1
|
Wu L, Coletta DK. Obesity and type 2 diabetes mellitus: insights from skeletal muscle extracellular matrix remodeling. Am J Physiol Cell Physiol 2025; 328:C1752-C1763. [PMID: 40244268 DOI: 10.1152/ajpcell.00154.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/23/2024] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are metabolic diseases at epidemic proportions. The economic burden for these diseases is at an all-time high, and as such, there is an urgent need for advancements in identifying targets for treating these complex disorders. The extracellular matrix (ECM), comprising collagen, fibronectin, laminin, elastin, and proteoglycan, surrounds skeletal muscles and plays a critical role in maintaining tissue homeostasis by providing structural support and facilitating cell-to-cell communication. Disruption of the ECM signaling results in changes to its micro/macroenvironment, thereby modifying tissue homeostasis. Skeletal muscle ECM remodeling has been shown to be associated with insulin resistance, an underlying feature of obesity and T2DM. This narrative review explores the critical components of skeletal muscle ECM and its accumulation and remodeling in metabolic diseases. In addition, we discuss potential treatments to mitigate the effects of ECM remodeling in skeletal muscle. We conclude that targeting ECM remodeling in skeletal muscle represents a promising yet underexplored therapeutic avenue in the management of metabolic disorders.
Collapse
Affiliation(s)
- Linda Wu
- Department of Physiology, University of Arizona, Tucson, Arizona, United States
| | - Dawn K Coletta
- Department of Physiology, University of Arizona, Tucson, Arizona, United States
- Division of Endocrinology, Department of Medicine, University of Arizona, Tucson, Arizona, United States
- Center for Disparities in Diabetes, Obesity, and Metabolism, University of Arizona, Tucson, Arizona, United States
| |
Collapse
|
2
|
Kapellou A, Salata E, Vrachnos DM, Papailia S, Vittas S. Gene-Diet Interactions in Diabetes Mellitus: Current Insights and the Potential of Personalized Nutrition. Genes (Basel) 2025; 16:578. [PMID: 40428400 PMCID: PMC12111186 DOI: 10.3390/genes16050578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Revised: 05/08/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) remaina significant global health challenge, with its increasing prevalence and associated complications contributing to high morbidity and economic burden. Genetic factors play a crucial role in T2DM susceptibility, yet individual responses to dietary interventions vary widely, emphasizing the importance of gene-diet (G × D) interactions. This review synthesizes the current literature on the genetic basis of T2DM and the role of G × D interactions in shaping individual responses to diet. We examine the genetics implication in T2DM risk and modulation by dietary factors, with a focus on the potential of Nutrigenetics in guiding personalized nutrition (PN) strategies. Moreover, the clinical implications of these interactions for the personalized prevention and management of T2DM are explored, highlighting the promise of tailoring dietary recommendations based on genetic profiles. Critical research gaps, including the need for diverse and longitudinal studies, the integration of multi-omic data, and the inclusion of digital health technologies in PN are discussed. Finally, future directions for the field are outlined, advocating for more inclusive, large-scale studies to optimize PN approaches for diverse populations and improve the efficacy of T2DM prevention and management. This review underscores the potential of an individualized, genetically informed dietary approach in modulating the global burden of T2DM.
Collapse
Affiliation(s)
| | | | | | | | - Spiros Vittas
- iDNA Laboratories, 7 Kavalieratou Taki, 14564 Athens, Greece; (A.K.); (E.S.); (D.M.V.); (S.P.)
| |
Collapse
|
3
|
Kong D, Kowalik O, Garratt E, Godfrey KM, Chan SY, Teo AKK. Genetics and epigenetics in gestational diabetes contributing to type 2 diabetes. Trends Endocrinol Metab 2025:S1043-2760(25)00074-8. [PMID: 40280863 DOI: 10.1016/j.tem.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/21/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025]
Abstract
Gestational diabetes mellitus (GDM) is a common pregnancy complication and a risk factor for the subsequent development of type 2 diabetes (T2D) in mothers and of several metabolic diseases in offspring. However, the molecular underpinnings of these risks are not well understood. Genome-wide association studies (GWAS) and epigenetic studies may provide complementary insights into the causal relationships between GDM exposure and maternal/offspring metabolic outcomes. In this review we discuss the potential pathophysiological roles of specific genetic variants and commonly reported differentially methylated loci in GDM development, and their link to the progression to T2D in both the mother and the offspring in later life, pointing to the potential for tailored interventional strategies based on these genetic and epigenetic mechanisms.
Collapse
Affiliation(s)
- Dewei Kong
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore; Dean's Office, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Oliwia Kowalik
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore; NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK; School of Human Development and Health, University of Southampton, Southampton, UK
| | - Emma Garratt
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK; School of Human Development and Health, University of Southampton, Southampton, UK
| | - Keith M Godfrey
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK; School of Human Development and Health, University of Southampton, Southampton, UK; Medical Research Council Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Shiao-Yng Chan
- Institute for Human Development and Potential (IHDP), A*STAR, Singapore 117609, Singapore; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Adrian Kee Keong Teo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; Precision Medicine Translational Research Programme (TRP), National University of Singapore, Singapore 119228, Singapore.
| |
Collapse
|
4
|
Wei H, Kappler C, Green E, Jiang H, Yeung T, Wang H. GRP94 is indispensable for definitive endoderm specification of human induced pluripotent stem cells. Mol Ther 2025:S1525-0016(25)00299-0. [PMID: 40254879 DOI: 10.1016/j.ymthe.2025.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/24/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025] Open
Abstract
Human induced pluripotent stem cell (hiPSC)-derived insulin-producing β cell therapy shows promise in treating type 1 diabetes and potentially type 2 diabetes. Understanding the genetic factors controlling hiPSC differentiation could optimize this therapy. In this study, we investigated the role of glucose-regulated protein 94 (GRP94) in human β cell development by generating HSP90B1/GRP94 knockout (KO) hiPSCs, re-expressing GRP94 in the mutants and inducing their β cell differentiation. Our results revealed that GRP94 depletion hindered β cell generation by promoting cell death induced by endoplasmic reticulum (ER) stress and other stressors during definitive endoderm (DE) differentiation. Moreover, GRP94 deletion resulted in decreased activation of WNT/β-catenin signaling, which is critical for DE specification. Re-expression of GRP94 in GRP94 KO iPSCs partially reversed DE differentiation deficiency and alleviated cell death. These findings highlight the previously unrecognized indispensable role of GRP94 in human DE formation and consequent β cell development from hiPSCs. GRP94 mitigates ER stress-induced cell death and regulates the WNT/β-catenin signaling pathway, which is both crucial for successful β cell differentiation. These results provide new insights into the molecular mechanisms underlying β cell differentiation from hiPSCs and suggest that targeting GRP94 pathways could enhance the efficiency of hiPSC-derived insulin-producing cell therapies for diabetes treatment.
Collapse
Affiliation(s)
- Hua Wei
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Christiana Kappler
- Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Erica Green
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Hanna Jiang
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Tiffany Yeung
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA; Ralph H Johnson VA Medical Center, Charleston, SC, USA.
| |
Collapse
|
5
|
Zhong X, Wei Q, Tiwari A, Wang Q, Tan Y, Chen R, Yan Y, Cox NJ, Li B. A Genetics-guided Integrative Framework for Drug Repurposing: Identifying Anti-hypertensive Drug Telmisartan for Type 2 Diabetes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.22.25324223. [PMID: 40166562 PMCID: PMC11957187 DOI: 10.1101/2025.03.22.25324223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Drug development is a long and costly process, and repurposing existing drugs for use toward a different disease or condition may serve as a cost-effective alternative. As drug targets with genetic support have a doubled success rate, genetics-informed drug repurposing holds promise in translating genetic findings into therapeutics. In this study, we developed a Genetics Informed Network-based Drug Repurposing via in silico Perturbation (GIN-DRIP) framework and applied the framework to repurpose drugs for type-2 diabetes (T2D). In GIN-DRIP for T2D, it integrates multi-level omics data to translate T2D GWAS signals into a genetics-informed network that simultaneously encodes gene importance scores and a directional effect (up/down) of risk genes for T2D; it then bases on the GIN to perform signature matching with drug perturbation experiments to identify drugs that can counteract the effect of T2D risk alleles. With this approach, we identified 3 high-confidence FDA-approved candidate drugs for T2D, and validated telmisartan, an anti-hypertensive drug, in our EHR data with over 3 million patients. We found that telmisartan users were associated with a reduced incidence of T2D compared to users of other anti-hypertensive drugs and non-users, supporting the therapeutic potential of telmisartan for T2D. Our framework can be applied to other diseases for translating GWAS findings to aid drug repurposing for complex diseases.
Collapse
Affiliation(s)
- Xue Zhong
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN
| | - Qiang Wei
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN
| | - Anshul Tiwari
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN
| | - Quan Wang
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN
| | - Yuting Tan
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN
| | - Rui Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN
| | - Yan Yan
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Nancy J Cox
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN
| | - Bingshan Li
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN
| |
Collapse
|
6
|
Rout M, Ramu D, Mariana M, Koshy T, Venkatesan V, Lopez-Alvarenga JC, Arya R, Ravichandran U, Sharma SK, Lodha S, Ponnala AR, Sharma KK, Shaik MV, Resendez RG, Venugopal P, R P, S N, Ezeilo JA, Almeida M, Paralta J, Mummidi S, Natesan C, Mehra NK, Singh JR, Wander GS, Ralhan S, Blackett PR, Blangero J, Medicherla KM, Thanikachalam S, Panchatcharam TS, K DK, Gupta R, Paul SFD, Ghosh AK, Aston CE, Duggirala R, Sanghera DK. Excess of rare noncoding variants in several type 2 diabetes candidate genes among Asian Indian families. COMMUNICATIONS MEDICINE 2025; 5:47. [PMID: 39987249 PMCID: PMC11846969 DOI: 10.1038/s43856-025-00750-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/23/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) etiology is highly complex due to its multiple roots of origin. Polygenic risk scores (PRS) based on genome-wide association studies (GWAS) can partially explain T2D risk. Asian Indian people have up to six times higher risk of developing T2D than European people, and underlying causes of this disparity are unknown. METHODS We have performed targeted sequencing of ten T2D GWAS/candidate regions using endogamous Punjabi Sikh families and replication studies using unrelated Sikh people and families from three other Indian endogamous ethnic groups (EEGs). RESULTS We detect rare and ultra-rare variants (RVs) in KCNJ11-ABCC8 and HNF4A (MODY genes) cosegregated with late-onset T2D. We also identify RV enrichment in two new genes, SLC38A11 and ANPEP, associated with T2D. Gene-burden analysis reveals the highest RV burden contributed by HNF4A (p = 0.0003), followed by KCNJ11/ABCC8 (p = 0.0061) and SLC38A11 (p = 0.03). Some RVs detected in Sikh people are also found in Agarwals from Jaipur, both from Northern India, but were monomorphic in other two EEGs from South Indian people. Despite carrying a high burden of T2D and RVs, most families have a significantly lower burden of PRS. Functional studies show that an intronic regulatory variant (RV) in ABCC8 affects the binding of Pax4 and NF-kB transcription factors, influencing downstream gene regulation. CONCLUSIONS The high burden of T2D in these families may stem from the enrichment of noncoding RVs in a small number of major known genes (including MODY genes) with oligogenic inheritance alongside RVs from genes associated with polygenic susceptibility. These findings highlight the need to conduct deeper evaluations of families from non-European ancestries to identify potential novel therapeutics and implement preventative strategies.
