1
|
Shin J, Bressler J, Grove ML, Brown M, Selvin E, Pankow JS, Fornage M, Morrison AC, Sarnowski C. DNA methylation markers of insulin resistance surrogate measures in the Atherosclerosis Risk in Communities (ARIC) study. Epigenetics 2025; 20:2498857. [PMID: 40327844 PMCID: PMC12064056 DOI: 10.1080/15592294.2025.2498857] [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: 10/23/2024] [Revised: 04/03/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025] Open
Abstract
Insulin resistance (IR) is a risk factor for cardiovascular diseases and type 2 diabetes. Associations between DNA methylation (DNAm) and IR have been less studied in African ancestry (AA) populations than those of European ancestry (EA). We aimed to identify associations between whole blood DNAm and IR in up to 1,811 AA and 964 EA participants from the Atherosclerosis Risk in Communities (ARIC) study. We quantified IR using three surrogate measures: the homeostasis model assessment of insulin resistance (HOMA-IR), the triglyceride-glucose index (TyG), and the triglyceride glucose-body mass index (TyG-BMI). We used ancestry-stratified linear regression models to conduct epigenome-wide association studies of IR, adjusting for batch effects and relevant covariates. Among 484,436 tested CpG sites, 39 were significantly associated with IR, of which 31% (10 in AA and two in EA) were associated with TyG-BMI and not previously reported for IR or related traits. These include a positive association at cg18335991-SEMA7A in AA. SEMA7A inhibits adipogenesis of preadipocytes and lipogenesis of mature adipocytes. DNAm levels at cg18335991 have been reported to be negatively associated with SEMA7A expression in blood. After additionally adjusting for smoking and drinking status, 15 of the 39 significant CpG sites remained significant or suggestive. Our study identified novel IR-associated CpG sites, contributing to a broader understanding of the epigenetic mechanisms underlying IR in diverse populations.
Collapse
Affiliation(s)
- Jeewoen Shin
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston, Houston, School of Public Health, TX, USA
| | - Jan Bressler
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston, Houston, School of Public Health, TX, USA
| | - Megan L. Grove
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston, Houston, School of Public Health, TX, USA
| | - Michael Brown
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston, Houston, School of Public Health, TX, USA
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - James S. Pankow
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston, Houston, School of Public Health, TX, USA
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alanna C. Morrison
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston, Houston, School of Public Health, TX, USA
| | - Chloé Sarnowski
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston, Houston, School of Public Health, TX, USA
| |
Collapse
|
2
|
Liu N, Wang B, Zhang G, Shen M, Cheng P, Guo Z, Zuo L, Yang J, Guo M, Wang M, Liu Z, Wu J. Waist-to-hip ratio better reflect beta-cell function and predicts diabetes risk in adult with overweight or obesity. Ann Med 2025; 57:2462447. [PMID: 39921368 PMCID: PMC11809193 DOI: 10.1080/07853890.2025.2462447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/09/2025] [Accepted: 01/24/2025] [Indexed: 02/10/2025] Open
Abstract
OBJECTIVE Controversy remains as to which obesity measures better predict type 2 diabetes (T2D) risk in overweight or obese individuals. The objective of this study is to determine which commonly used obesity measures better reflect beta cell function and predict T2D risk in participants with overweight or obesity and to validate the findings using prospective cohort data. PATIENTS AND METHODS Cross-sectional data from the Obesity Clinic of the Xiangya Hospital of the Central South University and prospective cohort from UK Biobank. BMI, waist circumference (WC), waist-to-hip ratio (WHpR), and waist-to-height ratio (WHtR) were measured. Primary outcomes included beta cell function indices in the cross-sectional study and the occurrence of diabetes obtained from UK Biobank data. RESULTS One thousand four hundred and ninety-seven participants with overweight or obesity (median age 29 years, 41% males) and 322,023 UK Biobank participants without diabetes at baseline (mean age 56.83 years, 50.4% males) were studied. WHpR had a stronger association with beta cell function and central body fat distribution than the other three obesity measures irrespective of glucometabolic states. WHpR associated positively with diabetes risk in participants using the hazard ratio scale (HR per SD increase of WHpR, 2.311, 95% CI 2.250-2.374). CONCLUSIONS WHpR is a superior index in reflecting central body obesity, estimating beta cell function, and predicting T2D risk in people with overweight or obesity compared to BMI, WC, and WHtR.
Collapse
Affiliation(s)
- Na Liu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Obesity and Its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bian Wang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Obesity and Its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guanxiong Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Minxue Shen
- Department of Social Medicine and Health Management, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Peng Cheng
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Obesity and Its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhanjun Guo
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Obesity and Its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Linhui Zuo
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Obesity and Its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Junya Yang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Min Guo
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Min Wang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Obesity and Its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenqi Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jing Wu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Obesity and Its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Lennartsson AK, Jonsdottir IH, Jansson PA, Sjörs Dahlman A. Study of glucose homeostasis in burnout cases using an oral glucose tolerance test. Stress 2025; 28:2438699. [PMID: 39688015 DOI: 10.1080/10253890.2024.2438699] [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: 06/05/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
Burnout is caused by long term psychosocial stress and has, besides the fatigue and mental health burden, been associated with increased risk of adverse physical health, such as for example type 2 diabetes. This study aims to investigate the glucose and insulin levels in individuals with stress related burnout, by assessing these metabolic markers in response to a standard oral glucose tolerance test (OGTT). 38 cases with burnout (13 men and 25 women) and 35 healthy controls (13 men and 22 women) in the age 24-55 were included in the study. The burnout group overall did not differ from healthy controls in glucose or insulin levels during the OGTT. However, the burnout cases who reported more severe burnout symptoms exhibited significantly higher levels of both glucose and insulin levels during the OGTT compared to burnout cases reporting lower severity of symptoms. Furthermore, the group of burnout cases who reported symptoms of depression exhibited higher insulin levels during OGTT compared to the burnout cases without depressive symptoms. The observed higher levels in the burnout cases with most severe symptoms indicate an increased diabetic risk in these patients and it may be of importance to follow glucose and insulin levels in individuals with more severe symptoms of burnout i.e. to perform an OGTT.
Collapse
Affiliation(s)
- Anna-Karin Lennartsson
- The Institute of Stress Medicine, Region Västra Götaland, Gothenburg, Sweden
- School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Ingibjörg H Jonsdottir
- The Institute of Stress Medicine, Region Västra Götaland, Gothenburg, Sweden
- School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Per-Anders Jansson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Sjörs Dahlman
- Swedish National Road and Transport Research Institute (VTI), Linköping, Sweden
- Department of Electrical Engineering, and SAFER Vehicle and Traffic Safety Centre, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
4
|
Jardon KM, Umanets A, Gijbels A, Trouwborst I, Hul GB, Siebelink E, Vliex LM, Bastings JJ, Argamasilla R, Chenal E, Venema K, Afman LA, Goossens GH, Blaak EE. Distinct gut microbiota and metabolome features of tissue-specific insulin resistance in overweight and obesity. Gut Microbes 2025; 17:2501185. [PMID: 40336254 PMCID: PMC12064058 DOI: 10.1080/19490976.2025.2501185] [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: 09/20/2024] [Revised: 02/24/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Insulin resistance (IR) is an early marker of cardiometabolic deterioration which may develop heterogeneously in key metabolic organs, including the liver (LIR) and skeletal muscle (MIR). This tissue-specific IR is characterized by distinct metabolic signatures, but the role of the gut microbiota in its etiology remains unclear. Here, we profiled the gut microbiota, its metabolites and the plasma metabolome in individuals with either a LIR or MIR phenotype (n = 233). We observed distinct microbial community structures LIR and MIR, and higher short-chain fatty acid (SCFA) producing bacteria, fecal SCFAs and branched-chain fatty acids and a higher postprandial plasma glucagon-like-peptide-1 response in LIR. In addition, we found variations in metabolome profiles and phenotype-specific associations between microbial taxa and functional metabolite groups. Overall, our study highlights association between gut microbiota and its metabolites composition with IR heterogeneity that can be targeted in precision-based strategies to improve cardiometabolic health. Clinicaltrials.gov registration: NCT03708419.
Collapse
Affiliation(s)
- Kelly M. Jardon
- TiFN, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Alexander Umanets
- Centre for Healthy Eating & Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
- Chair Group Youth Food and Health, Faculty of Science and Engineering, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Anouk Gijbels
- TiFN, Wageningen, The Netherlands
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Inez Trouwborst
- TiFN, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Gabby B. Hul
- TiFN, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Els Siebelink
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Lars M.M. Vliex
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Jacco J.A.J. Bastings
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | | | | | - Koen Venema
- Centre for Healthy Eating & Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Lydia A. Afman
- TiFN, Wageningen, The Netherlands
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Gijs H. Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Ellen E. Blaak
- TiFN, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
5
|
Ahmed EI, Moneam Shamardl HA, Elsayed AM, Sadik SA. Evolocumab ameliorates myocardial fibrosis and improves metabolic syndrome-induced cardiac dysfunction in rats via inhibiting PCSK9/NLRP3 inflammasome and Caspase-1 / IL-1β pathways. Eur J Pharmacol 2025; 998:177499. [PMID: 40064223 DOI: 10.1016/j.ejphar.2025.177499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/19/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Affiliation(s)
- Eman Ibrahim Ahmed
- Pharmacology and Therapeutics Department, College of Medicine, Jouf University, Sakaka, Saudi Arabia; Medical Pharmacology Department, Faculty of Medicine, Fayoum University, Fayoum, Egypt.
| | | | - Asmaa Mohammed Elsayed
- Histology and Cell Biology Department, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Sawsan A Sadik
- Medical Pharmacology Department, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| |
Collapse
|
6
|
Mansour MF, Behairy A, Mostafa M, Khamis T, Alsemeh AE, Ahmed NMQ, El-Emam MMA. Quercetin-loaded PEGylated liposomes alleviate testicular dysfunction in alloxan-induced diabetic rats: The role of Kisspeptin/Neurokinin B/Dynorphin pathway. Toxicol Appl Pharmacol 2025; 499:117337. [PMID: 40239742 DOI: 10.1016/j.taap.2025.117337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Diabetes mellitus (DM) is a chronic metabolic disorder that can lead to serious complications, including testicular dysfunction. This dysfunction is considered a significant cause of male infertility. Quercetin (Que), a naturally existing flavonoid with versatile biological functions, has limited water solubility and low bioavailability. The current study was designed to develop a bioavailable formulation of Que. via encapsulating it in PEGylated liposomes (Que-PEG-Lip) and determine whether this formulation is effective in the treatment of alloxan-induced testicular injury via targeting Kisspeptin/Neurokinin B/Dynorphin/steroidogenesis signaling pathway. Thirty-two male Sprague Dawley rats were randomly divided into four groups: Control, alloxan-induced diabetes with testicular dysfunction (ALX), ALX + metformin (MET) and ALX + Que-PEG-Lip. The results showed that treatment of ALX group with Que-PEG-Lip significantly improved the alteration of glycemic index, serum reproductive hormones, testicular antioxidant status, testicular Kiss-1, androgen receptor (AR), and proliferation marker protein (ki67) immunoexpression in compared to ALX group. Moreover, the treatment of ALX group with Que-PEG-Lip regulated the Kisspeptin/Neurokinin B/Dynorphin/steroidogenesis pathway gene expression. Interestingly, the outcomes of the molecular docking analysis revealed a strong agonistic effect of Que. on the kisspeptin, neurokinin, and dynorphin receptors. In conclusion, Que-PEG-Lip mitigated the testicular dysfunction in alloxan-induced diabetic rats via regulation of hypothalamic-pituitary-gonadal axis signaling pathway and alleviation the testicular oxidative stress.
Collapse
Affiliation(s)
- Mohamed Fouad Mansour
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt.
| | - Amany Behairy
- Department of Physiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Mahmoud Mostafa
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Amira Ebrahim Alsemeh
- Department of Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | | | - Mahran Mohamed Abd El-Emam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
7
|
Wagh R, Hatem G, Andersson J, Kunte P, Bandyopadhyay S, Yajnik CS, Prasad RB. Parent-of-origin effects in the life-course evolution of cardiometabolic traits. Diabetologia 2025; 68:1298-1314. [PMID: 40175764 DOI: 10.1007/s00125-025-06396-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 01/22/2025] [Indexed: 04/04/2025]
Abstract
AIMS/HYPOTHESIS Cardiometabolic traits are heritable, and some display parent-of-origin effects, which indicates preferential inheritance from one parent or parental bias. Most studies of these phenomena have focused on adult populations. We aimed to investigate the heritability and parent-of-origin effects on cardiometabolic traits in a birth cohort with serial measurements to determine whether these patterns emerged early in life. METHODS The Pune Maternal Nutrition Study comprises a birth cohort in which offspring and parents were studied from birth and followed up for 24 years. We investigated parent-of-origin effects on cardiometabolic traits cross-sectionally at available timepoints using linear regression, and longitudinally across the life course using mixed-effect regression. Maternal and paternal effects on offspring phenotype were modelled after adjusting for age, sex and BMI. Parent-of-origin effects were calculated based on the difference between maternal and paternal effects. We also investigated these effects in another birth cohort, that of the Pune Children's Study. Genetic parent-of-origin effects were assessed using generalised estimating equations after taking the parental origin of the alleles into account. RESULTS Birthweight showed a maternal parent-of-origin effect. At 24 years, maternal bias was seen for some obesity-related traits for daughters, while paternal bias was seen for WHR in sons. A shift from paternal bias at 6 years to maternal bias at 24 years for the skinfold thickness was observed in daughters. Fasting glucose and lipids showed maternal bias at 6, 12 and 24 years. For fasting insulin and HOMA2-S, a negative maternal effect at 6 years transitioned to a positive one at 12 years. For HOMA2-B, a paternal effect at 6 years transitioned to a maternal one at 12 years, and this remained so at 24 years. Some of these findings were also observed in the cohort from the Pune Children's Study. Longitudinal modelling revealed stronger paternal effects over time for fasting insulin and HOMA indices but maternal effects for glucose and lipids, reflecting their cumulative effect over time. Genetic variants at the KCNQ1 locus showed a maternal parent-of-origin effect on birthweight, on HOMA2-B at 12 years, and on lipids at 6 and 12 years. CONCLUSIONS/INTERPRETATION Our study provides proof of concept of the existence of parent-of-origin effects on cardiometabolic traits from birth, through childhood and puberty, until adult age. Our results indicate a predominantly maternal influence on intrauterine, pubertal and reproductive-age metabolism in the offspring. While the longitudinal analysis indicated a maternal bias for the macronutrients (glucose and lipids), and a paternal bias for glucose-insulin metabolism, the cross-sectional analysis revealed a transition between parental influence across physiological stages. This dynamic relationship may have its origins in the life-history theory of evolution, and could inform strategies for primordial prevention aimed at curbing the rising burden of cardiometabolic disease. Further studies are needed to determine the mechanisms underlying such effects.
Collapse
Affiliation(s)
- Rucha Wagh
- Diabetes Unit, Kamalnayan Bajaj Diabetology Research Centre, King Edward Memorial Hospital and Research Centre, Pune, India
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Gad Hatem
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University, Malmö, Sweden
| | - Jonas Andersson
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University, Malmö, Sweden
| | - Pooja Kunte
- Diabetes Unit, Kamalnayan Bajaj Diabetology Research Centre, King Edward Memorial Hospital and Research Centre, Pune, India
| | | | - Chittaranjan S Yajnik
- Diabetes Unit, Kamalnayan Bajaj Diabetology Research Centre, King Edward Memorial Hospital and Research Centre, Pune, India
| | - Rashmi B Prasad
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University, Malmö, Sweden.
- Institute of Molecular Medicine Finland, Helsinki University, Helsinki, Finland.
| |
Collapse
|
8
|
Pejovic S, Shang Y, Vgontzas AN, Fernandez‐Mendoza J, He F, Li Y, Kong L. C-reactive protein improves the ability to detect hypertension and insulin resistance in mild-to-moderate obstructive sleep apnea: Age effect. J Sleep Res 2025; 34:e14386. [PMID: 39462147 PMCID: PMC12069758 DOI: 10.1111/jsr.14386] [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: 08/25/2024] [Revised: 10/09/2024] [Accepted: 10/14/2024] [Indexed: 10/29/2024]
Abstract
C-reactive protein (CRP) appears to improve the ability to detect cardiometabolic risk in young and middle-aged adults with mild-to-moderate obstructive sleep apnea (mmOSA). The aim of this study is to assess utility of CRP in identifying the risk of hypertension and insulin resistance across a wide age range including older patients with mmOSA. Adults (n = 216) of a wide age range (28-90 years old, mean age 52.64 ± 12.74) with mmOSA (5 ≤ AHI < 30) completed in-lab polysomnography or home sleep apnea testing, physical examination including blood pressure (BP) measures, structured medical history questionnaire, and blood draw for CRP and fasting glucose and insulin levels. In adults < 60 years, lnCRP but not the apnea-hypopnea index (AHI) was associated with greater odds for hypertension (odds ratio [OR] = 2.40, 95% CI = 1.20-4.84, p = 0.01; OR = 1.00, 95% CI = 0.92-1.08, p = 0.92, respectively) and with higher average systolic and diastolic BP. Also, in adults < 60 years lnCRP but not AHI, was associated with higher lnHOMA values. In contrast, in adults > 60 years neither lnCRP nor AHI were associated with greater odds for hypertension, average systolic and diastolic BP, and lnHOMA. Receiver-operating characteristics curves revealed that adding CRP to standard clinical factors (age, sex, and BMI) yielded moderately good risk models for hypertension in patients < 60 years (AUC = 0.721). In conclusion, CRP improves the ability to detect cardiometabolic risk in young and middle-aged, but not older adults with mmOSA, suggesting that inflammation may be a primary pathogenetic mechanism in younger patients with OSA.
