Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal.
| |
Collapse
|
36
|
Lépine G, Mariotti F, Tremblay-Franco M, Courrent M, Verny MA, David J, Mathé V, Jame P, Anchisi A, Lefranc-Millot C, Perreau C, Guérin-Deremaux L, Chollet C, Castelli F, Chu-Van E, Huneau JF, Rémond D, Pickering G, Fouillet H, Polakof S. Increasing plant protein in the diet induces changes in the plasma metabolome that may be beneficial for metabolic health. A randomized crossover study in males. Clin Nutr 2024; 43:146-157. [PMID: 39454458 DOI: 10.1016/j.clnu.2024.10.009] [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: 07/12/2024] [Revised: 09/27/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND & AIM Dietary shifts replacing animal protein (AP) with plant protein (PP) sources have been associated with lowering cardiometabolic risk (CMR), but underlying mechanisms are poorly characterized. This nutritional intervention aims to characterize the metabolic changes induced by diets containing different proportions of AP and PP sources in males at CMR. DESIGN This study is a 4-week, crossover, randomized, controlled-feeding trial in which 19 males with CMR followed two diets providing either 36 % for the control diet (CON-D) or 64 % for the flexitarian diet (FLEX-D) of total protein intake from PP sources. Plasma nontargeted metabolomes (LC-MS method) were measured in the fasted state and after a high-fat challenge meal at the end of each intervention arm. Lipogenesis and protein synthesis fluxes, flow-mediated dilatation (FMD) and gluco-lipidic responses were assessed after the challenge meal. Data were analyzed with mixed models, and univariate and multivariate models for metabolomics data. RESULTS In both arms CMR improved with time, with decreased body weight (-0.9 %), insulin resistant (-34 %, HOMA-IR, Homeostatic Model Assessment for Insulin Resistance) and low-density lipoproteins (LDL)-cholesterol (-11 %). Diet had no effect on FMD or metabolic fluxes, but a trend (0.05 CONCLUSIONS Despite little changes in risk factors after 4 wk, this study evidenced subtle metabolic adaptations in amino acids and lipid metabolism and gut microbiota activity occurring after higher PP source intake that may be beneficial to CMR. CLINICALTRIALS GOV STUDY IDENTIFIER NCT04236518. CLINICAL TRIAL REGISTRY NCT04236518 on ClinicalTrials.gov.
Collapse
Affiliation(s)
- Gaïa Lépine
- Université Clermont Auvergne, INRAE, UNH, 63000, Clermont-Ferrand, France; Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 91120, Palaiseau, France
| | - François Mariotti
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 91120, Palaiseau, France
| | - Marie Tremblay-Franco
- Toxalim - Research Centre in Food Toxicology, Toulouse University, INRAE, ENVT, INP-Purpan, UT3, F-31300, Toulouse, France; MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, 31077, France
| | | | - Marie-Anne Verny
- Université Clermont Auvergne, INRAE, UNH, 63000, Clermont-Ferrand, France
| | - Jérémie David
- Université Clermont Auvergne, INRAE, UNH, 63000, Clermont-Ferrand, France
| | - Véronique Mathé
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 91120, Palaiseau, France
| | - Patrick Jame
- Universite Claude Bernard Lyon 1, CNRS, ISA, UMR5280, 5 rue de la Doua, F-69100 Villeurbanne, France
| | - Anthony Anchisi
- Universite Claude Bernard Lyon 1, CNRS, ISA, UMR5280, 5 rue de la Doua, F-69100 Villeurbanne, France
| | | | | | | | - Céline Chollet
- CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Université Paris-Saclay, 91191 Gif-sur-Yvette, France
| | - Florence Castelli
- CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Université Paris-Saclay, 91191 Gif-sur-Yvette, France
| | - Emeline Chu-Van
- CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Université Paris-Saclay, 91191 Gif-sur-Yvette, France
| | - Jean-François Huneau
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 91120, Palaiseau, France
| | - Didier Rémond
- Université Clermont Auvergne, INRAE, UNH, 63000, Clermont-Ferrand, France
| | | | - Hélène Fouillet
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 91120, Palaiseau, France.
| | - Sergio Polakof
- Université Clermont Auvergne, INRAE, UNH, 63000, Clermont-Ferrand, France.
| |
Collapse
|
37
|
Lin B, Coleman RL, Bragg F, Maddaloni E, Holman RR, Adler AI. Younger-onset compared with later-onset type 2 diabetes: an analysis of the UK Prospective Diabetes Study (UKPDS) with up to 30 years of follow-up (UKPDS 92). Lancet Diabetes Endocrinol 2024; 12:904-914. [PMID: 39461360 DOI: 10.1016/s2213-8587(24)00242-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/19/2024] [Accepted: 07/30/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND Younger-onset type 2 diabetes is associated with accelerated complications. We assessed whether complications and mortality rates differed for younger age compared with older age at diagnosis over 30 years of follow-up. METHODS In this study, we used data from the UKPDS, collected between 1977 and 2007, of participants aged 25-65 years with newly diagnosed type 2 diabetes with younger-onset (younger than 40 years) or later-onset (40 years or older), and without diabetes autoantibodies. We analysed standardised mortality ratios (SMR) using UK general population data, and incidence rates of prespecified outcomes by 10-year age intervals at diagnosis. FINDINGS Of 4550 participants testing negative to all measured autoantibodies, 429 (9·4%) had younger-onset type 2 diabetes. 2704 (59·4%) were male, and mean HbA1c was 76 mmol/mol (SD 24·6). The median follow-up was 17·5 years (IQR 12·7-20·8). SMR for younger-onset type 2 diabetes was higher (3·72 [95% CI 2·98-4·64]) compared with later-onset type 2 diabetes (1·54 [1·47-1·61]). The incidence rate was higher for all outcomes in later-onset type 2 diabetes, except for microvascular disease (younger-onset 14·5 (11·9-17·7) vs later-onset 12·1 (11·3-13·0) per 1000 person-years). However, at any given age, the 5-year incidence of any diabetes-related endpoint, all-cause mortality, microvascular disease, and myocardial infarction was higher with younger age at diagnosis. Annual mean HbA1c was higher in the first 20 years in younger-onset compared with later-onset type 2 diabetes. Among participants randomised to intensive versus conventional glycaemic control, we observed no interactions by subgroup of younger-onset versus later-onset type 2 diabetes for any outcome. INTERPRETATION The risk of dying relative to the general population is even greater for people diagnosed with type 2 diabetes at younger ages. The increased risk of complications and poorer glycaemic control in younger-onset type 2 diabetes calls for the development of services to identify and manage these individuals. FUNDING National Institute of Health and Care Research's Biomedical Research Centre.