Collapse
Affiliation(s)
- Madhusmita Rout
- Department of Pediatrics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Deepika Ramu
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Mendez Mariana
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Teena Koshy
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Vettriselvi Venkatesan
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Juan C Lopez-Alvarenga
- Department of Population Health & Biostatistics, University of Texas Rio Grande Valley (UTRGV), Harlingen, TX, USA
| | - Rector Arya
- Department of Health and Behavioral Sciences, Texas A&M University-San Antonio, San Antonio, TX, US
| | - Umarani Ravichandran
- Department of Medicine, Rajah Muthiah Medical College Hospital, Annamalai University, Chidambaram, India
| | | | - Sailesh Lodha
- Departments of Preventive Cardiology, Internal Medicine and Endocrinology, Eternal Heart Care Centre and Research Institute, Mount Sinai New York Affiliate, Jaipur, India
| | - Amaresh Reddy Ponnala
- Department of Endocrinology, Krishna Institute of Medical Sciences (KIMS) Hospital, Nellore, India
| | - Krishna Kumar Sharma
- Department of Pharmacology, Lal Bahadur Shastri College of Pharmacy, Rajasthan University of Health Sciences, Jaipur, India
| | - Mahaboob Vali Shaik
- Department of Endocrinology, Narayana Medical College and Hospital, Nellore, India
| | - Roy G Resendez
- Department of Health and Behavioral Sciences, Texas A&M University-San Antonio, San Antonio, TX, US
| | - Priyanka Venugopal
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Parthasarathy R
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Noelta S
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Juliet A Ezeilo
- Department of Health and Behavioral Sciences, Texas A&M University-San Antonio, San Antonio, TX, US
| | - Marcio Almeida
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley (UTRGV), Brownsville, TX, USA
| | - Juan Paralta
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley (UTRGV), Brownsville, TX, USA
| | - Srinivas Mummidi
- Department of Health and Behavioral Sciences, Texas A&M University-San Antonio, San Antonio, TX, US
| | - Chidambaram Natesan
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Narinder K Mehra
- All India Institute of Medical Sciences and Research, New Delhi, India
| | | | | | - Sarju Ralhan
- Hero Dayanand Medical College and Heart Institute, Ludhiana, India
| | - Piers R Blackett
- Department of Pediatrics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley (UTRGV), Brownsville, TX, USA
| | | | - Sadagopan Thanikachalam
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Thyagarajan Sadras Panchatcharam
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Dileep Kumar K
- Department of Endocrinology, Narayana Medical College and Hospital, Nellore, India
| | - Rajeev Gupta
- Departments of Preventive Cardiology, Internal Medicine and Endocrinology, Eternal Heart Care Centre and Research Institute, Mount Sinai New York Affiliate, Jaipur, India
| | - Solomon Franklin D Paul
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Asish K Ghosh
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Christopher E Aston
- Department of Pediatrics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Ravindranath Duggirala
- Department of Health and Behavioral Sciences, Texas A&M University-San Antonio, San Antonio, TX, US
| | - Dharambir K Sanghera
- Department of Pediatrics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
7
|
Lim EB, Cho YS. Identification of genetic loci enriched in obese or lean T2D cases in the Korean population. Genes Genomics 2025; 47:235-243. [PMID: 39693004 DOI: 10.1007/s13258-024-01602-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Obesity causes many complex diseases including type 2 diabetes (T2D). Obesity increases the risk of T2D in Europeans, but there are many non-obese (lean) T2D patients in East Asia. OBJECTIVE To discover genetic factors enriched in obese or lean T2D patients, we conducted a genome-wide association (GWA) analysis for T2D stratified by BMI in the Korean population. METHODS In the discovery stage, 654 and 247 individuals classified as obese (BMI > 25) and lean (BMI < 23) T2D patients, respectively, were compared with 3,842 control subjects for GWA analysis. Several BMI-stratified T2D variants detected in the discovery stage were further tested in the replication stage, which included 402 obese and 220 lean T2D cases, and 3,615 controls. RESULTS Meta-analysis combining the discovery and replication stages detected two variants with genome-wide significance: rs2356138 [P = 2.8 × 10-8, OR = 2.06 (1.59-2.65)] in obese T2D subjects and rs9295478 [P = 2.5 × 10-9, OR = 1.61 (1.38-1.88)] in lean ones. The SNP rs9295478 is located in CDKAL1, a well-known T2D gene previously identified in several GWA studies. Meanwhile, the SNP rs2356138 is a previously unknown variant located in PKP4. CONCLUSION We discovered genetic loci enriched in obese or lean T2D patients in the Korean population. Our findings should facilitate more effective control of T2D in Koreans.
Collapse
Affiliation(s)
- Eun Bi Lim
- Department of Biomedical Science, Hallym University, Chuncheon, Gangwon State, 24252, Republic of Korea
| | - Yoon Shin Cho
- Department of Biomedical Science, Hallym University, Chuncheon, Gangwon State, 24252, Republic of Korea.
- Department of Neuroscience, Hallym University College of Medicine, Chuncheon, Gangwon State, 24252, Republic of Korea.
- GenoMax Co., Ltd, Humanities Building 2, 4314-4, Hallymdaehakgil 1, Chuncheon, Gangwon State, Republic of Korea.
| |
Collapse
|
8
|
Parikh RM, Saboo B, Misra A, Basit A, Aravind SR, Bhowmik B, Schwarz P, Dhatariya K, Khunti K, Joshi S, Gupta S, Gupta A, Chawla M, Phatak S, Kalra S, Khan A, Mohan V. Ahmedabad declaration: A framework to combat growing epidemic of young-onset type 2 diabetes in Asia. Diabetes Metab Syndr 2025; 19:103205. [PMID: 40054119 PMCID: PMC11972441 DOI: 10.1016/j.dsx.2025.103205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 03/18/2025]
Abstract
AIM Rising prevalence of Type 2 Diabetes (T2D) among young Asians has emerged as a public health crisis that threatens the long-term health, economic stability, and productivity of nations across Asia (1). Early-onset T2D poses unique challenges, including higher rates of undiagnosed cases, more aggressive disease progression, an increased risk of chronic complications and higher mortality (2). Hyperglycemia during the reproductive age especially among the female population can potentially have transgenerational impact through epigenetic changes. METHODS A comprehensive search was conducted on PubMed with a combination of relevant keywords. A preliminary draft prepared after review of literature was electronically circulated among a panel of 64 experts from various parts of the region and representatives of the participating organizations - Diabetes India (www.diabetesindia.org.in) and the Diabetes Asia Study Group (DASG, www.da-sg.org). RESULTS This Ahmedabad Declaration outlines the scale of the problem, its root causes, and a comprehensive action plan for Asian populations. The objectives of this declaration include raising awareness, addressing systemic barriers, and advocating for evidence-based policies and interventions, limited to people with T2D. Through collaborative efforts, we aim to mitigate the growing burden of diabetes in young Asians and secure a healthier future.
Collapse
Affiliation(s)
- Rakesh M Parikh
- Diabecity Clinic, Jaipur, India; C K S Hospital, Jaipur, India.