Collapse
Affiliation(s)
- Slobodanka Pejovic
- Sleep Research & Treatment Center, Penn State Health Milton S. Hershey Medical CenterPennsylvania State University, College of MedicineHersheyPennsylvaniaUSA
| | - Yimeng Shang
- Department of Public Health SciencesPennsylvania State University College of MedicineHersheyPennsylvaniaUSA
| | - Alexandros N. Vgontzas
- Sleep Research & Treatment Center, Penn State Health Milton S. Hershey Medical CenterPennsylvania State University, College of MedicineHersheyPennsylvaniaUSA
| | - Julio Fernandez‐Mendoza
- Sleep Research & Treatment Center, Penn State Health Milton S. Hershey Medical CenterPennsylvania State University, College of MedicineHersheyPennsylvaniaUSA
| | - Fan He
- Department of Public Health SciencesPennsylvania State University College of MedicineHersheyPennsylvaniaUSA
| | - Yun Li
- Department of Sleep Medicine, Shantou University Mental Health CenterShantou University Medical CollegeShantouChina
- Sleep Medicine CenterShantou University Medical CollegeShantouChina
| | - Lan Kong
- Department of Public Health SciencesPennsylvania State University College of MedicineHersheyPennsylvaniaUSA
| |
Collapse
|
9
|
Sardar MA, Abbasian S, Moghavemi H, Karabi M. HIIT may ameliorate inter-organ crosstalk between liver and hypothalamus of HFD-induced MAFLD rats; A two-phase study to investigate the effect of exercise intensity as a stressor. Brain Res 2025; 1856:149591. [PMID: 40120709 DOI: 10.1016/j.brainres.2025.149591] [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: 01/08/2025] [Revised: 02/25/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Previous studies demonstrate that GDF15 and its related signaling activators may be affected by exercise training, leading to the suppression of inflammatory factors and the promotion of immune-metabolic balance. Therefore, the purpose of the study was to evaluate the effect of high-intensity interval training (HIIT) on amelioration of inter-organ crosstalk between liver and hypothalamus of the high-fat diet (HFD)-induced metabolic dysfunction-associated fatty liver disease (MAFLD) rats in a two-phase study. In this regard, rats were initially divided into two groups, the normal diet-inactive (NS) and the HFD groups. HFD course lasted 12 weeks to induce MAFLD in the latter group. After ensuring the induction of MAFLD, 25 rats were divided into three groups: the HFD-inactive group (HS), the HFD-HIIT group (HH), as well as the HFD-aerobic group (HA). The training interventions were consistently applied over a period of eight weeks, five days a week, with each session lasting 40-60 min, and the duration of the whole research was 21 weeks. The results of this study displayed that HIIT intervention promotes hypothalamic Gdf15 gene expression and there were similar alterations in genes expression of Foxo1 and Akt2. Moreover, our results confirmed that HIIT ameliorated hypothalamic NFKB gene expression and there was a similar trend in genes expression of Tnfa and Il1b following both HIIT as well as aerobic training protocols. Taking these findings together, it is concluded that interventions, particularly exercise training, uniquely contribute to the reduction of hypothalamic-associated inflammatory responses that result in prolonged and chronic increases in GDF15.
Collapse
Affiliation(s)
- Mohammad Ali Sardar
- Department of General Courses, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sadegh Abbasian
- Department of Physical Education, Farhangian University, P.O. Box 14665-889, Tehran, Iran.
| | - Hamid Moghavemi
- Department of Exercise Physiology, Faculty of Sport Sciences, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mina Karabi
- Department of Sport Sciences, Khavaran Institute of Higher Education, Mashhad, Iran
| |
Collapse
|
10
|
Mead LC, Hill AM, Carter S, Coates AM. Effects of energy-restricted diets with or without nuts on weight, body composition and glycaemic control in adults: a scoping review. Nutr Res Rev 2025; 38:202-218. [PMID: 38389450 DOI: 10.1017/s0954422424000106] [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: 02/24/2024]
Abstract
Energy-restricted (ER) diets promote weight loss and improve body composition and glycaemic control. Nut consumption also improves these parameters. However, less is known about the combined benefit of these two strategies. This scoping review implemented a systematic search of Medline, Embase and Scopus to identify randomised controlled trials evaluating the effect of ER diets with or without nuts on body mass, body composition and glycaemic control in adults. After reviewing titles and abstracts, twenty-nine full-text articles were screened, resulting in seven studies reported in eight papers that met the inclusion criteria. Energy restriction was achieved by prescribing a set energy target or reducing intake by 1000-4200 kJ from daily energy requirements. Interventions ranged from 4 to 52 weeks in duration and contained 42-84 g/d of almonds, peanuts, pistachios or walnuts. While all studies reported that energy restriction resulted in significant weight loss, the addition of nuts to ER diets demonstrated significantly greater weight loss in only approximately half of the included studies (4/7 studies). There was limited evidence to support additional benefits from nuts for body composition measures or glycaemic control. Although improvements in weight loss and glycaemia were not consistent when nuts were included in ER diets, no study revealed an adverse effect of nut consumption on health outcomes. Future studies could explore the effect of consuming different types and amounts of nuts, combined with various levels of energy restriction on weight, body composition and glycaemic control.
Collapse
Affiliation(s)
- Lauren C Mead
- Alliance for Research in Exercise, Nutrition and Activity (ARENA), Allied Health and Human Performance, University of South Australia, Adelaide, Australia
| | - Alison M Hill
- Alliance for Research in Exercise, Nutrition and Activity (ARENA), Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Sharayah Carter
- Alliance for Research in Exercise, Nutrition and Activity (ARENA), Allied Health and Human Performance, University of South Australia, Adelaide, Australia
| | - Alison M Coates
- Alliance for Research in Exercise, Nutrition and Activity (ARENA), Allied Health and Human Performance, University of South Australia, Adelaide, Australia
| |
Collapse
|
11
|
Zhang K, Huang C, Li J, Mai P, Xu S, Huang F, He W, Zhang H, Liu Y, Feng W. Association of long-term insulin variability before the onset of diabetes with cardiovascular outcomes in later life: Findings from the coronary artery risk development in young adults (CARDIA) study. Am J Prev Cardiol 2025; 22:100952. [PMID: 40166419 PMCID: PMC11957604 DOI: 10.1016/j.ajpc.2025.100952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 02/16/2025] [Accepted: 02/22/2025] [Indexed: 04/02/2025] Open
Abstract
Background The important effects of variability of some physiological/biological characteristics (such as LDL cholesterol, blood pressure) on cardiovascular outcomes have been elucidated, while the role of insulin variability is undefined. Objectives To investigate the associations of long-term fasting insulin variability during young adulthood before the onset of diabetes with subsequent cardiovascular outcomes in middle age. Methods We included 3,983 CARDIA (Coronary Artery Risk Development Study in Young Adults) participants aged 18 to 30 years with at least three fasting insulin measurements. Intra-individual fasting insulin variability was defined by the average real variability (ARV) of insulin and standard deviation (SD) of insulin during 30-year follow-up. The presence and the degree of coronary artery calcification (CAC) were assessed by computed tomography at year 25. Incident cardiovascular disease (CVD) and all-cause mortality were adjudicated. Results After multivariable adjustment, comparing high versus low tertile of insulin ARV, the hazard of CVD increased by 65 % (HR, 1.65; 95 % CI, 1.13-2.39) and all-cause mortality by 97 % (HR, 1.97; 95 % CI, 1.38-2.82). Higher tertile of insulin ARV was associated with significantly worse degree of CAC (β =0.1; 95 % CI, 0.03-0.18) but not with the presence of CAC (P = 0.197). Similar results were also observed in insulin SD. Conclusion High long-term insulin variability in young adulthood before the onset of diabetes was associated with an increased risk of CVD and all-cause mortality in later life, independent of average FG, HOMA-IR and other established cardiovascular risk factors. Long-term insulin variability was associated with the degree but not the presence of CAC.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Cardiology, The Seventh Affiliated Hospital of Sun Yat-Sen University, 628 Zhenyuan Road, Shenzhen 518107, China
| | - Chunlan Huang
- Department of Neurology, The Second Affiliated Hospital, University of South China, 30 Jiefang Road, Hengyang 421001, China
| | - Junping Li
- Department of Urology, Guangdong Second Provincial General Hospital, 466 Xingang Middle Road, Guangzhou 510120, China
| | - Peibiao Mai
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences (Shenzhen Sun Yat-sen Cardiovascular Hospital), Shenzhen 518000, China
| | - Shuwan Xu
- Department of Cardiology, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing 100191, China
| | - Feifei Huang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang Road, Guangzhou 510120, China
| | - Wanbing He
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang Road, Guangzhou 510120, China
| | - Huanji Zhang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025 Shennan Middle Road, Shenzhen 518033, China
| | - Yang Liu
- Department of Cardiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Weijing Feng
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Cardiac Function and Microcirculation, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, 1838 Guangzhou North Road, Guangzhou 510515, China
| |
Collapse
|
12
|
Chebii VJ, Wade AN, Crowther NJ, Nonterah EA, Agongo G, Simayi Z, Boua PR, Kisiangani I, Ramsay M, Choudhury A, Sengupta D. Genome-wide association study identifying novel risk variants associated with glycaemic traits in the continental African AWI-Gen cohort. Diabetologia 2025; 68:1184-1196. [PMID: 40025146 PMCID: PMC12069158 DOI: 10.1007/s00125-025-06395-6] [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: 07/02/2024] [Accepted: 01/24/2025] [Indexed: 03/04/2025]
Abstract
AIMS/HYPOTHESIS Glycaemic traits such as high fasting glucose levels and insulin resistance are positively associated with the risk of type 2 diabetes and other cardiometabolic diseases. Genetic association studies have identified hundreds of associations for each glycaemic trait, yet very few studies have involved continental African populations. We report the results of genome-wide association studies (GWASs) in a pan-African cohort for four glycaemic traits, namely fasting glucose, fasting insulin, insulin resistance (HOMA-IR) and beta cell function (HOMA-B), which are quantitative variables that affect the risk of developing type 2 diabetes. METHODS GWASs for the four traits were conducted in approximately 10,000 individuals from the Africa Wits-INDEPTH Partnership for Genomics Studies (AWI-Gen) cohort, with participants from Burkina Faso, Ghana, Kenya and South Africa. Association testing was performed using linear mixed models implemented in BOLT-LMM, with age, sex, BMI and principal components as covariates. Replication, fine mapping and functional annotation were performed using standard approaches. RESULTS We identified a novel signal (rs574173815) in the intron of the ankyrin repeat domain 33B (ANKRD33B) gene associated with fasting glucose, and a novel signal (rs114029796) in the intronic region of the WD repeat domain 7 (WDR7) gene associated with fasting insulin. SNPs in WDR7 have been shown to be associated with type 2 diabetes. A variant (rs74806991) in the intron of ADAM metallopeptidase with thrombospondin type 1 motif 16 (ADAMTS16) and another variant (rs6506934) in the β-1,4-galactosyltransferase 6 gene (B4GALT6) are associated with HOMA-IR. Both ADAMTS16 and B4GALT6 are implicated in the development of type 2 diabetes. In addition, our study replicated several well-established fasting glucose signals in the GCK-YTK6, SLC2A2 and THORLNC gene regions. CONCLUSIONS/INTERPRETATION Our findings highlight the importance of performing GWASs for glycaemic traits in under-represented populations, especially continental African populations, to discover novel associated variants and broaden our knowledge of the genetic aetiology of glycaemic traits. The limited replication of well-known signals in this study hints at the possibility of a unique genetic architecture of these traits in African populations. DATA AVAILABILITY The dataset used in this study is available in the European Genome-Phenome Archive (EGA) database ( https://ega-archive.org/ ) under study accession code EGAS00001002482. The phenotype dataset accession code is EGAD00001006425 and the genotype dataset accession code is EGAD00010001996. The availability of these datasets is subject to controlled access by the Data and Biospecimen Access Committee of the H3Africa Consortium. GWAS summary statistics are accessible through the NHGRI-EBI GWAS Catalog ( https://www.ebi.ac.uk/gwas/ ).
Collapse
Affiliation(s)
- Vivien J Chebii
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Alisha N Wade
- Department of Internal Medicine, School of Clinical Medicine, University of the Witwatersrand, Johannesburg, South Africa
- Research in Metabolism and Endocrinology, Division of Endocrinology, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- MRC/Wits Rural Public Health and Health Transitions Research Unit, School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nigel J Crowther
- Department of Chemical Pathology, National Health Laboratory Service, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Engelbert A Nonterah
- Navrongo Health Research Centre, Ghana Health Service, Navrongo, Ghana
- Department of Epidemiology, School of Public Health, C.K. Tedam University of Technology and Allied Sciences, Navrongo, Ghana
- Julius Global Health, Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Godfred Agongo
- Department of Biochemistry and Forensic Sciences, School of Chemical and Biochemical Sciences, C.K. Tedam University of Technology and Applied Sciences, Navrongo, Ghana
| | - Z Simayi
- Department of Pathology, Faculty of Health Sciences, University of Limpopo, Polokwane, South Africa
| | - Palwende R Boua
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Clinical Research Unit of Nanoro, Institut de Recherche en Sciences de la Santè, Nanoro, Burkina Faso
- MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | | | - Michèle Ramsay
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ananyo Choudhury
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Dhriti Sengupta
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
13
|
Cariou B, Thys A, Oliveira AR, Letertre MPM, Guyomarch B, Carpentier M, Cannet C, Morcel P, Ernould A, Flet L, Giraudeau P, Hadjadj S, Le May C, Croyal M. Effect of alirocumab on postprandial hyperlipidaemia in patients with type 2 diabetes: A randomized, double-blind, placebo-controlled, cross-over trial. Diabetes Obes Metab 2025; 27:3006-3016. [PMID: 40045751 DOI: 10.1111/dom.16305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/11/2025] [Accepted: 02/21/2025] [Indexed: 05/04/2025]
Abstract
AIMS Postprandial hyperlipidaemia (PPL), characterized by elevated triglyceride (TG) concentrations after a meal, is common in type 2 diabetes (T2D) and is often recognized as an independent cardiovascular risk factor. Here, we aimed to assess the effect of proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition by alirocumab on PPL in patients with T2D. MATERIALS AND METHODS EUTERPE is a randomized, double-blind, placebo-controlled cross-over trial conducted in male patients with T2D. Participants received sequentially two sequences of 10-week treatment (alirocumab 75 mg Q2W or placebo s/c) with a wash-out period of 10 weeks. The primary end-point was the percentage reduction in plasma TG response after an oral fat load (incremental area under the curve [iAUC]0-8h TG). Secondary end-points included mass spectrometry-based apolipoprotein measurements and nuclear magnetic resonance (NMR)-based lipoprotein profiling. RESULTS Fourteen participants were included: age 59 ± 9 years, BMI 32.8 ± 5.5 kg/m2, HbA1C 6.7 ± 0.5%. Compared to placebo, alirocumab did not reduce PPL (iAUC0-8h TG: -5% [CI 95%: -28, +25], p = 0.68). Alirocumab decreased fasting non-HDL cholesterol (-38.5 ± 5.6%, p = 0.0003), remnant cholesterol (-20.0 ± 13.3%, p = 0.04), apoB100 (-21.2 ± 6.4%, p = 0.004) and apoE (-15.3 ± 6.6%, p = 0.02) concentrations. NMR analyses showed that alirocumab decreased both postprandial VLDL2 cholesterol (-42% [-55, -25], p < 0.001) and IDL cholesterol (-26% [-38, -12], p = 0.0007), without effect on VLDL1 cholesterol or TG concentrations. CONCLUSIONS Inhibition of PCSK9 by alirocumab did not reduce PPL in T2D, confirming that PCSK9 controls remnant cholesterol catabolism rather than intestinal chylomicron production.
Collapse
Affiliation(s)
- Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, Nantes, France
- CHU Nantes, Inserm, CIC1413, l'institut du thorax, Nantes, France
| | - An Thys
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, Nantes, France
- CHU Nantes, Inserm, CIC1413, l'institut du thorax, Nantes, France
| | | | | | - Béatrice Guyomarch
- Plateforme de Méthodologie et Biostatistique, CHU Nantes, Nantes, France
| | - Maxime Carpentier
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, Nantes, France
- Department of Biochemistry, CHU Nantes, Nantes, France
| | | | - Pierre Morcel
- CHU Nantes, Inserm, CIC1413, l'institut du thorax, Nantes, France
| | - Audrey Ernould
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, Nantes, France
- CHU Nantes, Inserm, CIC1413, l'institut du thorax, Nantes, France
| | - Laurent Flet
- Department of Pharmacy, CHU Nantes, Nantes Université, Nantes, France
| | | | - Samy Hadjadj
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, Nantes, France
- CHU Nantes, Inserm, CIC1413, l'institut du thorax, Nantes, France
| | - Cédric Le May
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, Nantes, France
| | - Mikaël Croyal
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, Nantes, France
- Nantes Université, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes, France
| |
Collapse
|
14
|
Cefalo CMA, Riccio A, Fiorentino TV, Succurro E, Perticone M, Cassano V, Sciacqua A, Andreozzi F, Sesti G. The triglyceride glucose (TyG) index is associated with decreased myocardial mechano-energetic efficiency in individuals with different glucose tolerance status. Eur J Clin Invest 2025; 55:e70013. [PMID: 40007083 PMCID: PMC12066897 DOI: 10.1111/eci.70013] [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: 12/27/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND This investigation had two main objectives: (1) to compare the triglyceride-glucose (TyG) index with the homeostasis model assessment of insulin resistance (HOMA-IR) in relation to insulin-stimulated myocardial glucose metabolic rate (MrGlu), measured by a dynamic positron emission tomography (PET) scan using 18F-fluorodeoxyglucose (18F-FDG) coupled with a euglycemic-hyperinsulinemic clamp; and (2) to assess whether the TyG index correlates with myocardial mechano-energetic efficiency (MEE). METHODS We evaluated MrGlu in 46 participants who had no prior diagnosis of coronary heart disease. Myocardial MrGlu was quantified by 18F-FDG PET during a euglycemic-hyperinsulinemic clamp. In a larger cohort of 1820 individuals, myocardial MEE per gram of left ventricular mass (MEEi) was measured echocardiographically. The TyG index was computed as the Ln [fasting triglycerides (mg/dL) × fasting glucose (mg/dL)/2]. RESULTS When compared to HOMA-IR, the TyG index exhibited a stronger correlation with myocardial MrGlu (Pearson's r = -.566 for TyG vs. -.471 for HOMA-IR). Within the larger cohort, individuals in the highest TyG quartile showed significantly reduced MEEi compared to those in the lowest quartile (p < .001). Stepwise multivariate linear regression confirmed that the TyG index was the most significant determinant of MEEi, independent of traditional cardio-metabolic risk factors. CONCLUSIONS Our findings suggest that the TyG index is superior to HOMA-IR as an indicator of cardiac insulin resistance and that it independently correlates with MEEi. Thus, the TyG index may serve as a valuable, readily available tool to identify subjects at elevated cardiovascular risk.