Collapse
Affiliation(s)
- Beryl Lin
- Faculty of Medicine, University of Sydney, Sydney, NSW, Australia; Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Ruth L Coleman
- Faculty of Medicine, University of Sydney, Sydney, NSW, Australia
| | - Fiona Bragg
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK; Health Data Research UK Oxford, University of Oxford, Oxford, UK
| | - Ernesto Maddaloni
- Experimental Medicine Department, Sapienza University of Rome, Rome, Italy
| | - Rury R Holman
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Amanda I Adler
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
38
|
Ku CW, Zheng RT, Tan HY, Lim JYQ, Chen LW, Cheung YB, Godfrey KM, Chan JKY, Yap F, Lek N, Loy SL. Early continuous glucose monitoring-derived glycemic patterns are associated with subsequent insulin resistance and gestational diabetes mellitus development during pregnancy. Diabetol Metab Syndr 2024; 16:271. [PMID: 39538266 PMCID: PMC11562738 DOI: 10.1186/s13098-024-01508-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) and insulin resistance (IR) increase the risk of adverse pregnancy outcomes. We aimed to examine the relationship of interstitial glucose assessed by continuous glucose monitoring (CGM) at early gestation, and the subsequent development of IR and GDM, and to determine 24-h interstitial glucose centile distributions in women with normal (non-IR and non-GDM) and suboptimal glycemic status (IR and/or GDM). METHODS CGM measurements were taken for 3-10 days at 18-24 weeks' gestation, followed by fasting serum insulin and oral glucose tolerance testing at 24-28 weeks' gestation. IR and GDM were determined by the updated Homeostasis Model Assessment of IR score of ≥ 1.22 and 2013 World Health Organization criteria, respectively. Risks of IR and GDM were estimated using modified Poisson models, and hourly interstitial glucose centiles determined using Generalized Additive Models for Location, Scale and Shape. RESULTS This prospective cohort study involved 167 pregnant women in Singapore, with a mean age of 31.7 years, body mass index of 22.9 kg/m2, and gestation of 20.3 weeks. 25% of women exhibited IR and 18% developed GDM. After confounders adjustment, women with suboptimal glycemic control, indicated by higher mean daily glucose (risk ratio 1.42; 95% confidence interval 1.16, 1.73), glucose management indicator (1.08; 1.03, 1.12), and J-index (1.04; 1.02, 1.06), as well as those with greater glycemic variability, indicated by higher standard deviation (1.69; 1.37, 2.09), coefficient of variation (1.03; 1.00, 1.06), and mean amplitude of glycemic excursions (1.4; 1.14, 1.35) derived from CGM in early gestation were associated with higher risks of developing IR in later gestation. These associations were similarly observed for the development of GDM. Centile curves showed that, compared to those with normal glycemic status, women with suboptimal glycemic status had higher glucose levels, with greater fluctuations throughout 24 h. CONCLUSIONS In pregnant women who subsequently developed IR and GDM, interstitial glucose levels assessed by CGM were elevated and varied greatly. This supports the potential use of CGM to screen for glycemic changes early in pregnancy.
Collapse
Affiliation(s)
- Chee Wai Ku
- Department of Reproductive Medicine, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore, 229899, Singapore
- Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Ruther Teo Zheng
- Endocrinology Service, Department of Pediatrics, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore, 229899, Singapore
| | - Hong Ying Tan
- Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore, 117597, Singapore
| | - Jamie Yong Qi Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Experimental Medicine Building, Singapore, 636921, Singapore
| | - Ling-Wei Chen
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, No. 17 Xu-Zhou Road, Taipei, 10055, Taiwan
- Master of Public Health Program, College of Public Health, National Taiwan University, No. 17 Xu-Zhou Road, Taipei, 10055, Taiwan
| | - Yin Bun Cheung
- Program in Health Services & Systems Research, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
- Tampere Centre for Child, Adolescent and Maternal Health Research, Tampere University, 33014, Tampere, Finland
| | - Keith M Godfrey
- Medical Research Council Lifecourse Epidemiology Centre, University of Southampton, Southampton, SO16 6YD, UK
- National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, SO16 6YD, UK
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore, 229899, Singapore
- Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Fabian Yap
- Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
- Endocrinology Service, Department of Pediatrics, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore, 229899, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Experimental Medicine Building, Singapore, 636921, Singapore
| | - Ngee Lek
- Endocrinology Service, Department of Pediatrics, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore, 229899, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore, 117597, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Experimental Medicine Building, Singapore, 636921, Singapore
| | - See Ling Loy
- Department of Reproductive Medicine, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore, 229899, Singapore.
- Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore.
| |
Collapse
|
39
|
Nguyen MLT, Bui KC, Ngo TH, Nguyen TS, Nham PLT, Pham C, Hoang TM, Huynh QT, Tang TT, Dang TC, Dang TL, Nguyen LT, Can VM. Early impaired insulin tolerance among Vietnamese diabetes with or without dyslipidemia. Medicine (Baltimore) 2024; 103:e40212. [PMID: 39495996 PMCID: PMC11537581 DOI: 10.1097/md.0000000000040212] [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: 08/05/2024] [Accepted: 10/04/2024] [Indexed: 11/06/2024] Open
Abstract
This study aims to evaluate impaired insulin tolerance among Vietnamese diabetes with or without dyslipidemia. Diabetes mellitus (DM) remains the serious global health and social burden that has increased over the past few decades. It progresses silently to vascular injury and disability of injured vascular-perfused tissues/organs. Insulin intolerance and dyslipidemia exacerbate and accelerate the implications of DM. Thus, early detection and more evidence of early insulin intolerance and dyslipidemia is needed for proactive management. This cross-sectional descriptive study recruited 100 healthy control (HC) and 297 DM patients in Military Hospital 103 from 2021 to 2023. Patients with DM were subgrouped into lipid metabolism disorder (LMD, n = 98) and non-LMD (NLMD, n = 99). The biochemists' serum levels were measured automatically and the accuracy of the test result was strictly controlled. Insulin tolerance indices (HOMA2-IR, HOMA2-%S and HOMA2-%B) were compared between HC, DM with or without dyslipidemia as well as correlated with lipid ingredients (total Cholesterol, triglyceride, LDL-C and HDL-C). Among DM patients, HOMA2-IR was significantly high and HOMA2-%S and HOMA2-%B were significantly low. HOMA2-IR was higher and HOMA2-%S and HOMA2-%B were lower in DM with LMD than in DM without LMD. In addition, HOMA2-IR was positively correlated with serum cholesterol, triglyceride and LDL-C concentration, and negatively correlated to HDL-C concentration. In contrast, HOMA2-%S and HOMA2-%B was negatively correlated with serum cholesterol, triglyceride and LDL-C, and positively correlated with HDL-C. Impaired insulin intolerance occurred in early stage of DM, and more serious among DM with LMD, compared to DM with NLMD.
Collapse
Affiliation(s)
- Mai Ly Thi Nguyen
- Department of Biochemistry, Military Hospital 103, Vietnam Military Medical University Hanoi, Vietnam
| | - Khac Cuong Bui
- Laboratory Animal Research Center, Vietnam Military Medical University, Hanoi, Vietnam
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
- Vietnamese-German Center for Medical Research (VG-CARE), 108 Military Central Hospital, Hanoi, Vietnam
| | - Thu Hang Ngo
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| | - Tai Suc Nguyen
- Laboratory Animal Research Center, Vietnam Military Medical University, Hanoi, Vietnam
| | - Phuong Linh Thi Nham
- Laboratory Animal Research Center, Vietnam Military Medical University, Hanoi, Vietnam
| | - Chi Pham
- Laboratory Animal Research Center, Vietnam Military Medical University, Hanoi, Vietnam
| | - Thi Minh Hoang
- Department of Biochemistry, Military Hospital 103, Vietnam Military Medical University Hanoi, Vietnam
| | - Quang Thuan Huynh
- Department of Biochemistry, Military Hospital 103, Vietnam Military Medical University Hanoi, Vietnam
| | - Thanh Thuy Tang
- Department of Biochemistry, Military Hospital 103, Vietnam Military Medical University Hanoi, Vietnam
| | - Thanh Chung Dang
- Department of Pathology, Military Hospital 103, Vietnam Military Medical University Hanoi, Vietnam
| | - Thuy Linh Dang
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| | - Linh Toan Nguyen
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
- Vietnamese-German Center for Medical Research (VG-CARE), 108 Military Central Hospital, Hanoi, Vietnam
| | - Van Mao Can
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| |
Collapse
|
40
|
Fiorenza M, Onslev J, Henríquez-Olguín C, Persson KW, Hesselager SA, Jensen TE, Wojtaszewski JFP, Hostrup M, Bangsbo J. Reducing the mitochondrial oxidative burden alleviates lipid-induced muscle insulin resistance in humans. SCIENCE ADVANCES 2024; 10:eadq4461. [PMID: 39475607 PMCID: PMC11524190 DOI: 10.1126/sciadv.adq4461] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024]
Abstract
Preclinical models suggest mitochondria-derived oxidative stress as an underlying cause of insulin resistance. However, it remains unknown whether this pathophysiological mechanism is conserved in humans. Here, we used an invasive in vivo mechanistic approach to interrogate muscle insulin action while selectively manipulating the mitochondrial redox state in humans. To this end, we conducted insulin clamp studies combining intravenous infusion of a lipid overload with intake of a mitochondria-targeted antioxidant (mitoquinone). Under lipid overload, selective modulation of mitochondrial redox state by mitoquinone enhanced insulin-stimulated glucose uptake in skeletal muscle. Mechanistically, mitoquinone did not affect canonical insulin signaling but augmented insulin-stimulated glucose transporter type 4 (GLUT4) translocation while reducing the mitochondrial oxidative burden under lipid oversupply. Complementary ex vivo studies in human muscle fibers exposed to high intracellular lipid levels revealed that mitoquinone improves features of mitochondrial bioenergetics, including diminished mitochondrial H2O2 emission. These findings provide translational and mechanistic evidence implicating mitochondrial oxidants in the development of lipid-induced muscle insulin resistance in humans.