| | - Banshi Saboo
- Diacare Diabetes Care & Hormone Clinic, Ahmedabad, India
| | - Anoop Misra
- Fortis C-DOC Center of Excellence for Diabetes, Metabolic Diseases and Endocrinology, New Delhi, India; National Diabetes Obesity and Cholesterol Foundation, New Delhi, India; Diabetes Foundation (India), New Delhi, India
| | - Abdul Basit
- Indus Diabetes and Endocrinology Center, Indus Hospital and Health Network, Pakistan; Diabetic Association of Pakistan, Pakistan; Health Promotion Foundation, Pakistan; Diabetes in Asia Study Group, Doha, Qatar; Health Research Advisory Board, Pakistan
| | - S R Aravind
- Diacon Hospital, Bangalore, India; Research Trust of Diabetes India, Ahmedabad, India
| | - Bishwajit Bhowmik
- Centre for Global Health Research, Dhaka, Bangladesh; Diabetic Association of Bangladesh, Dhaka, Bangladesh
| | - Peter Schwarz
- International Diabetes Federation (IDF), Avenue Herrmann-Debroux 54., B-1160, Brussels, Belgium; Department for Prevention and Care of Diabetes, Faculty of Medicine, Carl Gustav Carus at the Technische Universität/TU Dresden, Dresden, Germany; Paul Langerhans Institute Dresden of Helmholtz Zentrum München at University Hospital and Faculty of Medicine, TU Dresden, 01307, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, 85764, Germany
| | - Ketan Dhatariya
- Elsie Bertram Diabetes Centre, Norfolk & Norwich University Hospitals NHS Foundation Trust, Norwich, NR4 7UY, UK; Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, UK
| | - Kamlesh Khunti
- Diabetes Research Centre, University of Leicester, Leicester, LE5 4PW, UK; Leicester Real World Evidence Unit, Diabetes Research Centre, University of Leicester, Leicester, LE5 4PW, UK; NIHR ARC East Midlands, University of Leicester, UK
| | - Shashank Joshi
- Department of Diabetology & Endocrinology, Lilavati Hospital and Research Centre, Mumbai, Maharashtra, India
| | - Sunil Gupta
- Sunil's Diabetes Care and Research Centre, Nagpur, India
| | - Amit Gupta
- Centre for Diabetes Care, Greater Noida, Uttar Pradesh, India
| | - Manoj Chawla
- Lina Diabetes Care and Mumbai Diabetes Research Centre, Mumbai, India
| | - Sanjeev Phatak
- Department of Diabetes and Metabolism, MARENGO CIMS Hospital, Ahmedabad, India; VIjayratna Diabetes Centre, Ahmedabad, India; J S Thakershy Hospital, Ahmedabad, India
| | - Sanjay Kalra
- Bharti Hospital, Karnal, Haryana, India; University Centre for Research and Development, Chandigarh University, Mohali, India
| | - Azad Khan
- Centre for Global Health Research, Dhaka, Bangladesh; Diabetic Association of Bangladesh, Dhaka, Bangladesh; Department of Public Health, Bangladesh University of Health Sciences, Dhaka, Bangladesh
| | - Viswanathan Mohan
- Madras Diabetes Research Foundation [ICMR- Collaborating Centre of Excellence (ICMR-CCoE)], India; Dr. Mohan's Diabetes Specialties Centre (IDF Centre of Excellence in Diabetes Care), Chennai, India
| |
Collapse
|
9
|
O CK, Fan B, Tsoi STF, Tam CHT, Wan R, Lau ESH, Shi M, Lim CKP, Yu G, Ho JPY, Chow EYK, Kong APS, Ozaki R, So WY, Ma RCW, Luk AOY, Chan JCN. A polygenic risk score derived from common variants of monogenic diabetes genes is associated with young-onset type 2 diabetes and cardiovascular-kidney complications. Diabetologia 2025; 68:367-381. [PMID: 39579208 PMCID: PMC11732898 DOI: 10.1007/s00125-024-06320-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/16/2024] [Indexed: 11/25/2024]
Abstract
AIMS/HYPOTHESIS Monogenic diabetes is caused by rare mutations in genes usually implicated in beta cell biology. Common variants of monogenic diabetes genes (MDG) may jointly influence the risk of young-onset type 2 diabetes (YOD, diagnosed before the age of 40 years) and cardiovascular and kidney events. METHODS Using whole-exome sequencing data, we constructed a weighted polygenic risk score (wPRS) consisting of 135 common variants (minor allele frequency >0.01) of 34 MDG based on r2>0.2 for linkage disequilibrium in a discovery case-control cohort of 453 adults with YOD (median [IQR] age 39.7 [34.9-46.9] years) and 405 without YOD (median [IQR] age 56.7 [50.3-61.0] years), followed by validation in an independent cross-sectional cohort with array-based genotyping for YOD and a prospective cohort of individuals with type 2 diabetes for cardiovascular and kidney events. RESULTS In the discovery cohort, the OR of the 135 common variants for YOD ranged from 1.00 to 2.61. In the validation cohort (920 YOD and 4910 non-YOD), top-10%-wPRS was associated with an OR of 1.42 (95% CI 1.03, 1.95, p=0.033) for YOD compared with bottom-10%-wPRS. In 2313 individuals with type 2 diabetes (median [IQR]: age 53.4 [45.4-61.7] years; disease duration 4.0 [1.0-9.0] years) observed for a median (IQR) of 17.5 (14.4-21.8) years, standardised wPRS was associated with increased HR for incident cardiovascular events (1.16 [95% CI 1.06, 1.27], p=0.001), kidney events (1.09 [95% CI 1.02, 1.16], p=0.013) and cardiovascular-kidney events (1.10 [95% CI 1.03, 1.16], p=0.003). Using the 'bottom-20%-wPRS plus baseline disease duration <5 years' group as referent, the 'top-20%-wPRS plus baseline disease duration 5 to <10 years' group had unadjusted and adjusted HR of 1.60 (95% CI 1.17, 2.19, p=0.003) and 1.62 (95% CI 1.16, 2.26, p=0.005), respectively, for cardiovascular-kidney events compared with 1.38 (95% CI 0.97, 1.98, p=0.075) and 1.06 (95% CI 0.72, 1.57, p=0.752) in the 'bottom-20%-wPRS plus baseline disease duration ≥10 years' group. CONCLUSIONS/INTERPRETATION Common variants of MDG increased risk for YOD and cardiovascular-kidney events.
Collapse
Affiliation(s)
- Chun-Kwan O
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Baoqi Fan
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Sandra T F Tsoi
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Claudia H T Tam
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Raymond Wan
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Eric S H Lau
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Mai Shi
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Cadmon K P Lim
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Gechang Yu
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Jane P Y Ho
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Elaine Y K Chow
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Alice P S Kong
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Risa Ozaki
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Wing Yee So
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Andrea O Y Luk
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China.
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China.
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China.
| |
Collapse
|
10
|
Korvyakova Y, Azarova I, Klyosova E, Postnikova M, Makarenko V, Bushueva O, Solodilova M, Polonikov A. The link between the ANPEP gene and type 2 diabetes mellitus may be mediated by the disruption of glutathione metabolism and redox homeostasis. Gene 2025; 935:149050. [PMID: 39489227 DOI: 10.1016/j.gene.2024.149050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Aminopeptidase N (ANPEP), a membrane-associated ectoenzyme, has been identified as a susceptibility gene for type 2 diabetes (T2D) by genome-wide association and transcriptome studies; however, the mechanisms by which this gene contributes to disease pathogenesis remain unclear. The aim of this study was to determine the comprehensive contribution of ANPEP polymorphisms to T2D risk and annotate the underlying mechanisms. A total of 3206 unrelated individuals including 1579 T2D patients and 1627 controls were recruited for the study. Twenty-three common functional single nucleotide polymorphisms (SNP) of ANPEP were genotyped by the MassArray-4 system. Six polymorphisms, rs11073891, rs12898828, rs12148357, rs9920421, rs7111, and rs25653, were found to be associated with type 2 diabetes (Pperm ≤ 0.05). Common haplotype rs9920421G-rs4932143G-rs7111T was strongly associated with increased risk of T2D (Pperm = 5.9 × 10-12), whereas two rare haplotypes such as rs9920421G-rs4932143C-rs7111T (Pperm = 6.5 × 10-40) and rs12442778A-rs12898828A-rs6496608T-rs11073891C (Pperm = 1.0 × 10-7) possessed strong protection against disease. We identified 38 and 109 diplotypes associated with T2D risk in males and females, respectively (FDR ≤ 0.05). ANPEP polymorphisms showed associations with plasma levels of fasting blood glucose, aspartate aminotransferase, total protein and glutathione (P < 0.05), and several haplotypes were strongly associated with the levels of reactive oxygen species and uric acid (P < 0.0001). A deep literature analysis has facilitated the formulation of a hypothesis proposing that increased plasma levels of ANPEP as well as liver enzymes such as aspartate aminotransferase, alanine aminotransferase and gammaglutamyltransferase serve as an adaptive response directed towards the restoration of glutathione deficiency in diabetics by stimulating the production of amino acid precursors for glutathione biosynthesis.
Collapse
Affiliation(s)
- Yaroslava Korvyakova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation; Research Centre for Medical Genetics, 1 Moskvorechie St., Moscow 115522, Russian Federation.
| | - Iuliia Azarova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation; Department of Biological Chemistry, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation.
| | - Elena Klyosova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation; Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation.
| | - Maria Postnikova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation.
| | - Victor Makarenko
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation
| | - Olga Bushueva
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation; Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation.
| | - Maria Solodilova
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation.
| | - Alexey Polonikov
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation; Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation.
| |
Collapse
|
11
|
Lee Jia Jia I, Zampetti S, Pozzilli P, Buzzetti R. Type 2 diabetes in children and adolescents: Challenges for treatment and potential solutions. Diabetes Res Clin Pract 2024; 217:111879. [PMID: 39369858 DOI: 10.1016/j.diabres.2024.111879] [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: 08/26/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Historically perceived as a disease mainly affecting adults, the prevalence of type 2 diabetes mellitus (T2DM) among children and adolescents has been rising, mirroring the increasing rates of childhood obesity. Currently, youth-onset T2DM poses a significant public health challenge globally. Treating youth-onset T2DM poses numerous critical challenges, namely limited and inadequate therapeutic options, and difficulties with conducting therapeutic studies. As a result, current treatment guidelines are based on adult studies and expert consensus. Few prominent guidelines on the treatment of youth-onset T2DM have been published recently, i.e., by the American Diabetes Association (ADA) 2024, National Institute for Healthcare and Excellence United Kingdom (NICE UK) 2023, International Society Paediatric and Adolescents Diabetes (ISPAD) 2022, Australasian Paediatric Endocrine Group (APEG) 2020 and Diabetes Canada 2018. This review first explores the unique aspects of youth-onset T2DM. It then summarises the different treatment guidelines, discusses the different treatment modalities based on available evidence and identifies any gaps. The review also explores challenges in the treatment of youth-onset T2DM with potential solutions and discusses recent trials on the treatment of youth-onset T2DM. Continued research aims to optimise treatment, improve outcomes, and alleviate the burden of T2DM on youths.
Collapse
Affiliation(s)
- Ivy Lee Jia Jia
- Barts and the London School of Medicine, Queen Mary University of London, London, UK
| | - Simona Zampetti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Paolo Pozzilli
- Centre of Immunobiology, Blizard Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK; Endocrinology and Diabetes, University Campus Bio-Medico, Rome, Italy
| | - Raffaella Buzzetti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
12
|
Jabeen A, Riaz S, Usman M, Parveen A, Mukhtar M, Wajid A, Hanif A, Batool A. Association of polymorphism of NLRP3, ICAM-1, PTPN22, INS genes in childhood onset type 1 diabetes in a Pakistani population. Mol Biol Rep 2024; 51:1070. [PMID: 39425874 DOI: 10.1007/s11033-024-09983-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 10/05/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Type 1 diabetes (T1D) is an organ-specific autoimmune disorder characterized by the destruction of pancreatic β cells, leading to absolute insulin deficiency. The genes NLRP3, ICAM-1, PTPN22, and INS are reportedly associated with T1D in other populations. However, the genetic pattern of T1D in the Pakistani population is not clear. This study aimed to find the association of polymorphisms in the PTPN22, INS, NLRP3, and ICAM-1 genes with T1D susceptibility in the Pakistani population. METHODOLOGY This case-control study includes 100 T1D patients (3-14 years), recruited randomly from the pediatric endocrinology department of Fatima Memorial Hospital, Lahore, Pakistan and 100 age-matched healthy controls were selected from different localities of the same population. The polymorphisms in PTPN22 (rs601, rs33996649, rs2488457), INS (rs80356664), NLRP3 (rs10754558, rs35829419), and ICAM-1 (rs1799969, rs5498) genes were genotyped by Sanger sequencing. The genotypic and allelic frequencies, haplotypes, and linkage disequilibrium were computed using the genetic toolset PLINK to investigate their relationship to T1D. RESULTS The results indicate that the occurrence of the GT genotype of the rs33996649 variant is significantly higher in children with T1D compared to a control group of healthy individuals (P = 0.001, OR: 2.0, 95% CI = 0.15-0.45). Furthermore, the CT genotype of rs2488457 was notably associated with T1D patients (P = 0.007, OR: 2.8, 95% CI = 0.56-0.67). The CG genotype of rs80356664 showed a slight association with T1D (P = 0.03, OR: 1.9, 95% CI = 0.35-0.59). The prevalence of the AT genotype of rs10754558 showed a strong association with T1D (P = 0.005, OR: 3.4, 95% CI = 0.45-0.69). The TG genotype of rs5498 was also strongly associated with T1D (P = 0.009, OR: 2.8, 95% CI = 0.75-0.89). CONCLUSION The present study provides evidence that SNPs in the PTPN22, INS, NLRP3, and ICAM-1 genes are associated with the development of T1D. Further research is needed to explore their potential use in genetic screening and personalized medication.