Collapse
Affiliation(s)
- Chiara M. A. Cefalo
- Department of Clinical and Molecular MedicineUniversity of Rome‐SapienzaRomeItaly
| | - Alessia Riccio
- Department of Clinical and Molecular MedicineUniversity of Rome‐SapienzaRomeItaly
| | | | - Elena Succurro
- Department of Medical and Surgical SciencesUniversity Magna Graecia of CatanzaroCatanzaroItaly
| | - Maria Perticone
- Department of Medical and Surgical SciencesUniversity Magna Graecia of CatanzaroCatanzaroItaly
| | - Velia Cassano
- Department of Medical and Surgical SciencesUniversity Magna Graecia of CatanzaroCatanzaroItaly
| | - Angela Sciacqua
- Department of Medical and Surgical SciencesUniversity Magna Graecia of CatanzaroCatanzaroItaly
| | - Francesco Andreozzi
- Department of Medical and Surgical SciencesUniversity Magna Graecia of CatanzaroCatanzaroItaly
| | - Giorgio Sesti
- Department of Clinical and Molecular MedicineUniversity of Rome‐SapienzaRomeItaly
| |
Collapse
|
15
|
Fossa AJ, Hall AM, Papandonatos GD, Arbuckle TE, Ashley-Martin J, Borghese MM, Bruin J, Chen A, Fisher M, Krzeczkowski JE, Lanphear BP, MacFarlane AJ, Manz KE, Morrison KM, Oulhote Y, Palaniyandi J, Palmert MR, Pennell KD, Vuong AM, Walker DI, Weiler HA, Braun JM. Prenatal PFAS exposures and cardiometabolic health in middle childhood in the MIREC cohort. ENVIRONMENTAL RESEARCH 2025; 274:121330. [PMID: 40057105 DOI: 10.1016/j.envres.2025.121330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 05/04/2025]
Abstract
Studies on prenatal exposure to per- and polyfluoroalkyl substances (PFAS) and cardiometabolic health in childhood have produced inconsistent results. In this study, we evaluated associations between prenatal PFAS exposures, individually and as a mixture, and cardiometabolic outcomes including insulin resistance, beta cell function, blood lipids, blood pressure and central adiposity during middle childhood (7-9 years of age) in a Canadian maternal-child cohort (n = 281). We also explored effect measure modification based on child sex and physical activity. We quantified maternal second trimester plasma concentrations of six PFAS and measured 11 offspring cardiometabolic outcomes at a 7-9-year follow-up. In single-exposure models, ten-fold higher prenatal PFDA (β: -0.82, 95% CI: -1.36, -0.28), PFNA (β: -0.8, 95% CI: -1.41, -0.19), and PFOA (β: -0.69, 95% CI: -1.18, -0.19) concentrations were associated with lower diastolic blood pressure z-scores. This association did not persist when considering PFAS exposures as a mixture using quantile g-computation. Associations between PFAS exposures, individually or as a mixture, and other cardiometabolic outcomes were null. We observed no effect measure modification by child sex or physical activity (p-values for interaction ≥0.2). Our results contradict existing studies that suggest prenatal PFAS exposures are associated with adverse childhood cardiometabolic outcomes. Future studies should consider alternative markers of cardiometabolic health, trajectories in cardiometabolic health throughout childhood, and further explore potentially protective health behaviors.
Collapse
Affiliation(s)
- Alan J Fossa
- Department of Epidemiology, Brown University, Providence, RI, United States
| | - Amber M Hall
- Department of Epidemiology, Brown University, Providence, RI, United States
| | | | - Tye E Arbuckle
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, Canada
| | | | - Michael M Borghese
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, Canada
| | - Jenny Bruin
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| | - Aimin Chen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Mandy Fisher
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, Canada
| | | | - Bruce P Lanphear
- Faculty of Health Sciences, Simon Fraser University, Vancouver, BC, Canada
| | - Amanda J MacFarlane
- Nutrition Research Division, Bureau of Nutritional Sciences, Food and Nutrition Directorate, Health Products and Food Branch, Health Canada, Canada
| | - Katherine E Manz
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Katherine M Morrison
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Youssef Oulhote
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Jana Palaniyandi
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| | - Mark R Palmert
- Division of Endocrinology, Hospital for Sick Children: Departments of Pediatrics and Physiology, University of Toronto, Canada
| | - Kurt D Pennell
- School of Engineering, Brown University, Providence, RI, United States
| | - Ann M Vuong
- Department of Epidemiology and Biostatistics, University of Nevada Las Vegas, School of Public Health, Las Vegas, NV, United States
| | - Douglas I Walker
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Hope A Weiler
- Nutrition Research Division, Bureau of Nutritional Sciences, Food and Nutrition Directorate, Health Products and Food Branch, Health Canada, Canada
| | - Joseph M Braun
- Department of Epidemiology, Brown University, Providence, RI, United States.
| |
Collapse
|
16
|
Vadivelan A, Doyle E, Carson S, Denton C, Veluswamy S, Hofstra T, Coates T, Wood JC. Prevalence and Significance of Pancreatic Iron in Transfusion-Dependent Sickle Cell Disease. Pediatr Blood Cancer 2025:e31780. [PMID: 40356049 DOI: 10.1002/pbc.31780] [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: 10/23/2024] [Revised: 04/22/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Chronically transfused patients with sickle cell disease (SCD) and beta thalassemia major (TM) develop iron overload. OBJECTIVE Determine the impact of iron overload on glucose regulation in SCD. METHODS Prospective study of 28 patients with SCD and 38 patients with TM who underwent liver and pancreas R2* measurements and oral glucose tolerance tests. RESULTS Impaired fasting glucose (2 vs. 9, p = 0.27) and impaired glucose tolerance (1 vs. 11, p = 0.019) were less common in patients with SCD compared with patients with TM. No SCD patient had diabetes. CONCLUSION Iron-mediated glucose dysregulation is present but less common in SCD patients.
Collapse
Affiliation(s)
- Akhila Vadivelan
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, UCLA Mattel Children's Hospital, Los Angeles, California, USA
| | - Eamon Doyle
- Department of Clinical Radiology, USC Keck School of Medicine, Children's Hospital of Los Angeles, Los Angeles, California, USA
| | - Susan Carson
- Department of Pediatrics, Cancer and Blood Disease Institute, Section of Hematology, USC Keck School of Medicine, Children's Hospital of Los Angeles, Los Angeles, California, USA
| | - Christopher Denton
- Department of Pediatrics, Cancer and Blood Disease Institute, Section of Hematology, USC Keck School of Medicine, Children's Hospital of Los Angeles, Los Angeles, California, USA
| | - Saranya Veluswamy
- Department of Pediatrics, Cancer and Blood Disease Institute, Section of Hematology, USC Keck School of Medicine, Children's Hospital of Los Angeles, Los Angeles, California, USA
| | - Thomas Hofstra
- Department of Pediatrics, Cancer and Blood Disease Institute, Section of Hematology, USC Keck School of Medicine, Children's Hospital of Los Angeles, Los Angeles, California, USA
| | - Thomas Coates
- Department of Pediatrics, Cancer and Blood Disease Institute, Section of Hematology, USC Keck School of Medicine, Children's Hospital of Los Angeles, Los Angeles, California, USA
| | - John C Wood
- Department of Pediatrics, Division of Cardiology, USC Keck School of Medicine, Children's Hospital of Los Angeles, Los Angeles, California, USA
| |
Collapse
|
17
|
Thomas J, Filleron T, Auriol F, Laurens C, Pasquet M. Study protocol of an early randomized intervention trial assessing the metabolic effects of two levels of exercise intensity in children undergoing cancer treatment: the APACIS study. BMC Cancer 2025; 25:850. [PMID: 40346598 PMCID: PMC12065348 DOI: 10.1186/s12885-025-14235-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Insulin sensitivity is a key factor of the development of metabolic diseases, highly prevalent in adult survivors of childhood cancers. The aim of the Adapted Physical Activity for children treated for Cancer and Insulin-Sensitivity (APACIS) study is to investigate the effects of two exercise programs started as early as diagnosis on metabolic profile and physical health. METHODS APACIS is a trial that includes children at diagnosis of all pediatric cancers which are randomly allocated to the Soft group - for low intensity physical activity - or to the Strong group - for mixed, high intensity exercise. Both programs are done at least twice weekly for 30 to 60 min over 6 months, adapted to the health status of the children, with a follow-up of 18 months. The primary objective is the change in insulin sensitivity measured by the Homeostasis Model Assessment of Insulin Resistance (HOMA-IR) at 0, 3, 6, 12 and 24 months between the two groups. The secondary objectives include changes in cholesterol, triglycerides and cortisol blood levels, state of undernourishment, cardiorespiratory fitness (based on peak rate of oxygen uptake of the 6 Minute Walk Test), flexibility (sit and reach flexibility test), fat mass distribution (Waist-to-Hip Ratio) and level of physical activity assessed by questionnaire at 0, 3, 6, 12 and 24 months. On-therapy metabolic adaptation in the different patient groups will also be evaluated by an integrated pipeline combining the detection of 150 metabolites, with metabolic pathway enrichment and network mapping. This approach will be complemented by an analysis of intestinal and oral microbiota, to identify the species impacted by treatments and the influence of exercise on these toxicities. DISCUSSION The APACIS study investigates the metabolic, motor, and nutritional effects in children with cancers performing low versus high intensity exercise with an innovative approach consisting of early practice since diagnosis. It will contribute to better personalize physical activity prescription during treatment of pediatric cancers. TRIAL REGISTRATION The study has been registered on ClinicalTrials.gov (NCT05383092) the 7 of November, 2022.
Collapse
Affiliation(s)
- Justine Thomas
- Department of Pediatric Hematology and Immunology, Toulouse University Hospital Center, 339 Avenue de Grande Bretagne, Toulouse, 31059, France
- University of Toulouse, Institute of Metabolic and Cardiovascular Diseases, UMR 1297 Inserm, Team Metadiab, Toulouse, France
| | - Thomas Filleron
- Department of Biostatistics, Claudius-Regaud Institute, Toulouse University Cancer Institute Oncopole, Toulouse, France
| | - Françoise Auriol
- Pediatric Clinical Research Unit, CIC1436, Children Hospital, Toulouse University Hospital Center, Toulouse, France
| | - Claire Laurens
- University of Toulouse, Institute of Metabolic and Cardiovascular Diseases, UMR 1297 Inserm, Team Metadiab, Toulouse, France
| | - Marlène Pasquet
- Department of Pediatric Hematology and Immunology, Toulouse University Hospital Center, 339 Avenue de Grande Bretagne, Toulouse, 31059, France.
- University of Toulouse, Toulouse Cancer Research Center, UMR1037 Inserm, UMR5077 CNRS, Toulouse, France.
| |
Collapse
|
18
|
Yuan L, Wang S, Wang D, Wang E. Association of cardiometabolic index with gallstone disease and insulin resistance based on NHANES data. BMC Gastroenterol 2025; 25:354. [PMID: 40346457 PMCID: PMC12063462 DOI: 10.1186/s12876-025-03950-8] [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: 01/15/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Cardiometabolic index (CMI) is an index integrating visceral obesity and dyslipidemia. This study intends to scrutinize the connection between CMI and gallstone disease (GSD) and to elucidate the association between CMI and insulin resistance (IR) in patients with GSD. METHODS To explore the potential nonlinear association and determine the inflection point, a restricted cubic spline (RCS) analysis was performed. Following categorization of CMI based on the identified inflection point, multivariate logistic regression models, subgroup analyses, and interaction tests were utilized to assess the connection between CMI and GSD, as well as between CMI and IR in GSD patients. The homeostasis model assessment for IR (HOMA-IR) and triglyceride-glucose (TyG) index was applied to evaluate IR. Spearman analysis was implemented to investigate the connection between CMI and HOMA-IR. The predictive performance of each indicator was evaluated by the receiver operating characteristic (ROC) curve and the area under the curve (AUC). RESULTS The study included 2311 individuals, with a GSD prevalence of 10.90%. RCS analysis revealed a nonlinear positive correlation between CMI and GSD (nonlinear P < 0.001), as well as between CMI and IR (nonlinear P < 0.001). In the fully adjusted multivariable logistic regression analysis of covariates, compared with the low-category CMI group, the high-category CMI was significantly associated with the risk of GSD (OR = 1.547, 95% CI: 1.143-2.092, P = 0.005), IR (OR = 4.990, 95% CI: 2.517-9.892, P < 0.001). Subgroup analysis demonstrated that the correlation between CMI and GSD was stronger in females. Spearman correlation analysis showed a positive association between CMI and HOMA-IR in GSD patients (r = 0.548, P < 0.001). The ROC curve demonstrated the predictive performance of the CMI model for GSD (AUC = 0.743), which was superior to conventional indicators such as Body Mass Index and Waist Circumference; the predictive performance of CMI (AUC = 0.772) for IR was consistent with that of TyG (AUC = 0.772). CONCLUSION Our research demonstrates that CMI exhibits a nonlinear positive correlation with the incidence of GSD and IR. This suggests that CMI may serve as a novel and valuable indicator for further investigating the intricate relationships among metabolic syndrome, obesity, and GSD.
Collapse
Affiliation(s)
- Liu Yuan
- China Medical University, Shenyang, China
- Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital), Dalian, China
| | - Shuqi Wang
- Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital), Dalian, China
| | - Dong Wang
- Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital), Dalian, China
| | - Enbo Wang
- Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital), Dalian, China.
| |
Collapse
|
19
|
Koay YC, McIntosh B, Ng YH, Cao Y, Wang XS, Han Y, Tomita S, Bai AY, Hunter B, Misra A, Loughrey CM, Bannon PG, Lal S, Lusis AJ, Kaye DM, Larance M, O’Sullivan JF. The Heart Has Intrinsic Ketogenic Capacity that Mediates NAD + Therapy in HFpEF. Circ Res 2025; 136:1113-1130. [PMID: 40211954 PMCID: PMC12063684 DOI: 10.1161/circresaha.124.325550] [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: 09/13/2024] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) has overtaken heart failure with reduced ejection fraction as the leading type of heart failure globally and is marked by high morbidity and mortality rates, yet with only a single approved pharmacotherapy: SGLT2i (sodium-glucose co-transporter 2 inhibitor). A prevailing theory for the mechanism underlying SGLT2i is nutrient deprivation signaling, of which ketogenesis is a hallmark. However, it is unclear whether the canonical ketogenic enzyme, HMGCS2 (3-hydroxy-3-methylglutaryl-coenzyme A synthase 2), plays any cardiac role in HFpEF pathogenesis or therapeutic response. METHODS We used human myocardium, human HFpEF and heart failure with reduced ejection fraction transcardiac blood sampling, an established murine model of HFpEF, ex vivo Langendorff perfusion, stable isotope tracing in isolated cardiomyocytes, targeted metabolomics, proteomics, lipidomics, and a novel cardiomyocyte-specific conditional HMGCS2-deficient model that we generated. RESULTS We demonstrate, for the first time, the intrinsic capacity of the human heart to produce ketones via HMGCS2. We found that increased acetylation of HMGCS2 led to a decrease in the enzyme's specific activity. However, this was overcome by an increase in the steady-state levels of protein. Oxidized form of nicotinamide adenine dinucleotide repletion restored HMGCS2 function via deacetylation, increased fatty acid oxidation, and rescued cardiac function in HFpEF. Critically, using a conditional, cardiomyocyte-specific HMGCS2 knockdown murine model, we revealed that the oxidized form of nicotinamide adenine dinucleotide is unable to rescue HFpEF in the absence of cardiomyocyte HMGCS2. CONCLUSIONS The canonical ketogenic enzyme, HMGCS2, mediates the therapeutic effects of the oxidized form of nicotinamide adenine dinucleotide repletion in HFpEF by restoring normal lipid metabolism and mitochondrial function.
Collapse
Affiliation(s)
- Yen Chin Koay
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Cardiometabolic Medicine (Y.C.K., B.M., Y.H.N., X.W., Y.H., P.G.B., S.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
| | - Bailey McIntosh
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Cardiometabolic Medicine (Y.C.K., B.M., Y.H.N., X.W., Y.H., P.G.B., S.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
| | - Yann Huey Ng
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Cardiometabolic Medicine (Y.C.K., B.M., Y.H.N., X.W., Y.H., P.G.B., S.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
| | - Yang Cao
- Division of Life Sciences and Medicine, Department of Cardiology, The First Affiliated Hospital of USTC (Y.C.), University of Science and Technology of China (USTC), Hefei
- Division of Life Sciences and Medicine, School of Basic Medical Sciences (Y.C.), University of Science and Technology of China (USTC), Hefei
| | - Xiao Suo Wang
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Cardiometabolic Medicine (Y.C.K., B.M., Y.H.N., X.W., Y.H., P.G.B., S.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
| | - Yanchuang Han
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Cardiometabolic Medicine (Y.C.K., B.M., Y.H.N., X.W., Y.H., P.G.B., S.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
| | - Saki Tomita
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
| | - Angela Yu Bai
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
| | - Benjamin Hunter
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Precision Cardiovascular Laboratory (B.H., S.L.), The University of Sydney, New South Wales, Australia
| | - Ashish Misra
- Heart Research Institute (A.M.), The University of Sydney, New South Wales, Australia
| | - Christopher M. Loughrey
- School of Cardiovascular and Metabolic Health and School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom (C.M.L.)
| | - Paul G. Bannon
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Cardiometabolic Medicine (Y.C.K., B.M., Y.H.N., X.W., Y.H., P.G.B., S.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Department of Cardiothoracic Surgery (P.G.B., J.F.O.), Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- The Baird Institute for Applied Heart and Lung Surgical Research, Sydney, New South Wales, Australia (P.G.B., S.L., J.F.O.)
| | - Sean Lal
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Cardiometabolic Medicine (Y.C.K., B.M., Y.H.N., X.W., Y.H., P.G.B., S.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Precision Cardiovascular Laboratory (B.H., S.L.), The University of Sydney, New South Wales, Australia
- Department of Cardiology (S.L., J.F.O.), Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- The Baird Institute for Applied Heart and Lung Surgical Research, Sydney, New South Wales, Australia (P.G.B., S.L., J.F.O.)
| | - Aldons J. Lusis
- Department of Medicine, Microbiology and Human Genetics, University of California, Los Angeles (A.J.L.)
| | - David M. Kaye
- Department of Cardiology, Alfred Hospital, Melbourne, Victoria, Australia (D.M.K.)
