1
|
Yurchishin ML, Fowler LA, Goss AM, Garvey WT, Gower BA. Predictability of genetic risk score for insulin resistance is influenced by both BMI and race. Obesity (Silver Spring) 2025; 33:788-795. [PMID: 39994503 PMCID: PMC11937860 DOI: 10.1002/oby.24238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/05/2024] [Accepted: 12/19/2024] [Indexed: 02/26/2025]
Abstract
OBJECTIVE The study objective was to determine whether associations between a genetic risk score (GRS) for insulin resistance (IR) and measures of insulin sensitivity differ by race and/or BMI status in African American (AA) and European American (EA) adults without diabetes. METHODS Fifty-three AA and 54 EA participants were classified into "high" or "low" BMI groups using the sample median (25.9 kg/m2) as the cut point. The GRS was derived from 52 previously identified genetic variants. Skeletal muscle insulin sensitivity was measured with the hyperinsulinemic-euglycemic clamp. The homeostasis model assessment of insulin resistance (HOMA-IR) and the Matsuda index of insulin sensitivity were calculated from oral glucose tolerance test values to determine hepatic and whole-body insulin sensitivity, respectively. Linear regression models, stratified by race, assessed interactions between BMI status and GRS on measures of insulin sensitivity. RESULTS In EA participants, associations of GRS with HOMA-IR and the Matsuda index differed by BMI status, where the GRS was associated with IR in the high-BMI group only. In AA participants, associations from the clamp differed by BMI status, but an association was observed only in the low-BMI group. CONCLUSIONS These results highlight the heterogeneity of IR and support the hypothesis that the relationship between genetic predisposition for IR and obesity is race- and tissue-specific.
Collapse
Affiliation(s)
- Marian L. Yurchishin
- Department of Nutrition SciencesUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Lauren A. Fowler
- Department of Nutrition SciencesUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Amy M. Goss
- Department of Nutrition SciencesUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - William T. Garvey
- Department of Nutrition SciencesUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Barbara A. Gower
- Department of Nutrition SciencesUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
2
|
Lee H, Rhee TM, Choi JM, Choi SY, Kim DW. The Close Link Between Obesity and Cardiovascular Disease: Current Insights and Remaining Challenges. Endocrinol Metab Clin North Am 2025; 54:175-192. [PMID: 39919874 DOI: 10.1016/j.ecl.2024.10.005] [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: 02/09/2025]
Abstract
Obesity is a global public health crisis, contributing to chronic disease development and poor prognosis. A large body of evidence consistently demonstrates that increased adiposity leads to many cardiovascular diseases (CVDs) and complications, such as coronary artery disease, heart failure, and arrhythmias, via direct and indirect mechanisms. Therefore, weight management is crucial to reduce and prevent cardiovascular risk. The recent emergence of glucose-like peptide-1 receptor agonists shows remarkable weight reduction and cardiovascular prevention. Despite the clear benefits, controversies and challenges on obesity-related CVD remain. This review aims to provide a comprehensive understanding of obesity-related CVD and explore current remaining tasks.
Collapse
Affiliation(s)
- Heesun Lee
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; Division of Cardiology, Department of Internal Medicine, Healthcare System Gangnam Centre, Seoul National University Hospital, 39th Floor, Gangnam Finance Center, 152 Teheran-ro, Gangnam-gu, Seoul 06236, Republic of Korea.
| | - Tae-Min Rhee
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; Division of Cardiology, Department of Internal Medicine, Healthcare System Gangnam Centre, Seoul National University Hospital, 39th Floor, Gangnam Finance Center, 152 Teheran-ro, Gangnam-gu, Seoul 06236, Republic of Korea
| | - Ji Min Choi
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; Division of Gastroenterology, Department of Internal Medicine, Healthcare System Gangnam Centre, Seoul National University Hospital, 39th Floor, Gangnam Finance Center, 152 Teheran-ro, Gangnam-gu, Seoul 06236, Republic of Korea
| | - Su-Yeon Choi
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; Division of Cardiology, Department of Internal Medicine, Healthcare System Gangnam Centre, Seoul National University Hospital, 39th Floor, Gangnam Finance Center, 152 Teheran-ro, Gangnam-gu, Seoul 06236, Republic of Korea
| | - Dong Wook Kim
- Division of Endocrinology, Diabetes & Hypertension, Brigham and Women's Hospital, 221 Longwood Avenue, RFB490, Boston, MA 02115, USA
| |
Collapse
|
3
|
Weeldreyer NR, De Guzman JC, Paterson C, Allen JD, Gaesser GA, Angadi SS. Cardiorespiratory fitness, body mass index and mortality: a systematic review and meta-analysis. Br J Sports Med 2025; 59:339-346. [PMID: 39537313 PMCID: PMC11874340 DOI: 10.1136/bjsports-2024-108748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE The purpose of this review was to assess the joint relationship of cardiorespiratory fitness (CRF) and Body Mass Index (BMI) on both cardiovascular disease (CVD) and all-cause mortality risk. DESIGN A systematic review and meta-analysis was conducted. Pooled HR and 95% CI were calculated using a three-level restricted maximum likelihood estimation random-effects model with robust variance estimation. The reference group was normal weight-fit and was compared with normal weight-unfit, overweight-unfit and fit, and obese-unfit and fit. DATA SOURCES Electronic databases (PubMed/MEDLINE, Web of Science and SportDiscus) were searched following registration on PROSPERO. ELIGIBILITY CRITERIA Articles meeting the following criteria were included: (1) published between January 1980 and February 2023, (2) prospective cohort study, (3) CRF assessed using a maximal or VO2peak exercise test, (4) BMI reported and directly measured, (5) joint impact of CRF and BMI on all-cause mortality or CVD mortality were analysed, and (6) the reference group was normal weight, fit individuals. RESULTS 20 articles were included in the analysis resulting in a total of 398 716 observations. Compared with the reference group, overweight-fit (CVD HR (95% CI): 1.50 (0.82-2.76), all-cause HR: 0.96 (0.61-1.50)) and obese-fit (CVD: 1.62 (0.87-3.01), all-cause: 1.11 (0.88-1.40)) did not have a statistically different risk of mortality. Normal weight-unfit (CVD: 2.04 (1.32-3.14), all-cause: 1.92 (1.43-2.57)), overweight-unfit (CVD: 2.58 (1.48-4.52), all-cause: 1.82 (1.47-2.24)) and obese-unfit (CVD: 3.35 (1.17-9.61), all-cause: 2.04 (1.54-2.71)) demonstrated 2-3-fold greater mortality risks. CONCLUSIONS CRF is a strong predictor of CVD and all-cause mortality and attenuates risks associated with overweight and obesity. These data have implications for public health and risk mitigation strategies.
Collapse
Affiliation(s)
- Nathan R Weeldreyer
- Department of Kinesiology, School of Education and Human Development, University of Virginia, Charlottesville, Virginia, USA
| | - Jeison C De Guzman
- Department of Kinesiology, School of Education and Human Development, University of Virginia, Charlottesville, Virginia, USA
| | - Craig Paterson
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jason D Allen
- Department of Kinesiology, School of Education and Human Development, University of Virginia, Charlottesville, Virginia, USA
| | - Glenn A Gaesser
- College of Health Solutions, Arizona State University, Phoenix, Arizona, USA
| | - Siddhartha S Angadi
- Department of Kinesiology, School of Education and Human Development, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
4
|
Xu W, Mesa-Eguiagaray I, Morris DM, Wang C, Gray CD, Sjöström S, Papanastasiou G, Badr S, Paccou J, Li X, Timmers PRHJ, Timofeeva M, Farrington SM, Dunlop MG, Semple SI, MacGillivray T, Theodoratou E, Cawthorn WP. Deep learning and genome-wide association meta-analyses of bone marrow adiposity in the UK Biobank. Nat Commun 2025; 16:99. [PMID: 39747859 PMCID: PMC11697225 DOI: 10.1038/s41467-024-55422-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025] Open
Abstract
Bone marrow adipose tissue is a distinct adipose subtype comprising more than 10% of fat mass in healthy humans. However, the functions and pathophysiological correlates of this tissue are unclear, and its genetic determinants remain unknown. Here, we use deep learning to measure bone marrow adiposity in the femoral head, total hip, femoral diaphysis, and spine from MRI scans of approximately 47,000 UK Biobank participants, including over 41,000 white and over 6300 non-white participants. We then establish the heritability and genome-wide significant associations for bone marrow adiposity at each site. Our meta-GWAS in the white population finds 67, 147, 134, and 174 independent significant single nucleotide polymorphisms, which map to 54, 90, 43, and 100 genes for the femoral head, total hip, femoral diaphysis, and spine, respectively. Transcriptome-wide association studies, colocalization analyses, and sex-stratified meta-GWASes in the white participants further resolve functional and sex-specific genes associated with bone marrow adiposity at each site. Finally, we perform a multi-ancestry meta-GWAS to identify genes associated with bone marrow adiposity across the different bone regions and across ancestry groups. Our findings provide insights into BMAT formation and function and provide a basis to study the impact of BMAT on human health and disease.
Collapse
Affiliation(s)
- Wei Xu
- Centre for Global Health and Molecular Epidemiology, Usher Institute, University of Edinburgh, Edinburgh, UK
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh, UK
| | - Ines Mesa-Eguiagaray
- Centre for Global Health and Molecular Epidemiology, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - David M Morris
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh, UK
| | - Chengjia Wang
- Edinburgh Imaging, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh, UK
- School of Mathematics and Computer Sciences, Heriot-Watt University, Edinburgh, UK
| | - Calum D Gray
- Edinburgh Imaging, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh, UK
| | - Samuel Sjöström
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh, UK
| | - Giorgos Papanastasiou
- Edinburgh Imaging, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh, UK
- Archimedes Unit, Athena Research Centre, Marousi, Greece
| | - Sammy Badr
- Univ. Lille, CHU Lille, Marrow Adiposity and Bone Laboratory (MABlab) ULR 4490, Department of Rheumatology, Lille, France
| | - Julien Paccou
- Univ. Lille, CHU Lille, Marrow Adiposity and Bone Laboratory (MABlab) ULR 4490, Department of Rheumatology, Lille, France
| | - Xue Li
- Department of Big Data in Health Science, School of Public Health and The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Paul R H J Timmers
- Medical Research Council Human Genetics Unit, Medical Research Council Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Maria Timofeeva
- Medical Research Council Human Genetics Unit, Medical Research Council Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Danish Institute for Advanced Study (DIAS), Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Susan M Farrington
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Colon Cancer Genetics Group, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Malcolm G Dunlop
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Colon Cancer Genetics Group, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Scott I Semple
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh, UK
| | - Tom MacGillivray
- Centre for Clinical Brain Sciences, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh, UK
| | - Evropi Theodoratou
- Centre for Global Health and Molecular Epidemiology, Usher Institute, University of Edinburgh, Edinburgh, UK.
- Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.
| | - William P Cawthorn
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh, UK.
| |
Collapse
|
5
|
Grannes H, Sharma A, Suntharalingam A, Michelsen AE, Aukrust P, Ueland T, Birkeland KI, Gregersen I, Lee-Ødegård S, Halvorsen B. Plasma soluble TIM-3 is increased in normoglycemic South Asian women compared to Nordic women after gestational diabetes mellitus and associated with markers of metaflammation. Heliyon 2024; 10:e40339. [PMID: 39641064 PMCID: PMC11617222 DOI: 10.1016/j.heliyon.2024.e40339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/10/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024] Open
Abstract
Background Women with South Asian ethnicity have a higher risk of developing type 2 diabetes mellitus (T2DM) compared with white women of European descent, especially after gestational diabetes mellitus (GDM). Central obesity and adipose tissue dysfunction have been linked to their higher risk of T2DM, but the mechanisms are not known. We hypothesize that low-grade, persistent immune cell activation is involved in metabolic disturbances following GDM with different influence according to ethnicity. Methods We measured plasma levels of T cell exhaustion marker soluble T cell immunoglobin mucin domain 3 (sTIM-3), sCD25, sCD27 and soluble lymphocyte activation gene (sLAG)-3 in 266 women of South Asian (n = 160) and white Nordic (n = 106) ethnic background with a history of GDM. Results Baseline plasma concentration of sTIM-3 was higher in South Asian women compared to Nordic women (p < 0.001). This difference was driven by higher sTIM-3 in South Asian women with NGT, compared to their Nordic counterparts (p = 0.005) but there were no significant differences comparing Nordic and South Asian women with altered glucose tolerance (AGT). Soluble TIM-3 correlated positively with waist-height ratio (WHtR) and body mass index across all groups, but whereas sTIM-3 correlated moderately and consistently with markers of metaflammation in South Asians, this pattern was not found in Nordic women. Mediation analysis indicated that 15 % of the difference found in adipose insulin resistance between ethnicities could be mediated by sTIM-3, and that 33 % of the difference in sTIM-3 concentrations could be mediated by WHtR. Moreover, T cell markers sCD27 and sLAG3 were also increased in South Asian women compared with Nordic women, further supporting involvement of T cell activation in these women. Conclusion We found increased levels of sTIM-3, as well as additional markers of T cell activation/exhaustion, in a population of normoglycemic South Asian women with previous gestational diabetes as compared to women of Nordic descent. The possible causal relationship between T cell activation and metabolic dysfunction in high-risk South Asian women is however still elusive and merits further investigation.
Collapse
Affiliation(s)
- Helene Grannes
- Research Institute for Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Archana Sharma
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Endocrinology, Akershus University Hospital, University of Oslo, 1478, Lørenskog, Norway
| | - Anita Suntharalingam
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Dep. of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Annika E. Michelsen
- Research Institute for Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute for Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Thor Ueland
- Research Institute for Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Thrombosis Research and Expertise Centre, University of Tromsø, Tromsø, Norway
| | - Kåre I. Birkeland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Dep. of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Ida Gregersen
- Research Institute for Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Sindre Lee-Ødegård
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Dep. of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Bente Halvorsen
- Research Institute for Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Deutsch AJ, Smith K, Udler MS. Genetic variants explain ancestry-related differences in type 2 diabetes risk. Clin Transl Med 2024; 14:e70076. [PMID: 39500627 PMCID: PMC11537768 DOI: 10.1002/ctm2.70076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 10/05/2024] [Indexed: 11/09/2024] Open
Affiliation(s)
- Aaron J. Deutsch
- Diabetes UnitEndocrine DivisionDepartment of MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Center for Genomic MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Kirk Smith
- Diabetes UnitEndocrine DivisionDepartment of MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Center for Genomic MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
| | - Miriam S. Udler
- Diabetes UnitEndocrine DivisionDepartment of MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Center for Genomic MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
7
|
Börgeson E, Tavajoh S, Lange S, Jessen N. The challenges of assessing adiposity in a clinical setting. Nat Rev Endocrinol 2024; 20:615-626. [PMID: 39009863 DOI: 10.1038/s41574-024-01012-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 07/17/2024]
Abstract
To tackle the burden of obesity-induced cardiometabolic disease, the scientific community relies on accurate and reproducible adiposity measurements in the clinic. These measurements guide our understanding of underlying biological mechanisms and clinical outcomes of human trials. However, measuring adiposity and adipose tissue distribution in a clinical setting can be challenging, and different measurement methods pose important limitations. BMI is a simple and high-throughput measurement, but it is associated relatively poorly with clinical outcomes when compared with waist-to-hip and sagittal abdominal diameter measurements. Body composition measurements by dual energy X-ray absorptiometry or MRI scans would be ideal due to their high accuracy, but are not high-throughput. Another important consideration is that adiposity measurements vary between men and women, between adults and children, and between people of different ethnic backgrounds. In this Perspective article, we discuss how these critical challenges can affect our interpretation of research data in the field of obesity and the design and implementation of clinical guidelines.