Collapse
Affiliation(s)
- Anjum Jabeen
- Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Samreen Riaz
- Institute of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan
| | - Muhammad Usman
- Fatima Memorial Medical and Dental College, Lahore, Pakistan
| | - Asia Parveen
- Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Maryam Mukhtar
- Institute of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan
| | - Abdul Wajid
- Department of Biotechnology, Faculty of Life Sciences and Informatics, Engineering and Management Sciences, Baluchistan University of Information Technology, Quetta, Pakistan
| | - Atif Hanif
- Department of Botany & Microbiology, King Saud University, Riaz, Saudi Arabia
| | - Andleeb Batool
- Department of Zoology, Government College University, Lahore, 54000, Pakistan.
| |
Collapse
|
13
|
Hajj J, Sizemore B, Singh K. Impact of Epigenetics, Diet, and Nutrition-Related Pathologies on Wound Healing. Int J Mol Sci 2024; 25:10474. [PMID: 39408801 PMCID: PMC11476922 DOI: 10.3390/ijms251910474] [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: 07/30/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Chronic wounds pose a significant challenge to healthcare. Stemming from impaired wound healing, the consequences can be severe, ranging from amputation to mortality. This comprehensive review explores the multifaceted impact of chronic wounds in medicine and the roles that diet and nutritional pathologies play in the wound-healing process. It has been well established that an adequate diet is crucial to proper wound healing. Nutrients such as vitamin D, zinc, and amino acids play significant roles in cellular regeneration, immune functioning, and collagen synthesis and processing. Additionally, this review discusses how patients with chronic conditions like diabetes, obesity, and nutritional deficiencies result in the formation of chronic wounds. By integrating current research findings, this review highlights the significant impact of the genetic make-up of an individual on the risk of developing chronic wounds and the necessity for adequate personalized dietary interventions. Addressing the nutritional needs of individuals, especially those with chronic conditions, is essential for improving wound outcomes and overall patient care. With new developments in the field of genomics, there are unprecedented opportunities to develop targeted interventions that can precisely address the unique metabolic needs of individuals suffering from chronic wounds, thereby enhancing treatment effectiveness and patient outcomes.
Collapse
Affiliation(s)
- John Hajj
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.H.); (B.S.)
| | - Brandon Sizemore
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.H.); (B.S.)
| | - Kanhaiya Singh
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.H.); (B.S.)
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
14
|
Rabby MG, Suzauddula M, Hasan MS, Dewan MA, Islam MN. In-silico identification and functional characterization of common genes associated with type 2 diabetes and hypertension. Heliyon 2024; 10:e36546. [PMID: 39262940 PMCID: PMC11388505 DOI: 10.1016/j.heliyon.2024.e36546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
Type 2 diabetes (T2D) and hypertension are global public health concerns and major metabolic disorders in humans. Experimental evidence indicates considerable hereditary influences on the etiology of T2D and hypertension, but the molecular basis of these diseases is still limited. Thus, the current study analyzed 185 (132 T2D and 53 hypertension) GWAS catalog datasets and identified 83 common genes linked to T2D and hypertension pathogenesis. These genes were further examined using various bioinformatics approaches to elucidate their molecular mechanisms underlying the pathophysiology of T2D and hypertension. Gene ontology (GO) analysis revealed the biological, cellular, and molecular functions of these genes, which were also linked to different T2D and hypertension pathways. Specifically, seven genes were found to be crucial for T2D, and nine were directly associated with hypertension. Protein-protein interaction (PPI) analysis identified 28 candidate genes and seven hub genes through 11 topological methods. Among 231 miRNAs, seven were significant in interacting with the hub genes, and nine transcription factors (TFs) out of 36 were linked to these hub genes. Additionally, two of the seven hub genes were downregulated by 43 FDA-approved drugs. These findings elucidate the molecular processes underlying T2D and hypertension, suggesting that targeting these genes could lead to future drug development and therapeutic strategies to treat T2D and hypertension.
Collapse
Affiliation(s)
- Md Golam Rabby
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Md Suzauddula
- College of Agriculture and Natural Resources, National Chung Hsing University, Taichung City, 40227, Taiwan
| | - Md Sakib Hasan
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Mahbubur Alam Dewan
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Md Numan Islam
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
- Department of Food Science and Technology, University of Nebraska Lincoln, USA
| |
Collapse
|
15
|
Sleigh JW, Morgan PG. Are Genome-wide Association Studies Worth the Trouble? Anesthesiology 2024; 141:214-216. [PMID: 38980159 PMCID: PMC11239121 DOI: 10.1097/aln.0000000000005074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Affiliation(s)
- Jamie W Sleigh
- Department of Anaesthesiology, Waikato Clinical Campus, Faculty of Medical and Health Sciences, University of Auckland, Hamilton, NZ
| | - Philip G Morgan
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
16
|
O CK, Fan YN, Fan B, Lim C, Lau ESH, Tsoi STF, Wan R, Lai WY, Poon EW, Ho J, Ho CCY, Fung C, Lee EK, Wong SY, Wang M, Ozaki R, Cheung E, Ma RCW, Chow E, Kong APS, Luk A, Chan JCN. Precision Medicine to Redefine Insulin Secretion and Monogenic Diabetes-Randomized Controlled Trial (PRISM-RCT) in Chinese patients with young-onset diabetes: design, methods and baseline characteristics. BMJ Open Diabetes Res Care 2024; 12:e004120. [PMID: 38901858 PMCID: PMC11212116 DOI: 10.1136/bmjdrc-2024-004120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/29/2024] [Indexed: 06/22/2024] Open
Abstract
INTRODUCTION We designed and implemented a patient-centered, data-driven, holistic care model with evaluation of its impacts on clinical outcomes in patients with young-onset type 2 diabetes (T2D) for which there is a lack of evidence-based practice guidelines. RESEARCH DESIGN AND METHODS In this 3-year Precision Medicine to Redefine Insulin Secretion and Monogenic Diabetes-Randomized Controlled Trial, we evaluate the effects of a multicomponent care model integrating use of information and communication technology (Joint Asia Diabetes Evaluation (JADE) platform), biogenetic markers and patient-reported outcome measures in patients with T2D diagnosed at ≤40 years of age and aged ≤50 years. The JADE-PRISM group received 1 year of specialist-led team-based management using treatment algorithms guided by biogenetic markers (genome-wide single-nucleotide polymorphism arrays, exome-sequencing of 34 monogenic diabetes genes, C-peptide, autoantibodies) to achieve multiple treatment goals (glycated hemoglobin (HbA1c) <6.2%, blood pressure <120/75 mm Hg, low-density lipoprotein-cholesterol <1.2 mmol/L, waist circumference <80 cm (women) or <85 cm (men)) in a diabetes center setting versus usual care (JADE-only). The primary outcome is incidence of all diabetes-related complications. RESULTS In 2020-2021, 884 patients (56.6% men, median (IQR) diabetes duration: 7 (3-12) years, current/ex-smokers: 32.5%, body mass index: 28.40±5.77 kg/m2, HbA1c: 7.52%±1.66%, insulin-treated: 27.7%) were assigned to JADE-only (n=443) or JADE-PRISM group (n=441). The profiles of the whole group included positive family history (74.7%), general obesity (51.4%), central obesity (79.2%), hypertension (66.7%), dyslipidemia (76.4%), albuminuria (35.4%), estimated glomerular filtration rate <60 mL/min/1.73 m2 (4.0%), retinopathy (13.8%), atherosclerotic cardiovascular disease (5.2%), cancer (3.1%), emotional distress (26%-38%) and suboptimal adherence (54%) with 5-item EuroQol for Quality of Life index of 0.88 (0.87-0.96). Overall, 13.7% attained ≥3 metabolic targets defined in secondary outcomes. In the JADE-PRISM group, 4.5% had pathogenic/likely pathogenic variants of monogenic diabetes genes; 5% had autoantibodies and 8.4% had fasting C-peptide <0.2 nmol/L. Other significant events included low/large birth weight (33.4%), childhood obesity (50.7%), mental illness (10.3%) and previous suicide attempts (3.6%). Among the women, 17.3% had polycystic ovary syndrome, 44.8% required insulin treatment during pregnancy and 17.3% experienced adverse pregnancy outcomes. CONCLUSIONS Young-onset diabetes is characterized by complex etiologies with comorbidities including mental illness and lifecourse events. TRIAL REGISTRATION NUMBER NCT04049149.