- Heart Failure Group, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.M.K.)
- Faculty of Medicine, Nursing, and Health Sciences, Central Clinical School, Monash University, Melbourne, Victoria, Australia (D.M.K.)
| | - Mark Larance
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
| | - John F. O’Sullivan
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Cardiometabolic Medicine (Y.C.K., B.M., Y.H.N., X.W., Y.H., P.G.B., S.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Department of Cardiothoracic Surgery (P.G.B., J.F.O.), Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Department of Cardiology (S.L., J.F.O.), Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- The Baird Institute for Applied Heart and Lung Surgical Research, Sydney, New South Wales, Australia (P.G.B., S.L., J.F.O.)
- Faculty of Medicine, Technische Universität Dresden, Germany (J.F.O.)
| |
Collapse
|
20
|
Chen W, Luo M, Guo J, Wang S, Yan D, Feng X, Huang Y, Zeng T, Shen L, Zhang R, Yan J, Hu C, Zhang W, Yu X. Metabolic pathways mediating insulin resistance and gestational diabetes mellitus discovered by high-dimensional systematic Mendelian randomization. Cardiovasc Diabetol 2025; 24:195. [PMID: 40346526 PMCID: PMC12065323 DOI: 10.1186/s12933-025-02746-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/18/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM), characterized by insulin resistance (IR) and β-cell dysfunction, is one of the most common complications of pregnancy with unmet needs of prevention methods. OBJECTIVE To investigate the causal role of insulin resistance and metabolic pathways in the pathogenesis of GDM with our proposed high-dimensional systematic Mendelian randomization (hdsMR) framework. METHODS Cases with GDM and controls with normal glucose tolerance were recruited at the University of Hong Kong-Shenzhen Hospital from 2015 to 2018. A total of 566 participants (aged > 18 years), including 274 with GDM, were enrolled after excluding subjects with major chronic diseases or long-term use of medications affecting glycolipid metabolism. Clinical characteristics and serum samples were collected during the GDM screening stage, and the genome and metabolome were tested. A novel hdsMR framework was proposed to estimate the causal role of IR index (Homeostasis Model Assessment of Insulin Resistance, HOMA-IR) and metabolic pathways in the pathogenesis of GDM. RESULTS Our hdsMR method confirmed that HOMA-IR was causal to GDM (odds ratio, 1.17; 95% confidence interval, 1.04-1.32) and revealed that two metabolic pathways (glyoxylate and dicarboxylate metabolism pathway and lysine degradation pathway) mediated 14.6% and 8.4%, respectively, between HOMA-IR and GDM. In an independent validation cohort comprising 255 pre-diabetic individuals, we showed that both pathways could be intervened through diet (P < 0.05). Furthermore, glyoxylate and dicarboxylate metabolism pathway was significantly associated with adverse pregnancy outcomes in GDM. CONCLUSIONS These results indicated that targeting specific metabolic pathways through dietary intervention is worth exploring as a possible GDM prevention approach, and hdsMR is more efficient in finding causal mediating metabolic pathways than traditional MR methods.
Collapse
Affiliation(s)
- Wei Chen
- Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingjuan Luo
- Department of Endocrinology and Metabolism, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jingyi Guo
- Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Suna Wang
- Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dandan Yan
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Diabetes Institute, Shanghai Clinical Center of Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Xiaohui Feng
- Department of Endocrinology and Metabolism, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yunting Huang
- Department of Endocrinology and Metabolism, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Tao Zeng
- Guangzhou National Laboratory, Guangzhou, China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| | - Li Shen
- Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Zhang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Yan
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Weituo Zhang
- School of Public Health, Clinical Research Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiangtian Yu
- Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
21
|
Zeng H, Liu C, Wan L, Peng L, Wang K, Zhou F, Fang W, Wen S, Bai Q, Yang X, Liu L, Zeng J, Huang J, Liu Z. Epigallocatechin gallate prevents and alleviates type 2 diabetes mellitus (T2DM) through gut microbiota and multi-organ interactions in Wistar healthy rats and GK T2DM rats. J Adv Res 2025:S2090-1232(25)00296-6. [PMID: 40349958 DOI: 10.1016/j.jare.2025.05.002] [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: 03/06/2025] [Revised: 04/21/2025] [Accepted: 05/02/2025] [Indexed: 05/14/2025] Open
Abstract
INTRODUCTION As the main active ingredient of the first FDA-approved phytochemical drug, epigallocatechin gallate (EGCG) can effectively alleviate glucolipid metabolic disorders. However, existing studies mainly focuses on the treatment of EGCG in disease models, with limited focus on its preventive effect on diseases in healthy models. OBJECTIVES This study investigated how EGCG prevents and alleviates T2DM through gut microbiota and multi-organ interactions in Wistar healthy rats and GK T2DM rats. METHODS The GK T2DM rat strain was established through repeated selective breeding of Wistar rats with glucose intolerance. Whether and how EGCG prevents and alleviates T2DM were evaluated, including glucose production and absorption efficiency, glucose transport, glucose metabolism, glucose excretion, T2DM-related tissue damage, gut microbiota, and liver transcriptome. RESULTS The health benefits of EGCG are primarily reliant on the involvement of the gut microbiota. Our study showed that although the specific microbial communities involved differ, the bidirectional interaction between EGCG and gut microbiota is widespread in healthy rats and T2DM rats. EGCG intervention elevated the relative abundance of specific microbial communities, which in turn promoted the metabolic processing of EGCG in the gut, producing numerous EGCG metabolites that may contribute to preventing and alleviating T2DM. In healthy rats, EGCG intervention selectively enhanced insulin secretion and serum insulin levels to prevent T2DM. In T2DM rats, EGCG intervention selectively lowered blood glucose levels, improved insulin resistance, delayed glucose production and absorption, and promoted urinary glucose excretion to alleviate T2DM. In both healthy and T2DM rats, EGCG intervention universally reduced gut microbiota-derived lipopolysaccharides, maintained systemic oxidative stress homeostasis, alleviated liver and kidney damage, increased muscle glycogen content, and promoted beige thermogenesis in white fat, thus demonstrating potential for preventing and alleviating T2DM. CONCLUSION As a natural active ingredient, EGCG could prevent and alleviate T2DM through gut microbiota and multi-organ interactions.
Collapse
Affiliation(s)
- Hongzhe Zeng
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Changwei Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Liwei Wan
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Liyuan Peng
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Kuofei Wang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Fang Zhou
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Wenwen Fang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Shuai Wen
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Qixian Bai
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Xiaomei Yang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Linmei Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Jie Zeng
- Micangshan Tea Industry Research Institute, Sichuan Wangcang Vocational Middle School, Sichuan 628200, China
| | - Jian'an Huang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China.
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
22
|
Choi J, Kim J, Oh HS. Relationship between insulin resistance surrogate markers with diabetes and dyslipidemia: A Bayesian network analysis of Korean adults. PLoS One 2025; 20:e0323329. [PMID: 40341273 PMCID: PMC12061414 DOI: 10.1371/journal.pone.0323329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/04/2025] [Indexed: 05/10/2025] Open
Abstract
Insulin resistance (IR) can be optimally assessed using the euglycemic clamp, but practical clinical limitations necessitate surrogate markers. This study leveraged the Bayesian network analysis to evaluate three established IR markers: the Homeostatic Model Assessment of IR (HOMA-IR) using insulin level and fasting blood glucose (FBG), TG-Glucose (TyG) index using triglycerides (TG) and FBG, and TG-to-HDL ratio (TG/HDL ratio) using TG and high-density lipoprotein (HDL), based on the Korean National Health and Nutrition Examination Survey data (2019-2021). Our analysis revealed a sequential association pattern (TG/HDL ratio → TyG index → HOMA-IR), positioning the TyG index as a central connecting marker. The HOMA-IR exhibited strong predictive power for diabetes, while the TG/HDL ratio was most effective for assessing dyslipidemia. However, both had limited crossover utility. In contrast, the TyG index bridged this gap, demonstrating robust predictive capability for both conditions. The Markov blanket analysis illuminated the distinctive metabolic signatures of each marker: The TyG index displayed balanced glucose-lipid metabolic contributions, the HOMA-IR predominantly reflected glucose metabolism and obesity characteristics, and the TG/HDL ratio emphasized lipid metabolism. Notably, the TyG index's predictive performance showed significant enhancement when integrated with obesity information, contrasting with the HOMA-IR's minimal response owing to its inherent incorporation of obesity characteristics. These findings position the TyG index as a superior clinical marker, offering both comprehensive predictive capability and enhanced performance through synergistic integration with obesity measures. While each marker demonstrated reliability, the TyG index's unique combination of versatility and scalability establishes it as an effective tool for comprehensive metabolic risk assessment.
Collapse
Affiliation(s)
- Jaeyeop Choi
- Department of Applied Statistics, Gachon University, Seongnam-si, Gyeonggi-do, Korea
| | - Jonghyun Kim
- Department of Applied Statistics, Gachon University, Seongnam-si, Gyeonggi-do, Korea
| | - Hyun Sook Oh
- Department of Applied Statistics, Gachon University, Seongnam-si, Gyeonggi-do, Korea
| |
Collapse
|
23
|
Romeo S, Vidal-Puig A, Husain M, Ahima R, Arca M, Bhatt DL, Diehl AM, Fontana L, Foo R, Frühbeck G, Kozlitina J, Lonn E, Pattou F, Plat J, Quaggin SE, Ridker PM, Rydén M, Segata N, Tuttle KR, Verma S, Roeters van Lennep J, Benn M, Binder CJ, Jamialahmadi O, Perkins R, Catapano AL, Tokgözoğlu L, Ray KK. Clinical staging to guide management of metabolic disorders and their sequelae: a European Atherosclerosis Society consensus statement. Eur Heart J 2025:ehaf314. [PMID: 40331343 DOI: 10.1093/eurheartj/ehaf314] [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] [Indexed: 05/08/2025] Open
Abstract
Obesity rates have surged since 1990 worldwide. This rise is paralleled by increases in pathological processes affecting organs such as the heart, liver, and kidneys, here termed systemic metabolic disorders (SMDs). For clinical management of SMD, the European Atherosclerosis Society proposes a pathophysiology-based system comprising three stages: Stage 1, where metabolic abnormalities such as dysfunctional adiposity and dyslipidaemia occur without detectable organ damage; Stage 2, which involves early organ damage manifested as Type 2 diabetes, asymptomatic diastolic dysfunction, metabolic-associated steatohepatitis (MASH), and chronic kidney disease (CKD); and Stage 3, characterized by more advanced organ damage affecting multiple organs. Various forms of high-risk obesity, driven by maintained positive energy balance, are the most common cause of SMD, leading to ectopic lipid accumulation and insulin resistance. This progression affects various organs, promoting comorbidities such as hypertension and atherogenic dyslipidaemia. Genetic factors influence SMD susceptibility, and ethnic disparities in SMD are attributable to genetic and socioeconomic factors. Key SMD features include insulin resistance, inflammation, pre-diabetes, Type 2 diabetes, MASH, hypertension, CKD, atherogenic dyslipidaemia, and heart failure. Management strategies involve lifestyle changes, pharmacotherapy, and metabolic surgery in severe cases, with emerging treatments focusing on genetic approaches. The staging system provides a structured approach to understanding and addressing the multi-faceted nature of SMD, which is crucial for improving health outcomes. Categorization of SMD abnormalities by presence and progression is aimed to improve awareness of a multi-system trait and encourage a tailored and global approach to treatment, ultimately aiming to reduce the burden of obesity-related comorbidities.
Collapse
Affiliation(s)
- Stefano Romeo
- Department of Medicine, H7 Medicin, Huddinge, H7 Endokrinologi och Diabetes Romeo, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Endocrinology, Karolinska University Hospital Huddinge, 141 57 Huddinge, Stockholm, Sweden
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Viale Europa, 88100 Catanzaro, Italy
| | - Antonio Vidal-Puig
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
- Centro de Investigacion Principe Felipe, C/ d'Eduardo Primo Yufera, 3, 46012 Valencia, Spain
- Cambridge University Nanjing Centre of Technology and Innovation, No. 23, Rongyue Road, Jiangbei New Area, Nanjing, Jiangsu, China
| | - Mansoor Husain
- Ted Rogers Centre for Heart Research, Department of Medicine, University of Toronto, 661 University Avenue, Toronto, ON, Canada M5G 1M1
| | - Rexford Ahima
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marcello Arca
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
- Unit of Internal Medicine and Metabolic Diseases, Hospital Policlinico Umberto I, Rome, Italy
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, NC, USA
| | - Luigi Fontana
- Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Roger Foo
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, National University Health Systems, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Gema Frühbeck
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
- Metabolic Research Laboratory, CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | - Julia Kozlitina
- The Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eva Lonn
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada
| | - Francois Pattou
- Department of Endocrine and Metabolic Surgery, CHU Lille, University of Lille, Inserm, Institut Pasteur Lille, Lille, France
| | - Jogchum Plat
- Department of Nutrition and Movement Sciences, NUTRIM School of Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Susan E Quaggin
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Nephrology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Katherine R Tuttle
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA, USA
- Providence Medical Research Center, Providence Inland Northwest Health, Spokane, WA, USA
| | - Subodh Verma
- Division of Cardiac Surgery, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Jeanine Roeters van Lennep
- Department of Internal Medicine, Cardiovascular Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marianne Benn
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, Centre of Diagnostic Investigation, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Oveis Jamialahmadi
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Rosie Perkins
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Alberico L Catapano
- Center for the Study of Atherosclerosis, IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Lale Tokgözoğlu
- Department of Cardiology, Hacettepe University Medical Faculty, Ankara, Turkey
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College, London, UK
| |
Collapse
|
24
|
Amirkhizi F, Taghizadeh M, Hamedi-Shahraki S, Asghari S. Association of dietary phytochemical index with metabolic markers, serum asymmetric dimethylarginine and atherogenic indices in patients with polycystic ovary syndrome. Nutr Metab (Lond) 2025; 22:39. [PMID: 40336098 PMCID: PMC12060492 DOI: 10.1186/s12986-025-00932-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is associated with an increased risk of cardiovascular diseases (CVD). Plant-based diets are associated with reduced CVD risk factors. This study aimed to explore the associations between dietary phytochemical index (DPI) and asymmetric dimethylarginine (ADMA), lipid profile, atherogenic indices, and other metabolic biomarkers in women with PCOS. METHODS In this cross-sectional study, 150 females aged 18-45 years diagnosed with PCOS were recruited. An interviewer-administered questionnaire was applied to gather the relevant demographic characteristics, detailed clinical information, and lifestyle habits of participants. A validated semi-quantitative food frequency questionnaire was used to assess dietary intake, and DPI was calculated accordingly. We used multiple linear regression to determine the association between serum concentrations of ADMA, total testosterone, sex hormone-binding globulin (SHBG), fasting serum glucose (FSG), insulin, and lipid profile, as well as atherogenic indices across quartiles of DPI. RESULTS There was a negative correlation between the DPI and serum levels of ADMA (p-trend = 0.022), triglycerides (TG) (p-trend = 0.003), oxidized low-density lipoprotein cholesterol (ox-LDL) (p-trend = 0.001), insulin (p-trend = 0.045) and Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) (p-trend = 0.018). Moreover, there was a tendency for visceral adiposity index (VAI) (p-trend = 0.005) and atherogenic index of plasma (AIP) (p-trend = 0.001) to decrease as the quartile categories of DPI increased. No significant regular trend was found for serum levels of FSG, SHBG, total testosterone, other lipid profiles, and lipid accumulation product (LAP). CONCLUSIONS These findings suggest that adherence to a phytochemical-rich diet decrease the CVD risk factors in PCOS patients.
Collapse
Affiliation(s)
- Farshad Amirkhizi
- Department of Nutrition, School of Public Health, Zabol University of Medical Sciences, Zabol, Iran
| | - Mahdiyeh Taghizadeh
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoust St., Naderi St., Keshavarz Blvd, Tehran, 141556117, Iran
| | - Soudabeh Hamedi-Shahraki
- Department of Epidemiology and Biostatistics, Faculty of Public Health, Zabol University of Medical Sciences, Bagheri St., Shahid Rajaei St, Zabol, 9861615881, Iran.
| | - Somayyeh Asghari
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoust St., Naderi St., Keshavarz Blvd, Tehran, 141556117, Iran.
| |
Collapse
|
25
|
Zhang S, Wang Y, Wang X, Leng M, Liu R, Li Z, Li J, Xiao J, Hou D, Gao X, Li C. Effect of hypoglycemic agents with weight loss effect plus a high protein diet and moderate exercise on diabetes remission in adults with obesity and type 2 diabetes: a randomized controlled trial. BMC Med 2025; 23:270. [PMID: 40336033 PMCID: PMC12060396 DOI: 10.1186/s12916-025-04072-4] [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: 06/24/2024] [Accepted: 04/10/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND This study aimed to explore the effects of hypoglycemic agents with weight loss effect plus a high protein diet and moderate exercise on weight loss and diabetes remission in adults with obesity and newly diagnosed prediabetes/type 2 diabetes (T2D). METHODS Participants with obesity and newly diagnosed prediabetes or T2D (n = 61) were randomly allocated to standard treatment group (conventional medication and lifestyle guidance for 12 months) and intensive treatment group (in addition to conventional medication, a high protein diet and moderate exercise were given for 12 months). RESULTS By month 12, 60 (98.4%) participants completed the 12-month follow-up visit. In the intensive treatment group, 73.33% patients in the prediabetes subgroup returned to normoglycemia and the diabetes remission rate was 86.67% in the diabetes subgroup, which were much higher than the remission rate of prediabetes subgroup (7.69%) and diabetes subgroup (16.67%) in the standard treatment group (P < 0.001). The mean weight change was - 19.29 kg (95% CI, - 22.95 to - 15.63) in the intensive treatment group and - 1.52 kg (95% CI, - 5.12 to 2.07) in the standard treatment group from baseline after intervention. The weight change between the two groups was significantly different (net difference, - 17.77 kg; 95% CI, - 22.90 to - 12.64; P < 0.001). Percent of body fat, visceral fat area, and hepatic controlled attenuation parameter value reduced significantly in the intensive treatment group compared to the standard treatment group (P < 0.001). CONCLUSIONS Hypoglycemic agents with weight loss effect plus a high protein diet and moderate exercise could lead to a considerable proportion of patients with diabetes achieving diabetes remission. TRIAL REGISTRATION chictr.org.cn ChiCTR2100044305.