Collapse
Affiliation(s)
- Matteo Fiorenza
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Johan Onslev
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Carlos Henríquez-Olguín
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
- Exercise Science Laboratory, Faculty of Medicine, Universidad Finis Terrae, Santiago 1509, Chile
| | - Kaspar W. Persson
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Sofie A. Hesselager
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Thomas E. Jensen
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jørgen F. P. Wojtaszewski
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Morten Hostrup
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jens Bangsbo
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| |
Collapse
|
41
|
Wang J, Gao B, Wang J, Liu W, Yuan W, Chai Y, Ma J, Ma Y, Kong G, Liu M. Identifying subtypes of type 2 diabetes mellitus based on real-world electronic medical record data in China. Diabetes Res Clin Pract 2024; 217:111872. [PMID: 39332534 DOI: 10.1016/j.diabres.2024.111872] [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: 06/21/2024] [Revised: 09/02/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
AIMS To replicate the European subtypes of type 2 diabetes mellitus (T2DM) in the Chinese diabetes population and investigate the risk of complications in different subtypes. METHODS A diabetes cohort using real-world patient data was constructed, and clustering was employed to subgroup the T2DM patients. Kaplan-Meier analysis and the Cox models were used to analyze the association between diabetes subtypes and the risk of complications. RESULTS A total of 2,652 T2DM patients with complete clustering data were extracted. Among them, 466 (17.57 %) were classified as severe insulin-deficient diabetes (SIDD), 502 (18.93 %) as severe insulin-resistant diabetes (SIRD), 672 (25.34 %) as mild obesity-related diabetes (MOD), and 1,012 (38.16 %) as mild age-related diabetes (MARD). The risk of chronic kidney disease (CKD) and diabetic retinopathy (DR) were different in the four subtypes. Compared with MARD, SIRD had a higher risk of CKD (HR 2.40 [1.16, 4.96]), and SIDD had a higher risk of DR (HR 2.16 [1.11, 4.20]). The risk of stroke and coronary events had no difference. CONCLUSIONS The European T2DM subtypes can be replicated in the Chinese diabetes population. The risk of CKD and DR varied among different subtypes, indicating that proper interventions can be taken to prevent specific complications in different subtypes.
Collapse
Affiliation(s)
- Jiayu Wang
- National Institute of Health Data Science, Peking University, Beijing 100191, China; Advanced Institute of Information Technology, Peking University, Hangzhou 314201, Zhejiang, China; Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China
| | - Bixia Gao
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Jinwei Wang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Wenwen Liu
- National Institute of Health Data Science, Peking University, Beijing 100191, China
| | - Weijia Yuan
- Department of Computer Application and Management, Chinese PLA General Hospital, Beijing 100039, China
| | - Yangfan Chai
- Peking University Chongqing Research Institute of Big Data, Chongqing 100871, China
| | - Jun Ma
- National Institute of Health Data Science, Peking University, Beijing 100191, China
| | - Yangyang Ma
- Department of Computer Application and Management, Chinese PLA General Hospital, Beijing 100039, China
| | - Guilan Kong
- National Institute of Health Data Science, Peking University, Beijing 100191, China; Advanced Institute of Information Technology, Peking University, Hangzhou 314201, Zhejiang, China; Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China.
| | - Minchao Liu
- Department of Computer Application and Management, Chinese PLA General Hospital, Beijing 100039, China.
| |
Collapse
|
42
|
Palmer ER, Morales-Muñoz I, Perry BI, Marwaha S, Warwick E, Rogers JC, Upthegrove R. Trajectories of Inflammation in Youth and Risk of Mental and Cardiometabolic Disorders in Adulthood. JAMA Psychiatry 2024; 81:1130-1137. [PMID: 39167392 PMCID: PMC11339695 DOI: 10.1001/jamapsychiatry.2024.2193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 06/06/2024] [Indexed: 08/23/2024]
Abstract
Importance Research suggests that low-grade, nonresolving inflammation may predate adult mental and physical illness. However, evidence to date is largely cross-sectional or focuses on single disorder outcomes. Objectives To examine trajectories of inflammation as measured by C-reactive protein (CRP) levels in a large sample of children and adolescents, and to explore associations between different identified trajectories and mental and related cardiometabolic health outcomes in early adulthood. Design, Setting, and Participants In a longitudinal cohort study using data from the large UK-based Avon Longitudinal Study of Parents and Children (ALSPAC), latent class growth analysis (LCGA) was used to explore different trajectories of inflammation, with logistic regression exploring association with mental and physical health outcomes. Participants with measurable CRP data and associated mental and cardiometabolic health outcomes recorded were included in the analysis. Data analysis was performed from May 1, 2023, to March 30, 2024. Exposures Inflammation was assessed via CRP levels at ages 9, 15, and 17 years. LCGA was used to identify different trajectories of inflammation. Main Outcomes and Measures Outcomes assessed at age 24 years included psychotic disorders, depressive disorders, anxiety disorders, hypomania, and, as a measure of insulin resistance, Homeostasis Model Assessment (HOMA2) score. Results A total of 6556 participants (3303 [50.4%] female) were included. Three classes of inflammation were identified: persistently low CRP levels (reference class, n = 6109); persistently raised CRP levels, peaking at age 9 years (early peak, n = 197); and persistently raised CRP levels, peaking at age 17 years (late peak, n = 250). Participants in the early peak group were associated with a higher risk of psychotic disorder (odds ratio [OR], 4.60; 95% CI, 1.81-11.70; P = .008), a higher risk of severe depression (OR, 4.37; 95% CI, 1.64-11.63; P = .02), and higher HOMA2 scores (β = 0.05; 95% CI, 0.01-0.62, P = .04) compared with participants with persistently low CRP. The late peak group was not associated with any outcomes at age 24 years. Conclusions and Relevance Low-grade systemic inflammation peaking in midchildhood was associated with specific mental and cardiometabolic disorders in young adulthood. These findings suggest that low-grade persistent inflammation in early life may be an important shared common factor for mental-physical comorbidity and so could be relevant to future efforts of patient stratification and risk profiling.