Collapse
Affiliation(s)
- Chun Kwan O
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Ying Nan Fan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Baoqi Fan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Cadmon Lim
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Eric S H Lau
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Sandra T F Tsoi
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Raymond Wan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Wai Yin Lai
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Emily Wm Poon
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Jane Ho
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Cherry Cheuk Yee Ho
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Chloe Fung
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Eric Kp Lee
- JC School of Public Health & Primary Care, The Chinese University of Hong Kong Faculty of Medicine, Hong Kong Special Administrative Region, People's Republic of China
| | - Samuel Ys Wong
- JC School of Public Health & Primary Care, The Chinese University of Hong Kong Faculty of Medicine, Hong Kong Special Administrative Region, People's Republic of China
| | - Maggie Wang
- JC School of Public Health & Primary Care, The Chinese University of Hong Kong Faculty of Medicine, Hong Kong Special Administrative Region, People's Republic of China
| | - Risa Ozaki
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Elaine Cheung
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Ronald Ching Wan Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Elaine Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Alice Pik Shan Kong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Andrea Luk
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| |
Collapse
|
17
|
Armstrong ND, Patki A, Srinivasasainagendra V, Ge T, Lange LA, Kottyan L, Namjou B, Shah AS, Rasmussen-Torvik LJ, Jarvik GP, Meigs JB, Karlson EW, Limdi NA, Irvin MR, Tiwari HK. Variant level heritability estimates of type 2 diabetes in African Americans. Sci Rep 2024; 14:14009. [PMID: 38890458 PMCID: PMC11189523 DOI: 10.1038/s41598-024-64711-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Type 2 diabetes (T2D) is caused by both genetic and environmental factors and is associated with an increased risk of cardiorenal complications and mortality. Though disproportionately affected by the condition, African Americans (AA) are largely underrepresented in genetic studies of T2D, and few estimates of heritability have been calculated in this race group. Using genome-wide association study (GWAS) data paired with phenotypic data from ~ 19,300 AA participants of the Reasons for Geographic and Racial Differences in Stroke (REGARDS) study, Genetics of Hypertension Associated Treatments (GenHAT) study, and the Electronic Medical Records and Genomics (eMERGE) network, we estimated narrow-sense heritability using two methods: Linkage-Disequilibrium Adjusted Kinships (LDAK) and Genome-Wide Complex Trait Analysis (GCTA). Study-level heritability estimates adjusting for age, sex, and genetic ancestry ranged from 18% to 34% across both methods. Overall, the current study narrows the expected range for T2D heritability in this race group compared to prior estimates, while providing new insight into the genetic basis of T2D in AAs for ongoing genetic discovery efforts.
Collapse
Affiliation(s)
- Nicole D Armstrong
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Amit Patki
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Tian Ge
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Leslie A Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Leah Kottyan
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Bahram Namjou
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Amy S Shah
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center &, The University of Cincinnati, Cincinnati, OH, USA
| | - Laura J Rasmussen-Torvik
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Gail P Jarvik
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - James B Meigs
- Division of General Internal Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Elizabeth W Karlson
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Mass General Brigham Personalized Medicine, Boston, MA, USA
| | - Nita A Limdi
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Marguerite R Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hemant K Tiwari
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
18
|
Elashi AA, Toor SM, Umlai UKI, Al-Sarraj YA, Taheri S, Suhre K, Abou-Samra AB, Albagha OME. Genome-wide association study and trans-ethnic meta-analysis identify novel susceptibility loci for type 2 diabetes mellitus. BMC Med Genomics 2024; 17:115. [PMID: 38685053 PMCID: PMC11059680 DOI: 10.1186/s12920-024-01855-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 03/28/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND The genetic basis of type 2 diabetes (T2D) is under-investigated in the Middle East, despite the rapidly growing disease prevalence. We aimed to define the genetic determinants of T2D in Qatar. METHODS Using whole genome sequencing of 11,436 participants (2765 T2D cases and 8671 controls) from the population-based Qatar Biobank (QBB), we conducted a genome-wide association study (GWAS) of T2D with and without body mass index (BMI) adjustment. RESULTS We replicated 93 known T2D-associated loci in a BMI-unadjusted model, while 96 known loci were replicated in a BMI-adjusted model. The effect sizes and allele frequencies of replicated SNPs in the Qatari population generally concurred with those from European populations. We identified a locus specific to our cohort located between the APOBEC3H and CBX7 genes in the BMI-unadjusted model. Also, we performed a transethnic meta-analysis of our cohort with a previous GWAS on T2D in multi-ancestry individuals (180,834 T2D cases and 1,159,055 controls). One locus in DYNC2H1 gene reached genome-wide significance in the meta-analysis. Assessing polygenic risk scores derived from European- and multi-ancestries in the Qatari population showed higher predictive performance of the multi-ancestry panel compared to the European panel. CONCLUSION Our study provides new insights into the genetic architecture of T2D in a Middle Eastern population and identifies genes that may be explored further for their involvement in T2D pathogenesis.
Collapse
Affiliation(s)
- Asma A Elashi
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha, P.O. Box 34110, Qatar
| | - Salman M Toor
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha, P.O. Box 34110, Qatar
| | - Umm-Kulthum Ismail Umlai
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha, P.O. Box 34110, Qatar
| | - Yasser A Al-Sarraj
- Qatar Genome Program (QGP), Qatar Foundation Research, Development and Innovation, Qatar Foundation (QF), Doha, P.O. Box 5825, Qatar
| | - Shahrad Taheri
- Qatar Metabolic Institute, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, P.O. Box 24144, Qatar
- Department of Biophysics and Physiology, Weill Cornell Medicine, 510065, New York, USA
| | | | - Omar M E Albagha
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha, P.O. Box 34110, Qatar.
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, EH4 2XU, Edinburgh, UK.
| |
Collapse
|
19
|
Mandla R, Schroeder P, Porneala B, Florez JC, Meigs JB, Mercader JM, Leong A. Polygenic scores for longitudinal prediction of incident type 2 diabetes in an ancestrally and medically diverse primary care physician network: a patient cohort study. Genome Med 2024; 16:63. [PMID: 38671457 PMCID: PMC11046943 DOI: 10.1186/s13073-024-01337-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The clinical utility of genetic information for type 2 diabetes (T2D) prediction with polygenic scores (PGS) in ancestrally diverse, real-world US healthcare systems is unclear, especially for those at low clinical phenotypic risk for T2D. METHODS We tested the association of PGS with T2D incidence in patients followed within a primary care practice network over 16 years in four hypothetical scenarios that varied by clinical data availability (N = 14,712): (1) age and sex; (2) age, sex, body mass index (BMI), systolic blood pressure, and family history of T2D; (3) all variables in (2) and random glucose; and (4) all variables in (3), HDL, total cholesterol, and triglycerides, combined in a clinical risk score (CRS). To determine whether genetic effects differed by baseline clinical risk, we tested for interaction with the CRS. RESULTS PGS was associated with incident T2D in all models. Adjusting for age and sex only, the Hazard Ratio (HR) per PGS standard deviation (SD) was 1.76 (95% CI 1.68, 1.84) and the HR of top 5% of PGS vs interquartile range (IQR) was 2.80 (2.39, 3.28). Adjusting for the CRS, the HR per SD was 1.48 (1.40, 1.57) and HR of the top 5% of PGS vs IQR was 2.09 (1.72, 2.55). Genetic effects differed by baseline clinical risk ((PGS-CRS interaction p = 0.05; CRS below the median: HR 1.60 (1.43, 1.79); CRS above the median: HR 1.45 (1.35, 1.55)). CONCLUSIONS Genetic information can help identify high-risk patients even among those perceived to be low risk in a clinical evaluation.
Collapse
Affiliation(s)
- Ravi Mandla
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Philip Schroeder
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Bianca Porneala
- Division of General Internal Medicine, Department of Medicine, Massachusetts General Hospital, 100 Cambridge St. Fl. 16, Boston, MA, 02114, USA
| | - Jose C Florez
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - James B Meigs
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of General Internal Medicine, Department of Medicine, Massachusetts General Hospital, 100 Cambridge St. Fl. 16, Boston, MA, 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Josep M Mercader
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Aaron Leong
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Division of General Internal Medicine, Department of Medicine, Massachusetts General Hospital, 100 Cambridge St. Fl. 16, Boston, MA, 02114, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
Liu X, Littlejohns TJ, Bešević J, Bragg F, Clifton L, Collister JA, Trichia E, Gray LJ, Khunti K, Hunter DJ. Incorporating polygenic risk into the Leicester Risk Assessment score for 10-year risk prediction of type 2 diabetes. Diabetes Metab Syndr 2024; 18:102996. [PMID: 38608567 PMCID: PMC11913737 DOI: 10.1016/j.dsx.2024.102996] [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: 09/05/2023] [Revised: 02/22/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024]
Abstract
AIMS We evaluated whether incorporating information on ethnic background and polygenic risk enhanced the Leicester Risk Assessment (LRA) score for predicting 10-year risk of type 2 diabetes. METHODS The sample included 202,529 UK Biobank participants aged 40-69 years. We computed the LRA score, and developed two new risk scores using training data (80% sample): LRArev, which incorporated additional information on ethnic background, and LRAprs, which incorporated polygenic risk for type 2 diabetes. We assessed discriminative and reclassification performance in a test set (20% sample). Type 2 diabetes was ascertained using primary care, hospital inpatient and death registry records. RESULTS Over 10 years, 7,476 participants developed type 2 diabetes. The Harrell's C indexes were 0.796 (95% Confidence Interval [CI] 0.785, 0.806), 0.802 (95% CI 0.792, 0.813), and 0.829 (95% CI 0.820, 0.839) for the LRA, LRArev and LRAprs scores, respectively. The LRAprs score significantly improved the overall reclassification compared to the LRA (net reclassification index [NRI] = 0.033, 95% CI 0.015, 0.049) and LRArev (NRI = 0.040, 95% CI 0.024, 0.055) scores. CONCLUSIONS Polygenic risk moderately improved the performance of the existing LRA score for 10-year risk prediction of type 2 diabetes.
Collapse
Affiliation(s)
- Xiaonan Liu
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | | | - Jelena Bešević
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Fiona Bragg
- Nuffield Department of Population Health, University of Oxford, Oxford, UK; MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Lei Clifton
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | | | - Eirini Trichia
- Nuffield Department of Population Health, University of Oxford, Oxford, UK; MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Laura J Gray
- Department of Population Health Sciences, University of Leicester, UK
| | - Kamlesh Khunti
- Diabetes Research Centre, University of Leicester, Leicester, UK
| | - David J Hunter
- Nuffield Department of Population Health, University of Oxford, Oxford, UK; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
| |
Collapse
|
21
|
Imamura M, Maeda S. Perspectives on genetic studies of type 2 diabetes from the genome-wide association studies era to precision medicine. J Diabetes Investig 2024; 15:410-422. [PMID: 38259175 PMCID: PMC10981147 DOI: 10.1111/jdi.14149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/24/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Genome-wide association studies (GWAS) have facilitated a substantial and rapid increase in the number of confirmed genetic susceptibility variants for complex diseases. Approximately 700 variants predisposing individuals to the risk for type 2 diabetes have been identified through GWAS until 2023. From 2018 to 2022, hundreds of type 2 diabetes susceptibility loci with smaller effect sizes were identified through large-scale GWAS with sample sizes of 200,000 to >1 million. The clinical translation of genetic information for type 2 diabetes includes the development of novel therapeutics and risk predictions. Although drug discovery based on loci identified in GWAS remains challenging owing to the difficulty of functional annotation, global efforts have been made to identify novel biological mechanisms and therapeutic targets by applying multi-omics approaches or searching for disease-associated coding variants in isolated founder populations. Polygenic risk scores (PRSs), comprising up to millions of associated variants, can identify individuals with higher disease risk than those in the general population. In populations of European descent, PRSs constructed from base GWAS data with a sample size of approximately 450,000 have predicted the onset of diseases well. However, European GWAS-derived PRSs have limited predictive performance in non-European populations. The predictive accuracy of a PRS largely depends on the sample size of the base GWAS data. The results of GWAS meta-analyses for multi-ethnic groups as base GWAS data and cross-population polygenic prediction methodology have been applied to establish a universal PRS applicable to small isolated ethnic populations.