Collapse
Affiliation(s)
- Shi Zhang
- Department of Health Management Center, Endocrinology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Yi Wang
- Department of Health Management Center, Endocrinology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Xincheng Wang
- Department of Health Management Center, Endocrinology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Mingxin Leng
- Department of Health Management Center, Endocrinology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Renjiao Liu
- Department of Health Management Center, Endocrinology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Zhouhuiling Li
- Department of Health Management Center, Endocrinology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Jing Li
- Department of Health Management Center, Endocrinology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Jixuan Xiao
- Department of Health Management Center, Endocrinology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Dangmin Hou
- Department of Health Management Center, Endocrinology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Xinying Gao
- Department of Health Management Center, Endocrinology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Chunjun Li
- Department of Health Management Center, Endocrinology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China.
| |
Collapse
|
26
|
Yamashita A, Kaku M, Ideguchi T, Nishida S, Kinoshita H, Nishikawa T. Association between gestational weight gain and pregnancy outcomes, neonatal birth weight, and maternal postpartum glucose tolerance in Japanese gestational diabetes mellitus patients: comparison of old and new gestational weight gain standards. Endocr J 2025; 72:509-524. [PMID: 39909435 DOI: 10.1507/endocrj.ej24-0486] [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] [Indexed: 02/07/2025] Open
Abstract
In Japan, the guidelines for gestational weight gain (GWG) were revised in 2021. Under the new guidelines, pregnant women are recommended to increase their GWG. The aim of this study was to compare the incidence of adverse pregnancy outcomes (APOs), large for gestational age (LGA), and postpartum glucose tolerance in gestational diabetes mellitus (GDM) patients before and after the revised GWG standards. This retrospective cohort study enrolled 1,021 GDM patients who underwent prenatal glycemic control and a postpartum 75-g oral glucose tolerance test. The endpoint was the incidence of APOs, LGA, and postpartum impaired glucose tolerance (IGT) and diabetes mellitus (DM). There was no significant difference in the incidence of APOs and postpartum IGT and DM in GDM patients before and after the revised GWG standards. On the other hand, when the new GWG standards were applied to GDM patients, the incidence of LGA increased (adjusted odds ratio [aOR]; 1.764, 95% confidence interval [CI]; 1.180-2.637). In particular, when classified by pre-pregnancy body mass index, the incidence of LGA increased in the obese group (aOR; 5.944, 95% CI; 1.847-19.129). Future prospective cohort studies are needed to verify the efficacy and safety of appropriate GWG in Japanese GDM patients.
Collapse
Affiliation(s)
- Akiho Yamashita
- Department of Nutrition, NHO Kumamoto Medical Center, Kumamoto 860-0008, Japan
- Department of International Medical Cooperation, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Masayuki Kaku
- Department of Nutrition, NHO Kumamoto Medical Center, Kumamoto 860-0008, Japan
| | - Takuya Ideguchi
- Department of Diabetes and Endocrinology, NHO Kumamoto Medical Center, Kumamoto 860-0008, Japan
| | - Shuhei Nishida
- Department of Diabetes and Endocrinology, NHO Kumamoto Medical Center, Kumamoto 860-0008, Japan
| | - Hiroyuki Kinoshita
- Department of Diabetes and Endocrinology, NHO Kumamoto Medical Center, Kumamoto 860-0008, Japan
| | - Takeshi Nishikawa
- Department of International Medical Cooperation, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
- Department of Diabetes and Endocrinology, NHO Kumamoto Medical Center, Kumamoto 860-0008, Japan
| |
Collapse
|
27
|
Al-Dallal R, Thomas K, Lee M, Chaudhri A, Davis E, Vaidya P, Lee M, McCormick JB, Fisher-Hoch SP, Gutierrez AD. The Association of Resistin with Metabolic Health and Obesity in a Mexican-American Population. Int J Mol Sci 2025; 26:4443. [PMID: 40362681 DOI: 10.3390/ijms26094443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
Research on the relationship between resistin levels, metabolic health, and obesity has produced inconsistent findings across different ethnic groups, making it unclear whether lower resistin levels are associated with these conditions in Mexican-Americans. This cross-sectional study investigated the relationship between resistin, metabolic health, and obesity in an adult Mexican-American cohort (n = 1511) using multivariable linear regression analysis. Related adipokines (leptin and adiponectin) were measured simultaneously. Participants were categorized into four groups by metabolic health (healthy/unhealthy) and obesity (obese/non-obese) status. "Metabolically unhealthy" was defined as ≥2 cardiometabolic abnormalities. Obesity was defined as a BMI ≥ 30 kg/m2. We also investigated the associations of related proinflammatory cytokines, demographic/anthropometric variables, and medications with each outcome variable of interest. The results showed no statistically significant differences in resistin levels between the groups. Leptin was higher and adiponectin was lower in groups with obesity and/or metabolically unhealthy status. The resistin findings contrast studies in other populations, while other leptin and adiponectin findings confirm those seen in many ethnic groups. Thiazolidinedione use was associated with lower resistin, confirming earlier research. These findings suggest that resistin's role in metabolic health may be different in Mexican-Americans compared to other populations.
Collapse
Affiliation(s)
- Reem Al-Dallal
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - Keziah Thomas
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - MinJae Lee
- Department of Health Data Science & Biostatistics, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern, Dallas, TX 75390, USA
| | - Aysha Chaudhri
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Eleanor Davis
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - Priyanka Vaidya
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - Miryoung Lee
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Brownsville Regional Campus, Brownsville, TX 78520, USA
| | - Joseph B McCormick
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Brownsville Regional Campus, Brownsville, TX 78520, USA
| | - Susan P Fisher-Hoch
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Brownsville Regional Campus, Brownsville, TX 78520, USA
| | - Absalon D Gutierrez
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, The University of Texas Health Science Center, Houston, TX 77030, USA
| |
Collapse
|
28
|
Suleman S, Ängquist L, Linneberg A, Hansen T, Grarup N. Exploring the genetic intersection between obesity-associated genetic variants and insulin sensitivity indices. Sci Rep 2025; 15:15761. [PMID: 40328835 PMCID: PMC12056085 DOI: 10.1038/s41598-025-98507-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
Insulin sensitivity (IS) is a key determinant of metabolic health and may share genetic factors with obesity-related traits. Previous large-scale genetic studies have identified variants associated with IS as well as obesity related traits like body mass index (BMI) and waist-to-hip ratio (WHR). Notably, many of these associations are shared across traits, indicating a potential genetic overlap. However, the genetic intersection between IS and obesity-related traits remains underexplored. To explore this gap, we investigated associations between six IS indices, including fasting and post-glucose load measures, and genetic variants linked to BMI and WHR to determine their influence on IS and related cardiometabolic traits. To achieve this, we calculated six IS indices using fasting and oral glucose tolerance test (OGTT) data from 5,007 non-diabetic individuals, grouping them into fasting, OGTT0,120, and OGTT0,30,120 categories. A total of 678 BMI-associated and 265 WHR-associated genetic variants were analysed using linear regression, adjusting for age and sex, with sex-specific analyses for WHR. Analyses were conducted with and without BMI adjustments and corrected for multiple testing (padj). Additionally, we explored the relationship between IS-linked variants and their associations with type 2 diabetes (T2D), coronary artery disease (CAD) and stroke. Among the 678 BMI-associated variants, 100 showed nominal associations (p < 0.05) with at least one IS index; and 20 remained significant after multiple testing correction (padj < 0.05) when not adjusting for BMI. After adjusting for BMI, 70 variants retained nominal associations, and six remained significant (padj < 0.05). In sex-specific analyses of the 265 WHR-associated variants, 12 variants were associated in females when adjusted for BMI, whereas no significant associations were observed in males. Furthermore, BMI- and WHR-associated variants linked to decreased IS, such as those in FTO and VPS13C loci, were also associated with increased T2D and stroke risk, whereas IS-increasing variants, including those in VPS13C and PPARG, were linked to lower T2D and stroke risk, with some, like THADA, showing opposing effects on CAD. This study offers insights into genetic variants that influence both IS and obesity-related traits, revealing BMI- and WHR-associated variants with both positive and negative effects on IS and their potential impact on cardiometabolic health.
Collapse
Affiliation(s)
- Sufyan Suleman
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedicine, Human Genetics, Aarhus University, Aarhus, 8000, Denmark
| | - Lars Ängquist
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
29
|
Wu C, Ke Y, Nianogo RA. Trends in Hyperinsulinemia and Insulin Resistance Among Nondiabetic US Adults, NHANES, 1999-2018. J Clin Med 2025; 14:3215. [PMID: 40364246 DOI: 10.3390/jcm14093215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/23/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction: Hyperinsulinemia and insulin resistance are strong predictors of cardiometabolic diseases, which disproportionately affect individuals across gender, racial/ethnic, and socioeconomic groups. We aim to estimate and test the temporal trends in the prevalence of hyperinsulinemia and insulin resistance (IR) by sociodemographic groups among nondiabetic adults in the United States from 1999 to 2018. Methods: We used data from the National Health and Nutrition Examination Survey (NHANES) from 1999 to 2018. We fitted linear and joinpoint logistic regression models to test the sample weighted and age-standardized time trends for linear and nonlinear trends in the prevalence of hyperinsulinemia and IR, respectively. Results: We included 17,310 nondiabetic men and nonpregnant women aged 20 years or older. The age-standardized prevalence of hyperinsulinemia increased from 28.2% in 1999-2000 to 41.4% in 2017-2018, with IR prevalence similarly rising from 24.8% in 1999-2000 to 38.4% in 2017-2018. Across the entire period examined, individuals who were male; non-Hispanic Black; Hispanic; or had a lower educational level or lower family income consistently had a higher prevalence of hyperinsulinemia and IR than other groups. We found increasing temporal trends in the prevalence of hyperinsulinemia and IR for all the sociodemographic subgroups, at least in some periods from 1999 to 2018. Conclusions: There was an increased age-standardized prevalence of hyperinsulinemia and IR among nondiabetic adults in the US across each defined sociodemographic group from 1999 to 2018. The difference in prevalence across subgroups underscores the need for designing personalized and targeted interventions to address disparities.
Collapse
Affiliation(s)
- Chuyue Wu
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Yixun Ke
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Roch A Nianogo
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA
- California Center for Population Research, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
30
|
Tiwari V, Kamboj A, Sheoran B, Chaudhary E, Yadav M, Kumari A, Krishania M, Ali U, Tiwari A, Garg M, Bhatnagar A. Anthocyanin-rich black wheat as a functional food for managing type 2 diabetes mellitus: a study on high fat diet-streptozotocin-induced diabetic rats. Food Funct 2025; 16:3273-3295. [PMID: 39688703 DOI: 10.1039/d4fo05065g] [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: 12/18/2024]
Abstract
Background: Type 2 Diabetes Mellitus (T2DM) is associated with insulin resistance, hyperglycemia, and hyperlipidemia. Anthocyanins, which are natural antioxidants, have been reported to manage T2DM-related complications. However, the potential of anthocyanin-rich black wheat as a functional food for managing diabetes remains unexplored. Aim: This study aimed to investigate the effects of anthocyanin-rich black wheat on glucose metabolism, insulin sensitivity, lipid profile, oxidative stress, inflammation, and organ protection in high fat diet-streptozotocin (HFD-STZ) induced T2DM rats. Methods: T2DM was induced in rats using HFD-STZ. The rats were fed with either white wheat or anthocyanin-rich black wheat chapatti. Glucose metabolism, insulin sensitivity, lipid profile, antioxidant enzymes, inflammatory markers, and glucose transporters were assessed. Histopathological analysis of the liver, kidneys, and spleen was performed. Results: Compared to white wheat chapatti, black wheat chapatti exhibited higher α-amylase and α-glucosidase inhibitory activities. Black wheat chapatti consumption significantly reduced blood glucose and HbA1c levels, and improved insulin sensitivity, oral glucose tolerance, and insulin tolerance. Antioxidant enzyme (superoxide dismutase and catalase) activities were enhanced. Atherogenic dyslipidemia was attenuated, with improved high-density lipoprotein cholesterol levels. Inflammatory markers (TNF-α, IL-1β, leptin, resistin and cortisol) were reduced, while adiponectin (Acrp-30) levels increased. Black wheat chapatti activated adiponectin-AMPK and PI3K-AKT pathways, upregulating glucose transporters (GLUT-2 and GLUT-4). Histopathology revealed protective effects on the liver, kidneys, and spleen. Conclusions: Anthocyanin-rich black wheat chapatti ameliorates insulin resistance and associated complications in HFD-STZ-induced T2DM rats. It modulates key signaling pathways and glucose transporters, demonstrating its potential as a functional food for managing T2DM and its complications.
Collapse
Affiliation(s)
- Vandita Tiwari
- Department of Biochemistry, Panjab University, Chandigarh, India
- National Agri-Food Biotechnology Institute (NABI), Mohali, Punjab, India.
| | - Akhil Kamboj
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - Bhawna Sheoran
- National Agri-Food Biotechnology Institute (NABI), Mohali, Punjab, India.
- Regional Centre for Biotechnology, Faridabad, Haryana (NCR), Delhi, India
| | - Era Chaudhary
- National Agri-Food Biotechnology Institute (NABI), Mohali, Punjab, India.
- Regional Centre for Biotechnology, Faridabad, Haryana (NCR), Delhi, India
| | - Mona Yadav
- National Agri-Food Biotechnology Institute (NABI), Mohali, Punjab, India.
- Regional Centre for Biotechnology, Faridabad, Haryana (NCR), Delhi, India
| | - Anita Kumari
- National Agri-Food Biotechnology Institute (NABI), Mohali, Punjab, India.
| | - Meena Krishania
- Center of Innovative and Applied Bioprocessing, Mohali, Punjab, India
| | - Usman Ali
- National Agri-Food Biotechnology Institute (NABI), Mohali, Punjab, India.
| | - Apoorv Tiwari
- National Agri-Food Biotechnology Institute (NABI), Mohali, Punjab, India.
| | - Monika Garg
- National Agri-Food Biotechnology Institute (NABI), Mohali, Punjab, India.
| | | |
Collapse
|
31
|
Chaisungnern K, Rattananupong T, Klinhom R, Nanta S, Banchuen K, Itharat A, Kuropakornpong P, Supasiri T, Nootim P, Jiamjarasrangsi W. Efficacy of Hibiscus sabdariffa L. extract on metabolic parameters in participants with abdominal obesity and mild metabolic syndrome in Bangkok, Thailand: A double-blind, randomized, placebo-controlled trial. Complement Ther Med 2025:103185. [PMID: 40334927 DOI: 10.1016/j.ctim.2025.103185] [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: 12/22/2024] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Hibiscus sabdariffa L. (HS) has been investigated as an alternative treatment for metabolic syndrome (MetS), as it affects all MetS components with low side effects simultaneously; however, clinical evidence regarding its efficacy compared with placebo is inconsistent. This study assessed how the aqueous calyx extract of HS influences insulin resistance and MetS parameters and examined the safety effects on liver, kidney, and hematological indexes in participants with abdominal obesity and mild MetS symptoms. METHODS In this double-blind, randomized, placebo-controlled trial, 108 participants with MetS were randomly assigned to take 1000-mg HS (45.04mg/day in total polyphenols) or placebo daily for 12 weeks. Insulin resistance (HOMA-IR), glycemic markers, body mass index (BMI), waist circumference (WC), lipid profiles, and blood pressure were assessed at baseline, 6 weeks, and 12 weeks. Additionally, liver and kidney function indicators along with hematological parameters were evaluated. RESULTS Compared with placebo, HS did not significantly affect HOMA-IR, glycemic markers, BMI, WC, lipid profile, or blood pressure. Although HS did not significantly alter the lipid profile overall, serum low-density lipoprotein (LDL) levels decreased significantly at 12 weeks compared with baseline (-7.98mg/dL, [95% CI, -14.80, -1.15]). Additionally, HS did not cause significant liver or kidney function or hematological changes compared with placebo. CONCLUSION Taking 1000-mg HS daily for 12 weeks seems to be safe. Placebo and HS groups showed good clinical results, and the extract was not associated with improved metabolic parameters in individuals with abdominal obesity and mild MetS symptoms, with the exception of lower serum LDL.