Collapse
Affiliation(s)
- Emma Börgeson
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark.
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Saeideh Tavajoh
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Stephan Lange
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Cardiology, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Niels Jessen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
8
|
Liu Y, Li D, Liang Y. Comorbidities and mortality risk in COVID-19 patients with congestive heart failure: A comprehensive analysis. Heliyon 2024; 10:e35746. [PMID: 39170371 PMCID: PMC11336877 DOI: 10.1016/j.heliyon.2024.e35746] [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: 11/09/2023] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
The COVID-19 pandemic has posed unprecedented challenges to global healthcare systems, resulting in alarming incidence and mortality rates among patients with comorbidities, including heart failure. Understanding the characteristics of heart failure and other comorbidities during the COVID-19 pandemic is crucial for effective prevention and treatment. However, the current understanding of these characteristics among different racial groups remains incomplete. In this study, we investigated a cohort of 4711 patients, classifying them into congestive heart failure (CHF) and non-CHF groups. Biomarker analysis revealed noteworthy variations in blood urea nitrogen, aspartate aminotransferase, and white blood cell levels based on the presence or absence of CHF. Stratified by three racial groups, univariate logistic regression analysis identified significant differences in multiple variables, including CHF. Subsequent univariate Cox regression and Kaplan-Meier analysis demonstrated variations in mortality factors among distinct populations, with age and comorbidity playing prominent roles. This study utilized a large-scale database to investigate the characteristics of heart failure and related variables during the COVID-19 pandemic. The findings revealed distinctive mortality risk factors among various racial groups, emphasizing the significance of customized risk assessment and management approaches for diverse populations. These findings also provide a valuable resource for the development of targeted interventions and the promotion of equitable healthcare outcomes in the context of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Yi Liu
- College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan, 030024, China
- Key Laboratory of Big Data Fusion Analysis and Application of Shanxi Province, Taiyuan, 030024, China
- Intelligent Perception Engineering Technology Center of Shanxi, Taiyuan, 030024, China
| | - Dengao Li
- College of Computer Science and Technology (College of Data Science), Taiyuan University of Technology, Taiyuan, 030024, China
- Key Laboratory of Big Data Fusion Analysis and Application of Shanxi Province, Taiyuan, 030024, China
- Intelligent Perception Engineering Technology Center of Shanxi, Taiyuan, 030024, China
| | - Yuchen Liang
- Shanxi Cardiovascular Hospital, Taiyuan, 030027, China
| |
Collapse
|
9
|
Minato-Inokawa S, Honda M, Tsuboi-Kaji A, Takeuchi M, Kitaoka K, Kurata M, Wu B, Kazumi T, Fukuo K. Associations of adipose insulin resistance index with pancreatic β cell function (inverse) and glucose excursion (positive) in young Japanese women. Sci Rep 2024; 14:18590. [PMID: 39127728 PMCID: PMC11316777 DOI: 10.1038/s41598-024-69181-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
The relationship of adipose tissue insulin resistance (AT-IR, a product of fasting insulin and free fatty acids) and homeostasis-model assessment-insulin resistance (HOMA-IR) to β-cell function was studied cross-sectionally in the setting of subtle glucose dysregulation. Associations of AT-IR and HOMA-IR with fasting and post-glucose glycemia and β-cell function inferred from serum insulin kinetics during a 75 g oral glucose tolerance test were studied in 168 young female Japanese students. β-cell function was evaluated by disposition index calculated as a product of the insulinogenic index (IGI) and Matsuda index. AT-IR, not HOMA-IR, showed positive associations with post-glucose glycemia and area under the glucose response curve although both indices were associated with fasting glycemia. HOMA-IR, not AT-IR, was associated positively with log IGI whereas both indices were inversely associated with Matsuda index. AT-IR, not HOMA-IR, showed inverse associations with log disposition index. Associations of adipose tissue insulin resistance with β-cell function (inverse) and glucose excursion in young Japanese women may suggest that lipotoxicity to pancreatic β-cells for decades may be associated with β cell dysfunction found in Japanese patients with type 2 diabetes. Positive association of HOMA-IR with insulinogenic index may be associated with compensatory increased insulin secretion.
Collapse
Affiliation(s)
- Satomi Minato-Inokawa
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-Cho, Nishinomiya, Hyogo, 663-8558, Japan
- Laboratory of Community Health and Nutrition, Department of Bioscience, Graduate School of Agriculture, Ehime University, Matsuyama, Ehime, Japan
| | - Mari Honda
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Health, Sports, and Nutrition, Faculty of Health and Welfare, Kobe Women's University, Kobe, Hyogo, Japan
| | - Ayaka Tsuboi-Kaji
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-Cho, Nishinomiya, Hyogo, 663-8558, Japan
- Department of Nutrition, Osaka City Juso Hospital, Osaka, Japan
| | - Mika Takeuchi
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-Cho, Nishinomiya, Hyogo, 663-8558, Japan
| | - Kaori Kitaoka
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-Cho, Nishinomiya, Hyogo, 663-8558, Japan
- Department of Advanced Epidemiology, Noncommunicable Disease (NCD) Epidemiology Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Miki Kurata
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-Cho, Nishinomiya, Hyogo, 663-8558, Japan
- Department of Food Sciences and Nutrition, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Bin Wu
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Endocrinology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Tsutomu Kazumi
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-Cho, Nishinomiya, Hyogo, 663-8558, Japan.
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan.
- Department of Medicine, Kohan Kakogawa Hospital, Kakogawa, Hyogo, Japan.
| | - Keisuke Fukuo
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-Cho, Nishinomiya, Hyogo, 663-8558, Japan
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Food Sciences and Nutrition, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| |
Collapse
|
10
|
Mai TP, Luong BA, Ma PT, Tran TV, Dinh Ngo TT, Hoang CK, Van Tran L, Le BH, Vu HA, Le LHG, Le KT, Truong S, Tran NQ, Do MD. Genome-wide association and polygenic risk score estimation of type 2 diabetes mellitus in Kinh Vietnamese-A pilot study. J Cell Mol Med 2024; 28:e18526. [PMID: 38957036 PMCID: PMC11220366 DOI: 10.1111/jcmm.18526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 07/04/2024] Open
Abstract
A genome-wide association study (GWAS) is a powerful tool in investigating genetic contribution, which is a crucial factor in the development of complex multifactorial diseases, such as type 2 diabetes mellitus. Type 2 diabetes mellitus is a major healthcare burden in the Western Pacific region; however, there is limited availability of genetic-associated data for type 2 diabetes in Southeast Asia, especially among the Kinh Vietnamese population. This lack of information exacerbates global healthcare disparities. In this study, 997 Kinh Vietnamese individuals (503 with type 2 diabetes and 494 controls) were prospectively recruited and their clinical and paraclinical information was recorded. DNA samples were collected and whole genome genotyping was performed. Standard quality control and genetic imputation using the 1000 Genomes database were executed. A polygenic risk score for type 2 diabetes was generated in different models using East Asian, European, and mix ancestry GWAS summary statistics as training datasets. After quality control and genetic imputation, 107 polymorphisms reached suggestive statistical significance for GWAS (≤5 × 10-6) and rs11079784 was one of the potential markers strongly associated with type 2 diabetes in the studied population. The best polygenic risk score model predicting type 2 diabetes mellitus had AUC = 0.70 (95% confidence interval = 0.62-0.77) based on a mix of ancestral GWAS summary statistics. These data show promising results for genetic association with a polygenic risk score estimation in the Kinh Vietnamese population; the results also highlight the essential role of population diversity in a GWAS of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Thao Phuong Mai
- Department of Physiology‐Pathophysiology‐Immunology, Faculty of MedicineUniversity of Medicine and Pharmacy at Ho Chi Minh CityHo Chi Minh CityVietnam
| | - Bac An Luong
- Center for Molecular BiomedicineUniversity of Medicine and Pharmacy at Ho Chi Minh CityHo Chi Minh CityVietnam
| | - Phat Tung Ma
- Department of Endocrinology, Faculty of MedicineUniversity of Medicine and Pharmacy at Ho Chi Minh CityHo Chi Minh CityVietnam
- Department of EndocrinologyUniversity Medical Center Ho Chi Minh CityHo Chi Minh CityVietnam
| | - Thang Viet Tran
- Department of Endocrinology, Faculty of MedicineUniversity of Medicine and Pharmacy at Ho Chi Minh CityHo Chi Minh CityVietnam
- Department of EndocrinologyUniversity Medical Center Ho Chi Minh CityHo Chi Minh CityVietnam
| | - Tat Thang Dinh Ngo
- Department of EndocrinologyUniversity Medical Center Ho Chi Minh CityHo Chi Minh CityVietnam
| | - Chi Khanh Hoang
- Department of EndocrinologyUniversity Medical Center Ho Chi Minh CityHo Chi Minh CityVietnam
| | - Luong Van Tran
- Department of EndocrinologyUniversity Medical Center Ho Chi Minh CityHo Chi Minh CityVietnam
| | - Bao Hoang Le
- Department of EndocrinologyUniversity Medical Center Ho Chi Minh CityHo Chi Minh CityVietnam
| | - Hoang Anh Vu
- Center for Molecular BiomedicineUniversity of Medicine and Pharmacy at Ho Chi Minh CityHo Chi Minh CityVietnam
| | - Linh Hoang Gia Le
- Center for Molecular BiomedicineUniversity of Medicine and Pharmacy at Ho Chi Minh CityHo Chi Minh CityVietnam
| | - Khuong Thai Le
- Center for Molecular BiomedicineUniversity of Medicine and Pharmacy at Ho Chi Minh CityHo Chi Minh CityVietnam
| | - Steven Truong
- MIT Department of Biological EngineeringCambridgeMassachusettsUSA
| | - Nam Quang Tran
- Department of Endocrinology, Faculty of MedicineUniversity of Medicine and Pharmacy at Ho Chi Minh CityHo Chi Minh CityVietnam
- Department of EndocrinologyUniversity Medical Center Ho Chi Minh CityHo Chi Minh CityVietnam
| | - Minh Duc Do
- Center for Molecular BiomedicineUniversity of Medicine and Pharmacy at Ho Chi Minh CityHo Chi Minh CityVietnam
| |
Collapse
|
11
|
Abraham A, Cule M, Thanaj M, Basty N, Hashemloo MA, Sorokin EP, Whitcher B, Burgess S, Bell JD, Sattar N, Thomas EL, Yaghootkar H. Genetic Evidence for Distinct Biological Mechanisms That Link Adiposity to Type 2 Diabetes: Toward Precision Medicine. Diabetes 2024; 73:1012-1025. [PMID: 38530928 PMCID: PMC11109787 DOI: 10.2337/db23-1005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/22/2024] [Indexed: 03/28/2024]
Abstract
We aimed to unravel the mechanisms connecting adiposity to type 2 diabetes. We used MR-Clust to cluster independent genetic variants associated with body fat percentage (388 variants) and BMI (540 variants) based on their impact on type 2 diabetes. We identified five clusters of adiposity-increasing alleles associated with higher type 2 diabetes risk (unfavorable adiposity) and three clusters associated with lower risk (favorable adiposity). We then characterized each cluster based on various biomarkers, metabolites, and MRI-based measures of fat distribution and muscle quality. Analyzing the metabolic signatures of these clusters revealed two primary mechanisms connecting higher adiposity to reduced type 2 diabetes risk. The first involves higher adiposity in subcutaneous tissues (abdomen and thigh), lower liver fat, improved insulin sensitivity, and decreased risk of cardiometabolic diseases and diabetes complications. The second mechanism is characterized by increased body size and enhanced muscle quality, with no impact on cardiometabolic outcomes. Furthermore, our findings unveil diverse mechanisms linking higher adiposity to higher disease risk, such as cholesterol pathways or inflammation. These results reinforce the existence of adiposity-related mechanisms that may act as protective factors against type 2 diabetes and its complications, especially when accompanied by reduced ectopic liver fat. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Angela Abraham
- Joseph Banks Laboratories, College of Health and Science, University of Lincoln, Lincoln, U.K
| | | | - Marjola Thanaj
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, U.K
| | - Nicolas Basty
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, U.K
| | - M. Amin Hashemloo
- Department of Life Sciences, Brunel University London, Uxbridge, U.K
| | | | - Brandon Whitcher
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, U.K
- MRI Unit, Department of Radiology, The Royal Marsden National Health Service Foundation Trust, London, U.K
| | - Stephen Burgess
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, U.K
| | - Jimmy D. Bell
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, U.K
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| | - E. Louise Thomas
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, U.K
| | - Hanieh Yaghootkar
- Joseph Banks Laboratories, College of Health and Science, University of Lincoln, Lincoln, U.K
| |
Collapse
|
12
|
Bayoumi R, Farooqi M, Alawadi F, Hassanein M, Osama A, Mukhopadhyay D, Abdul F, Sulaiman F, Dsouza S, Mulla F, Ahmed F, AlSharhan M, Khamis A. Etiologies underlying subtypes of long-standing type 2 diabetes. PLoS One 2024; 19:e0304036. [PMID: 38805513 PMCID: PMC11132508 DOI: 10.1371/journal.pone.0304036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/05/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Attempts to subtype, type 2 diabetes (T2D) have mostly focused on newly diagnosed European patients. In this study, our aim was to subtype T2D in a non-white Emirati ethnic population with long-standing disease, using unsupervised soft clustering, based on etiological determinants. METHODS The Auto Cluster model in the IBM SPSS Modeler was used to cluster data from 348 Emirati patients with long-standing T2D. Five predictor variables (fasting blood glucose (FBG), fasting serum insulin (FSI), body mass index (BMI), hemoglobin A1c (HbA1c) and age at diagnosis) were used to determine the appropriate number of clusters and their clinical characteristics. Multinomial logistic regression was used to validate clustering results. RESULTS Five clusters were identified; the first four matched Ahlqvist et al subgroups: severe insulin-resistant diabetes (SIRD), severe insulin-deficient diabetes (SIDD), mild age-related diabetes (MARD), mild obesity-related diabetes (MOD), and a fifth new subtype of mild early onset diabetes (MEOD). The Modeler algorithm allows for soft assignments, in which a data point can be assigned to multiple clusters with different probabilities. There were 151 patients (43%) with membership in cluster peaks with no overlap. The remaining 197 patients (57%) showed extensive overlap between clusters at the base of distributions. CONCLUSIONS Despite the complex picture of long-standing T2D with comorbidities and complications, our study demonstrates the feasibility of identifying subtypes and their underlying causes. While clustering provides valuable insights into the architecture of T2D subtypes, its application to individual patient management would remain limited due to overlapping characteristics. Therefore, integrating simplified, personalized metabolic profiles with clustering holds greater promise for guiding clinical decisions than subtyping alone.