Collapse
Affiliation(s)
- Minako Imamura
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of MedicineUniversity of the RyukyusNishihara‐ChoJapan
- Division of Clinical Laboratory and Blood TransfusionUniversity of the Ryukyus HospitalNishihara‐ChoJapan
| | - Shiro Maeda
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of MedicineUniversity of the RyukyusNishihara‐ChoJapan
- Division of Clinical Laboratory and Blood TransfusionUniversity of the Ryukyus HospitalNishihara‐ChoJapan
| |
Collapse
|
22
|
Bansal V, Winkelmann BR, Dietrich JW, Boehm BO. Whole-exome sequencing in familial type 2 diabetes identifies an atypical missense variant in the RyR2 gene. Front Endocrinol (Lausanne) 2024; 15:1258982. [PMID: 38444585 PMCID: PMC10913019 DOI: 10.3389/fendo.2024.1258982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 01/10/2024] [Indexed: 03/07/2024] Open
Abstract
Genome-wide association studies have identified several hundred loci associated with type 2 diabetes mellitus (T2DM). Additionally, pathogenic variants in several genes are known to cause monogenic diabetes that overlaps clinically with T2DM. Whole-exome sequencing of related individuals with T2DM is a powerful approach to identify novel high-penetrance disease variants in coding regions of the genome. We performed whole-exome sequencing on four related individuals with T2DM - including one individual diagnosed at the age of 33 years. The individuals were negative for mutations in monogenic diabetes genes, had a strong family history of T2DM, and presented with several characteristics of metabolic syndrome. A missense variant (p.N2291D) in the type 2 ryanodine receptor (RyR2) gene was one of eight rare coding variants shared by all individuals. The variant was absent in large population databases and affects a highly conserved amino acid located in a mutational hotspot for pathogenic variants in Catecholaminergic polymorphic ventricular tachycardia (CPVT). Electrocardiogram data did not reveal any cardiac abnormalities except a lower-than-normal resting heart rate (< 60 bpm) in two individuals - a phenotype observed in CPVT individuals with RyR2 mutations. RyR2-mediated Ca2+ release contributes to glucose-mediated insulin secretion and pathogenic RyR2 mutations cause glucose intolerance in humans and mice. Analysis of glucose tolerance testing data revealed that missense mutations in a CPVT mutation hotspot region - overlapping the p.N2291D variant - are associated with complete penetrance for glucose intolerance. In conclusion, we have identified an atypical missense variant in the RyR2 gene that co-segregates with diabetes in the absence of overt CPVT.
Collapse
Affiliation(s)
- Vikas Bansal
- Department of Pediatrics, University of California San Diego, La Jolla, CA, United States
- Institute of Genomic Medicine, University of California San Diego, La Jolla, CA, United States
| | | | - Johannes W Dietrich
- Diabetes, Endocrinology and Metabolism Section, Department of Internal Medicine I, St. Josef Hospital, Ruhr University Hospitals, Bochum, Germany
- Diabetes Center Bochum-Hattingen, St. Elisabeth-Hospital Blankenstein, Hattingen, Germany
- Center for Rare Endocrine Diseases, Ruhr Center for Rare Diseases (CeSER), Ruhr University Bochum and Witten/Herdecke University, Bochum, Germany
- Center for Diabetes Technology, Catholic Hospitals Bochum, Bochum, Germany
| | - Bernhard O Boehm
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
23
|
Chaudhary N, Alawadhi F, Al-Serri A, Al-Temaimi R. TCF7L2 and FTO Polymorphisms Are Associated with Type 2 Diabetes Mellitus Risk in Kuwait. Med Princ Pract 2024; 33:157-163. [PMID: 38228106 PMCID: PMC11045214 DOI: 10.1159/000536229] [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: 09/14/2023] [Accepted: 01/10/2024] [Indexed: 01/18/2024] Open
Abstract
OBJECTIVE Despite the high prevalence of type 2 diabetes mellitus (T2DM) and obesity in the region, reports are limited on genetic risk factors associated with T2DM risk in Kuwait. Our aim was to investigate the association of reported FTO and TCF7L2 T2DM genetic risk variants in Kuwaiti T2DM patients. SUBJECTS AND METHODS FTO rs9939609 and TCF7L2 rs7903146 variants were genotyped in 203 T2DM patients and 162 healthy controls. Data analysis included Fisher's exact test, χ2 test, and linear and logistic regression analyses. RESULTS FTO rs9939609 (AA) and TCF7L2 rs7903146 (TT) genotypes associated with T2DM risk among Kuwaitis (p = 0.0016 and p < 0.0001; respectively). Both variants had the strongest association with T2DM risk in an autosomal recessive inheritance model (FTO rs9939609A: odds ratio (OR) 2.136, 95% confidence interval (CI): 1.21-3.67, p = 0.0075; TCF7L2 rs7903146T: OR 3.283, 95% CI: 1.92-5.76, p < 0.0001). Moreover, rs7903146T associated with risk of peripheral neuropathy (β = 0.735, 95% CI: 0.514-0.96, p < 0.001) and risk of myocardial infarction (β = 0.36, 95% CI: 0.024-0.7, p = 0.036) in T2DM patients. CONCLUSION The increased susceptibility of Kuwaitis to T2DM is influenced by the same common genetic factors found in other T2DM populations. Further investigations of other T2DM genetic risk factors in Kuwait should refine and further support the clinical utility of a genetic risk score in predicting T2DM risk in a high-risk population such as Kuwait.
Collapse
Affiliation(s)
- Nawal Chaudhary
- Undergraduate Medical Program, Department of Pathology, College of Medicine, Kuwait University, Jabriya, Kuwait
| | - Faye Alawadhi
- Undergraduate Medical Program, Department of Pathology, College of Medicine, Kuwait University, Jabriya, Kuwait
| | - Ahmad Al-Serri
- Human Genetics Unit, Department of Pathology, College of Medicine, Kuwait University, Jabriya, Kuwait
| | - Rabeah Al-Temaimi
- Human Genetics Unit, Department of Pathology, College of Medicine, Kuwait University, Jabriya, Kuwait
| |
Collapse
|
24
|
Mehra A, Mittal A, Vishwakarma PK. Prediction of Protein-Drug Interactions, Pharmacophore Modeling, and Toxicokinetics of Novel Leads for Type 2 Diabetes Treatment. Curr Drug Metab 2024; 25:355-380. [PMID: 39108115 DOI: 10.2174/0113892002321919240801065905] [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: 04/21/2024] [Revised: 06/23/2024] [Accepted: 07/05/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Small heterocyclic compounds have been crucial in pioneering advances in type 2 diabetes treatment. There has been a dramatic increase in the pharmacological development of novel heterocyclic derivatives aimed at stimulating the activation of Glucokinase (GK). A pharmaceutical intervention for diabetes is increasingly targeting GK as a legitimate target. Diabetes type 2 compromises Glucokinase's function, an enzyme vital for maintaining the balance of blood glucose levels. Medicinal substances strategically positioned to improve type 2 diabetes management are used to stimulate the GK enzyme using heterocyclic derivatives. OBJECTIVE The research endeavor aimed to craft novel compounds, drawing inspiration from the inherent coumarin nucleus found in nature. The goal was to evoke the activity of the glucokinase enzyme, offering a tailored approach to mitigate the undesired side effects typically associated with conventional therapies employed in the treatment of type 2 diabetes. METHODS Coumarin, sourced from nature's embrace, unfolds as a potent and naturally derived ally in the quest for innovative antidiabetic interventions. Coumarin was extracted from a variety of botanical origins, including Artemisia keiskeana, Mallotus resinosus, Jatropha integerrima, Ferula tingitana, Zanthoxylum schinifolium, Phebalium clavatum, and Mammea siamensis. This inclusive evaluation was conducted on Muybridge's digital database containing 53,000 hit compounds. The presence of the coumarin nucleus was found in 100 compounds, that were selected from this extensive repository. Utilizing Auto Dock Vina 1.5.6 and ChemBioDraw Ultra, structures generated through this process underwent docking analysis. Furthermore, these compounds were accurately predicted online log P using the Swiss ADME algorithm. A predictive analysis was conducted using PKCSM software on the primary compounds to assess potential toxicity. RESULTS Using Auto Dock Vina 1.5.6, 100 coumarin derivatives were assessed for docking. Glucokinase (GK) binding was significantly enhanced by most of these compounds. Based on superior binding characteristics compared with Dorzagliatin (standard GKA) and MRK (co-crystallized ligand), the top eight molecules were identified. After further evaluation through ADMET analysis of these eight promising candidates, it was confirmed that they met the Lipinski rule of five and their pharmacokinetic profile was enhanced. The highest binding affinity was demonstrated by APV16 at -10.6 kcal/mol. A comparison between the APV16, Dorzagliatin and MRK in terms of toxicity predictions using PKCSM indicated that the former exhibited less skin sensitization, AMES toxicity, and hepatotoxicity. CONCLUSION Glucokinase is most potently activated by 100 of the compound leads in the database of 53,000 compounds that contain the coumarin nucleus. APV12, with its high binding affinity, favorable ADMET (adjusted drug metabolic equivalents), minimal toxicity, and favorable pharmacokinetic profile warrants consideration for progress to in vitro testing. Nevertheless, to uncover potential therapeutic implications, particularly in the context of type 2 diabetes, thorough investigations and in-vivo evaluations are necessary for benchmarking before therapeutic use, especially experiments involving the STZ diabetic rat model.