Collapse
Affiliation(s)
- Kanchaporn Chaisungnern
- Health Research and Management Program, Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| | - Thanapoom Rattananupong
- Health Research and Management Program, Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| | - Rossukon Klinhom
- Institute of Thai Traditional Medicine, Department of Thai Traditional and Alternative Medicine, Ministry of Public Health, Thailand.
| | - Srisuphak Nanta
- Institute of Thai Traditional Medicine, Department of Thai Traditional and Alternative Medicine, Ministry of Public Health, Thailand.
| | - Kamonwan Banchuen
- Institute of Thai Traditional Medicine, Department of Thai Traditional and Alternative Medicine, Ministry of Public Health, Thailand.
| | - Arunporn Itharat
- Center of Excellence in Applied Thai Traditional Medicine Research, Thammasat University, Thailand.
| | - Pranporn Kuropakornpong
- Center of Excellence in Applied Thai Traditional Medicine Research, Thammasat University, Thailand.
| | - Thanan Supasiri
- Health Research and Management Program, Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Center of Excellence in Preventive and Integrative Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| | - Preecha Nootim
- Institute of Thai Traditional Medicine, Department of Thai Traditional and Alternative Medicine, Ministry of Public Health, Thailand.
| | - Wiroj Jiamjarasrangsi
- Health Research and Management Program, Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
32
|
Choi MJ, Yu J. Menopause and Diabetes Risk Along with Trajectory of β-Cell Function and Insulin Sensitivity: A Community-Based Cohort Study. Healthcare (Basel) 2025; 13:1062. [PMID: 40361840 DOI: 10.3390/healthcare13091062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND The relationship between menopause and diabetes risk is unclear, with some studies indicating a weak association. This study examined changes in diabetes risk, β-cell function, and insulin sensitivity in relation to menopause. METHODS In this community-based cohort study, data from 6684 visits to 1224 women over a 16-year follow-up were analyzed. Diabetes risk changes were assessed in relation to the different menopausal phases: premenopausal (≥3 years before menopause), perimenopausal (2 years before to 1 year after menopause), and postmenopausal (≥2 years after menopause). Changes in β-cell function and insulin sensitivity indices were tracked, and their relationship with diabetes risk was assessed. Generalized estimating equations and linear mixed models were used, adjusting for covariates including age at menopause and obesity. RESULTS Diabetes incidence was 18.6% among participants. The odds ratio (OR) of diabetes increased by 1.03 times annually during the premenopausal period (OR 1.03; 95% CI 1.02-1.04) and decreased during the postmenopausal period (OR 0.96; 95% CI 0.95-0.97). The incident diabetes groups showed a decline in insulin sensitivity and β-cell function, resulting in a decrease in the disposition indices over time. A large change in insulin sensitivity, especially during the period immediately before the onset of diabetes, increased the risk of diabetes (OR 1.88; 95% CI 1.33-2.67). CONCLUSIONS This study indicates an increased diabetes risk during the premenopausal periods, compared with that in the postmenopausal period, independent of age at menopause and obesity. Additionally, a decrease in insulin sensitivity followed by a subsequent decrease in β-cell function depending on the time of onset was related to the risk of diabetes. These findings enhance the understanding of diabetes risk and associated changes in insulin indices in relation to menopause, emphasizing the importance of health management and diabetes prevention for women in menopausal transition.
Collapse
Affiliation(s)
- Mi Jin Choi
- College of Nursing, Gyeongsang National University, Jinju-si 52727, Republic of Korea
| | - Juyoun Yu
- Department of Nursing, Changwon National University, Changwon-si 51140, Republic of Korea
| |
Collapse
|
33
|
Mohamed RA, Abdallah DM, El-Abhar HS. Chaperone-mediated autophagy, heat shock protein 70, and serotonin: novel targets of beta-hydroxybutyrate in HFFD/LPS-induced sporadic Alzheimer's disease model. Inflammopharmacology 2025:10.1007/s10787-025-01754-6. [PMID: 40319428 DOI: 10.1007/s10787-025-01754-6] [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: 02/18/2025] [Accepted: 04/04/2025] [Indexed: 05/07/2025]
Abstract
Sporadic Alzheimer's disease (AD), which accounts for the majority of cases, is sturdily influenced by lifestyle factors such as dietary habits, obesity, and diabetes, leading to its classification as Type 3 diabetes. To model this pathological link, our AD-like model was developed by feeding Wistar male rats a high-fat diet with fructose in drinking water (HFFD) for 8 weeks, followed by a single dose of lipopolysaccharide (LPS). This group was compared with a normal control group fed a standard diet and a β-hydroxybutyrate (BHB)-treated group (125 mg/kg, p.o.), administered starting 3 h after LPS and continuing for 1 week. The results demonstrate that BHB treatment illuminated cognitive gains, as indicated by the Y-maze, Morris water maze, and novel object recognition tests. In addition, it preserved hippocampal cytoarchitecture, reduced neurodegeneration, and attenuated amyloid plaques and phosphorylated Tau deposition. Cellularly, BHB restored critical molecular mechanisms, including increased lysosomal-associated membrane protein 2A (LAMP2A) hippocampal content as the main marker of chaperone-mediated autophagy (CMA), along with the chaperon protein Hsp70. Moreover, BHB alleviated neuroinflammation by inhibiting the nucleotide-binding domain, leucine-rich-containing family, and pyrin domain-containing-3 (NLRP3) inflammasome activation alongside the downstream targets cleaved caspase-1 and IL-1β/IL-18 cytokines. BHB also reduced pyroptotic markers, caspase-11 and gasdermin-N, and microglia-induced inflammation as it shifted microglial polarization toward the neuroprotective M2 phenotype. Finally, BHB normalized hippocampal neurotransmitter levels of the inhibited acetylcholine and serotonin. These findings support BHB as a promising, multifaceted treatment for AD, highlighting the roles of CMA, Hsp70, and 5-HT in slowing disease progression and improving cognitive function.
Collapse
Affiliation(s)
- Reem A Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Science and Arts (MSA), Cairo, 12566, Egypt.
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hanan S El-Abhar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
34
|
Sarabi M, Torshizian A, Khorasani ZM, Firoozi A, Majd HM, Khoshhal N, Saeidi N, AkbariRad M. Impact of Probiotics Administration on the VEGF, Adiponectin, and Glycolipid Metabolism, in Prediabetic Patients: A Randomized, Double-Blinded, Placebo-Controlled Clinical Trial. Food Sci Nutr 2025; 13:e70146. [PMID: 40321607 PMCID: PMC12045928 DOI: 10.1002/fsn3.70146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/21/2024] [Accepted: 03/27/2025] [Indexed: 05/08/2025] Open
Abstract
It is debated that probiotics can improve glycolipid metabolism and slow the progression of prediabetes to diabetes mellitus. This study aimed to evaluate the effect of probiotics on lipid profile, glucose homeostasis, serum level of resistin, adiponectin, and vascular endothelial growth factor (VEGF) in prediabetic patients. This double-blind, randomized, placebo-controlled clinical trial was conducted on prediabetic patients in the Endocrinology clinic of Ghaem Hospital. Patients were randomly divided into two groups: the probiotics group was prescribed a daily 500-mg capsule of probiotics (109 colony-forming units), while the other received a placebo capsule with the same appearance. After 3 months, the effect of probiotic administration on laboratory parameters indicative of glycolipid metabolism, resistin, adiponectin, VEGF, body mass index (BMI), and blood pressure was compared between groups. This study was registered in the Iranian Registry of Clinical Trials (IRCT 20190801044405 N2). Fifty-two patients were included in the final analysis, with 26 patients in each group. The mean age of patients was 43.75 ± 8.45. At the beginning, both groups were similar in all demographic characteristics and measured serum levels of investigated biomarkers (p > 0.05 for all parameters). Both groups exhibited significant changes in BMI and fasting blood sugar (FBS). However, regarding FBS, the magnitude of change was significantly greater in patients treated with probiotics (p = 0.022). Our findings also revealed a significant increase in HDL (p = 0.001), adiponectin (p < 0.001), and VEGF (p = 0.024) serum levels and a significant decrease in HbA1c (p = 0.034), LDL (p = 0.002), TG (p < 0.001), and total cholesterol (p = 0.001) exclusively in the probiotics group. Probiotic supplementation efficiently improved glycolipid metabolism, adiponectin, and VEGF serum levels.
Collapse
Affiliation(s)
- Mehrdad Sarabi
- Student Research Committee, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Ashkan Torshizian
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
| | - Zahra Mazloum Khorasani
- Department of Endocrinology, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | | | - Hassan Mehrad Majd
- Molecular Medicine, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Nastaran Khoshhal
- Student Research Committee, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Nikoo Saeidi
- Student Research CommitteeIslamic Azad UniversityMashhadIran
| | - Mina AkbariRad
- Department of Internal Medicine, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
35
|
Sørensen KV, Justesen JM, Ängquist L, Bork-Jensen J, Hartmann B, Jørgensen NR, Rungby J, Sørensen HT, Vaag A, Nielsen JS, Holst JJ, Pedersen O, Linneberg A, Hansen T, Grarup N. Rare MTNR1B variants causing diminished MT2 signalling associate with elevated HbA 1c levels but not with type 2 diabetes. Diabetologia 2025; 68:1016-1030. [PMID: 40064676 PMCID: PMC12021717 DOI: 10.1007/s00125-025-06381-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 12/10/2024] [Indexed: 04/25/2025]
Abstract
AIMS/HYPOTHESIS An intronic variant (rs10830963) in MTNR1B (encoding the melatonin receptor type 2 [MT2]) has been shown to strongly associate with impaired glucose regulation and elevated type 2 diabetes prevalence. However, MTNR1B missense variants have shown conflicting results on type 2 diabetes. Thus, we aimed to gain further insights into the impact of MTNR1B coding variants on type 2 diabetes prevalence and related phenotypes. METHODS We conducted a cross-sectional study, performing MTNR1B variant burden testing of glycaemic phenotypes (N=248,454, without diabetes), other cardiometabolic phenotypes (N=330,453) and type 2 diabetes prevalence (case-control study; N=263,739) in the UK Biobank. Similar burden testing with glycaemic phenotypes was performed in Danish Inter99 participants without diabetes (N=5711), and type 2 diabetes prevalence (DD2 cohort serving as cases [N=2930] and Inter99 serving as controls [N=4243]). Finally, we evaluated the effects of MTNR1B variants on the melatonin-induced glucose regulation response in a recall-by-genotype study of individuals without diabetes. RESULTS In the UK Biobank, MTNR1B variants were not associated with cardiometabolic phenotypes, including type 2 diabetes prevalence, except that carriers of missense MTNR1B variants causing impaired MT2 signalling exhibited higher HbA1c levels compared with non-carriers (effect size, β, 0.087 SD [95% CI 0.039, 0.135]). Similarly, no significant associations were observed with phenotypes associated with glycaemic phenotypes in the Inter99 population. However, carriers of variants impairing MT2 signalling demonstrated a nominally significant lower glucose-stimulated insulin response (β -0.47 SD [95% CI -0.82, -0.11]). A reduced insulin response was also observed in carriers of variants impairing MT2 signalling (β -476.0 [95% CI -928.6, -24.4]) or the rs10830963 variant (β -390.8 [95% CI -740.1, -41.6]) compared with non-carriers after melatonin treatment. CONCLUSIONS/INTERPRETATION The higher type 2 diabetes prevalence previously observed in carriers of missense MTNR1B variants causing impairment in MT2 signalling was not replicated in the UK Biobank, yet carriers had elevated HbA1c levels. DATA AVAILABILITY Data (Inter99 cohort and recall-by-genotype study) are available on reasonable request from the corresponding author. Requests for DD2 data are through the application form at https://dd2.dk/forskning/ansoeg-om-data . Access to UK Biobank data can be requested through the UK Biobank website ( https://www.ukbiobank.ac.uk/enable-your-research ).
Collapse
Affiliation(s)
- Kimmie V Sørensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Johanne M Justesen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars Ängquist
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jette Bork-Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niklas R Jørgensen
- Department of Clinical Biochemistry, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Translational Research Centre, Rigshospitalet, Copenhagen, Denmark
| | - Jørgen Rungby
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Herlev Hospital, Herlev, Denmark
| | - Henrik T Sørensen
- Department of Clinical Epidemiology, Aarhus University, Aarhus, Denmark
- Department of Epidemiology, Boston University, Boston, MA, USA
| | - Allan Vaag
- Steno Diabetes Center Copenhagen, Herlev Hospital, Herlev, Denmark
- Lund University Diabetes Care, Lund University, Malmö, Sweden
- Department of Endocrinology, Skåne University Hospital, Malmö, Sweden
| | - Jens S Nielsen
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jens J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Clinical Metabolic Research, Herlev-Gentofte Hospital, Copenhagen, Denmark
| | - Allan Linneberg
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Clinical Research and Prevention, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
36
|
Wu Z, Liang G, Zhang Y, Li R. Risk Factors for Metabolic Dysfunction-Associated Steatotic Liver Disease in Patients With Polycystic Ovary Syndrome in East Asia: A Review and Meta-Analysis. Endocr Pract 2025; 31:668-676. [PMID: 39947624 DOI: 10.1016/j.eprac.2025.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/02/2024] [Accepted: 01/24/2025] [Indexed: 05/09/2025]
Abstract
OBJECTIVES The incidence of metabolic dysfunction-associated steatotic liver disease (MASLD) is increasing in women with polycystic ovary syndrome (PCOS). Epidemiologic literature regarding the risk factors for MASLD in PCOS women in East Asia is inconsistent. Studies of PCOS and MASLD in East Asia are restricted by limited data and various biases. Therefore, this meta-analysis was conducted. METHODS This meta-analysis followed the MOOSE statement. Relevant studies published before July 13, 2023 were retrieved from the PubMed, Embase, Cochrane Library, Web of Science, CNKI, VIP, Wan Fang, KISS, and Japan medical online databases. The related data were extracted, and the weighted mean difference, odds ratios and 95% confidence intervals were calculated. RESULTS Twenty-four studies were included. Through univariate analysis, age, body mass index, waist-to-hip ratio (WHR), blood pressure, alanine transaminase, aspartate transaminase, fasting blood glucose, fasting insulin, homeostatic model assessment for insulin resistance (HOMA-IR), 2-hour postprandial blood glucose, 2 hour insulin, HBA1C, low-density lipoprotein cholesterol, triglyceride, total cholesterol, and testosterone were notably higher in PCOS women with MASLD, with high-density lipoprotein cholesterol markedly lower in PCOS women with MASLD. According to the pooled multivariate analysis, the WHR (P < .001), testosterone (P = .034), and HOMA-IR (P = .02) were substantially greater in PCOS women with MASLD. CONCLUSION MASLD is associated with obesity, IR, and hyperandrogenemia among PCOS women in East Asia. The abnormality of WHR, HOMA-IR, and testosterone suggested early screening of MASLD in this population.
Collapse
Affiliation(s)
- Zhao Wu
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China; Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Guining Liang
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China; Department of Clinical Medicine, Nan Shan School of Guangzhou Medical University, Guangzhou, China
| | - Ying Zhang
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China; Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China; Department of Clinical Medicine, Nan Shan School of Guangzhou Medical University, Guangzhou, China
| | - Renyuan Li
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China; Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China; Department of Clinical Medicine, Nan Shan School of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
37
|
Xiao Q, Feng Q, Rutter MK, Albalak G, Wang H, Noordam R. Associations between the timing of 24 h physical activity and diabetes mellitus: results from a nationally representative sample of the US population. Diabetologia 2025; 68:1005-1015. [PMID: 39982484 PMCID: PMC12021934 DOI: 10.1007/s00125-025-06368-9] [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: 08/22/2024] [Accepted: 12/05/2024] [Indexed: 02/22/2025]
Abstract
AIMS/HYPOTHESIS Growing evidence suggests that timing may be an important aspect of physical activity that influences cardiometabolic health. However, the current literature is inconclusive regarding the time of day that physical activity offers the greatest metabolic advantages. We investigated associations between hourly physical activity levels and diabetes mellitus and glycaemic biomarkers in a cross-sectional and nationally representative sample of US adults. METHODS We studied 7074 adults (mean age 48 years; 52% women) from the National Health and Nutrition Examination Survey (2011-2014). Physical activity was measured by actigraphy. A monitor-independent movement summary (MIMS) unit was used to derive the total activity level (divided into quintiles) for hourly windows that were defined relative to sleep timing and according to clock time. The primary outcome was prevalent diabetes, and secondary outcomes included fasting glucose, fasting insulin, HOMA-IR and 2 h OGTT results. RESULTS Physical activity levels in late morning and late afternoon were associated with lower adjusted odds of diabetes. Specifically, in late morning (8:01-9:00 h after the sleep midpoint), the highest quintile of activity was associated with a 35% decrease (OR 0.65; 95% CI 0.44, 0.96) in the odds of diabetes when compared with the lowest quintile, while in late afternoon (11:01-17:00 h after the sleep midpoint), the highest quintiles were associated with 56% and 36% lower odds (OR 0.44; 95% CI 0.29, 0.69 and OR 0.64; 95% CI 0.43, 0.95). Higher night-time activity was associated with higher odds of diabetes. Similar patterns of results were observed with OGTT data and across subgroups of age, gender, race/ethnicity, chronotype and sleep duration. CONCLUSIONS/INTERPRETATION Our findings suggest that the timing of physical activity may modulate its metabolic effects.
Collapse
Affiliation(s)
- Qian Xiao
- Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Center for Spatial‑temporal Modeling for Applications in Population Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Qiuyu Feng
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Martin K Rutter
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Diabetes, Endocrinology and Metabolism Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester, UK
| | - Gali Albalak
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Heming Wang
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands.
- Health Campus the Hague/Public Health and Primary Care, Leiden University Medical Center, the Hague, the Netherlands.
| |
Collapse
|
38
|
Mora-Rodriguez R, Moreno-Cabañas A, Alvarez-Jimenez L, Mora-Gonzalez D, Morales-Palomo F. High-Intensity Intervallic Exercise (HIIE) Is Superior to Isocaloric Moderate-Intensity Continuous Exercise (MICE) at Reducing Postprandial Hyperglycemia. Med Sci Sports Exerc 2025; 57:1019-1031. [PMID: 39661748 DOI: 10.1249/mss.0000000000003625] [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: 12/13/2024]
Abstract
PURPOSE We investigated if a bout of high-intensity intervallic exercise (HIIE) is more efficacious at reducing postprandial hyperglycemia than an isocaloric bout of moderate-intensity continuous exercise (MICE). METHODS Nineteen healthy physically active individuals (21% women) completed three trials in a randomized order: i ) HIIE cycling consisting of five bouts of 4 min at 83 ± 9% of subjects' maximal oxygen consumption ( O 2MAX ) with active recoveries at 53 ± 8%, for a total of 50 min; ii ) MICE cycling at 65 ± 8% of O 2max for 50 min; and iii ) CONTROL no exercise. All trials were followed by a standard oral glucose tolerance test (OGTT) ingesting 74 g of glucose traced with 1 g of uniformly labeled [ 13 C]-glucose. Plasma glucose and insulin concentrations, and plasma glucose kinetics ([6,6 2 H 2 ] glucose infusion) were measured before exercise, during exercise, and during the OGTT. Insulin sensitivity was estimated by the Matsuda index (ISI). Energy expenditure and carbohydrate oxidation (CHOxid) were monitored. RESULTS At rest, blood glucose, insulin concentrations, and CHOxid were similar in all three trials. During exercise, energy expenditure was similar in HIIE versus MICE (548 ± 131 vs 560 ± 125 kcal; P = 0.340). However, CHOxid, plasma glucose concentration, and its rates of appearance in plasma (Ra) were higher in HIIE versus MICE (Ra glucose 34.3 ± 9.8 vs 28.9 ± 6.8 μmol·kg -1 ·min -1 ; P = 0.021). During the OGTT, plasma glucose and insulin concentrations were lower, and insulin sensitivity was higher in HIIE versus CONTROL (ISI MATSUDA ; 12.4 ± 4.7 vs 10.8 ± 4.7 au; P = 0.007). Exercise delayed blood incorporation of [ 13 C]-glucose into blood ( P = 0.023). Early during the OGTT, glucose clearance rates were higher in HIIE versus CONTROL (7.1 ± 3.1 vs 5.5 ± 3.0 mL·kg -1 ·min -1 ; P = 0.015). CONCLUSIONS HIIE is more effective than MICE to reduce hyperglycemia and hyperinsulinemia after glucose ingestion. HIIE improves glycemic control by increasing splanchnic glucose retention and glucose clearance rates.