Collapse
Affiliation(s)
- Edward R. Palmer
- Institute for Mental Health, School of Psychology, University of Birmingham, Birmingham, United Kingdom
- Early Intervention Service, Birmingham Women’s and Children’s NHS Trust, Birmingham, United Kingdom
| | - Isabel Morales-Muñoz
- Institute for Mental Health, School of Psychology, University of Birmingham, Birmingham, United Kingdom
| | - Benjamin I. Perry
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, United Kingdom
| | - Steven Marwaha
- Institute for Mental Health, School of Psychology, University of Birmingham, Birmingham, United Kingdom
- The Barberry National Centre for Mental Health, Birmingham and Solihull Mental Health NHS Trust, Birmingham, United Kingdom
| | - Ella Warwick
- Institute for Mental Health, School of Psychology, University of Birmingham, Birmingham, United Kingdom
| | - Jack C. Rogers
- Institute for Mental Health, School of Psychology, University of Birmingham, Birmingham, United Kingdom
| | - Rachel Upthegrove
- Institute for Mental Health, School of Psychology, University of Birmingham, Birmingham, United Kingdom
- Early Intervention Service, Birmingham Women’s and Children’s NHS Trust, Birmingham, United Kingdom
| |
Collapse
|
43
|
Cree JME, Brennan NM, Poppitt SD, Miles-Chan JL. The Effect of the Oral Contraceptive Pill on Acute Glycaemic Response to an Oral Glucose Bolus in Healthy Young Women: A Randomised Crossover Study. Nutrients 2024; 16:3490. [PMID: 39458485 PMCID: PMC11510745 DOI: 10.3390/nu16203490] [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: 09/11/2024] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objective: The oral contraceptive pill (OCP) is widely used by women worldwide, yet the influence of the OCP on carbohydrate metabolism remains under-investigated, with existing studies being few and largely cross-sectional. The study objective was to assess, for the first time, the effect of the combined OCP on postprandial glycaemic response to an oral glucose bolus, using a randomised crossover design. Methods: The effect of a combined monophasic OCP phase on glucose homeostasis and metabolic profile was investigated in 21 healthy young women, who were regular users of either androgenic or anti-androgenic OCP formulations. Plasma glycaemic markers (glucose, insulin and C-peptide) were assessed prior to a 60 g glucose drink (fasting) and for a further 4 h postprandially; once during the "active" (hormone-containing) pill phase and once during the "inactive" (hormone-free) pill phase of the OCP usage cycle. Results: Despite no change in fasting values, in androgenic pill users, postprandial glucose and insulin responses to an oral glucose bolus were ~100% and ~50% greater, respectively, during the active versus inactive phase. In contrast, in anti-androgenic pill users there was no significant change in response between the two OCP usage cycle phases. Conclusions: These findings highlight an acute, but potentially detrimental, influence of the combined OCP on glucose homeostasis, particularly in users of formulations containing androgenic progestogens. Given the high global prevalence of OCP use and increasingly common prolonged active pill regimens, which may continue for months, years or even decades, potential cumulative effects of such changes on metabolic risk demand further investigation.
Collapse
Affiliation(s)
- Julia M. E. Cree
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand (S.D.P.)
| | - Niamh M. Brennan
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand (S.D.P.)
| | - Sally D. Poppitt
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand (S.D.P.)
- High Value Nutrition, National Science Challenge, Auckland 1023, New Zealand
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand
| | - Jennifer L. Miles-Chan
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand (S.D.P.)
- High Value Nutrition, National Science Challenge, Auckland 1023, New Zealand
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand
| |
Collapse
|
44
|
Magodoro IM, Aluoch A, Claggett B, Nyirenda MJ, Siedner MJ, Wilkinson KA, Wilkinson RJ, Ntusi NAB. Association Between Mycobacterium tuberculosis Sensitization and Insulin Resistance Among US Adults Screened for Type 2 Diabetes Mellitus. Open Forum Infect Dis 2024; 11:ofae568. [PMID: 39469603 PMCID: PMC11518572 DOI: 10.1093/ofid/ofae568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 10/06/2024] [Indexed: 10/30/2024] Open
Abstract
Background Type 2 diabetes mellitus (T2DM) may be a long-term sequela of infection with Mycobacterium tuberculosis (Mtb) by mechanisms that remain to be fully explained. We evaluated the association between Mtb sensitization and T2DM and, via mediation analysis, the extent to which it is mediated by insulin resistance and/or β-cell failure. Methods Adults were assessed for T2DM by fasting plasma glucose, 2-hour oral glucose tolerance testing, and hemoglobin A1c; β-cell dysfunction and insulin resistance by homoeostasis model assessment 2; and Mtb sensitization by tuberculin skin testing. Associations between Mtb sensitization and T2DM were modeled with probit regression and decomposed into indirect effects of β-cell dysfunction and insulin resistance. Analyses were adjusted for sociodemographic, behavioral, and clinical characteristics. Results We included 1843 adults. Individuals with Mtb sensitization were older than those without Mtb (median [IQR], 54 [39-64] vs 47 [33-62] years). As compared with being uninfected, Mtb sensitization was associated with T2DM (adjusted absolute risk difference, 9.34% [95% CI, 2.38%-15.0%]; P < .001) and increased insulin resistance (adjusted median difference, 0.16 [95% CI, .03-.29]; P = .014) but not β-cell dysfunction (adjusted median difference, -3.1 [95% CI, -10.4 to 4.3]; P = .42). In mediation analyses, insulin resistance mediated 18.3% (95% CI, 3.29%-36.0%; P = .020) of the total effect of the association between Mtb sensitization and T2DM. Conclusions Definitive prospective studies examining incident T2DM following tuberculosis are warranted. Notwithstanding, our findings suggest that exposure to Mtb may be a novel risk factor for T2DM, likely driven by an increase in insulin resistance.