Collapse
Affiliation(s)
- Riad Bayoumi
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | | | - Fatheya Alawadi
- Endocrinology Department, Dubai Hospital, Dubai Health, Dubai, UAE
| | | | - Aya Osama
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Debasmita Mukhopadhyay
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Fatima Abdul
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Fatima Sulaiman
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Stafny Dsouza
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Fahad Mulla
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Fayha Ahmed
- Pathology Department, Dubai Hospital, Dubai Health, Dubai, UAE
| | - Mouza AlSharhan
- Pathology Department, Dubai Hospital, Dubai Health, Dubai, UAE
| | - Amar Khamis
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| |
Collapse
|
13
|
Chiang YTT, Kassotis CD. Molecular Assessment of Proadipogenic Effects for Common-Use Contraceptives and Their Mixtures. Endocrinology 2024; 165:bqae050. [PMID: 38648498 PMCID: PMC11081078 DOI: 10.1210/endocr/bqae050] [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: 12/20/2023] [Revised: 03/12/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
Hormonal contraceptives are widely prescribed due to their effectiveness and convenience and have become an integral part of family planning strategies worldwide. In the United States, approximately 65% of reproductive-aged women are estimated to be using contraceptive options, with approximately 33% using one or a combination of hormonal contraceptives. While these methods have undeniably contributed to improved reproductive health, recent studies have raised concerns regarding their potential effect on metabolic health. Despite widespread anecdotal reports, epidemiological research has been mixed as to whether hormonal contraceptives contribute to metabolic health effects. As such, the goals of this study were to assess the adipogenic activity of common hormonal contraceptive chemicals and their mixtures. Five different models of adipogenesis were used to provide a rigorous assessment of metabolism-disrupting effects. Interestingly, every individual contraceptive (both estrogens and progestins) and each mixture promoted significant adipogenesis (eg, triglyceride accumulation and/or preadipocyte proliferation). These effects appeared to be mediated in part through estrogen receptor signaling, particularly for the contraceptive mixtures, as cotreatment with fulvestrant acted to inhibit contraceptive-mediated proadipogenic effects on triglyceride accumulation. In conclusion, this research provides valuable insights into the complex interactions between hormonal contraceptives and adipocyte development. The results suggest that both progestins and estrogens within these contraceptives can influence adipogenesis, and the specific effects may vary based on the receptor disruption profiles. Further research is warranted to establish translation of these findings to in vivo models and to further assess causal mechanisms underlying these effects.
Collapse
Affiliation(s)
- Yu-Ting Tiffany Chiang
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA
| | - Christopher D Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
14
|
Smith K, Deutsch AJ, McGrail C, Kim H, Hsu S, Huerta-Chagoya A, Mandla R, Schroeder PH, Westerman KE, Szczerbinski L, Majarian TD, Kaur V, Williamson A, Zaitlen N, Claussnitzer M, Florez JC, Manning AK, Mercader JM, Gaulton KJ, Udler MS. Multi-ancestry polygenic mechanisms of type 2 diabetes. Nat Med 2024; 30:1065-1074. [PMID: 38443691 PMCID: PMC11175990 DOI: 10.1038/s41591-024-02865-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/06/2024] [Indexed: 03/07/2024]
Abstract
Type 2 diabetes (T2D) is a multifactorial disease with substantial genetic risk, for which the underlying biological mechanisms are not fully understood. In this study, we identified multi-ancestry T2D genetic clusters by analyzing genetic data from diverse populations in 37 published T2D genome-wide association studies representing more than 1.4 million individuals. We implemented soft clustering with 650 T2D-associated genetic variants and 110 T2D-related traits, capturing known and novel T2D clusters with distinct cardiometabolic trait associations across two independent biobanks representing diverse genetic ancestral populations (African, n = 21,906; Admixed American, n = 14,410; East Asian, n =2,422; European, n = 90,093; and South Asian, n = 1,262). The 12 genetic clusters were enriched for specific single-cell regulatory regions. Several of the polygenic scores derived from the clusters differed in distribution among ancestry groups, including a significantly higher proportion of lipodystrophy-related polygenic risk in East Asian ancestry. T2D risk was equivalent at a body mass index (BMI) of 30 kg m-2 in the European subpopulation and 24.2 (22.9-25.5) kg m-2 in the East Asian subpopulation; after adjusting for cluster-specific genetic risk, the equivalent BMI threshold increased to 28.5 (27.1-30.0) kg m-2 in the East Asian group. Thus, these multi-ancestry T2D genetic clusters encompass a broader range of biological mechanisms and provide preliminary insights to explain ancestry-associated differences in T2D risk profiles.
Collapse
Affiliation(s)
- Kirk Smith
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aaron J Deutsch
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Carolyn McGrail
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Hyunkyung Kim
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL, USA
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Sarah Hsu
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Alicia Huerta-Chagoya
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ravi Mandla
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Philip H Schroeder
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kenneth E Westerman
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Lukasz Szczerbinski
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Timothy D Majarian
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
- Vertex Pharmaceuticals, Boston, MA, USA
| | - Varinderpal Kaur
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alice Williamson
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Noah Zaitlen
- Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Melina Claussnitzer
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jose C Florez
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Alisa K Manning
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Josep M Mercader
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kyle J Gaulton
- Department of Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Miriam S Udler
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
Xu F, Earp JE, Riebe D, Delmonico MJ, Lofgren IE, Greene GW. The relationship between fat distribution and diabetes in US adults by race/ethnicity. Front Public Health 2024; 12:1373544. [PMID: 38450122 PMCID: PMC10916687 DOI: 10.3389/fpubh.2024.1373544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 02/07/2024] [Indexed: 03/08/2024] Open
Abstract
Introduction This study examined the relationship between fat distribution and diabetes by sex-specific racial/ethnic groups. Methods A secondary data analysis of National Health and Nutrition Examination Survey 2011-2018 data (n = 11,972) was completed. Key variables examined were visceral adipose tissue area (VATA), subcutaneous fat area (SFA), diabetes prevalence, and race/ethnicity. The association of VATA and SFA and diabetes prevalence was examined separately and simultaneously using multiple logistic regression. Bonferroni corrections were applied to all multiple comparisons between racial/ethnic groups. All analyses were adjusted for demographics and muscle mass. Results VATA was positively associated with diabetes in both sexes (p < 0.001) and across all racial/ethnic groups (p < 0.05) except Black females. No statistically significant relationships were observed between SFA and diabetes while accounting for VATA with the exception of White females (p = 0.032). When comparing racial/ethnic groups, the relationship between VATA and diabetes was stronger in White and Hispanic females than in Black females (p < 0.005) while the relationship between SFA and diabetes did not differ between any racial/ethnic groups. Conclusion This study found that VATA is associated with diabetes for both sexes across almost all racial/ethnic groups independent of SFA whereas the only significant relationship between SFA and diabetes, independent of VATA, was observed in White females. The findings indicated that visceral fat was more strongly associated with diabetes than subcutaneous. Additionally, there are health disparities in sex-specific racial/ethnic groups thus further study is warranted.
Collapse
Affiliation(s)
- Furong Xu
- College of Education, University of Rhode Island, Kingston, RI, United States
| | - Jacob E. Earp
- Department of Kinesiology, University of Connecticut, Storrs, CT, United States
| | - Deborah Riebe
- Department of Kinesiology, University of Rhode Island, Kingston, RI, United States
| | - Matthew J. Delmonico
- Department of Kinesiology, University of Rhode Island, Kingston, RI, United States
| | - Ingrid E. Lofgren
- Department of Nutrition, University of Rhode Island, Kingston, RI, United States
| | - Geoffrey W. Greene
- Department of Nutrition, University of Rhode Island, Kingston, RI, United States
| |
Collapse
|
16
|
Zhang H, Che X, Jing H, Su Y, Yang W, Wang R, Zhang G, Meng J, Yuan W, Wang J, Gao W. A New Potent Inhibitor against α-Glucosidase Based on an In Vitro Enzymatic Synthesis Approach. Molecules 2024; 29:878. [PMID: 38398628 PMCID: PMC10893485 DOI: 10.3390/molecules29040878] [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: 01/19/2024] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Inhibiting the activity of intestinal α-glucosidase is considered an effective approach for treating type II diabetes mellitus (T2DM). In this study, we employed an in vitro enzymatic synthesis approach to synthesize four derivatives of natural products (NPs) for the discovery of therapeutic drugs for T2DM. Network pharmacology analysis revealed that the betulinic acid derivative P3 exerted its effects in the treatment of T2DM through multiple targets. Neuroactive ligand-receptor interaction and the calcium signaling pathway were identified as key signaling pathways involved in the therapeutic action of compound P3 in T2DM. The results of molecular docking, molecular dynamics (MD) simulations, and binding free energy calculations indicate that compound P3 exhibits a more stable binding interaction and lower binding energy (-41.237 kcal/mol) with α-glucosidase compared to acarbose. In addition, compound P3 demonstrates excellent characteristics in various pharmacokinetic prediction models. Therefore, P3 holds promise as a lead compound for the development of drugs for T2DM and warrants further exploration. Finally, we performed site-directed mutagenesis to achieve targeted synthesis of betulinic acid derivative. This work demonstrates a practical strategy of discovering novel anti-hyperglycemic drugs from derivatives of NPs synthesized through in vitro enzymatic synthesis technology, providing potential insights into compound P3 as a lead compound for anti-hyperglycemic drug development.
Collapse
Affiliation(s)
- Huanyu Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China; (H.Z.); (Y.S.); (W.Y.); (R.W.); (G.Z.); (J.M.); (W.Y.)
- Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, Tianjin 300072, China
| | - Xiance Che
- School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301600, China; (X.C.); (H.J.)
| | - Hongyan Jing
- School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301600, China; (X.C.); (H.J.)
| | - Yaowu Su
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China; (H.Z.); (Y.S.); (W.Y.); (R.W.); (G.Z.); (J.M.); (W.Y.)
- Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, Tianjin 300072, China
| | - Wenqi Yang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China; (H.Z.); (Y.S.); (W.Y.); (R.W.); (G.Z.); (J.M.); (W.Y.)
- Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, Tianjin 300072, China
| | - Rubing Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China; (H.Z.); (Y.S.); (W.Y.); (R.W.); (G.Z.); (J.M.); (W.Y.)
- Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, Tianjin 300072, China
| | - Guoqi Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China; (H.Z.); (Y.S.); (W.Y.); (R.W.); (G.Z.); (J.M.); (W.Y.)
- Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, Tianjin 300072, China
| | - Jie Meng
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China; (H.Z.); (Y.S.); (W.Y.); (R.W.); (G.Z.); (J.M.); (W.Y.)
- Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, Tianjin 300072, China
| | - Wei Yuan
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China; (H.Z.); (Y.S.); (W.Y.); (R.W.); (G.Z.); (J.M.); (W.Y.)
- Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, Tianjin 300072, China
| | - Juan Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China; (H.Z.); (Y.S.); (W.Y.); (R.W.); (G.Z.); (J.M.); (W.Y.)
- Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, Tianjin 300072, China
| | - Wenyuan Gao
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China; (H.Z.); (Y.S.); (W.Y.); (R.W.); (G.Z.); (J.M.); (W.Y.)
- Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, Tianjin 300072, China
| |
Collapse
|
17
|
Su Z, Efremov L, Mikolajczyk R. Differences in the levels of inflammatory markers between metabolically healthy obese and other obesity phenotypes in adults: A systematic review and meta-analysis. Nutr Metab Cardiovasc Dis 2024; 34:251-269. [PMID: 37968171 DOI: 10.1016/j.numecd.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/28/2023] [Accepted: 09/04/2023] [Indexed: 11/17/2023]
Abstract
AIMS The aim of this study was to systematically review and analyze differences in the levels of C-reactive protein (CRP), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α) comparing metabolically healthy but obese (MHO) with metabolically healthy non-obese (MHNO), metabolically unhealthy non-obese (MUNO), and metabolically unhealthy obese (MUO) subjects. DATA SYNTHESIS We searched PubMed, Embase, Web of Science, and Scopus for studies that matched the relevant search terms. Differences in inflammatory marker levels between MHO and the other three phenotypes were pooled as standardized mean differences (SMD) or differences of medians (DM) using a random-effects model. We included 91 studies reporting data on 435,007 individuals. The CRP levels were higher in MHO than in MHNO subjects (SMD = 0.63, 95% CI: 0.49, 0.76; DM = 0.83 mg/L, 95% CI: 0.56, 1.11). The CRP levels were higher in MHO than in MUNO subjects (SMD = 0.16, 95% CI: 0.05, 0.28; DM = 0.39 mg/L, 95% CI: 0.09, 0.69). The CRP levels were lower in MHO than in MUO individuals (SMD = -0.43, 95% CI: -0.54, -0.31; DM = -0.82 mg/L, 95% CI: -1.16, -0.48). The IL-6 levels in MHO were higher than in MHNO while lower than in MUO subjects. The TNF-α levels in MHO were higher than in MHNO individuals. CONCLUSIONS This review provides evidence that CRP levels in MHO are higher than in MHNO and MUNO subjects but lower than in MUO individuals. Additionally, IL-6 levels in MHO are higher than in MHNO but lower than in MUO subjects, and TNF-α levels in MHO are higher than in MHNO individuals. SYSTEMATIC REVIEW REGISTRATION PROSPERO number: CRD42021234948.
Collapse
Affiliation(s)
- Zhouli Su
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Center for Health Sciences, Martin-Luther-University Halle-Wittenberg, D-06112 Halle (Saale), Germany
| | - Ljupcho Efremov
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Center for Health Sciences, Martin-Luther-University Halle-Wittenberg, D-06112 Halle (Saale), Germany; Department of Radiation Oncology, Martin-Luther-University Halle-Wittenberg, D-06120 Halle (Saale), Germany
| | - Rafael Mikolajczyk
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Center for Health Sciences, Martin-Luther-University Halle-Wittenberg, D-06112 Halle (Saale), Germany.