Collapse
Affiliation(s)
- Anuradha Mehra
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road (NH-1), Phagwara (Punjab), 144411, India
| | - Amit Mittal
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road (NH-1), Phagwara (Punjab), 144411, India
| | - Prakhar Kumar Vishwakarma
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road (NH-1), Phagwara (Punjab), 144411, India
| |
Collapse
|
25
|
Szczerbinski L, Florez JC. Precision medicine in diabetes - current trends and future directions. Is the future now? COMPREHENSIVE PRECISION MEDICINE 2024:458-483. [DOI: 10.1016/b978-0-12-824010-6.00021-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
26
|
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
|
27
|
Kakutani-Hatayama M, Kadoya M, Morimoto A, Miyoshi A, Kosaka-Hamamoto K, Kanzaki A, Konishi K, Kusunoki Y, Syoji T, Koyama H. Excessive daytime napping independently associated with decreased insulin sensitivity in cross-sectional study - Hyogo Sleep Cardio-Autonomic Atherosclerosis cohort study. Front Endocrinol (Lausanne) 2023; 14:1211705. [PMID: 38027100 PMCID: PMC10656607 DOI: 10.3389/fendo.2023.1211705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Background Although excessive daytime napping has been shown to be involved in diabetes occurrence, its impact on insulin secretion and sensitivity has not been elucidated. It is speculated that excessive napping disrupts the sleep-wake rhythm and increases sympathetic nerve activity during the day, resulting in decreased insulin sensitivity, which may be a mechanism leading to development of diabetes. We previously conducted a cross-sectional study that showed an association of autonomic dysfunction with decreased insulin sensitivity, though involvement of autonomic function in the association between napping and insulin sensitivity remained unclear. Furthermore, the effects of napping used to supplement to short nighttime sleep on insulin secretion and sensitivity are also unknown. In the present cross-sectional study, we examined the relationships of daytime nap duration and autonomic function with insulin secretion and sensitivity in 436 subjects enrolled in the Hyogo Sleep Cardio-Autonomic Atherosclerosis (HSCAA) Cohort Study who underwent a 75-g oral glucose tolerance test (75-g OGTT), after excluding those already diagnosed with diabetes. Methods Daytime nap duration was objectively measured using actigraphy, with the subjects divided into the short (≤1 hour) and long (>1 hour) nap groups. Insulin secretion and sensitivity were determined using 75-g OGTT findings. Standard deviation of normal to normal R-R interval (SDNN), a measure of autonomic function, was also determined based on heart rate variability. Subgroup analysis was performed for the associations of napping with insulin secretion and sensitivity, with the results stratified by nighttime sleep duration of less or greater than six hours. Results Subjects in the long nap group exhibited lower insulin sensitivity parameters (QUICKI: β=-0.135, p<0.01; Matsuda index: β=-0.119, p<0.05) independent of other clinical factors. In contrast, no associations with insulin secretion were found in either group. Furthermore, the association of long nap duration with insulin sensitivity was not confounded by SDNN. Specific subgroup analyses revealed more prominent associations of long nap habit with lower insulin sensitivity in subjects with a short nighttime sleep time (β=-0.137, p<0.05). Conclusion Long daytime nap duration may be a potential risk factor for decreased insulin sensitivity.
Collapse
Affiliation(s)
| | - Manabu Kadoya
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Thongsroy J, Mutirangura A. The inverse association between DNA gaps and HbA1c levels in type 2 diabetes mellitus. Sci Rep 2023; 13:18987. [PMID: 37923892 PMCID: PMC10624909 DOI: 10.1038/s41598-023-46431-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 10/31/2023] [Indexed: 11/06/2023] Open
Abstract
Naturally occurring DNA gaps have been observed in eukaryotic DNA, including DNA in nondividing cells. These DNA gaps are found less frequently in chronologically aging yeast, chemically induced senescence cells, naturally aged rats, D-galactose-induced aging model rats, and older people. These gaps function to protect DNA from damage, so we named them youth-associated genomic stabilization DNA gaps (youth-DNA-gaps). Type 2 diabetes mellitus (type 2 DM) is characterized by an early aging phenotype. Here, we explored the correlation between youth-DNA-gaps and the severity of type 2 DM. Here, we investigated youth-DNA-gaps in white blood cells from normal controls, pre-DM, and type 2 DM patients. We found significantly decreased youth-DNA-gap numbers in the type 2 DM patients compared to normal controls (P = 0.0377, P = 0.0018 adjusted age). In the type 2 DM group, youth-DNA-gaps correlate directly with HbA1c levels. (r = - 0.3027, P = 0.0023). Decreased youth-DNA-gap numbers were observed in patients with type 2 DM and associated with increased HbA1c levels. Therefore, the decrease in youth-DNA-gaps is associated with the molecular pathogenesis of high blood glucose levels. Furthermore, youth-DNA-gap number is another marker that could be used to determine the severity of type 2 DM.
Collapse
Affiliation(s)
- Jirapan Thongsroy
- School of Medicine, Walailak University, Nakhon Si Thammarat, 80160, Thailand.
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand.
| | - Apiwat Mutirangura
- Center for Excellence in Molecular Genetics of Cancer and Human Diseases, Chulalongkorn University, Bangkok, Thailand
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
29
|
Burton MA, Garratt ES, Hewitt MO, Sharkh HY, Antoun E, Westbury LD, Dennison EM, Harvey NC, Cooper C, MacIsaac JL, Kobor MS, Patel HP, Godfrey KM, Lillycrop KA. DNA methylation of insulin signaling pathways is associated with HOMA2-IR in primary myoblasts from older adults. Skelet Muscle 2023; 13:17. [PMID: 37898813 PMCID: PMC10612387 DOI: 10.1186/s13395-023-00326-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/09/2023] [Indexed: 10/30/2023] Open
Abstract
BACKGROUND While ageing is associated with increased insulin resistance (IR), the molecular mechanisms underlying increased IR in the muscle, the primary organ for glucose clearance, have yet to be elucidated in older individuals. As epigenetic processes are suggested to contribute to the development of ageing-associated diseases, we investigated whether differential DNA methylation was associated with IR in human primary muscle stem cells (myoblasts) from community-dwelling older individuals. METHODS We measured DNA methylation (Infinium HumanMethylationEPIC BeadChip) in myoblast cultures from vastus lateralis biopsies (119 males/females, mean age 78.24 years) from the Hertfordshire Sarcopenia Study extension (HSSe) and examined differentially methylated cytosine phosphate guanine (CpG) sites (dmCpG), regions (DMRs) and gene pathways associated with HOMA2-IR, an index for the assessment of insulin resistance, and levels of glycated hemoglobin HbA1c. RESULTS Thirty-eight dmCpGs (false discovery rate (FDR) < 0.05) were associated with HOMA2-IR, with dmCpGs enriched in genes linked with JNK, AMPK and insulin signaling. The methylation signal associated with HOMA2-IR was attenuated after the addition of either BMI (6 dmCpGs), appendicular lean mass index (ALMi) (7 dmCpGs), grip strength (15 dmCpGs) or gait speed (23 dmCpGs) as covariates in the model. There were 8 DMRs (Stouffer < 0.05) associated with HOMA2-IR, including DMRs within T-box transcription factor (TBX1) and nuclear receptor subfamily-2 group F member-2 (NR2F2); the DMRs within TBX1 and NR2F2 remained associated with HOMA2-IR after adjustment for BMI, ALMi, grip strength or gait speed. Forty-nine dmCpGs and 21 DMRs were associated with HbA1c, with cg13451048, located within exoribonuclease family member 3 (ERI3) associated with both HOMA2-IR and HbA1c. HOMA2-IR and HbA1c were not associated with accelerated epigenetic ageing. CONCLUSIONS These findings suggest that insulin resistance is associated with differential DNA methylation in human primary myoblasts with both muscle mass and body composition making a significant contribution to the methylation changes associated with IR.
Collapse
Affiliation(s)
- Mark A Burton
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Emma S Garratt
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Matthew O Hewitt
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Hanan Y Sharkh
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK
- Biological Sciences, University of Southampton, Southampton, UK
| | - Elie Antoun
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Leo D Westbury
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Elaine M Dennison
- NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital Southampton NHS Foundation Trust, Southampton, UK
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Nicholas C Harvey
- NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital Southampton NHS Foundation Trust, Southampton, UK
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Cyrus Cooper
- NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital Southampton NHS Foundation Trust, Southampton, UK
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Julia L MacIsaac
- Department of Medical Genetics, Faculty of Medicine, Edwin S.H. Leong Healthy Aging Program, University of British Columbia, Vancouver, Canada
| | - Michael S Kobor
- Department of Medical Genetics, Faculty of Medicine, Edwin S.H. Leong Healthy Aging Program, University of British Columbia, Vancouver, Canada
| | - Harnish P Patel
- NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital Southampton NHS Foundation Trust, Southampton, UK
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
- Faculty of Medicine, Academic Geriatric Medicine, University of Southampton, Southampton, UK
| | - Keith M Godfrey
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital Southampton NHS Foundation Trust, Southampton, UK
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Karen A Lillycrop
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK.
- NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital Southampton NHS Foundation Trust, Southampton, UK.
- Biological Sciences, University of Southampton, Southampton, UK.
| |
Collapse
|
30
|
Misra S, Ke C, Srinivasan S, Goyal A, Nyriyenda MJ, Florez JC, Khunti K, Magliano DJ, Luk A. Current insights and emerging trends in early-onset type 2 diabetes. Lancet Diabetes Endocrinol 2023; 11:768-782. [PMID: 37708901 DOI: 10.1016/s2213-8587(23)00225-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/01/2023] [Accepted: 07/19/2023] [Indexed: 09/16/2023]
Abstract
Type 2 diabetes diagnosed in childhood or early adulthood is termed early-onset type 2 diabetes. Cases of early-onset type 2 diabetes are increasing rapidly globally, alongside rising obesity. Compared with a diagnosis later in life, an earlier-onset diagnosis carries an unexplained excess risk of microvascular complications, adverse cardiovascular outcomes, and earlier death. Women with early-onset type 2 diabetes also have a higher risk of adverse pregnancy outcomes. The high burden of complications renders individuals with early-onset type 2 diabetes at future risk of multimorbidity and interventions to reverse these concerning trends should be a priority. Within the early-onset cohort, disease pathophysiology and interventions have been better studied in paediatric-onset (<19 years) type 2 diabetes compared to adults; however, young adults aged 19-39 years (a larger number proportionally) are not well characterised and are also invisible in the current evidence base supporting management, which is derived from trials in later-onset type 2 diabetes. Young adults with type 2 diabetes face challenges in self-management that older individuals are less likely to experience (being in education or of working age, higher diabetes distress, and possible obesity-related stigma and diabetes-related stigma). There is a major research gap as to the optimal strategies to deploy in managing type 2 diabetes in adolescents and young adults, given that current models of care appear to not work as well in this age group. In the face of manifold risk factors (obesity, female sex, social deprivation, non-White European ethnicity, and genetic risk factors) prevention strategies with tailored lifestyle interventions, where needed, are likely to have greater success, but more evidence is needed. In this Review, we draw on evidence from both adolescents and young adults to provide a contemporary update on the current insights and emerging trends in early-onset type 2 diabetes.