Collapse
Affiliation(s)
- Itai M Magodoro
- Department of Medicine, University of Cape Town, Observatory, Republic of South Africa
| | - Aloice Aluoch
- Piedmont Eastside Rheumatology, Snellville, Georgia, USA
| | | | - Moffat J Nyirenda
- Department of Non-communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Mark J Siedner
- Harvard Medical School, Boston, Massachusetts, USA
- Africa Health Research Institute, KwaZulu-Natal, South Africa
| | - Katalina A Wilkinson
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Republic of South Africa
- Francis Crick Institute, London, UK
| | - Robert J Wilkinson
- Department of Medicine, University of Cape Town, Observatory, Republic of South Africa
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Republic of South Africa
- Francis Crick Institute, London, UK
| | - Ntobeko A B Ntusi
- Department of Medicine, University of Cape Town, Observatory, Republic of South Africa
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Republic of South Africa
- South African Medical Research Council, Tygerberg, Republic of South Africa
- ARUA/Guild Cluster of Research Excellence on Noncommunicable Diseases and Associated Multimorbidity
| |
Collapse
|
45
|
Alvarez-Jimenez L, Morales-Palomo F, Moreno-Cabañas A, Mora-Gonzalez D, Turrillas MDCM, Mora-Rodriguez R. Time-course atherogenic blood lipid response to statin discontinuation in dyslipidemic adults. Nutr Metab Cardiovasc Dis 2024; 34:2334-2343. [PMID: 39013748 DOI: 10.1016/j.numecd.2024.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND AND AIMS Half of dyslipidemic patients sometimes discontinue statin medication. It is unclear if blood atherogenic risk increases right after statin discontinuation or if there is a lingering protective effect. We sought to determine if a legacy effect prevented blood lipid increases during the first stages of statin cessation. METHODS AND RESULTS Atherogenic blood lipid profile was measured in 10 overweight (BMI 31 ± 3 kg m-2) middle-aged males (62 ± 7 years old), statin users, while fasted and postprandially. Trials were conducted before (i.e., Day 0) and after 4, 7, 15, and 30 days of statin withdrawal and 20 days after statins reloading (Day 50). Four days after statin discontinuation, blood fasting LDL-c, total cholesterol (CHOL), and triglyceride (TG) concentrations increased by 30%, 18%, and 17%, respectively (P < 0.05). The increases in LDL-c, CHOL, and TG peaked after 7-15 days at 79%, 48%, and 34% of basal levels (P < 0.001), respectively. There were no significant correlations between the increases in blood lipids and the dose or years under statin treatment (P = 0.156-0.575). Twenty days after resuming statins, blood LDL-c (2.79 ± 1.06 vs 2.20 ± 0.50 mmol L-1; P = 0.568), CHOL (4.85 ± 1.41 vs 4.25 ± 0.83 mmol L-1; P = 0.747), and TG (1.47 ± 0.60 vs 1.50 ± 0.68 mmol L-1; P = 0.782), returned to basal levels. CONCLUSIONS Our data does not support a statin lingering/legacy effect in blood lipids since they dangerously increased after only 4 days of statin withdrawal in every patient, regardless of dose and years under treatment. Reloading statins restored blood lipids, evidencing a reproducible biological effect at the whole-body level.
Collapse
Affiliation(s)
| | | | - Alfonso Moreno-Cabañas
- Exercise Physiology Lab at Toledo, University of Castilla-La Mancha, Spain; Centre for Nutrition, Exercise, and Metabolism, University of Bath, Bath, United Kingdom
| | - Diego Mora-Gonzalez
- Department of Nursing, Physiotherapy, and Occupational Therapy, University of Castilla-La Mancha, Toledo, Spain
| | | | | |
Collapse
|
46
|
Hernández-Huerta MT, Martínez-Cruz R, Pérez-Campos Mayoral L, Pina-Canseco MDS, Solórzano-Mata CJ, Martínez-Cruz M, Vásquez Martínez IP, Zenteno E, Laguna Barrios LÁ, Matias-Cervantes CA, Pérez-Campos Mayoral E, Pérez-Campos E. Association between O-GlcNAc levels and platelet function in obese insulin-resistant subjects. Glycoconj J 2024; 41:291-300. [PMID: 39300054 DOI: 10.1007/s10719-024-10164-9] [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: 03/14/2024] [Revised: 07/22/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024]
Abstract
Obesity is an epidemic associated with platelet and vascular disorders. Platelet O-GlcNAcylation has been poorly studied in obese subjects. We aimed to evaluate O-linked N-acetyl-glucosamine (O-GlcNAc) levels and platelet activity in obese insulin-resistant (ObIR) subjects. Six healthy and six insulin-resistant obese subjects with a body mass index of 22.6 kg/m2 (SD ± 2.2) and 35.6 kg/m2 (SD ± 3.8), respectively, were included. Flow cytometry was used to measure markers of platelet activity, expression of P-selectin (CD62P antibody), glycoprotein IIb/IIIa (integrins αIIbβ3 binding to PAC-1 antibody), and thrombin stimulation. O-GlcNAc was determined in the platelets of all test subjects by cytofluometry, intracellular calcium, percentage of platelet aggregation, and immunofluorescence microscopy and Western blot were used to assess O-GlcNAc and OGT (O-GlcNAc transferase) in platelets. Platelets from ObIR subjects had on average 221.4 nM intracellular calcium, 81.89% PAC-1, 22.85% CD62P, 57.48% OGT, and 66.62% O-GlcNAc, while platelets from healthy subjects had on average 719.2 nM intracellular calcium, 4.99% PAC-1, 3.17% CD62P, 18.38% OGT, and 23.41% O-GlcNAc. ObIR subjects showed lower platelet aggregation than healthy subjects, 13.83% and 54%, respectively. The results show that ObIR subjects have increased O-GlcNAc, and increased intraplatelet calcium associated with platelet hyperactivity and compared to healthy subjects, suggesting that changes in platelet protein O-GlcNAcylation and platelet activity might serve as a possible prognostic tool for insulin resistance, prediabetes and its progression to type 2 diabetes mellitus.