| |
Collapse
|
18
|
Tian C, Ye Z, McCoy RG, Pan Y, Bi C, Gao S, Ma Y, Chen M, Yu J, Lu T, Hong LE, Kochunov P, Ma T, Chen S, Liu S. The causal effect of HbA1c on white matter brain aging by two-sample Mendelian randomization analysis. Front Neurosci 2024; 17:1335500. [PMID: 38274506 PMCID: PMC10808780 DOI: 10.3389/fnins.2023.1335500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/28/2023] [Indexed: 01/27/2024] Open
Abstract
Background Poor glycemic control with elevated levels of hemoglobin A1c (HbA1c) is associated with increased risk of cognitive impairment, with potentially varying effects between sexes. However, the causal impact of poor glycemic control on white matter brain aging in men and women is uncertain. Methods We used two nonoverlapping data sets from UK Biobank cohort: gene-outcome group (with neuroimaging data, (N = 15,193; males/females: 7,101/8,092)) and gene-exposure group (without neuroimaging data, (N = 279,011; males/females: 122,638/156,373)). HbA1c was considered the exposure and adjusted "brain age gap" (BAG) was calculated on fractional anisotropy (FA) obtained from brain imaging as the outcome, thereby representing the difference between predicted and chronological age. The causal effects of HbA1c on adjusted BAG were studied using the generalized inverse variance weighted (gen-IVW) and other sensitivity analysis methods, including Mendelian randomization (MR)-weighted median, MR-pleiotropy residual sum and outlier, MR-using mixture models, and leave-one-out analysis. Results We found that for every 6.75 mmol/mol increase in HbA1c, there was an increase of 0.49 (95% CI = 0.24, 0.74; p-value = 1.30 × 10-4) years in adjusted BAG. Subgroup analyses by sex and age revealed significant causal effects of HbA1c on adjusted BAG, specifically among men aged 60-73 (p-value = 2.37 × 10-8). Conclusion Poor glycemic control has a significant causal effect on brain aging, and is most pronounced among older men aged 60-73 years, which provides insights between glycemic control and the susceptibility to age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Cheng Tian
- Key Laboratory of Computing Power Network and Information Security, Ministry of Education, Shandong Computer Science Center (National Supercomputer Center in Jinan), Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
- Shandong Engineering Research Center of Big Data Applied Technology, Faculty of Computer Science and Technology, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
- Shandong Provincial Key Laboratory of Computer Networks, Shandong Fundamental Research Center for Computer Science, Jinan, China
| | - Zhenyao Ye
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, United States
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Rozalina G. McCoy
- Division of Endocrinology, Diabetes, & Nutrition, Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, United States
- University of Maryland Institute for Health Computing, Bethesda, MD, United States
| | - Yezhi Pan
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Chuan Bi
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Si Gao
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Yizhou Ma
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Mo Chen
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Jiaao Yu
- Department of Mathematics, University of Maryland, College Park, MD, United States
| | - Tong Lu
- Department of Mathematics, University of Maryland, College Park, MD, United States
| | - L. Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Tianzhou Ma
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD, United States
| | - Shuo Chen
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, United States
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Song Liu
- Key Laboratory of Computing Power Network and Information Security, Ministry of Education, Shandong Computer Science Center (National Supercomputer Center in Jinan), Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
- Shandong Engineering Research Center of Big Data Applied Technology, Faculty of Computer Science and Technology, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
- Shandong Provincial Key Laboratory of Computer Networks, Shandong Fundamental Research Center for Computer Science, Jinan, China
| |
Collapse
|
19
|
Yu T, Wong TJ, Chang JW, Lao XQ. Trajectories of body mass index before the diagnosis of type 2 diabetes in a cohort of Taiwanese adults. Obes Res Clin Pract 2024; 18:21-27. [PMID: 38331596 DOI: 10.1016/j.orcp.2024.02.002] [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: 08/01/2023] [Revised: 01/18/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND Although the prevalence of overweight/obesity is lower in Asian countries, the risk of type 2 diabetes (T2DM) is disproportionally higher. We identified and characterized the trajectory patterns of body mass index (BMI) before the onset of T2DM in a Taiwanese population. METHODS Using the Taiwan MJ cohort study, we sampled the health examination data of 22,934 participants, including 7618 cases of T2DM and 15,316 controls. We used latent class trajectory analysis to identify distinct groups of pre-disease BMI trajectory. To compare the trajectories of cardiometabolic risk factors among different groups, we used linear mixed-effects models. RESULTS These 22,934 participants included 13,074 men (57%) and 9860 women (43%) who were on average followed for 9.0 years. We identified three distinct pre-disease BMI trajectories in cases: "stable overweight" (n = 7016, 92.1%), "weight gain" (n = 333, 4.4%) and "obesity" (n = 269, 3.5%). The "stable overweight" group had a mean BMI of 24.6 kg/m2 at 15 years prior to diagnosis, had a 1.2 unit increase during follow-up, and had a mean BMI of 25.8 kg/m2 at the time of diagnosis. The "weight gain" group had the most increasing trends in blood pressure/low-density lipoprotein cholesterol over time. CONCLUSION The BMI trajectory patterns among individuals who later developed diabetes in Taiwan seemed comparable to that of Western populations, but our population developed T2DM at a much lower BMI. Given that most cases belong to the "stable overweight" group, we also support using a population-based strategy for diabetes prevention instead of focusing on the high risk individuals.
Collapse
Affiliation(s)
- Tsung Yu
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Tzu-Jung Wong
- Department of Healthcare Information and Management, School of Health and Medical Engineering, Ming Chuan University, Taoyuan, Taiwan
| | - Jen-Wen Chang
- Department of Pharmacy, Chi Mei Medical Center, Tainan, Taiwan
| | - Xiang-Qian Lao
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong; School of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
20
|
Abraham A, Yaghootkar H. Identifying obesity subtypes: A review of studies utilising clinical biomarkers and genetic data. Diabet Med 2023; 40:e15226. [PMID: 37704218 DOI: 10.1111/dme.15226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/15/2023]
Abstract
Obesity is a complex and multifactorial condition that poses significant health risks. Recent advancements in our understanding of obesity have highlighted the heterogeneity within this disorder. Identifying distinct subtypes of obesity is crucial for personalised treatment and intervention strategies. This review paper aims to examine studies that have utilised clinical biomarkers and genetic data to identify clusters or subtypes of obesity. The findings of these studies may provide valuable insights into the underlying mechanisms and potential targeted approaches for managing obesity-related health issues such as type 2 diabetes.
Collapse
Affiliation(s)
- Angela Abraham
- Joseph Banks Laboratories, College of Health and Science, University of Lincoln, Lincoln, Lincolnshire, UK
| | - Hanieh Yaghootkar
- Joseph Banks Laboratories, College of Health and Science, University of Lincoln, Lincoln, Lincolnshire, UK
| |
Collapse
|
21
|
Muniyappa R, Narayanappa SBK. Disentangling Dual Threats: Premature Coronary Artery Disease and Early-Onset Type 2 Diabetes Mellitus in South Asians. J Endocr Soc 2023; 8:bvad167. [PMID: 38178904 PMCID: PMC10765382 DOI: 10.1210/jendso/bvad167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Indexed: 01/06/2024] Open
Abstract
South Asian individuals (SAs) face heightened risks of premature coronary artery disease (CAD) and early-onset type 2 diabetes mellitus (T2DM), with grave health, societal, and economic implications due to the region's dense population. Both conditions, influenced by cardiometabolic risk factors such as insulin resistance, hypertension, and central adiposity, manifest earlier and with unique thresholds in SAs. Epidemiological, demographic, nutritional, environmental, sociocultural, and economic transitions in SA have exacerbated the twin epidemic. The coupling of premature CAD and T2DM arises from increased obesity due to limited adipose storage, early-life undernutrition, distinct fat thresholds, reduced muscle mass, and a predisposition for hepatic fat accumulation from certain dietary choices cumulatively precipitating a decline in insulin sensitivity. As T2DM ensues, the β-cell adaptive responses are suboptimal, precipitating a transition from compensatory hyperinsulinemia to β-cell decompensation, underscoring a reduced functional β-cell reserve in SAs. This review delves into the interplay of these mechanisms and highlights a prediabetes endotype tied to elevated vascular risk. Deciphering these mechanistic interconnections promises to refine stratification paradigms, surpassing extant risk-prediction strategies.
Collapse
Affiliation(s)
- Ranganath Muniyappa
- Clinical Endocrine Section, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Satish Babu K Narayanappa
- Department of Medicine, Sri Madhusudan Sai Institute of Medical Sciences and Research, Muddenahalli, Karnataka 562101, India
| |
Collapse
|
22
|
Niknejad A, Hosseini Y, Shamsnia HS, Kashani AS, Rostamian F, Momtaz S, Abdolghaffari AH. Sodium Glucose Transporter-2 Inhibitors (SGLT2Is)-TLRs Axis Modulates Diabetes. Cell Biochem Biophys 2023; 81:599-613. [PMID: 37658280 DOI: 10.1007/s12013-023-01164-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2023] [Indexed: 09/03/2023]
Abstract
Diabetes affects millions of people worldwide and is mainly associated with impaired insulin function. To date, various oral anti-diabetic drugs have been developed, of which, the sodium glucose transporter-2 inhibitors (SGLT2Is) are of the most recent classes that have been introduced. They differ from other classes in terms of their novel mechanism of actions and unique beneficial effects rather than just lowering glucose levels. SGLT2Is can protect body against cardiovascular events and kidney diseases even in non-diabetic individuals. SGLT2Is participate in immune cell activation, oxidative stress reduction, and inflammation mediation, thereby, moderating diabetic complications. In addition, toll like receptors (TLRs) are the intermediators of the immune system and inflammatory process, thus it's believed to play crucial roles in diabetic complications, particularly the ones that are related to inflammatory reactions. SGLT2Is are also effective against diabetic complications via their anti-inflammatory and oxidative properties. Given the anti-inflammatory properties of TLRs and SGLT2Is, this review investigates how SGLT2Is can affect the TLR pathway, and whether this could be favorable toward diabetes.
Collapse
Affiliation(s)
- Amirhossein Niknejad
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Yasamin Hosseini
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hedieh Sadat Shamsnia
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Ayeh Sabbagh Kashani
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Fatemeh Rostamian
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Saeideh Momtaz
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran.
- Department of Toxicology and Pharmacology, School of Pharmacy, and Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
23
|
Rivers JK, McGillivray W, Braun M, Bhogal M, Zheng S, Hickling M. Cryolipolysis of the Arms and Inner Thighs Shows Similar Treatment Outcomes in Chinese Individuals Compared to White Individuals Treated in a Prior Study: The XinCOOL Study. Aesthet Surg J Open Forum 2023; 5:ojad103. [PMID: 38108021 PMCID: PMC10725280 DOI: 10.1093/asjof/ojad103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
Abstract
Background Studies of predominantly White participants show that cryolipolysis reduces subcutaneous fat in the arms and inner thighs, but none have specifically tested for similar outcomes in participants of Chinese descent. Objectives This study assessed the safety and effectiveness of cryolipolysis treatment for noninvasive subcutaneous fat reduction of arms and inner thighs in participants of Chinese descent to assess equivalence to results seen in a prior study of White participants. Methods Replicating a similar study design, participants of first- or second-generation Chinese descent underwent cryolipolysis treatment of arms and/or inner thighs. Effectiveness was assessed using pretreatment and posttreatment photographic review by blinded, independent experts, investigator-assessed caliper measurements, and participant satisfaction 12 weeks posttreatment. Safety was assessed throughout. Results Among 50 enrolled participants, 48 completed the study. The majority of participants (97.9%) were female, with a mean age of 36.0 years and mean BMI of 24.16 kg/m2 (range 19.3-29.9 kg/m2). Overall, 76.4% and 70.0% of pretreatment photographs of arms and pairs of inner thighs, respectively, were correctly identified by at least 2 of 3 reviewers. The mean reduction from baseline in caliper-measured fat thickness was 6.5 mm for arms and 6.6 mm for inner thighs, and the majority of participants (>60%) were satisfied with the treatment. No adverse events were reported. Conclusions Cryolipolysis is a well-tolerated, effective means of noninvasive fat reduction of arms and inner thighs in participants of Chinese descent. The results from this study show similar effectiveness and safety in Chinese participants compared with White participants treated in a prior study. Level of Evidence 2
Collapse
Affiliation(s)
- Jason K Rivers
- Corresponding Author: Dr Jason K. Rivers, 200-2425 Hemlock St, Vancouver, BC, V6H 4E1, Canada. E-mail:
| | | | | | | | | | | |
Collapse
|
24
|
Liwin LK, Payne CF. How do sex-specific BMI trajectories shape diabetes risk? A longitudinal analysis of Indonesian adults. BMJ PUBLIC HEALTH 2023; 1:e000020. [PMID: 40017894 PMCID: PMC11812683 DOI: 10.1136/bmjph-2023-000020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 10/10/2023] [Indexed: 03/01/2025]
Abstract
Introduction Sex is a critical predictor of body mass index (BMI) trajectory over the life span, playing a role in disparities in the risk of diabetes. While there is some evidence of the effect of BMI on the risk of diabetes, little is known about sex differences in BMI trajectories over the life span and their later life association with diabetes, especially in low-income and middle-income settings. Methods Using panel data from the Indonesian Family Life Survey, this study examines the impact of an individual's BMI trajectory throughout adulthood on diabetes onset. Analysis for men and women were conducted separately. First, growth curve modelling estimates individuals' BMI trajectories over age. Second, the effect of BMI trajectories on diabetes is estimated using logistic regression adjusted for individual socioeconomic status. Finally, we perform relative dominance analysis to test the importance of BMI trajectories as a predictor of diabetes in later life against abdominal obesity measurements. Results BMI trajectories over adulthood strongly predict the probability of diabetes in both men and women. A rapidly increasing BMI significantly increases the probability of diabetes in men regardless of individuals' initial BMI. Among women, those who started out with an overweight/obese BMI and experienced a rapid loss in BMI over age had the highest risk of diabetes. Greater educational attainment is associated with an increase in the likelihood of diabetes in men, but higher education level is a protective factor from diabetes in women. Conclusion The findings suggest that maintaining BMI at normal cut-off over the life course can lower the likelihood of diabetes onset in later adulthood. This study emphasises that simple monitoring of BMI trajectory over adulthood could be a useful tool to identify the population at risk of diabetes in contexts with substantial underdiagnoses of diabetes.
Collapse
Affiliation(s)
- Lilipramawanty Kewok Liwin
- School of Demography, the Australian National University, Canberra, Australian Capital Territory, Australia
| | - Collin F Payne
- School of Demography, the Australian National University, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
25
|
Smith K, Deutsch AJ, McGrail C, Kim H, Hsu S, Mandla R, Schroeder PH, Westerman KE, Szczerbinski L, Majarian TD, Kaur V, Williamson A, Claussnitzer M, Florez JC, Manning AK, Mercader JM, Gaulton KJ, Udler MS. Multi-ancestry Polygenic Mechanisms of Type 2 Diabetes Elucidate Disease Processes and Clinical Heterogeneity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.28.23296294. [PMID: 37808749 PMCID: PMC10557820 DOI: 10.1101/2023.09.28.23296294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
We identified genetic subtypes of type 2 diabetes (T2D) by analyzing genetic data from diverse groups, including non-European populations. We implemented soft clustering with 650 T2D-associated genetic variants, capturing known and novel T2D subtypes with distinct cardiometabolic trait associations. The twelve genetic clusters were distinctively enriched for single-cell regulatory regions. Polygenic scores derived from the clusters differed in distribution between ancestry groups, including a significantly higher proportion of lipodystrophy-related polygenic risk in East Asian ancestry. T2D risk was equivalent at a BMI of 30 kg/m2 in the European subpopulation and 24.2 (22.9-25.5) kg/m2 in the East Asian subpopulation; after adjusting for cluster-specific genetic risk, the equivalent BMI threshold increased to 28.5 (27.1-30.0) kg/m2 in the East Asian group, explaining about 75% of the difference in BMI thresholds. Thus, these multi-ancestry T2D genetic subtypes encompass a broader range of biological mechanisms and help explain ancestry-associated differences in T2D risk profiles.