Collapse
Affiliation(s)
- Ricardo Mora-Rodriguez
- Exercise Physiology Lab at Toledo, Faculty of Sport Sciences, University of Castilla-La Mancha, Toledo, SPAIN
| | | | | | - Diego Mora-Gonzalez
- Grupo IMCU, Department of Nursing, Physiotherapy, and Occupational Therapy, University of Castilla-La Mancha, Toledo, SPAIN
| | - Felix Morales-Palomo
- Exercise Physiology Lab at Toledo, Faculty of Sport Sciences, University of Castilla-La Mancha, Toledo, SPAIN
| |
Collapse
|
39
|
Martín-Saladich Q, Simó R, Herance JR, Ballester MAG. MIRA: Myocardial insulin resistance app for clinical practice. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 263:108674. [PMID: 39978140 DOI: 10.1016/j.cmpb.2025.108674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/31/2025] [Accepted: 02/13/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND AND OBJECTIVE Type 2 diabetes (T2D) is a prevalent disease characterized by insulin resistance (IR), leading to energy disruptions in myocardial cells and increasing cardiovascular (CV) risk. Current diagnostic methods are either systemic, thus lacking tissue-specific information, or invasive. The hyperinsulinemic euglycemic clamp (HEC) combined with [18F]FDG-PET images, only used in clinical trials, allow to assess regional IR and identify phenotypes within T2D, linking myocardial IR to higher CV risk. However, phenotyping is not easily accessible, creating a need for alternative assessment tools. Thus, we propose a myocardial IR model to address these gaps and improve T2D management. METHODS The study included forty-two patients with T2D who enrolled in a clinical trial (NCT02248311) and who underwent biochemical analyses, anthropometric measurements and [18F]FDG PET/CT imaging before and after HEC with. Patients were phenotyped into mIR and mIS according to poor or good uptake after HEC, respectively. The proposed predictive model was based on stepwise regression including feature selection to provide an estimate of myocardial IS and thus IR=1/IS by using biochemical parameters in T2D. A software application, the myocardial IR app (MIRA), was developed using MATLAB. RESULTS MIRA was developed as a myocardial IR estimator (R2=0.97, p=7.1 × 10-7, error=1.24) for patients with T2D. Moreover, since HEC is not allowed in rutinary clinical practice, the application includes a prediction of the expected myocardial HEC [18F]FDG uptake from baseline uptake (r=0.52, p=5 × 10-4, R2=0.60). The app also yields the patient's phenotype, either mIR or mIS, according to poor or good uptake after HEC. Enhanced CV risk exposure due to altered T2D biomarkers and associated to mIR is also provided with highlighted features. CONCLUSIONS We hereby present MIRA, a myocardial IR calculation app to manage myocardial-specific affectation in T2D, as well as to provide with patient phenotyping and CV risk assessment.
Collapse
Affiliation(s)
- Queralt Martín-Saladich
- Medical Molecular Imaging Research Group, Vall d'Hebron Research Institute (VHIR), Nuclear Medicine, Radiology and Cardiology Departments, Vall d'Hebron University Hospital, Autonomous University Barcelona, Barcelona 08035, Spain; BCN Medtech, Department of Engineering, Universitat Pompeu Fabra, Barcelona 08018, Spain
| | - Rafael Simó
- Diabetes and Metabolism Research Group, VHIR, Endocrinology Department, Vall d'Hebron University Hospital, Autonomous University Barcelona, Barcelona 08035, Spain; CIBERDEM (Instituto de Salud Carlos III), Madrid 28029, Spain
| | - José Raul Herance
- Medical Molecular Imaging Research Group, Vall d'Hebron Research Institute (VHIR), Nuclear Medicine, Radiology and Cardiology Departments, Vall d'Hebron University Hospital, Autonomous University Barcelona, Barcelona 08035, Spain; CIBERBBN (Instituto de Salud Carlos III), Madrid 28029, Spain
| | - Miguel A González Ballester
- BCN Medtech, Department of Engineering, Universitat Pompeu Fabra, Barcelona 08018, Spain; Catalan Institution for Research and Advanced Studies ICREA, 08010 Barcelona, Spain.
| |
Collapse
|
40
|
Hao QY, Gao JW, Zeng YH, Zhang SL, Xiong ZC, Li SC, Lin ZW, Yang PZ, Liu PM, Li ZH. Roles of triglyceride-glucose index in aortic valve calcification progression: a prospective and Mendelian randomization analysis. Clin Radiol 2025; 84:106860. [PMID: 40106977 DOI: 10.1016/j.crad.2025.106860] [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: 10/20/2024] [Accepted: 02/11/2025] [Indexed: 03/22/2025]
Abstract
AIM The triglyceride-glucose (TyG) index, recognized as a surrogate marker for insulin resistance, is an established cardiovascular risk factor. We aimed to prospectively investigate the association between the TyG index and aortic valve calcific (AVC) progression, as well as its relationship with incident calcific aortic valve stenosis (CAVS). MATERIALS AND METHODS A post hoc analysis was conducted on 5589 participants from the Multi-Ethnic Study of Atherosclerosis (MESA) database. The TyG index was calculated using ln (fasting triglycerides [mg/dL] × fasting glucose [mg/dL]/2). Multivariate Cox regression assessed the association between baseline TyG index and AVC progression. Two-sample Mendelian randomization (MR) analysis was employed to evaluate the potential causality between the TyG index and CAVS. RESULTS Over a median 2.4 years follow up, 567 cases of AVC progression were idenrified. After adjusting for traditional cardiovascular risk factors, each 1-SD increase in the TyG index was associated with a 20.8% increased risk of AVC progression. Robustness was confirmed in sensitivity analyses and nearly all subgroups. Two sample MR analysis supported a causal relationship between a higher TyG index and increased risk of CAVS. CONCLUSION A higher TyG index independently predicts AVC progression and causally influences CAVS incidence in the general population.
Collapse
Affiliation(s)
- Q-Y Hao
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - J-W Gao
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Y-H Zeng
- Medical Apparatus and Equipment Deployment, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - S-L Zhang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Z-C Xiong
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - S-C Li
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Z-W Lin
- Department of Joint and Orthopedics, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - P-Z Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - P-M Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Z-H Li
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
41
|
Al-Qudah AA, Al-Hanaktah M, Albadaineh R. Effects of Exenatide plus Metformin versus Metformin alone on insulin resistance in women with Polycystic Ovary Syndrome: A systematic review and meta-analysis. J Obstet Gynaecol Res 2025; 51:e16296. [PMID: 40301112 DOI: 10.1111/jog.16296] [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: 01/28/2025] [Accepted: 04/08/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a common endocrine disorder among reproductive-age women, often accompanied by insulin resistance, obesity, and increased metabolic risk. While Metformin (MET) is commonly used to improve insulin sensitivity, its limited effect on postprandial glucose has led to interest in combination therapies. Exenatide (EX), a glucagon-like peptide-1 receptor agonist, may offer complementary benefits. OBJECTIVE To assess the effectiveness of EX combined with MET (EX + MET) versus MET alone (MET) in improving insulin resistance and metabolic outcomes in overweight and obese women with PCOS. METHODS This systematic review and meta-analysis included five randomized controlled trials (n = 339) and followed Preferred Reporting Items for Systematic reviews and Meta-Analysis Extension guidelines. The primary outcome was the change in insulin resistance (Homeostatic Model Assessment of Insulin Resistance [HOMA-IR]). Secondary outcomes included body mass index (BMI), 2-h oral glucose tolerance test (OGTT), lipid profile, and reproductive hormones. RESULTS Pooled analysis revealed that EX + MET significantly reduced HOMA-IR (mean difference [MD]: -0.9; p < 0.001), improved 2-h OGTT values (MD: -1.78; p < 0.001), reduced BMI (MD: -0.4; p = 0.03), with low heterogeneity. Combination therapy also improved triglyceride and total cholesterol levels. However, no significant effects were observed on reproductive hormones or low-density lipoprotein and high-density lipoprotein cholesterol. CONCLUSIONS While hormonal and some lipid changes were not significant, their inclusion highlights the multifaceted impact of PCOS and the need for longer-term studies. By improving insulin sensitivity and weight-related outcomes, EX + MET may be a valuable clinical option for metabolically high-risk PCOS patients.
Collapse
|
42
|
Vataja E, Viitanen M, Rinne JO, Lehtisalo J, Erlund I, Ngandu T, Koskinen S, Åberg F, Jula A, Ekblad L. Metabolic dysfunction-associated steatotic liver disease as a predictor of cognitive performance: An 11-year population-based follow-up study. Dig Liver Dis 2025; 57:585-595. [PMID: 39922744 DOI: 10.1016/j.dld.2025.01.197] [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: 07/03/2024] [Revised: 10/23/2024] [Accepted: 01/24/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND Prevalent manifestation of metabolic dysfunction, metabolic dysfunction-associated steatotic liver disease (MASLD), has been associated with poorer cognitive performance and greater decline in cognitive functions. AIM The aim of this study was to analyze whether MASLD, measured by fatty-liver-index (FLI), predicts decline in cognitive performance during 11 years. METHODS This study was based on the Finnish nationwide, population-based Health 2000 Health Examination Survey and its follow-up, Health 2011 Survey. Cognitive performance was assessed with verbal fluency, word-list learning (WLL), delayed word-list recall (both at baseline and at follow-up), and with simple reaction time and visual choice reaction time tests (only at baseline). Statistical analyses were performed using multivariate linear regression adjusted for age, sex, education, APOE ε4 genotype, hypercholesterolemia, diabetes mellitus, hypertension, depressive symptoms, physical activity smoking status, C-reactive protein and HOMA of insulin resistance. RESULTS Cross-sectionally, 5,139 (mean age 52.3 years) and longitudinally, 3,143 (mean age 49.3 years) participants were examined. Cross-sectionally, no associations between FLI and cognitive performance were found in the adjusted models. Longitudinally, baseline FLI > 60 predicted poorer WLL (p < 0.005) and a decline in WLL from baseline to follow-up (p < 0.04). CONCLUSIONS Our results suggest that MASLD is an independent predictor of decline in a test measuring working memory and learning.
Collapse
Affiliation(s)
- Emilia Vataja
- Department of Geriatric Medicine, University of Turku, Turku, Finland.
| | - Matti Viitanen
- Department of Geriatric Medicine, University of Turku, Turku, Finland; Division of Clinical Geriatrics, NVS, Karolinska Institutet, Stockholm, Sweden
| | - Juha O Rinne
- Turku PET Centre, University of Turku, Turku, Finland; Turku PET Centre, University Hospital of Turku, Turku, Finland; InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | | | - Iris Erlund
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Tiia Ngandu
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Seppo Koskinen
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Fredrik Åberg
- Department of Transplantation and Liver Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Antti Jula
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Laura Ekblad
- Department of Geriatric Medicine, University of Turku, Turku, Finland; Turku PET Centre, University of Turku, Turku, Finland; Turku PET Centre, University Hospital of Turku, Turku, Finland; Department of Geriatrics, Turku University Hospital, Wellbeing services county of Southwestern Finland, Finland
| |
Collapse
|
43
|
Amiri M, Raeisi-Dehkordi H, Steur M, Grisotto G, Rivadeneira F, Ikram MK, Kavousi M, Muka T, Voortman T. Dietary patterns derived using reduced rank regression in postmenopausal women and risk of mortality: A population-based study. Maturitas 2025; 196:108234. [PMID: 40090127 DOI: 10.1016/j.maturitas.2025.108234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/18/2025]
Abstract
OBJECTIVES The menopause transition increases the risk of chronic conditions in women. Given the crucial role of diet in health, we identified dietary patterns that explain variations in factors related to major health concerns in postmenopausal women. Also, we explored their association with all-cause and cardiovascular mortality. STUDY DESIGN This study was conducted on 1814 postmenopausal women from the population-based Rotterdam Study. MAIN OUTCOME MEASURES Dietary patterns were identified using reduced rank regression. Response variables included bone mineral density, body composition parameters, lipid profile markers, insulin resistance, systolic blood pressure, cognitive function, depression, and sleep quality. The associations with risk of mortality were assessed using Cox proportional hazard models. RESULTS The first dietary pattern, characterized by higher intake of vegetables, whole grains, legumes, nuts, coffee, tea, alcoholic beverages, and cheese, explained 2.95 % of the variation in responses, accounted for 12.11 % of the variation in general cognitive function captured by G-factor, 5.62 % in systolic blood pressure, and 4.13 % in bone mineral density, and was correlated with less adiposity, lower blood pressure, lipid markers, and insulin resistance. The second dietary pattern, characterized by higher intakes of processed meat, unprocessed red meat, poultry, eggs, and coffee, and lower intakes of sweets and tea, explained 1.54 % of the variation in responses, accounted for 5.45 % of variation in fat mass percentage, 3.47 % in lean mass index, and 3.29 % in bone mineral density, and was correlated with higher adiposity, insulin resistance, and lipid markers. No associations with mortality risk were identified after adjusting for confounders such as demographics, socioeconomic status, lifestyle, disease history, and medication use. CONCLUSIONS We identified dietary patterns explaining a range of variation in health factors related to postmenopausal health. While these dietary patterns explained a large variation in some of the individual factors, their combined explained variation across multiple risk factors simultaneously was limited and no significant association with mortality risk was observed. This study provides a foundation for future research aimed at identifying optimal dietary patterns, integrating diverse health aspects, to improve health in postmenopausal populations.
Collapse
Affiliation(s)
- Mojgan Amiri
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Hamidreza Raeisi-Dehkordi
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Marinka Steur
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, the Netherlands
| | - Giorgia Grisotto
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - M Kamran Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands; Department of Neurology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | | | - Trudy Voortman
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands; Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, USA.
| |
Collapse
|
44
|
Dai H, Zhang S, Tian M, Yang P, Yang G, Li L, Liu D, Li K, Li S, Yang M. Netrin 4 is a novel cytokine associated with oxidative stress and insulin resistance in obese individuals. Diabetes Res Clin Pract 2025; 223:112166. [PMID: 40204125 DOI: 10.1016/j.diabres.2025.112166] [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: 09/20/2024] [Revised: 03/27/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Netrin 4 (NTN4) has been reported to be involved in a variety of pathophysiological processes, such as the occurrence and development of tumors, viral replication and infection, and diabetic retinopathy. However, the relationships between NTN4 and metabolic diseases have not been reported. METHODS The PhenoScanner tool and R language were used for bioinformatics analysis. Serum NTN4 was measured by ELISA in 211 healthy women and 193 overweight/obesity (OW/OB) women. Alterations in the serum NTN4 level were examined during the cold-exposure tests, acute exercise, lipid infusion, OGTT and EHC. RESULTS GWAS and Bioinformatics analysis revealed that NTN4 was closely related to energy metabolism and OS. In a population-based cohort study, we observed that individuals with OW/OB and IR exhibited significantly elevated levels of circulating NTN4. In addition, the serum NTN4 concentration was found to be significantly correlated with indicators of obesity, IR, sex hormone and glucose/lipid metabolism. CONCLUSIONS NTN4 is associated with obesity-related IR. TRIAL REGISTRATION All research plans were approved by the Human Research Ethics Committee of Chongqing Medical University (No. (74)2012, No. (72)2014 and No. (74)2015).
Collapse
Affiliation(s)
- Han Dai
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Siliang Zhang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Mingyuan Tian
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ping Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Gangyi Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ling Li
- Key Laboratory of Diagnostic Medicine (Ministry of Education) and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, China
| | - Dongfang Liu
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ke Li
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Shengbing Li
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Mengliu Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
45
|
Che X, Shang X, WeiXu, Xing M, Wei H, Li W, Li Z, Teng X, Geng L. Selenium-enriched Lactiplantibacillus plantarum alleviates alkalinity stress-induced selective hepatic insulin resistance in common carp. Int J Biol Macromol 2025; 305:141204. [PMID: 39986514 DOI: 10.1016/j.ijbiomac.2025.141204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/18/2024] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Carbonate alkalinity is one of the primary factors limiting saline-alkaline water aquaculture, and high alkalinity can lead to respiratory alkalosis, which is hazardous to fish health. Selenium (Se) and Lactiplantibacillus plantarum (L. plantarum) can be used for the biosynthesis of organic selenium (selenium-enriched Lactiplantibacillus plantarum: SL), which has low toxicity, high bioavailability, and the promotion of metabolism. Additionally, it can be used as a feed additive in aquaculture. In the present study, we established a model of chronic alkalinity stress in common carp and added SL to the feed. We found that alkalinity stress can cause severe hepatic dysfunction in common carp, as well as disrupt the intestinal barrier, further contributing to the translocation of enterogenous lipopolysaccharides through portal circulation and exacerbating liver injury. SL alleviated glucose-lipid metabolism abnormalities of the liver while reducing serum LPS levels and reduction of enterogenous LPS translocation to the liver, thus significantly reducing the degree of intestinal villi damage, hepatocyte vacuolisation, and nuclear damage. The significantly increased activities of SOD, GSH-Px, CAT, and T-AOC revealed that SL improved the antioxidant capacity of common carp. SL inhibited the alkalinity stress-induced overexpression of genes related to lipid synthesis and gluconeogenesis by modulating the P13K/Akt/FoxO1 signalling pathway, thus alleviating selective hepatic insulin resistance. SL attenuated the inflammatory response by modulating the mRNA expression levels of IL-7, IL-6, TNF-α and IL-10. In addition, apparent increase in the abundance of pathogenic bacteria (Brevinema, Bosea, Luteolibacter, and Vibrio) and apparent reduction in the abundance of beneficial bacteria (Cetobacterium, ZOR0006, and Shewanella) were closely related to the hepato-intestinal circulation process in carp exposed to alkalinity stress. SL regulated the hepato-intestinal circulation, reduced the abundance of Brevinema, Bosea, Luteolibacter, and Vibrio, increased the abundance of Cetobacterium, ZOR0006, and Shewanella, alleviated alkalinity stress-induced damage to intestinal microvilli (villus height and width), and significantly restored normal liver and intestinal functions. This study reveals the physiological regulatory mechanism by which Se-enriched L. plantarum through liver-intestinal axis alleviates alkalinity stress-induced hepatic insulin resistance and may provide new ideas and a theoretical basis for protecting against alkalosis and treating insulin resistance.