Collapse
Affiliation(s)
| | - Ruth Martínez-Cruz
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca, 68020, México
| | - Laura Pérez-Campos Mayoral
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca, 68020, México
| | - María Del Socorro Pina-Canseco
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca, 68020, México
| | - Carlos Josué Solórzano-Mata
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca, 68020, México
- Facultad de Odontología, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca City, 68120, México
| | | | - Itzel Patricia Vásquez Martínez
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca, 68020, México
| | - Edgar Zenteno
- Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, 04360, México
| | - Luis Ángel Laguna Barrios
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca, 68020, México
| | | | - Eduardo Pérez-Campos Mayoral
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca, 68020, México
| | - Eduardo Pérez-Campos
- Tecnológico Nacional de México/IT de Oaxaca, Oaxaca, 68030, México.
- Laboratorio de Patología Clínica, "Dr. Eduardo Pérez Ortega,", Oaxaca, 68000, México.
| |
Collapse
|
47
|
Westcott FA, Nagarajan SR, Parry SA, Savic D, Green CJ, Marjot T, Johnson E, Cornfield T, Mózes FE, O’Rourke P, Mendall J, Dearlove D, Fielding B, Smith K, Tomlinson JW, Hodson L. Dissociation between liver fat content and fasting metabolic markers of selective hepatic insulin resistance in humans. Eur J Endocrinol 2024; 191:463-472. [PMID: 39353069 PMCID: PMC11497584 DOI: 10.1093/ejendo/lvae123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 08/16/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVE Fasting hyperglycemia and hypertriglyceridemia are characteristic of insulin resistance (IR) and rodent work has suggested this may be due to selective hepatic IR, defined by increased hepatic gluconeogenesis and de novo lipogenesis (DNL), but this has not been shown in humans. DESIGN Cross-sectional study in men and women across a range of adiposity. METHODS Medication-free participants (n = 177) were classified as normoinsulinemic (NI) or hyperinsulinemic (HI) and as having low (LF) or high (HF) liver fat content measured by magnetic resonance spectroscopy. Fractional gluconeogenesis (frGNG) and hepatic DNL were measured using stable isotope tracer methodology following an overnight fast. RESULTS Although HI and HF groups had higher fasting plasma glucose and triglyceride concentrations when compared to NI and LF groups respectively, there was no difference in frGNG. However, HF participants tended to have lower frGNG than LF participants. HI participants had higher DNL compared to NI participants but there was no difference observed between liver fat groups. CONCLUSIONS Taken together, we found no metabolic signature of selective hepatic IR in fasting humans. DNL may contribute to hypertriglyceridemia in individuals with HI but not those with HF. Glycogenolysis and systemic glucose clearance may have a larger contribution to fasting hyperglycemia than gluconeogenesis, especially in those with HF, and these pathways should be considered for therapeutic targeting.
Collapse
Affiliation(s)
- Felix A Westcott
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Shilpa R Nagarajan
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Sion A Parry
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
- Aston Medical School, Aston University, Birmingham, B4 7ET, United Kingdom
| | - Dragana Savic
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
- Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Charlotte J Green
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Thomas Marjot
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Elspeth Johnson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Thomas Cornfield
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Ferenc E Mózes
- Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Paige O’Rourke
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Jessica Mendall
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - David Dearlove
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Barbara Fielding
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom
| | - Kieran Smith
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
- OCDEM, National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital Trusts, Oxford, OX3 7LE, United Kingdom
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
- OCDEM, National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital Trusts, Oxford, OX3 7LE, United Kingdom
| |
Collapse
|
48
|
Peng X, Xian H, Ge N, Hou L, Tang T, Xie D, Gao L, Yue J. Effect of probiotics on glycemic control and lipid profiles in patients with type 2 diabetes mellitus: a randomized, double blind, controlled trial. Front Endocrinol (Lausanne) 2024; 15:1440286. [PMID: 39351535 PMCID: PMC11439702 DOI: 10.3389/fendo.2024.1440286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction This double-blind, placebo-controlled, randomized (1:1) clinical trial was conducted at the West China Hospital, Sichuan University, from March to September 2017. Methods Eligible participants included adults aged 18 years and older, living in the community, diagnosed with type 2 Diabetes Mellitus according to ADA guidelines, capable of self-managing their diabetes, and able to visit the study site for follow-up. The intervention group received 25 ml of a probiotic beverage containing with over 10^8 CFU/mL of Lactobacillus, administered four times daily. An equal volume of inactivated Lactobacillus was administered to the control group and the control group was administered the same volume of inactivated Lactobacillus. This study aimed to evaluate the effectiveness of probiotics on glycemic control and other diabetes-related outcomes in patients with type 2 diabetes patients. The primary outcomes were changes in HbA1c and FBG levels post-intervention. Investigators, participants, and study site personnel were blinded to the treatment allocation until the conclusion of the study. This double-blind, randomized, placebo-controlled clinical trial was registered in the Chinese Clinical Trial Registry (ChiCTR-POR-17010850). Results Of the 490 participants screened, 213 were randomized to either the probiotics group (n = 103) or the placebo group (n = 110). After 16 weeks of follow-up, the probiotic group showed reductions in HbA1c [-0.44 (-0.66 to -0.22)] and FBG [-0.97 (-1.49 to 0.46)] post-intervention, similar to the placebo group with reductions in HbA1c [-0.33 (-0.52 to -0.15)] and FBG [-0.90 (-1.32 to -0.47)], but these changes were not statistically significant in PP and ITT analyses (P>0.05). Adverse events were similarly distributed among groups, indicating comparable safety profiles. Discussion Overall, 16-week probiotic supplementation showed no beneficial effects on glycemic control, lipid profiles, or weight. Clinical Trial Registration https://www.chictr.org.cn/showproj.html?proj=18421, identifier ChiCTR-POR-17010850.