Collapse
Affiliation(s)
- Kirk Smith
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aaron J. Deutsch
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Carolyn McGrail
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Hyunkyung Kim
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL, USA
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Sarah Hsu
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Ravi Mandla
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Philip H. Schroeder
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kenneth E. Westerman
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Lukasz Szczerbinski
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Timothy D. Majarian
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Varinderpal Kaur
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alice Williamson
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Melina Claussnitzer
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jose C. Florez
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Alisa K. Manning
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Josep M. Mercader
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kyle J. Gaulton
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Miriam S. Udler
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
26
|
Cerdeña JP, Tsai JW, Warpinski C, Rosencrans RF, Gravlee CC. Racial, Gender, and Size Bias in a Medical Graphical Abstract Gallery: A Content Analysis. Health Equity 2023; 7:631-643. [PMID: 37786527 PMCID: PMC10541937 DOI: 10.1089/heq.2023.0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2023] [Indexed: 10/04/2023] Open
Abstract
Introduction Graphical abstracts may enhance dissemination of scientific and medical research but are also prone to reductionism and bias. We conducted a systematic content analysis of the Journal of Internal Medicine (JIM) Graphical Abstract Gallery to assess for evidence of bias. Materials and Methods We analyzed 140 graphical abstracts published by JIM between February 2019 and May 2020. Using a combination of inductive and deductive approaches, we developed a set of codes and code definitions for thematic, mixed-methods analysis. Results We found that JIM graphical abstracts disproportionately emphasized male (59.5%) and light-skinned (91.3%) bodies, stigmatized large body size, and overstated genetic and behavioral causes of disease, even relative to the articles they purportedly represented. Whereas 50.7% of the graphical surface area was coded as representing genetic factors, just 0.4% represented the social environment. Discussion Our analysis suggests evidence of bias and reductionism promoting normative white male bodies, linking large bodies with disease and death, conflating race with genetics, and overrepresenting genes while underrepresenting the environment as a driver of health and illness. These findings suggest that uncritical use of graphical abstracts may distort rather than enhance our understanding of disease; harm patients who are minoritized by race, gender, or body size; and direct attention away from dismantling the structural barriers to health equity. Conclusion We recommend that journals develop standards for mitigating bias in the publication of graphical abstracts that (1) ensure diverse skin tone and gender representation, (2) mitigate weight bias, (3) avoid racial or ethnic essentialism, and (4) attend to sociostructural contributors to disease.
Collapse
Affiliation(s)
- Jessica P. Cerdeña
- Department of Family Medicine, Middlesex Health, Middletown, Connecticut, USA
- Institute for Collaboration on Health, Implementation, and Policy (InCHIP), University of Connecticut, Storrs, Connecticut, USA
- Department of Anthropology, University of Connecticut, Storrs, Connecticut, USA
| | - Jennifer W. Tsai
- Department of Emergency Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Chloe Warpinski
- Department of Anthropology, University of Florida, Gainesville, Florida, USA
- MD-PhD Training Program, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Robert F. Rosencrans
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Clarence C. Gravlee
- Department of Anthropology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
27
|
Wu J, Feng Z, Duan J, Li Y, Deng P, Wang J, Yang Y, Meng C, Wang W, Wang A, Wang J. Global burden of type 2 diabetes attributable to non-high body mass index from 1990 to 2019. BMC Public Health 2023; 23:1338. [PMID: 37438808 DOI: 10.1186/s12889-023-15585-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/02/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND The prevalence of type 2 diabetes mellitus (T2DM) currently was increased in some countries of the world like China. However, the epidemiological trends of T2DM attributable to non-high body mass index (BMI) remain unclear. Thus, we aimed to describe the burden of T2DM attributable to non-high BMI. METHODS To estimate the burden of T2DM attributable to non-high BMI, data from the Global Burden of Disease Study 2019 were used to calculate the deaths and disability-adjusted life years (DALYs) by age, sex, year, and location. The estimated annual percentage change (EAPC) was applied in the analysis of temporal trends in T2DM from 1990 to 2019. RESULTS Globally in 2019, the number of death cases and DALYs of T2DM attributable to non-high BMI accounted for 57.9% and 48.1% of T2DM-death from all risks, respectively. Asia accounted for 59.5% and 63.6% of the global non-high-BMI-related death cases and DALYs of T2DM in 2019, respectively. From 1990 to 2019, regions in the low-income experienced a rise in DALYs attributable to non-high BMI. As compared to other age groups, older participants had higher deaths and DALYs of T2DM attributable to non-high BMI. The death and DALY rates of T2DM due to non-high BMI were higher in males and people in regions with low socio-demographic index (SDI) countries. CONCLUSIONS The burden of T2DM attributable to non-high BMI is higher in the elderly and in people in regions with low- and middle-SDI, resulting in a substantial burden on human health and the social cost of healthcare.
Collapse
Affiliation(s)
- Jingjing Wu
- Health Management Center, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha City, Hunan Province, 410013, China
- Clinical Research Center, Central South University, Changsha, Hunan, China
| | - Zeying Feng
- Clinical Trial Institution Office, Liuzhou Hospital of Guangzhou Women and Children's Medical Center, No. 50 Boyuan Avenue, Liuzhou City, Guangxi Province, 545001, China
| | - Jingwen Duan
- Health Management Center, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha City, Hunan Province, 410013, China
- Clinical Research Center, Central South University, Changsha, Hunan, China
| | - Yalan Li
- Health Management Center, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha City, Hunan Province, 410013, China
- Clinical Research Center, Central South University, Changsha, Hunan, China
| | - Peizhi Deng
- Health Management Center, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha City, Hunan Province, 410013, China
- Clinical Research Center, Central South University, Changsha, Hunan, China
| | - Jie Wang
- Health Management Center, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha City, Hunan Province, 410013, China
- Clinical Research Center, Central South University, Changsha, Hunan, China
| | - Yiping Yang
- Health Management Center, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha City, Hunan Province, 410013, China
- Clinical Research Center, Central South University, Changsha, Hunan, China
| | - Changjiang Meng
- Health Management Center, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha City, Hunan Province, 410013, China
- Clinical Research Center, Central South University, Changsha, Hunan, China
| | - Wei Wang
- Health Management Center, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha City, Hunan Province, 410013, China
- Clinical Research Center, Central South University, Changsha, Hunan, China
| | - Anli Wang
- Information Center of The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha City, Hunan Province, 410013, China.
| | - Jiangang Wang
- Health Management Center, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha City, Hunan Province, 410013, China.
| |
Collapse
|
28
|
Minato-Inokawa S, Hayashida Y, Honda M, Tsuboi-Kaji A, Takeuchi M, Kitaoka K, Kurata M, Wu B, Kazumi T, Fukuo K. Association between serum leptin concentrations and homeostasis model assessment-insulin resistance of 2.5 and higher in normal weight Japanese women. Sci Rep 2023; 13:8217. [PMID: 37217782 DOI: 10.1038/s41598-023-35490-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/18/2023] [Indexed: 05/24/2023] Open
Abstract
Normal weight insulin resistant phenotype was characterized in 251 Japanese female university students using homeostasis model assessment-insulin resistance. Birth weight, body composition at age 20, cardiometabolic traits and dietary intake were compared cross-sectionally between insulin sensitive (< 1.6, n = 194) and insulin resistant (2.5 and higher, n = 16) women. BMI averaged < 21 kg/m2 and waist < 72 cm and did not differ between two groups. The percentage of macrosomia and serum absolute and fat-mass corrected leptin concentrations were higher in insulin resistant women although there was no difference in birth weight, fat mass index, trunk/leg fat ratio and serum adiponectin. In addition, resting pulse rate, serum concentrations of free fatty acids, triglycerides and remnant-like particle cholesterol were higher in insulin resistant women although HDL cholesterol and blood pressure did not differ. In multivariate logistic regression analyses, serum leptin (odds ratio:1.68, 95% confidential interval:1.08-2.63, p = 0.02) was associated with normal weight insulin resistance independently of macrosomia, free fatty acids, triglycerides, remnant-like particle cholesterol and resting pulse rate. In conclusion, normal weight IR phenotype may be associated with increased plasma leptin concentrations and leptin to fat mass ratio in young Japanese women, suggesting higher leptin production by body fat unit.
Collapse
Affiliation(s)
- Satomi Minato-Inokawa
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-cho, Nishinomiya, Hyogo, 663-8558, Japan
- Laboratory of Community Health and Nutrition, Department of Bioscience, Graduate School of Agriculture, Ehime University, Matsuyama, Ehime, Japan
- Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| | - Yuuna Hayashida
- Department of Food Sciences and Nutrition, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Mari Honda
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Health, Sports, and Nutrition, Faculty of Health and Welfare, Kobe Women's University, Kobe, Hyogo, Japan
| | - Ayaka Tsuboi-Kaji
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-cho, Nishinomiya, Hyogo, 663-8558, Japan
- Department of Nutrition, Osaka City Juso Hospital, Osaka, Japan
| | - Mika Takeuchi
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-cho, Nishinomiya, Hyogo, 663-8558, Japan
| | - Kaori Kitaoka
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-cho, Nishinomiya, Hyogo, 663-8558, Japan
- Department of Advanced Epidemiology, Noncommunicable Disease (NCD) Epidemiology Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Miki Kurata
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-cho, Nishinomiya, Hyogo, 663-8558, Japan
- Department of Food Sciences and Nutrition, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Bin Wu
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Endocrinology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Tsutomu Kazumi
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-cho, Nishinomiya, Hyogo, 663-8558, Japan.
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan.
- Department of Medicine, Kohan Kakogawa Hospital, Kakogawa, Hyogo, Japan.
| | - Keisuke Fukuo
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-cho, Nishinomiya, Hyogo, 663-8558, Japan
- Department of Food Sciences and Nutrition, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| |
Collapse
|
29
|
Azizi N, Zangiabadian M, Seifi G, Davari A, Yekekhani E, Safavi-Naini SAA, Berger NA, Nasiri MJ, Sohrabi MR. Gastric Cancer Risk in Association with Underweight, Overweight, and Obesity: A Systematic Review and Meta-Analysis. Cancers (Basel) 2023; 15:2778. [PMID: 37345115 DOI: 10.3390/cancers15102778] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 04/28/2023] [Accepted: 04/28/2023] [Indexed: 06/23/2023] Open
Abstract
This study aimed to investigate the risk of gastric cancer (GC) in abnormal body mass index (BMI) groups. A systematic search was carried out on Embase, PubMed/Medline, and Scopus from January 2000 to January 2023. The pooled risk ratio (RR) with a 95% confidence interval (CI) was assessed using a random-effect model. Thirteen studies with total of 14,020,031 participants were included in this systematic review. The pooled RR of GC was 1.124 (95% CI, 0.968-1.304, I2: 89.08%) in underweight class, 1.155 (95% CI, 1.051-1.270, I2: 95.18%) in overweight class, and in 1.218 (95% CI, 1.070-1.386, I2: 97.65%) obesity class. There is no difference between cardia and non-cardia gastric cancer, while non-Asian race and female gender have higher risk of cancer, as Meta-regression of obesity and overweight classes showed. These findings suggest that there is a positive association between excess body weight and the risk of GC, with a higher impact in women than men and in non-Asian than Asian populations. Since abnormal weight is tied to various diseases, including GC, healthcare experts, and policymakers should continue interventions aiming to achieve a normal BMI range.
Collapse
Affiliation(s)
- Narges Azizi
- School of Medicine, Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Moein Zangiabadian
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman 7616913555, Iran
| | - Golnoosh Seifi
- School of Medicine, Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Afshan Davari
- School of Medicine, Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Elham Yekekhani
- School of Medicine, Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Seyed Amir Ahmad Safavi-Naini
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran
| | - Nathan A Berger
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Mohammad Javad Nasiri
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran
| | - Mohammad-Reza Sohrabi
- Community Medicine Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1983963113, Iran
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran 1983963113, Iran
| |
Collapse
|
30
|
Chew NWS, Ng CH, Tan DJH, Kong G, Lin C, Chin YH, Lim WH, Huang DQ, Quek J, Fu CE, Xiao J, Syn N, Foo R, Khoo CM, Wang JW, Dimitriadis GK, Young DY, Siddiqui MS, Lam CSP, Wang Y, Figtree GA, Chan MY, Cummings DE, Noureddin M, Wong VWS, Ma RCW, Mantzoros CS, Sanyal A, Muthiah MD. The global burden of metabolic disease: Data from 2000 to 2019. Cell Metab 2023; 35:414-428.e3. [PMID: 36889281 DOI: 10.1016/j.cmet.2023.02.003] [Citation(s) in RCA: 300] [Impact Index Per Article: 150.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 12/19/2022] [Accepted: 02/03/2023] [Indexed: 03/09/2023]
Abstract
Global estimates of prevalence, deaths, and disability-adjusted life years (DALYs) from the Global Burden of Diseases, Injuries, and Risk Factors Study 2019 were examined for metabolic diseases (type 2 diabetes mellitus [T2DM], hypertension, and non-alcoholic fatty liver disease [NAFLD]). For metabolic risk factors (hyperlipidemia and obesity), estimates were limited to mortality and DALYs. From 2000 to 2019, prevalence rates increased for all metabolic diseases, with the greatest increase in high socio-demographic index (SDI) countries. Mortality rates decreased over time in hyperlipidemia, hypertension, and NAFLD, but not in T2DM and obesity. The highest mortality was found in the World Health Organization Eastern Mediterranean region, and low to low-middle SDI countries. The global prevalence of metabolic diseases has risen over the past two decades regardless of SDI. Urgent attention is needed to address the unchanging mortality rates attributed to metabolic disease and the entrenched sex-regional-socioeconomic disparities in mortality.
Collapse
Affiliation(s)
- Nicholas W S Chew
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore, Singapore.
| | - Cheng Han Ng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Darren Jun Hao Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Gwyneth Kong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chaoxing Lin
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yip Han Chin
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wen Hui Lim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Daniel Q Huang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore; National University Centre for Organ Transplantation, National University Health System, Singapore, Singapore
| | - Jingxuan Quek
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Clarissa Elysia Fu
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jieling Xiao
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nicholas Syn
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Division of General Surgery, University Surgical Cluster, National University Hospital, Singapore, Singapore
| | - Roger Foo
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chin Meng Khoo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Division of Endocrinology, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Cardiovascular Research Institute, National University Heart Centre, National University Health System, Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Georgios K Dimitriadis
- Department of Endocrinology ASO/Easo COM, King's College Hospital NHS Foundation Trust, Denmark Hill, London, UK; Obesity, Type 2 Diabetes and Immunometabolism Research Group, Department of Diabetes, Faculty of Cardiovascular Medicine & Sciences, School of Life Course Sciences, King's College London, London, UK
| | - Dan Yock Young
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore; National University Centre for Organ Transplantation, National University Health System, Singapore, Singapore
| | - Mohammad Shadab Siddiqui
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Carolyn S P Lam
- National Heart Centre Singapore, Singapore, Singapore; Duke-NUS Medical School, Singapore, Singapore
| | - Yibin Wang
- Duke-NUS Medical School, Singapore, Singapore
| | - Gemma A Figtree
- Northern Clinical School, Kolling Institute of Medical Research, University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Mark Y Chan
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David E Cummings
- UW Medicine Diabetes Institute, VA Puget Sound Health Care System, University of Washington, Seattle, WA, USA
| | | | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong, China
| | - Ronald Ching Wan Ma
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong, China; Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China; Hong Kong Institute of Diabetes and Obesity, Chinese University of Hong Kong, Hong Kong, China
| | - Christos S Mantzoros
- Section of Endocrinology, Boston VA Healthcare System, Boston, MA, USA; Faculty of Medicine, Harvard University, Boston, MA, USA
| | - Arun Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Mark Dhinesh Muthiah
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore; National University Centre for Organ Transplantation, National University Health System, Singapore, Singapore
| |
Collapse
|
31
|
Li Z, Daniel S, Fujioka K, Umashanker D. Obesity among Asian American people in the United States: A review. Obesity (Silver Spring) 2023; 31:316-328. [PMID: 36695056 PMCID: PMC10108164 DOI: 10.1002/oby.23639] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/10/2022] [Accepted: 10/27/2022] [Indexed: 01/26/2023]
Abstract
Standard measures of obesity, i.e., body weight and BMI, suggest that Asian American people have a lower obesity prevalence than other racial groups in the United States. However, Asian American people face a unique challenge in their pattern of adiposity with central obesity, which raises the risk for multiple comorbidities, such as type 2 diabetes, metabolic syndrome, and cardiovascular disease, at a lower BMI compared with other populations. Several organizations recommend lower BMI cutoffs for obesity in Asian people (BMI ≥25.0 or ≥27.5 kg/m2 ) instead of the standard ≥30.0 kg/m2 threshold. The risks of obesity and related comorbidities in this population are further influenced by diet, physical activity, perceptions of health, and access to information and therapies. Asian-specific parameters for assessing obesity should become a standard part of clinical practice. Asian American people should equally be offered subgroup-specific tailored interventions owing to heterogeneity of this population. Access to medications and surgery should be improved, in part by updating US indications for therapies to reflect race-specific obesity thresholds and through inclusion of Asian American people of all subtypes with lower BMI values in clinical trials.