Collapse
Affiliation(s)
- Xinghua Che
- Key Open Laboratory of Cold Water Fish Germplasm Resources and Breeding of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Xinchi Shang
- Key Open Laboratory of Cold Water Fish Germplasm Resources and Breeding of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China; College of Life Science, Northeast Agricultural University, Harbin 150036, China
| | - WeiXu
- Key Open Laboratory of Cold Water Fish Germplasm Resources and Breeding of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Meiqi Xing
- Key Open Laboratory of Cold Water Fish Germplasm Resources and Breeding of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Haiju Wei
- Key Open Laboratory of Cold Water Fish Germplasm Resources and Breeding of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Wang Li
- Key Open Laboratory of Cold Water Fish Germplasm Resources and Breeding of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Zhengwei Li
- Fisheries Technology Extension Station of Heilongjiang Province, Daqing 166299, China
| | - Xiaohua Teng
- College of Life Science, Northeast Agricultural University, Harbin 150036, China.
| | - Longwu Geng
- Key Open Laboratory of Cold Water Fish Germplasm Resources and Breeding of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China.
| |
Collapse
|
46
|
Wanders L, Gijbels A, Hul GB, Feskens EJM, Afman LA, Blaak EE, Hopman MTE, Goossens GH, Thijssen DHJ. Impact of a 12-week personalized dietary intervention on vascular function and cardiovascular risk factors. Diabetes Obes Metab 2025; 27:2601-2612. [PMID: 40013435 PMCID: PMC11965023 DOI: 10.1111/dom.16261] [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: 12/18/2024] [Revised: 01/31/2025] [Accepted: 01/31/2025] [Indexed: 02/28/2025]
Abstract
AIMS Individuals with liver insulin-resistant (LIR) or muscle insulin-resistant (MIR) phenotypes may respond differently to dietary interventions. Given the interaction between insulin resistance and cardiovascular risk, this sub-analysis of the PERSON study examined whether a personalized diet according to MIR or LIR phenotypes improves vascular function and cardiovascular disease risk factors. MATERIALS AND METHODS We randomized 119 participants to a 12-week low-fat, high-protein, high-fibre diet (LFHP; may be optimal for LIR) or Mediterranean diet (high in monounsaturated fat, HMUFA; may be optimal for MIR). Randomization linked the insulin-resistant (IR) phenotype to the proposed optimal diet, leading to PhenoDiet A (MIR-HMUFA and LIR-LFHP) and PhenoDiet B (MIR-LFHP and LIR-HMUFA). Before and after the intervention, vascular function (carotid artery reactivity) and cardiovascular risk factors (blood pressure, total cholesterol, HDL-cholesterol and Framingham risk score) were examined. A 7-point oral glucose tolerance test was performed to determine insulin resistance (Matsuda index and HOMA-IR) and disposition index. RESULTS Following drop-out (n = 18), 101 participants finished the intervention (54 women, 61 ± 7 years, 27.6 [26.4;30.0] kg/m2), with n = 80 available for the primary outcome of vascular function. Overall, the dietary interventions significantly decreased blood pressure, total cholesterol, HDL-cholesterol and the Framingham risk score (all p < 0.05), while vascular function was not affected (p = 0.485). Insulin resistance (p ≤ 0.001), but not disposition index (p = 0.362), was significantly improved after intervention. The Matsuda index (p = 0.078) tended to increase more and total cholesterol (p = 0.052) tended to decrease more in PhenoDiet group B than A, but other changes in outcome parameters were not significantly different between PhenoDiet groups. The LFHP diet resulted in more pronounced improvements in cholesterol, diastolic blood pressure (DBP) and insulin resistance compared with the HMUFA diet (all p < 0.05). CONCLUSION A 12-week diet improves metabolic and cardiovascular outcomes, but not vascular function in insulin-resistant adults with overweight or obesity. Whilst the LFHP diet resulted in greater improvements in cardiometabolic risk markers than the HMUFA diet, we found no significant differences between the PhenoDiet groups.
Collapse
Affiliation(s)
- Lisa Wanders
- Radboud Institute for Health Sciences, Department of PhysiologyRadboud university medical centerNijmegenThe Netherlands
- TiFNWageningenThe Netherlands
| | - Anouk Gijbels
- TiFNWageningenThe Netherlands
- Division of Human Nutrition and HealthWageningen UniversityWageningenThe Netherlands
| | - Gabby B. Hul
- Department of Human BiologyInstitute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical CenterMaastrichtThe Netherlands
| | - Edith J. M. Feskens
- Division of Human Nutrition and HealthWageningen UniversityWageningenThe Netherlands
| | - Lydia A. Afman
- Division of Human Nutrition and HealthWageningen UniversityWageningenThe Netherlands
| | - Ellen E. Blaak
- Department of Human BiologyInstitute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical CenterMaastrichtThe Netherlands
| | - Maria T. E. Hopman
- Radboud Institute for Health Sciences, Department of PhysiologyRadboud university medical centerNijmegenThe Netherlands
| | - Gijs H. Goossens
- Department of Human BiologyInstitute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical CenterMaastrichtThe Netherlands
| | - Dick H. J. Thijssen
- Radboud Institute for Health Sciences, Department of PhysiologyRadboud university medical centerNijmegenThe Netherlands
- Research Institute for Sport and Exercise SciencesLiverpool John Moores UniversityLiverpoolUK
| |
Collapse
|
47
|
Shin S, Kim HY, Kim SY, Kim J. Clinical Characteristics of Korean Patients with Youth-Onset Type 2 Diabetes Mellitus in Remission. J Obes Metab Syndr 2025; 34:158-165. [PMID: 40194890 PMCID: PMC12067001 DOI: 10.7570/jomes24042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/12/2025] [Accepted: 03/26/2025] [Indexed: 04/09/2025] Open
Abstract
Background Improving β-cell function can lead to remission in some patients with type 2 diabetes mellitus (T2DM). However, research on pharmacotherapy-induced remission in youth-onset T2DM remains scarce. Our study aimed to identify the clinical characteristics of pediatric patients who experience remission. Methods We retrospectively reviewed 88 pediatric patients with T2DM followed for at least 1 year at Seoul National University Bundang Hospital between 2013 and 2023. Remission was defined as a glycosylated hemoglobin (HbA1c) level less than 6.5% for at least 3 months after ceasing glucose-lowering pharmacotherapy. Results Among 88 patients (60 males, 68.2%) diagnosed at an average age of 14.4±2.1 years, 19 patients (21.6%) achieved remission after a median duration of 1.4 years. The remission group had a larger proportion of males (89.5% vs. 62.3%, P=0.024) and a lower urinary albumin-to-creatinine ratio (ACR) at diagnosis (P=0.011). They also showed lower HbA1c levels at 1 year and more significant changes in HbA1c and body mass index (all P<0.05). Higher urinary ACR levels correlated with a longer duration to achieve remission (hazard ratio, 0.928; P=0.013). In three of the 19 remission patients (15.8%), recurrence occurred after a median of 1.5 years. Conclusion Among Korean youth with T2DM, 21.6% achieved remission after a median duration of 1.4 years. Those who experienced remission were predominantly male, had lower ACR at diagnosis, and had significant weight loss within the first year. Further investigation into the factors influencing remission and long-term outcomes is essential.
Collapse
Affiliation(s)
- Sohyun Shin
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hwa Young Kim
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Se Young Kim
- Department of Pediatrics, Bundang Jesaeng Hospital, Daejin Medical Center, Seongnam, Korea
| | - Jaehyun Kim
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
48
|
Herz CT, Kulterer OC, Prager M, Marculescu R, Prager G, Kautzky-Willer A, Hacker M, Trajanoski S, Köfeler HC, Gallé B, Haug AR, Berry D, Kiefer FW. Bariatric surgery promotes recruitment of brown fat linked to alterations in the gut microbiota. Eur J Endocrinol 2025; 192:603-611. [PMID: 40366070 DOI: 10.1093/ejendo/lvaf081] [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: 12/10/2024] [Revised: 03/10/2025] [Indexed: 05/15/2025]
Abstract
OBJECTIVE The mechanisms of bariatric surgery-induced weight loss and metabolic improvements are still incompletely understood and reach beyond malabsorption or calorie restriction. We sought to investigate the effect of bariatric surgery on brown adipose tissue (BAT) activity and a potential connection with changes in energy metabolism, the gut microbiota, and short-chain fatty acid (SCFA) composition. METHODS We included 32 subjects (25 females) with morbid obesity and analyzed their metabolic profile, gut microbiota composition, circulating SCFAs, energy expenditure, and cold-induced BAT activity using [18F]Fluorodeoxyglucose-positron emission tomography-computed tomography before and up to 1 year after bariatric surgery. RESULTS Twelve months after surgery, the percentage of individuals with active BAT had increased from 28% to 53%. The BAT-negative (BATneg) individuals who had an adverse metabolic profile at baseline compared with subjects with active BAT (BATpos) showed a greater metabolic benefit after surgery. While no changes in overall gut bacterial diversity were observed between BATpos and BATneg, the abundance of 3 specific bacterial families, including Akkermansiaceae, Pasteurellaceae, and Carnobacteriaceae, was distinctly regulated between BAT groups. The bacterial genera most strongly increased in BATpos vs BATneg subjects were all positively correlated with BAT volume and BAT activity. Finally, circulating concentrations of the SCFAs acetate, butyrate, and propionate rose after bariatric surgery and were related to bacterial genera such as Akkermansia, Dialister, and Lachnospiraceae FCS020 group, all known SCFA producers. CONCLUSIONS Bariatric surgery helps recruit active BAT in individuals with obesity and is linked to distinct alterations in the gut microbiome and SCFA composition. TRIAL REGISTRATION NUMBER ClinicalTrials.gov (NCT03168009).
Collapse
Affiliation(s)
- Carsten T Herz
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Oana C Kulterer
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Marlene Prager
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Rodrig Marculescu
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Gerhard Prager
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Alexandra Kautzky-Willer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Slave Trajanoski
- Core Facility Computational Bioanalytics, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Harald C Köfeler
- Core Facility Mass Spectrometry, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Birgit Gallé
- Core Facility Molecular Biology, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Alexander R Haug
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - David Berry
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
| | - Florian W Kiefer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
49
|
Feola T, De Alcubierre D, Puliani G, Cozzolino A, Sciarra F, Vincenzi L, Hasenmajer V, Sada V, Appetecchia M, Giannetta E, Sbardella E, Jaffrain-Rea ML, Venneri MA, Isidori AM. Circulating immune cell profile in patients with acromegaly: results from the PROMISE, a prospective clinical trial. Eur J Endocrinol 2025; 192:577-589. [PMID: 40341338 DOI: 10.1093/ejendo/lvaf085] [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: 01/31/2025] [Revised: 03/29/2025] [Indexed: 05/10/2025]
Abstract
OBJECTIVE Although GH and IGF-1 have long been proposed to play a role in immune-modulation, the circulating immune cell phenotype in acromegaly (ACRO) is poorly understood. DESIGN This observational, prospective, single-site clinical trial (NCT05069324) analyzed peripheral blood mononuclear cell (PBMC) subpopulations in ACRO, investigating the role of disease control, pharmacological treatments, and metabolic profile. METHODS Sixty consecutive patients with ACRO (34 males, mean age 54.7 ± 15) attending an outpatient visit between July 2020 and December 2024 were enrolled. Patients were compared with two populations: 40 healthy controls (CTRL) and 40 patients with type 2 diabetes (T2DM), with no significant differences in age or sex. An 8-week follow-up evaluation was performed and compared among patients according to the introduction or changes in their pharmacological treatment. RESULTS Compared with CTRL, ACRO patients had lower total monocytes with a decreased percentage of classical and an increased proportion of non-classical subsets. There were fewer NK cells, with higher CD56dim and lower CD56bright subpopulations, and similar T and B lymphocytes. Compared with T2DM, ACRO showed lower total monocytes, with higher classical and lower non-classical subsets, as well as lower NK cells and B lymphocytes. In the short-term, treatment appeared unable to restore immune cell profile but partially affected the distributions of innate immune cells subpopulations. CONCLUSIONS This is the first evidence of a distinct immunological pattern in ACRO that is independent of glucose metabolism. The immune signature may contribute to the persistence of cardio-metabolic and oncological risk observed in ACRO, even when the disease is adequately controlled by medical treatment.
Collapse
Affiliation(s)
- Tiziana Feola
- Department of Experimental Medicine, Sapienza University of Rome, Rome 00161, Italy
- Neuroendocrinology, Neuromed Institute, IRCCS, Pozzilli 86077, Italy
| | - Dario De Alcubierre
- Department of Experimental Medicine, Sapienza University of Rome, Rome 00161, Italy
- Neuroendocrinology, Neuromed Institute, IRCCS, Pozzilli 86077, Italy
| | - Giulia Puliani
- Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, Rome 00144, Italy
| | - Alessia Cozzolino
- Department of Experimental Medicine, Sapienza University of Rome, Rome 00161, Italy
| | - Francesca Sciarra
- Department of Experimental Medicine, Sapienza University of Rome, Rome 00161, Italy
| | - Ludovica Vincenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome 00161, Italy
| | - Valeria Hasenmajer
- Department of Experimental Medicine, Sapienza University of Rome, Rome 00161, Italy
| | - Valentina Sada
- Department of Experimental Medicine, Sapienza University of Rome, Rome 00161, Italy
| | - Marialuisa Appetecchia
- Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, Rome 00144, Italy
| | - Elisa Giannetta
- Department of Experimental Medicine, Sapienza University of Rome, Rome 00161, Italy
| | - Emilia Sbardella
- Department of Experimental Medicine, Sapienza University of Rome, Rome 00161, Italy
| | - Marie-Lise Jaffrain-Rea
- Neuroendocrinology, Neuromed Institute, IRCCS, Pozzilli 86077, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila 67100, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Rome 00161, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Rome 00161, Italy
- Policlinico Umberto I, Centre for Rare Diseases (Endo-ERN Accredited), Rome 00161, Italy
| |
Collapse
|
50
|
Dehzad MJ, Raja A, Moghdani Z, Sohrabi Z, Fararooei M, Famouri M, Askarpour M, Babajafari S. Effects of Yogurt Enriched with Konjac Glucomannan and Inulin on Insulin Sensitivity, Glycemic Control, Lipid Profiles, Anthropometric Measures and Oxidative Stress in Type 2 Diabetes Mellitus: A Randomized Controlled Trial. Prev Nutr Food Sci 2025; 30:120-131. [PMID: 40352303 PMCID: PMC12061536 DOI: 10.3746/pnf.2025.30.2.120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 05/14/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a prevalent metabolic disorder that requires effective dietary strategies for management. In this randomized, double-blind, placebo-controlled clinical trial, the effects of low-fat yogurt enriched with konjac glucomannan (KGM) and inulin on glycemic control, lipid profiles, anthropometric indices, and oxidative stress were investigated in patients with T2DM. Eighty participants were randomly assigned to consume either 150 g of yogurt enriched with 1.5 g of KGM and 1.5 g of inulin (n=40) or plain low-fat yogurt (n=40) daily for 8 weeks. The primary outcomes were fasting blood glucose and fasting insulin levels, insulin sensitivity indices [homeostasis model assessment for insulin resistance (HOMA-IR) and quantitative insulin sensitivity check index (QUICKI)], and glycated hemoglobin. Secondary outcomes included lipid profile [total cholesterol (TC), low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, and triglyceride (TG)], anthropometric indices (weight, body mass index, fat mass, skeletal muscle, and waist circumference), and oxidative stress markers. Compared to control group, the intervention significantly improved fasting insulin levels (-1.85 µIU/mL, P=0.042), HOMA-IR (-0.89, P=0.029), and QUICKI (0.11, P=0.032). Lipid profile analysis revealed reductions in TC (-18.51 mg/dL, P=0.049) and TG levels (-15.0 mg/dL, P=0.041). These findings suggest that daily consumption of yogurt fortified with KGM and inulin significantly enhances insulin sensitivity and lipid profiles in patients with T2DM over an 8-week period. This dietary intervention shows promise as a complementary strategy for T2DM management. Further studies are needed to assess the long-term outcomes, optimize doses, and elucidate the underlying mechanisms of this intervention.
Collapse
Affiliation(s)
- Mohammad Jafar Dehzad
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz 7153675500, Iran
| | - Ali Raja
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz 7153675500, Iran
| | - Zahra Moghdani
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz 7153675500, Iran
| | - Zahra Sohrabi
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz 7153675500, Iran
| | - Mohammad Fararooei
- Department of Epidemiology, School of Health, Shiraz University of Medical Sciences, Shiraz 7153675500, Iran
| | - Mandana Famouri
- Dairy Expert at Research and Development of Zarrin Ghazal Company (DAITY), Shiraz 7158188785, Iran
| | - Moein Askarpour
- Social Determinants of Health Research Center, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman 7616911320, Iran
| | - Siavash Babajafari
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz 7153675500, Iran
| |
Collapse
|