Collapse
Affiliation(s)
| | | | - Ning Ge
- Department of Geriatrics and National Clinical Research Center for Geriatrics, West
China Hospital of Sichuan University, Chengdu, China
| | | | | | | | | | - Jirong Yue
- Department of Geriatrics and National Clinical Research Center for Geriatrics, West
China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
49
|
Liu J, Jin X, Feng Z, Huang J. Using anthropometric parameters to predict insulin resistance among patients without diabetes mellitus. Sci Rep 2024; 14:21407. [PMID: 39271702 PMCID: PMC11399142 DOI: 10.1038/s41598-024-57020-2] [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: 06/30/2023] [Accepted: 03/13/2024] [Indexed: 09/15/2024] Open
Abstract
Anthropometric parameters are widely used in the clinical assessment of hypertension, type 2 diabetes, and cardiovascular disease. However, few studies have compared the association between different anthropometric parameters and insulin resistance (IR). This study was aimed at investigating the relationship between 6 indicators, including body mass index (BMI), calf circumference (CC), arm circumference (AC), thigh circumference (TC), waist circumference (WC), waist-height ratio (WHtR), and IR. Homeostasis Model Assessment of Insulin Resistance (HOMA-IR) was used to measure IR. Weighted linear regression was used to assess the relationship between different parameters and IR. The receiver operating characteristic curve (ROC) was employed to compare the strength of the relationship between different anthropometric parameters and IR. A total of 8069 participants were enrolled in our study, including 4873 without IR and 3196 with IR. The weighted linear regression results showed that BMI, CC, AC, TC and WC were significantly correlated with IR, except WHtR. After adjusting for multiple confounding factors, we found that BMI, AC and WC were significantly positively correlated with IR, while TC was significantly negatively correlated with IR. Logistic regression results showed that a larger TC was associated with a decreased risk of IR. In addition, BMI and WC had similar areas under the curve (AUC: 0.780, 95% CI 0.770-0.790; AUC: 0.774, 95% CI 0.763-0.784, respectively), which were higher than TC and AC (AUC: 0.698, 95% CI 0.687-0.710, AUC: 0.746, 95% CI 0.735-0.757, respectively). To our knowledge, this is the first study to report a negative correlation between TC and IR among patients without diabetes mellitus. Therefore, TC may be a new tool to guide public health and a clinical predictor of IR in non-diabetic patients.
Collapse
Affiliation(s)
- Jiajun Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xueshan Jin
- The Affiliated Jiangmen TCM Hospital, Jinan University, Jiangmen, Guangdong, China
| | - Ziyi Feng
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jieming Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
50
|
Amin NG, Rahim AA, Rohoma K, Elwafa RAA, Dabees HMF, Elrahmany S. The relation of mTOR with diabetic complications and insulin resistance in patients with type 2 diabetes mellitus. Diabetol Metab Syndr 2024; 16:222. [PMID: 39261960 PMCID: PMC11389252 DOI: 10.1186/s13098-024-01450-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Dysregulation of the mechanistic target of rapamycin (mTOR) has been related to several metabolic conditions, notably obesity and type 2 diabetes (T2DM). This study aimed to evaluate the role of mTOR in patients with T2DM, and its relationship with insulin resistance and microvascular complications. METHODS This case-control study was conducted on 90 subjects attending the Outpatient Internal Medicine Clinic in Damanhur Teaching Hospital. Subjects were divided into 3 groups, Group I: 20 healthy controls, Group II: 20 subjects with T2DM without complications, and Group III: 50 subjects with T2DM with microvascular complications. An Enzyme-linked immunosorbent assay was used to measure serum mTOR levels. T2DM and diabetic complications were defined according to the diagnostic criteria of the American Diabetes Association. RESULTS The results revealed significant positive correlations to HbA1c (r = 0.530, P < 0.001), fasting glucose (r = 0.508, P < 0.001), and HOMA- IR (r = 0.559, P < 0.001), and a significant negative correlation to eGFR (r=-0.370, P = 0.002). Multivariate analysis revealed an independent association of mTOR and HbA1c values with the presence of microvascular complications. The prediction of microvascular complications was present at a cutoff value of 8 ng/ml mTOR with a sensitivity of 100% and specificity of 95% with an AUC of 0.983 and a p-value < 0.001. CONCLUSION mTOR is a prognostic marker of diabetic microvascular and is associated with insulin resistance in patients with T2DM. TRIAL REGISTRATION The study was conducted following the Declaration of Helsinki, and approved by the Ethics Committee of Alexandria University (0201127, 19/7/2018).
Collapse
Affiliation(s)
- Noha G Amin
- Department of Internal Medicine (Diabetes, Lipidology & Metabolism), Faculty of Medicine, Alexandria University, 17, Champollion Street, El Messallah, Alexandria, Egypt.
| | - A Abdel Rahim
- Department of Internal Medicine (Diabetes, Lipidology & Metabolism), Faculty of Medicine, Alexandria University, 17, Champollion Street, El Messallah, Alexandria, Egypt
| | - Kamel Rohoma
- Department of Internal Medicine (Diabetes, Lipidology & Metabolism), Faculty of Medicine, Alexandria University, 17, Champollion Street, El Messallah, Alexandria, Egypt
| | - Reham A Abo Elwafa
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Hossam M F Dabees
- Department of Internal Medicine (Diabetes, Lipidology & Metabolism), Faculty of Medicine, Alexandria University, 17, Champollion Street, El Messallah, Alexandria, Egypt
| | - Shimaa Elrahmany
- Department of Internal Medicine (Diabetes, Lipidology & Metabolism), Faculty of Medicine, Alexandria University, 17, Champollion Street, El Messallah, Alexandria, Egypt
| |
Collapse
|