Collapse
Affiliation(s)
- Zhaoping Li
- Center for Human Nutrition, UCLA, Los Angeles, California, USA
| | | | - Ken Fujioka
- Scripps Clinic Del Mar, San Diego, California, USA
| | | |
Collapse
|
32
|
Salvatore T, Galiero R, Caturano A, Rinaldi L, Criscuolo L, Di Martino A, Albanese G, Vetrano E, Catalini C, Sardu C, Docimo G, Marfella R, Sasso FC. Current Knowledge on the Pathophysiology of Lean/Normal-Weight Type 2 Diabetes. Int J Mol Sci 2022; 24:ijms24010658. [PMID: 36614099 PMCID: PMC9820420 DOI: 10.3390/ijms24010658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
Since early times, being overweight and obesity have been associated with impaired glucose metabolism and type 2 diabetes (T2D). Similarly, a less frequent adult-onset diabetes in low body mass index (BMI) people has been known for many decades. This form is mainly found in developing countries, whereby the largest increase in diabetes incidence is expected in coming years. The number of non-obese patients with T2D is also on the rise among non-white ethnic minorities living in high-income Western countries due to growing migratory flows. A great deal of energy has been spent on understanding the mechanisms that bind obesity to T2D. Conversely, the pathophysiologic features and factors driving the risk of T2D development in non-obese people are still much debated. To reduce the global burden of diabetes, we need to understand why not all obese people develop T2D and not all those with T2D are obese. Moreover, through both an effective prevention and the implementation of an individualized clinical management in all people with diabetes, it is hoped that this will help to reduce this global burden. The purpose of this review is to take stock of current knowledge about the pathophysiology of diabetes not associated to obesity and to highlight which aspects are worthy of future studies.
Collapse
Affiliation(s)
- Teresa Salvatore
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, I–80138 Naples, Italy
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I–80138 Naples, Italy
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I–80138 Naples, Italy
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I–80138 Naples, Italy
| | - Livio Criscuolo
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I–80138 Naples, Italy
| | - Anna Di Martino
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I–80138 Naples, Italy
| | - Gaetana Albanese
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I–80138 Naples, Italy
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I–80138 Naples, Italy
| | - Christian Catalini
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I–80138 Naples, Italy
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I–80138 Naples, Italy
| | - Giovanni Docimo
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I–80138 Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I–80138 Naples, Italy
- Mediterrannea Cardiocentro, I–80122 Napoli, Italy
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I–80138 Naples, Italy
- Correspondence:
| |
Collapse
|
33
|
Chihota BV, Mandiriri A, Shamu T, Muula G, Nyamutowa H, Taderera C, Mwamba D, Chilengi R, Bolton‐Moore C, Bosomprah S, Egger M, Chimbetete C, Wandeler G. Metabolic syndrome among treatment-naïve people living with and without HIV in Zambia and Zimbabwe: a cross-sectional analysis. J Int AIDS Soc 2022; 25:e26047. [PMID: 36522287 PMCID: PMC9755006 DOI: 10.1002/jia2.26047] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Chronic viral replication has been linked to an increased risk of cardiovascular and metabolic diseases in people living with HIV (PLWH), but few studies have evaluated this association in Southern Africa. We explored the determinants of metabolic syndrome (MetS) among treatment-naïve adults living with and without HIV in Southern Africa. METHODS Treatment-naïve PLWH and people living without HIV (PLWOH) ≥30 years were consecutively enrolled from primary care clinics in Zambia and Zimbabwe. PLWOH were seronegative partners or persons presenting for HIV testing. We defined MetS as the presence of central obesity plus any two of the following: raised blood pressure, impaired fasting glucose, reduced high-density lipoprotein cholesterol and raised triglycerides, as defined by the International Diabetes Federation. We used logistic regression to determine factors associated with MetS. RESULTS Between August 2019 and March 2022, we screened 1285 adults and enrolled 420 (47%) PLWH and 481 (53%) PLWOH. The median age was similar between PLWH and PLWOH (40 vs. 38 years, p < 0.24). In PLWH, the median CD4+ count was 228 cells/mm3 (IQR 108-412) and the viral load was 24,114 copies/ml (IQR 277-214,271). Central obesity was present in 365/523 (70%) females and 57/378 males (15%). MetS was diagnosed in 172/901 (19%, 95% confidence interval [CI] 17-22%), and prevalence was higher among females than males (27% vs. 9%). In multivariable analyses, HIV status was not associated with MetS (adjusted odds ratio [aOR] 1.05, 95% CI 0.74-1.51). Risk factors for MetS included age older than 50 years (aOR 2.31, 95% CI 1.49-3.59), female sex (aOR 3.47, 95% CI 2.15-5.60), highest income (aOR 2.19, 95% CI 1.39-3.44) and less than World Health Organization recommended weekly physical activity (aOR 3.35, 95% CI 1.41-7.96). CONCLUSIONS We report a high prevalence of MetS and central obesity among females in urban Zambia and Zimbabwe. Lifestyle factors and older age appear to be the strongest predictors of MetS in our population, with no evident difference in MetS prevalence between treatment-naïve PLWH and PLWOH.
Collapse
Affiliation(s)
- Belinda V. Chihota
- Centre for Infectious Disease ResearchLusakaZambia,Graduate School of Health SciencesUniversity of BernBernSwitzerland,Institute of Social and Preventive MedicineUniversity of BernBernSwitzerland
| | | | - Tinei Shamu
- Graduate School of Health SciencesUniversity of BernBernSwitzerland,Institute of Social and Preventive MedicineUniversity of BernBernSwitzerland,Newlands ClinicHarareZimbabwe
| | - Guy Muula
- Centre for Infectious Disease ResearchLusakaZambia
| | | | | | | | | | - Carolyn Bolton‐Moore
- Centre for Infectious Disease ResearchLusakaZambia,Department of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Samuel Bosomprah
- Centre for Infectious Disease ResearchLusakaZambia,Department of BiostatisticsSchool of Public HealthUniversity of GhanaAccraGhana
| | - Matthias Egger
- Institute of Social and Preventive MedicineUniversity of BernBernSwitzerland,Centre for Infectious Disease Epidemiology and ResearchUniversity of Cape TownCape TownSouth Africa,Population Health SciencesBristol Medical School, University of BristolBristolUnited Kingdom
| | | | - Gilles Wandeler
- Institute of Social and Preventive MedicineUniversity of BernBernSwitzerland,Department of Infectious DiseasesBern University HospitalUniversity of BernBernSwitzerland
| | | |
Collapse
|
34
|
de Silva MHAD, Hewawasam RP, Kulatunge CR, Chamika RMA. The accuracy of fatty liver index for the screening of overweight and obese children for non-alcoholic fatty liver disease in resource limited settings. BMC Pediatr 2022; 22:511. [PMID: 36042456 PMCID: PMC9426280 DOI: 10.1186/s12887-022-03575-w] [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: 02/12/2021] [Accepted: 08/18/2022] [Indexed: 11/23/2022] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) has become the most common liver disease among children with a higher prevalence among obese. Fatty liver index (FLI), an algorithm derived to screen NAFLD using the waist circumference, body mass index, triglyceride and gamma glutamyl-transferase concentration was rarely investigated in Asian paediatric population. Since each component is routinely measured in clinical practice, it is feasible to measure FLI even in resource limited settings. Hence, we determined the accuracy of FLI for the screening of NAFLD in obese children in Southern Sri Lanka. Methods A descriptive cross sectional study was conducted on ninety five children (56 boys) aged 5–15 years with BMI ≥ 85th percentile for age and gender based on CDC 2000 growth charts recruited from the nutrition clinic at the Teaching Hospital, Karapitiya, Sri Lanka. NAFLD was diagnosed by ultrasonography. Factors associated with ultrasonographic fatty liver such as biochemical parameters and fatty liver index in either sex or the whole population were determined by a multivariate analysis. The ability of FLI to screen NAFLD was determined by the analysis of area under the receiver operator characteristic curve (AUROC) and the maximum Youden index analysis. Results Overweight and obese children with ultrasonographic fatty liver had a significantly higher FLI than those without fatty liver according to the multivariate analysis performed (Odds ratio 3.524; 95% CI 1.104–11.256, P = 0.033). AUROC of FLI for NAFLD was 0.692 (95% CI; 0.565–0.786) and the optimal cut off value for the screening of NAFLD was 30 (Maximum Youden index 0.2782, Sensitivity, 58.33%; Specificity, 69.49%). Conclusion FLI could accurately be used in resource limited community settings and in epidemiological studies to screen overweight and obese children for NAFLD.
Collapse
|
35
|
Paul SK, Shaw JE, Fenici P, Montvida O. Cardiorenal Complications in Young-Onset Type 2 Diabetes Compared Between White Americans and African Americans. Diabetes Care 2022; 45:1873-1881. [PMID: 35699938 DOI: 10.2337/dc21-2349] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 04/26/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To explore risks and associated mediation effects of developing chronic kidney disease (CKD) and heart failure (HF) in young- and usual-onset type 2 diabetes (T2D) between White Americans (WAs) and African Americans (AAs). RESEARCH DESIGN AND METHODS From U.S. medical records, 1,491,672 WAs and 31,133 AAs were identified and stratified by T2D age of onset (18-39, 40-49, 50-59, 60-70 years). Risks, mediation effects, and time to CKD and HF were evaluated, adjusting for time-varying confounders. RESULTS In the 18-39, 40-49, 50-59, 60-70 age-groups, the hazard ratios (of developing CKD and HF in AAs versus WAs were 1.21 (95% CI 1.17-1.26) and 2.21 (1.98-2.45), 1.25 (1.22-1.28) and 1.86 (1.75-1.97), 1.21 (1.19-1.24) and 1.54 (1.48-1.60), and 1.10 (1.08-1.12) and 1.11 (1.07-1.15), respectively. In AAs and WAs aged 18-39 years, time in years to CKD (8.7 [95% CI 8.2-9.1] and 9.7 [9.2-10.2]) and HF (10.3 [9.3-11.2] and 12.1 [10.6-13.5]) were, on average, 3.6 and 4.0 and 3.1 and 4.1 years longer compared with those diagnosed at age 60-70 years. Compared with females, AA males aged <60 years had an 11-49% higher CKD risk, while WA males aged <40 years had a 23% higher and those aged ≥50 years a 7-14% lower CKD risk, respectively. The mediation effects of CKD on the HF risk difference between ethnicities across age-groups (range 54-91%) were higher compared with those of HF on CKD risk difference between ethnicities across age-groups (13-39%). CONCLUSIONS Developing cardiorenal complications within an average of 10 years of young-onset T2DM and high mediation effects of CKD on HF call for revisiting guidelines on early diagnosis and proactive treatment strategies for effective management of cardiometabolic risk.
Collapse
Affiliation(s)
- Sanjoy K Paul
- Melbourne EpiCentre, University of Melbourne and Melbourne Health, Melbourne, Australia
| | | | - Peter Fenici
- Biomagnetism and Clinical Physiology International Center, Rome, Italy
| | - Olga Montvida
- Melbourne EpiCentre, University of Melbourne and Melbourne Health, Melbourne, Australia
| |
Collapse
|
36
|
Ke C, Narayan KMV, Chan JCN, Jha P, Shah BR. Pathophysiology, phenotypes and management of type 2 diabetes mellitus in Indian and Chinese populations. Nat Rev Endocrinol 2022; 18:413-432. [PMID: 35508700 PMCID: PMC9067000 DOI: 10.1038/s41574-022-00669-4] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/24/2022] [Indexed: 02/08/2023]
Abstract
Nearly half of all adults with type 2 diabetes mellitus (T2DM) live in India and China. These populations have an underlying predisposition to deficient insulin secretion, which has a key role in the pathogenesis of T2DM. Indian and Chinese people might be more susceptible to hepatic or skeletal muscle insulin resistance, respectively, than other populations, resulting in specific forms of insulin deficiency. Cluster-based phenotypic analyses demonstrate a higher frequency of severe insulin-deficient diabetes mellitus and younger ages at diagnosis, lower β-cell function, lower insulin resistance and lower BMI among Indian and Chinese people compared with European people. Individuals diagnosed earliest in life have the most aggressive course of disease and the highest risk of complications. These characteristics might contribute to distinctive responses to glucose-lowering medications. Incretin-based agents are particularly effective for lowering glucose levels in these populations; they enhance incretin-augmented insulin secretion and suppress glucagon secretion. Sodium-glucose cotransporter 2 inhibitors might also lower blood levels of glucose especially effectively among Asian people, while α-glucosidase inhibitors are better tolerated in east Asian populations versus other populations. Further research is needed to better characterize and address the pathophysiology and phenotypes of T2DM in Indian and Chinese populations, and to further develop individualized treatment strategies.
Collapse
Affiliation(s)
- Calvin Ke
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
- Department of Medicine, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada.
- Centre for Global Health Research, Unity Health Toronto, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada.
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
- Asia Diabetes Foundation, Shatin, Hong Kong SAR, China.
| | - K M Venkat Narayan
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Nutrition and Health Sciences Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Asia Diabetes Foundation, Shatin, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Prabhat Jha
- Centre for Global Health Research, Unity Health Toronto, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Baiju R Shah
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| |
Collapse
|
37
|
Real-world evaluation of sodium-glucose co-transporter-2 inhibitors and dipeptidyl peptidase-4 inhibitors for managing type 2 diabetes mellitus: a retrospective multi-ethnic cohort study. J Diabetes Metab Disord 2022; 21:521-555. [PMID: 35673518 PMCID: PMC9167339 DOI: 10.1007/s40200-022-01004-4] [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: 09/15/2021] [Accepted: 02/07/2022] [Indexed: 11/29/2022]
Abstract
Abstract Purpose Sodium-glucose co-transporter-2 (SGLT2) inhibitors and dipeptidyl peptidase-4 (DPP4) inhibitors are increasingly used as second-line therapies in patients with type 2 diabetes. The aim of this study was to assess the real-world effects of SGLT2 inhibitors in a multi-ethnic population in Singapore. Methods This retrospective cohort study examined patients diagnosed with and treated for diabetes from the Ministry of Health’s administrative database. Differences in outcomes between treatment groups were assessed using Poisson regression. Demographics, clinical characteristics, previous diagnoses and hospitalisations, and diabetes medication history were used for propensity score matching. Subgroup analyses by ethnicity were performed. Effect size was estimated using risk ratios (RRs) with 95% confidence intervals (CIs). Results Patients initiating SGLT2 inhibitors were more likely to achieve glycaemic control target than DPP4 inhibitor-treated patients (RR 1.09; 95% CI 1.04, 1.14). This was observed only in patients of Chinese ethnicity. A higher risk of diabetic ketoacidosis in SGLT2 inhibitor initiators was not observed. SGLT2 inhibitors were associated with reduced risk of hypoglycaemia (RR 0.69; 95% CI 0.59, 0.82) and urinary tract infection (RR 0.52; 95% CI 0.43, 0.63) but was not statistically significant for hypoglycaemia in Malay patients. Compared to DPP4 inhibitors, SGLT2 inhibitors were associated with 12% and 34% reduction in any-cause hospitalisation and all-cause mortality, respectively, potentially resulting in more than $50 million savings over 10 years. Conclusion SGLT2 inhibitors were associated with improvements in glycaemic control, reduced risk of complications, and was well tolerated. Ethnicity also plays a role and should be considered in future studies.
Collapse
|
38
|
Cheng A, Yeoh E, Moh A, Low S, Tan CH, Lam B, Sum CF, Subramaniam T, Lim SC. Roux-en-Y gastric bypass versus best medical treatment for type 2 diabetes mellitus in adults with body mass index between 27 and 32 kg/m 2: A 5-year randomized controlled trial. Diabetes Res Clin Pract 2022; 188:109900. [PMID: 35513159 DOI: 10.1016/j.diabres.2022.109900] [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: 09/02/2021] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 11/26/2022]
Abstract
AIMS To evaluate the effects of Roux-en-Y gastric bypass (RYGB) versus best medical treatment in Asians with type 2 diabetes mellitus (T2DM) and class I obesity. METHODS In this 5-year single-centre, open-label randomized controlled trial, participants were randomized to RYGB or medical treatment including newer classes of diabetes medications (ClinicalTrials.gov:NCT02041234). The primary endpoint was diabetes remission defined as HbA1c ≤ 6% (≤42 mmol/mol) and discontinuation of glucose-lowering medication at 12 months post-intervention and beyond. Glycaemia and weight changes were assessed. Continuous glucose monitoring was performed. RESULTS Of 28 subjects randomized, 26 were analyzed in the final cohort (14 medical, 12 RYGB; age:44 ± 10 years, 34.6% males, BMI:29.4 ± 1.6 kg/m2). At 12 months, 50% of RYGB subjects achieved diabetes remission; 83% stopped all glucose-lowering medications. By year 5, 42% were in remission. None attained diabetes remission in the medical group. Percentage declines in fasting plasma glucose, HbA1c and BMI were significantly greater in the RYGB arm (all P < 0.05). Early improvements in glycaemic variability and time in range were similar in both treatment arms. Hypoglycaemia and surgical complications were observed in some RYGB subjects. CONCLUSIONS Over 5 years, RYGB outperforms best medical treatment in glycemia and weight improvements for Asians with T2DM and class I obesity.
Collapse
Affiliation(s)
- Anton Cheng
- Department of General Surgery, Khoo Teck Puat Hospital, Singapore; Clinical Research Unit, Khoo Teck Puat Hospital, Singapore.
| | - Ester Yeoh
- Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore
| | - Angela Moh
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | - Serena Low
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore; Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore
| | - Chun Hai Tan
- Department of General Surgery, Khoo Teck Puat Hospital, Singapore; Integrated Centre for Obesity and Diabetes, Khoo Teck Puat Hospital, Singapore
| | - Benjamin Lam
- Integrated Centre for Obesity and Diabetes, Khoo Teck Puat Hospital, Singapore
| | - Chee Fang Sum
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore; Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore
| | - Tavintharan Subramaniam
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore; Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore
| | - Su Chi Lim
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore; Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore; Saw Swee Hock School of Public Health, National University Hospital, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
39
|
Lee K, Vakili S, Burden HJ, Adams S, Smith GC, Kulatea B, Wright-McNaughton M, Sword D, Watene-O'Sullivan C, Atiola RD, Paul RG, Plank LD, Kallingappa P, King F, Wilcox P, Merriman TR, Krebs JD, Hall RM, Murphy R, Merry TL, Shepherd PR. The minor allele of the CREBRF rs373863828 p.R457Q coding variant is associated with reduced levels of myostatin in males: Implications for body composition. Mol Metab 2022; 59:101464. [PMID: 35218947 PMCID: PMC8927835 DOI: 10.1016/j.molmet.2022.101464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 12/04/2022] Open
Abstract
OBJECTIVE The minor allele (A) of the rs373863828 variant (p.Arg457Gln) in CREBRF is restricted to indigenous peoples of the Pacific islands (including New Zealand Māori and peoples of Polynesia), with a frequency of up to 25% in these populations. This allele associates with a large increase in body mass index (BMI) but with significantly lower risk of type-2 diabetes (T2D). It remains unclear whether the increased BMI is driven by increased adiposity or by increased lean mass. METHODS We undertook body composition analysis using DXA in 189 young men of Māori and Pacific descent living in Aotearoa New Zealand. Further investigation was carried out in two orthologous Arg458Gln knockin mouse models on FVB/NJ and C57BL/6j backgrounds. RESULTS The rs373863828 A allele was associated with lower fat mass when adjusted for BMI (p < 0.05) and was associated with significantly lower circulating levels of the muscle inhibitory hormone myostatin (p < 0.05). Supporting the human data, significant reductions in adipose tissue mass were observed in the knockin mice. This was more significant in older mice in both backgrounds and appeared to be the result of reduced age-associated increases in fat mass. The older male knockin mice on C57BL/6j background also had increased grip strength (p < 0.01) and lower levels of myostatin (p < 0.05). CONCLUSION Overall, these results prove that the rs373863828 A-allele is associated with a reduction of myostatin levels which likely contribute to an age-dependent lowering of fat mass, at least in males.
Collapse
Affiliation(s)
- Kate Lee
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Sanaz Vakili
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Hannah J Burden
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Shannon Adams
- Discipline of Nutrition, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Greg C Smith
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Kensington, Australia
| | - Braydon Kulatea
- Discipline of Nutrition, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | | | - Danielle Sword
- Department of Medicine, University of Otago Wellington, Wellington, New Zealand
| | | | - Robert D Atiola
- Discipline of Nutrition, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Ryan G Paul
- Waikato Medical Research Centre, University of Waikato, Hamilton, New Zealand
| | - Lindsay D Plank
- Department of Surgery, School of Medicine, The University of Auckland, Auckland, New Zealand
| | - Prasanna Kallingappa
- Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Frances King
- Ngati Porou Hauora, Te Puia Springs, New Zealand
| | - Phillip Wilcox
- Department of Mathematics and Statistics, University of Otago, New Zealand
| | - Tony R Merriman
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Biochemistry, School of Biomedical Sciences, University of Otago, New Zealand; Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Alabama, United States
| | - Jeremy D Krebs
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Medicine, University of Otago Wellington, Wellington, New Zealand
| | - Rosemary M Hall
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Medicine, University of Otago Wellington, Wellington, New Zealand
| | - Rinki Murphy
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Medicine, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Troy L Merry
- Discipline of Nutrition, School of Medical Sciences, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Peter R Shepherd
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand; Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
40
|
Fowler LA, Fernández JR, Deemer SE, Gower BA. Genetic risk score prediction of leg fat and insulin sensitivity differs by race/ethnicity in early pubertal children. Pediatr Obes 2021; 16:e12828. [PMID: 34180151 PMCID: PMC10228538 DOI: 10.1111/ijpo.12828] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 04/08/2021] [Accepted: 06/09/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND In the United States, the underlying reasons for racial/ethnic disparities in type 2 diabetes risk remain unclear. However, differences in genetic risk for insulin resistance and peripheral adipose tissue distribution may be contributing factors. OBJECTIVE To investigate racial/ethnic differences in associations of genetic risk for insulin resistance with leg fat and insulin sensitivity in a cohort of American children. METHODS Participants were healthy European-American (n = 83), African-American (n = 79) and Hispanic-American (n = 74) children aged 7-12 years. Genetic risk scores were derived from published variants associated with insulin resistance phenotypes in European adults. Body composition was assessed using dual-energy X-ray absorptiometry. Insulin sensitivity was determined from the frequently sampled intravenous glucose tolerance test and minimal modelling. Statistical models were adjusted for age, sex, pubertal stage and body composition. RESULTS In the combined cohort, risk score was inversely associated with insulin sensitivity (p = 0.033) but not leg fat (p = 0.170). Within Hispanic Americans, risk score was inversely associated with insulin sensitivity (p = 0.027) and leg fat (p = 0.005), while associations were non-significant in European and African Americans (p > 0.200). CONCLUSIONS The higher type 2 diabetes risk observed among Hispanic Americans may have a genetic basis related to an inability to store lipid in peripheral adipose tissue.
Collapse
Affiliation(s)
- Lauren A Fowler
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - José R Fernández
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Sarah E Deemer
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Barbara A Gower
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
41
|
Loy SL, Lin J, Cheung YB, Sreedharan AV, Chin X, Godfrey KM, Tan KH, Shek LPC, Chong YS, Leow MKS, Khoo CM, Lee YS, Chan SY, Lek N, Chan JKY, Yap F. Influence of red blood cell indices on HbA1c performance in detecting dysglycaemia in a Singapore preconception cohort study. Sci Rep 2021; 11:20850. [PMID: 34675297 PMCID: PMC8531017 DOI: 10.1038/s41598-021-00445-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 10/12/2021] [Indexed: 12/16/2022] Open
Abstract
Abnormalities of red blood cell (RBC) indices may affect glycated haemoglobin (HbA1c) levels. We assessed the influence of haemoglobin (Hb) and mean corpuscular volume (MCV) on the performance of HbA1c in detecting dysglycaemia among reproductive aged women planning to conceive. Women aged 18-45 years (n = 985) were classified as normal (12 ≤ Hb ≤ 16 g/dL and 80 ≤ MCV ≤ 100 fL) and abnormal (Hb < 12 g/dL and/or MCV < 80 fL). The Area Under the Receiver Operating Characteristic (AUROC) curve was used to determine the performance of HbA1c in detecting dysglycaemic status (prediabetes and diabetes). There were 771 (78.3%) women with normal RBC indices. The AUROCs for the normal and abnormal groups were 0.75 (95% confidence interval 0.69, 0.81) and 0.80 (0.70, 0.90), respectively, and were not statistically different from one another [difference 0.04 (- 0.16, 0.08)]. Further stratification by ethnicity showed no difference between the two groups among Chinese and Indian women. However, Malay women with normal RBC indices displayed lower AUROC compared to those with abnormal RBC indices (0.71 (0.55, 0.87) vs. 0.98 (0.93, 1.00), p = 0.002). The results suggest that the performance of HbA1c in detecting dysglycaemia was not influenced by abnormal RBC indices based on low Hb and/or low MCV. However, there may be ethnic variations among them.
Collapse
Affiliation(s)
- See Ling Loy
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, 229899, Singapore.
- Duke-NUS Medical School, Singapore, 169857, Singapore.
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, 117609, Singapore.
| | - Jinjie Lin
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Yin Bun Cheung
- Program in Health Services and Systems Research and Center for Quantitative Medicine, Duke-NUS Medical School, Singapore, 169857, Singapore
- Tampere Center for Child, Adolescent and Maternal Health Research, Tampere University, 33014, Tampere, Finland
| | | | - Xinyi Chin
- Duke-NUS Medical School, Singapore, 169857, Singapore
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Keith M Godfrey
- Medical Research Council Lifecourse Epidemiology Unit, University of Southampton, Southampton, SO16 6YD, UK
- National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, SO16 6YD, UK
| | - Kok Hian Tan
- Duke-NUS Medical School, Singapore, 169857, Singapore
- Department of Maternal Fetal Medicine, KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Lynette Pei-Chi Shek
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, 117609, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore, 119074, Singapore
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, 117609, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore, 119228, Singapore
| | - Melvin Khee-Shing Leow
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, 117609, Singapore
- Cardiovascular and Metabolic Disorder Programme, Duke-NUS Medical School, Singapore, 169857, Singapore
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore, 308433, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore
| | - Chin Meng Khoo
- Cardiovascular and Metabolic Disorder Programme, Duke-NUS Medical School, Singapore, 169857, Singapore
- Department of Medicine, National University Hospital, Singapore, 119074, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yung Seng Lee
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, 117609, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore, 119074, Singapore
| | - Shiao-Yng Chan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, 117609, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Ngee Lek
- Duke-NUS Medical School, Singapore, 169857, Singapore
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, 229899, Singapore
- Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Fabian Yap
- Duke-NUS Medical School, Singapore, 169857, Singapore.
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore, 229899, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore.
| |
Collapse
|
42
|
Lee S, Lee DH, Lee JH, Shin SJ, Lee HS, Park EJ, Baik SH, Lee KY, Kang J. Association of Body Mass Index with Survival in Asian Patients with Colorectal Cancer. Cancer Res Treat 2021; 54:860-872. [PMID: 34665954 PMCID: PMC9296949 DOI: 10.4143/crt.2021.656] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/14/2021] [Indexed: 11/30/2022] Open
Abstract
Purpose The clinical significance of body mass index (BMI) on long-term outcomes has not been extensively investigated in Asian patients with colorectal cancer (CRC). This study aims to describe the association between BMI and survival, plus providing BMI cutoff value for predicting prognosis in CRC patients. Materials and Methods A total of 1,182 patients who had undergone surgery for stage I–III CRC from June 2004 to February 2014 were included. BMI was categorized into four groups based on the recommendation for Asian ethnicity. The optimal BMI cutoff value was determined to maximize overall survival (OS) difference. Results In multivariable analysis, underweight BMI was significantly associated with poor OS (hazard ratio [HR], 2.38; 95% confidence interval [CI], 1.55 to 3.71; p < 0.001) and obese BMI was associated with better OS (HR, 0.72; 95% CI, 0.53 to 0.97; p=0.036) compared with the normal BMI. Overweight and obese BMI were associated with better recurrence-free survival (HR, 0.64; 95% CI, 0.42 to 0.99; p=0.046 and HR, 0.58; 95% CI, 0.38 to 0.89; p=0.014, respectively) compared with the normal BMI group. BMI cutoff value was 20.44 kg/m2. Adding the BMI cutoff value to cancer staging could increase discriminatory performance in terms of integrated area under the curve and Harrell’s concordance index. Conclusion Compared to normal BMI, underweight BMI was associated with poor survival whereas obese BMI was associated with better survival. BMI cutoff value of 20.44 kg/m2 is a useful discriminator in Asian patients with CRC.
Collapse
Affiliation(s)
- Sangwon Lee
- Yonsei University College of Medicine, Seoul, Korea
| | - Dong Hee Lee
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jae-Hoon Lee
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Su-Jin Shin
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Jung Park
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Hyuk Baik
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kang Young Lee
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jeonghyun Kang
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
43
|
Tanabe H, Masuzaki H, Shimabukuro M. Novel strategies for glycaemic control and preventing diabetic complications applying the clustering-based classification of adult-onset diabetes mellitus: A perspective. Diabetes Res Clin Pract 2021; 180:109067. [PMID: 34563587 DOI: 10.1016/j.diabres.2021.109067] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 09/13/2021] [Accepted: 09/20/2021] [Indexed: 12/17/2022]
Abstract
Diabetes mellitus results from an interplay between insulin resistance and β-cell dysfunction. Since their relative contributions to its pathogenesis are difficult to quantify, therapeutic strategies for glycaemic control are determined primarily based on two limited metrics: plasma glucose and haemoglobin A1c. Recent attempts have been made to subclassify diabetes mellitus to better predict its associated pathology and plan appropriate therapeutic strategies. These classifications are based on data-driven cluster analysis using autoimmunity, age, obesity (metabolically unhealthy and healthy phenotypes), insulin secretory capacity and resistance, and ethnicity. This review addresses potential therapeutic strategies for the cluster-based classifications of adult-onset diabetes mellitus to achieve better glycaemic control and prevent or at least delay the concomitant complications.
Collapse
Affiliation(s)
- Hayato Tanabe
- Department of Diabetes, Endocrinology and Metabolism, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hiroaki Masuzaki
- Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), University of the Ryukyus, Okinawa 903-0215, Japan
| | - Michio Shimabukuro
- Department of Diabetes, Endocrinology and Metabolism, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan.
| |
Collapse
|
44
|
Mamtani M, Jaisinghani MT, Jaiswal SG, Pipal KV, Patel AA, Kulkarni H. Genetic association of anthropometric traits with type 2 diabetes in ethnically endogamous Sindhi families. PLoS One 2021; 16:e0257390. [PMID: 34506595 PMCID: PMC8432747 DOI: 10.1371/journal.pone.0257390] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 08/31/2021] [Indexed: 12/20/2022] Open
Abstract
Background Ethnically endogamous populations can shed light on the genetics of type 2 diabetes. Such studies are lacking in India. We conducted this study to determine the genetic and environmental contributions of anthropometric traits to type 2 diabetes risk in the Sindhi families in central India. Methods We conducted a family study in Indian Sindhi families with at least one case of type 2 diabetes. Variance components methods were used to quantify the genetic association of 18 anthropometric traits with eight type 2 diabetes related traits. Univariate and bivariate polygenic models were used to determine the heritability, genetic and environmental correlation of anthropometric traits with type 2 diabetes related traits. Results We included 1,152 individuals from 112 phenotyped families. The ascertainment-bias corrected prevalence of type 2 diabetes was 35%. Waist circumference, hip circumference and the biceps, triceps, subscapular and medial calf skinfold thicknesses were polygenically and significantly associated with type 2 diabetes. The range of heritability of the anthropometric traits and type 2 diabetes related traits was 0.27–0.73 and 0.00–0.39, respectively. Heritability of type 2 diabetes as a discrete trait was 0.35. Heritability curves demonstrated a substantial local influence of type 2 diabetes related traits. Bivariate trait analyses showed that biceps and abdominal skinfold thickness and all waist-containing indexes were strongly genetically correlated with type 2 diabetes. Conclusions In this first study of Sindhi families, we found evidence for genetic and environmental concordance of anthropometric traits with type 2 diabetes. Future studies need to probe into the genetics of type 2 diabetes in this population.
Collapse
Affiliation(s)
- Manju Mamtani
- Lata Medical Research Foundation, Nagpur, India
- M&H Research, LLC, San Antonio, Texas, United States of America
- * E-mail:
| | | | | | | | | | - Hemant Kulkarni
- Lata Medical Research Foundation, Nagpur, India
- M&H Research, LLC, San Antonio, Texas, United States of America
| |
Collapse
|
45
|
Martin S, Cule M, Basty N, Tyrrell J, Beaumont RN, Wood AR, Frayling TM, Sorokin E, Whitcher B, Liu Y, Bell JD, Thomas EL, Yaghootkar H. Genetic Evidence for Different Adiposity Phenotypes and Their Opposing Influences on Ectopic Fat and Risk of Cardiometabolic Disease. Diabetes 2021; 70:1843-1856. [PMID: 33980691 DOI: 10.2337/db21-0129] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/06/2021] [Indexed: 11/13/2022]
Abstract
To understand the causal role of adiposity and ectopic fat in type 2 diabetes and cardiometabolic diseases, we aimed to identify two clusters of adiposity genetic variants: one with "adverse" metabolic effects (UFA) and the other with, paradoxically, "favorable" metabolic effects (FA). We performed a multivariate genome-wide association study using body fat percentage and metabolic biomarkers from UK Biobank and identified 38 UFA and 36 FA variants. Adiposity-increasing alleles were associated with an adverse metabolic profile, higher risk of disease, higher CRP, and higher fat in subcutaneous and visceral adipose tissue, liver, and pancreas for UFA and a favorable metabolic profile, lower risk of disease, higher CRP and higher subcutaneous adipose tissue but lower liver fat for FA. We detected no sexual dimorphism. The Mendelian randomization studies provided evidence for a risk-increasing effect of UFA and protective effect of FA for type 2 diabetes, heart disease, hypertension, stroke, nonalcoholic fatty liver disease, and polycystic ovary syndrome. FA is distinct from UFA by its association with lower liver fat and protection from cardiometabolic diseases; it was not associated with visceral or pancreatic fat. Understanding the difference in FA and UFA may lead to new insights in preventing, predicting, and treating cardiometabolic diseases.
Collapse
Affiliation(s)
- Susan Martin
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Royal Devon & Exeter Hospital, Exeter, U.K
| | | | - Nicolas Basty
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, U.K
| | - Jessica Tyrrell
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Royal Devon & Exeter Hospital, Exeter, U.K
| | - Robin N Beaumont
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Royal Devon & Exeter Hospital, Exeter, U.K
| | - Andrew R Wood
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Royal Devon & Exeter Hospital, Exeter, U.K
| | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Royal Devon & Exeter Hospital, Exeter, U.K
| | | | - Brandon Whitcher
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, U.K
| | - Yi Liu
- Calico Life Sciences LLC, South San Francisco, CA
| | - Jimmy D Bell
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, U.K
| | - E Louise Thomas
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, U.K
| | - Hanieh Yaghootkar
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Royal Devon & Exeter Hospital, Exeter, U.K.
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, U.K
| |
Collapse
|
46
|
Andersson-Hall U, de Maré H, Askeli F, Börjesson M, Holmäng A. Physical activity during pregnancy and association with changes in fat mass and adipokines in women of normal-weight or with obesity. Sci Rep 2021; 11:12549. [PMID: 34131242 PMCID: PMC8206069 DOI: 10.1038/s41598-021-91980-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 06/03/2021] [Indexed: 11/20/2022] Open
Abstract
Adipose tissue and adipokine concentrations change markedly during pregnancy, but the effects of physical activity on these changes are rarely studied. We aimed to assess physical activity levels in pregnant women of normal-weight (NW) or with obesity (OB), and to determine the relation with changes in fat mass and adipokines. In each trimester, pregnant women (136 NW, 51 OB) were interviewed about their physical activity and had their body composition, leptin, soluble leptin receptor (sOB-R) and adiponectin determined. NW reported higher activity and more aerobic exercise than OB during early pregnancy. Both groups maintained training frequency but reduced overall activity as pregnancy progressed. NW women reporting aerobic and/or resistance exercise and OB women reporting aerobic exercise had greater sOB-R increases (independent of BMI or gestational weight gain). In NW, exercise also associated with lower fat mass and leptin increases. Higher activity levels associated with lower gestational weight gain in both groups. The relationship between physical activity and adiponectin differed between NW and OB. Maternal exercise may partly mediate its beneficial effects through regulation of leptin bioavailability, by enhancing pregnancy-induced increases in sOB-R. This could be of particular importance in OB with pre-gestational hyperleptinemia and leptin resistance.
Collapse
Affiliation(s)
- Ulrika Andersson-Hall
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden.
| | - Hanna de Maré
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Freja Askeli
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Mats Börjesson
- Department of Food and Nutrition, and Sport Science, Centre for Health and Performance, University of Gothenburg, Gothenburg, Sweden
- Department of Acute and Molecular Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of MGA, Sahlgrenska University Hospital, Region of Västra Götaland, Gothenburg, Sweden
| | - Agneta Holmäng
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| |
Collapse
|
47
|
Garcidueñas-Fimbres TE, Paz-Graniel I, Nishi SK, Salas-Salvadó J, Babio N. Eating Speed, Eating Frequency, and Their Relationships with Diet Quality, Adiposity, and Metabolic Syndrome, or Its Components. Nutrients 2021; 13:1687. [PMID: 34063439 PMCID: PMC8156274 DOI: 10.3390/nu13051687] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Excess body weight is a major global health concern, particularly due to its associated increased health risks. Several strategies have been proposed to prevent overweight and obesity onset. In the past decade, it has been suggested that eating speed/rate and eating frequency might be related to obesity. The main aim of this narrative review was to summarize existing evidence regarding the impact of eating speed/rate and eating frequency on adiposity, metabolic syndrome (MetS), or diet quality (DQ). For this purpose, a literature search of observational and interventional trials was conducted between June and September 2020 in PubMed and Web of Sciences databases, without any data filters and no limitations for publication date. Results suggest that children and adults with a faster eating speed/rate may be associated with a higher risk of developing adiposity, MetS or its components. Furthermore, a higher eating frequency could be associated with diet quality improvement, lower adiposity, and lower risk of developing MetS or its components. Further interventional trials are warranted to clarify the mechanism by which these eating behaviors might have a potential impact on health.
Collapse
Affiliation(s)
- Tany E. Garcidueñas-Fimbres
- Universitat Rovira i Virgili, Departament de Bioquimica i Biotecnologia, Unitat de Nutrició Humana, 43201 Reus, Spain; (T.E.G.-F.); (I.P.-G.); (S.K.N.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
- CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, 28029 Madrid, Spain
| | - Indira Paz-Graniel
- Universitat Rovira i Virgili, Departament de Bioquimica i Biotecnologia, Unitat de Nutrició Humana, 43201 Reus, Spain; (T.E.G.-F.); (I.P.-G.); (S.K.N.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
- CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, 28029 Madrid, Spain
| | - Stephanie K. Nishi
- Universitat Rovira i Virgili, Departament de Bioquimica i Biotecnologia, Unitat de Nutrició Humana, 43201 Reus, Spain; (T.E.G.-F.); (I.P.-G.); (S.K.N.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
- CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, 28029 Madrid, Spain
| | - Jordi Salas-Salvadó
- Universitat Rovira i Virgili, Departament de Bioquimica i Biotecnologia, Unitat de Nutrició Humana, 43201 Reus, Spain; (T.E.G.-F.); (I.P.-G.); (S.K.N.)
| | - Nancy Babio
- Universitat Rovira i Virgili, Departament de Bioquimica i Biotecnologia, Unitat de Nutrició Humana, 43201 Reus, Spain; (T.E.G.-F.); (I.P.-G.); (S.K.N.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
- CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, 28029 Madrid, Spain
| |
Collapse
|
48
|
Hattori H, Hanai Y, Oshima Y, Kataoka H, Eto N. Excessive Intake of High-Fructose Corn Syrup Drinks Induces Impaired Glucose Tolerance. Biomedicines 2021; 9:biomedicines9050541. [PMID: 34066196 PMCID: PMC8150719 DOI: 10.3390/biomedicines9050541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 12/19/2022] Open
Abstract
The number of patients with diabetes was approximately 463 million worldwide in 2019, with almost 57.6% of this population concentrated in Asia. Asians often develop type 2 diabetes (T2D), even if they are underweight and consume a smaller amount of food. Soft drinks contain large amounts of sweeteners, such as high-fructose corn syrup (HFCS). Excessive intake of HFCS drinks is considered to be one of the causes of T2D. In the present study, we investigated the effect of excessive consumption of HFCS-water on glucose tolerance and obesity under conditions of controlled caloric intake using a mouse model. Three-week-old male ICR mice were divided into two groups and given free access to 10% HFCS-water or deionized water. The caloric intake was adjusted to be the same in both groups using a standard rodent diet. The excess HFCS-water intake did not lead to obesity, but led to impaired glucose tolerance (IGT) due to insulin-secretion defect. It affected glucose and fructose metabolism; for example, it decreased the expression of glucokinases, ketohexokinase, and glucose transporter 2 in the pancreas. These results suggest that excessive consumption of HFCS drinks, such as soft drinks, without a proper diet, induces nonobese IGT due to insulin-secretion defect.
Collapse
Affiliation(s)
- Hidemi Hattori
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan; (Y.H.); (Y.O.); (N.E.)
- Correspondence: ; Tel.: +81-985-58-7255
| | - Yuma Hanai
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan; (Y.H.); (Y.O.); (N.E.)
| | - Yuto Oshima
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan; (Y.H.); (Y.O.); (N.E.)
| | - Hiroaki Kataoka
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan;
| | - Nozomu Eto
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan; (Y.H.); (Y.O.); (N.E.)
| |
Collapse
|
49
|
Osteopontin's relationship with malnutrition and oxidative stress in adolescents. A pilot study. PLoS One 2021; 16:e0249057. [PMID: 33765028 PMCID: PMC7993823 DOI: 10.1371/journal.pone.0249057] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 03/10/2021] [Indexed: 01/24/2023] Open
Abstract
Osteopontin (OPN) is a protein involved in inflammatory illnesses such as fibrosis and cancer; its overexpression in cardiovascular diseases promotes the biomineralization of blood vessels and other soft tissues. Moreover, there is an active component of oxidative stress related with those diseases. The present study relates serum OPN levels with nutritional condition and oxidative stress in a group of adolescents. Anthropometric measurements were performed, and fasting blood samples were analyzed to determine OPN concentrations, blood chemistry parameters (glucose, triglycerides, total cholesterol, urea, uric acid, and creatinine) and oxidative stress biomarkers (Paraoxonase-1, Glutathione S-Transferase, Catalase, NAD(P)H Quinone Oxidoreductase, free carbonyl groups and malondialdehyde). Adolescents were categorized according to body mass index (BMI) and metabolic syndrome (MetS) criteria. We found increased OPN serum concentrations in overweight and obese adolescents, as well as in adolescents with MetS. Rises in OPN correlated with arm circumference and biomarkers of lipid peroxidation; with regard to serum glucose there was a trend to positive correlation. Our results suggest that serum OPN is associated to nutritional status and could be considered as an early biomarker of low-grade inflammation and probably the early biomineralization of soft tissues in adolescence.
Collapse
|
50
|
Cousminer DL, Freathy RM. Genetics of early growth traits. Hum Mol Genet 2020; 29:R66-R72. [PMID: 32886111 PMCID: PMC7530515 DOI: 10.1093/hmg/ddaa149] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/04/2020] [Accepted: 07/09/2020] [Indexed: 12/11/2022] Open
Abstract
In recent years, genome-wide association studies have shed light on the genetics of early growth and its links with later-life health outcomes. Large-scale datasets and meta-analyses, combined with recently developed analytical methods, have enabled dissection of the maternal and fetal genetic contributions to variation in birth weight. Additionally, longitudinal approaches have shown differences between the genetic contributions to infant, childhood and adult adiposity. In contrast, studies of adult height loci have shown strong associations with early body length and childhood height. Early growth-associated loci provide useful tools for causal analyses: Mendelian randomization (MR) studies have provided evidence that early BMI and height are causally related to a number of adult health outcomes. We advise caution in the design and interpretation of MR studies of birth weight investigating effects of fetal growth on later-life cardiometabolic disease because birth weight is only a crude indicator of fetal growth, and the choice of genetic instrument (maternal or fetal) will greatly influence the interpretation of the results. Most genetic studies of early growth have to date centered on European-ancestry participants and outcomes measured at a single time-point, so key priorities for future studies of early growth genetics are aggregation of large samples of diverse ancestries and longitudinal studies of growth trajectories.
Collapse
Affiliation(s)
- Diana L Cousminer
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Rachel M Freathy
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| |
Collapse
|