Collapse
Affiliation(s)
- Shivani Misra
- Division of Metabolism, Digestion and Reproduction, Imperial College London, London, UK; Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, London, UK.
| | - Calvin Ke
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | - Shylaja Srinivasan
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of California at San Francisco, San Francisco, CA, USA
| | - Alpesh Goyal
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, New Delhi, India
| | - Moffat J Nyriyenda
- Medical Research Council-Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine, Uganda Research Unit, Entebbe, Uganda; London School of Hygiene and Tropical Medicine, London, UK
| | - Jose C Florez
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kamlesh Khunti
- Diabetes Research Centre, Leicester General Hospital, University of Leicester, Leicester, UK
| | - Dianna J Magliano
- Baker Heart and Diabetes Institute, Melbourne, Australia; School of Public Health and Prevention, Monash University Melbourne, Melbourne, Australia
| | - Andrea Luk
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
31
|
Mandla R, Schroeder P, Porneala B, Florez JC, Meigs JB, Mercader JM, Leong A. Polygenic Scores for Longitudinal Prediction of Incident Type 2 Diabetes in an Ancestrally and Medically Diverse Primary Care Network. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.08.23295276. [PMID: 37732255 PMCID: PMC10508788 DOI: 10.1101/2023.09.08.23295276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
OBJECTIVE The clinical utility of genetic information for type 2 diabetes (T2D) prediction with polygenic score (PGS) in ancestrally diverse, real-world US healthcare systems is unclear, especially for those at low clinical phenotypic risk for T2D. RESEARCH DESIGN AND METHODS We tested the association of PGS with T2D incidence in patients followed within a primary care practice network over 16 years in four hypothetical scenarios that varied by clinical data availability (N = 14,712): 1) age and sex, 2) age, sex, BMI, systolic blood pressure, and family history of diabetes; 3) all variables in (2) and random glucose; 4) all variables in (3), HDL, total cholesterol, and triglycerides, combined in a clinical risk score (CRS). To determine whether genetic effects differed by baseline clinical risk, we tested for interaction with the CRS. RESULTS PGS was associated with incident diabetes in all models. Adjusting for age and sex only, the Hazard Ratio (HR) per PGS standard deviation (SD) was 1.76 (95% CI 1.68, 1.84) and the HR of top 5% of PGS vs interquartile range (IQR) was 2.80 (2.39, 3.28). Adjusting for the CRS, the HR per SD was 1.48 (1.40, 1.57) and HR of top 5% of PGS vs IQR was 2.09 (1.72, 2.55). Genetic effects differed by baseline clinical risk [(PGS-CRS interaction p =0.05; CRS below the median: HR 1.60 (1.43, 1.79); CRS above the median: HR 1.45 (1.35, 1.55)]. CONCLUSIONS Genetic information can help identify high-risk patients even among those perceived to be low risk in a clinical evaluation.
Collapse
|
32
|
Zöller B, Manderstedt E, Lind‐Halldén C, Halldén C. Bioinformatic and rare-variant collapsing analyses for type 1 and type 2 diabetes in the UK Biobank reveal novel pleiotropic susceptibility loci. J Diabetes 2023; 15:799-802. [PMID: 37528512 PMCID: PMC10509505 DOI: 10.1111/1753-0407.13452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 07/19/2023] [Indexed: 08/03/2023] Open
Affiliation(s)
- Bengt Zöller
- Center for Primary Health Care Research, Department of Clinical SciencesLund University and Region SkåneMalmöSweden
| | - Eric Manderstedt
- Center for Primary Health Care Research, Department of Clinical SciencesLund University and Region SkåneMalmöSweden
| | - Christina Lind‐Halldén
- Department of Environmental Science and BioscienceKristianstad UniversityKristianstadSweden
| | - Christer Halldén
- Center for Primary Health Care Research, Department of Clinical SciencesLund University and Region SkåneMalmöSweden
| |
Collapse
|
33
|
Thongsroy J, Mutirangura A. Decreased Alu methylation in type 2 diabetes mellitus patients increases HbA1c levels. J Clin Lab Anal 2023; 37:e24966. [PMID: 37743692 PMCID: PMC10623537 DOI: 10.1002/jcla.24966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/20/2023] [Accepted: 09/13/2023] [Indexed: 09/26/2023] Open
Abstract
INTRODUCTION Alu hypomethylation is a common epigenetic process that promotes genomic instability with aging phenotypes, which leads to type 2 diabetes mellitus (type 2 DM). Previously, our results showed significantly decreased Alu methylation levels in type 2 DM patients. In this study, we aimed to investigate the longitudinal changes in Alu methylation levels in these patients. RESULTS We observed significantly decreased Alu methylation levels in type 2 DM patients compared with normal (p = 0.0462). Moreover, our findings demonstrated changes in Alu hypomethylation over a follow-up period within the same individuals (p < 0.0001). A reduction in Alu methylation was found in patients with increasing HbA1c levels (p = 0.0013) and directly correlated with increased HbA1c levels in type 2 DM patients (r = -0.2273, p = 0.0387). CONCLUSIONS Alu methylation in type 2 DM patients progressively decreases with increasing HbA1c levels. This observation suggests a potential association between Alu hypomethylation and the underlying molecular mechanisms of elevated blood glucose. Furthermore, monitoring Alu methylation levels may serve as a valuable biomarker for assessing the clinical outcomes of type 2 DM.
Collapse
Affiliation(s)
- Jirapan Thongsroy
- School of MedicineWalailak UniversityNakhon Si ThammaratThailand
- Research Center in Tropical PathobiologyWalailak UniversityNakhon Si ThammaratThailand
| | - Apiwat Mutirangura
- Center for Excellence in Molecular Genetics of Cancer and Human DiseasesChulalongkorn UniversityBangkokThailand
- Department of Anatomy, Faculty of MedicineChulalongkorn UniversityBangkokThailand
| |
Collapse
|
34
|
Shahisavandi M, Wang K, Ghanbari M, Ahmadizar F. Exploring Metabolomic Patterns in Type 2 Diabetes Mellitus and Response to Glucose-Lowering Medications-Review. Genes (Basel) 2023; 14:1464. [PMID: 37510368 PMCID: PMC10379356 DOI: 10.3390/genes14071464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
The spectrum of information related to precision medicine in diabetes generally includes clinical data, genetics, and omics-based biomarkers that can guide personalized decisions on diabetes care. Given the remarkable progress in patient risk characterization, there is particular interest in using molecular biomarkers to guide diabetes management. Metabolomics is an emerging molecular approach that helps better understand the etiology and promises the identification of novel biomarkers for complex diseases. Both targeted or untargeted metabolites extracted from cells, biofluids, or tissues can be investigated by established high-throughput platforms, like nuclear magnetic resonance (NMR) and mass spectrometry (MS) techniques. Metabolomics is proposed as a valuable tool in precision diabetes medicine to discover biomarkers for diagnosis, prognosis, and management of the progress of diabetes through personalized phenotyping and individualized drug-response monitoring. This review offers an overview of metabolomics knowledge as potential biomarkers in type 2 diabetes mellitus (T2D) diagnosis and the response to glucose-lowering medications.
Collapse
Affiliation(s)
- Mina Shahisavandi
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Kan Wang
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Fariba Ahmadizar
- Department of Data Science & Biostatistics, Julius Global Health, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
35
|
Rothman DL, Moore PB, Shulman RG. The impact of metabolism on the adaptation of organisms to environmental change. Front Cell Dev Biol 2023; 11:1197226. [PMID: 37377740 PMCID: PMC10291235 DOI: 10.3389/fcell.2023.1197226] [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: 03/30/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Since Jacob and Monod's discovery of the lac operon ∼1960, the explanations offered for most metabolic adaptations have been genetic. The focus has been on the adaptive changes in gene expression that occur, which are often referred to as "metabolic reprogramming." The contributions metabolism makes to adaptation have been largely ignored. Here we point out that metabolic adaptations, including the associated changes in gene expression, are highly dependent on the metabolic state of an organism prior to the environmental change to which it is adapting, and on the plasticity of that state. In support of this hypothesis, we examine the paradigmatic example of a genetically driven adaptation, the adaptation of E. coli to growth on lactose, and the paradigmatic example of a metabolic driven adaptation, the Crabtree effect in yeast. Using a framework based on metabolic control analysis, we have reevaluated what is known about both adaptations, and conclude that knowledge of the metabolic properties of these organisms prior to environmental change is critical for understanding not only how they survive long enough to adapt, but also how the ensuing changes in gene expression occur, and their phenotypes post-adaptation. It would be useful if future explanations for metabolic adaptations acknowledged the contributions made to them by metabolism, and described the complex interplay between metabolic systems and genetic systems that make these adaptations possible.
Collapse
Affiliation(s)
- Douglas L. Rothman
- Departments of Radiology, Yale University, New Haven, CT, United States
- Biomedical Engineering, Yale University, New Haven, CT, United States
- Yale Magnetic Resonance Research Center, Yale University School of Medicine, New Haven, CT, United States
| | - Peter B. Moore
- Department of Molecular Biology and Biophysics, Yale University, New Haven, CT, United States
- Department of Chemistry, Yale University, New Haven, CT, United States
| | - Robert G. Shulman
- Yale Magnetic Resonance Research Center, Yale University School of Medicine, New Haven, CT, United States
- Department of Molecular Biology and Biophysics, Yale University, New Haven, CT, United States
| |
Collapse
|
36
|
Beydag-Tasöz BS, Yennek S, Grapin-Botton A. Towards a better understanding of diabetes mellitus using organoid models. Nat Rev Endocrinol 2023; 19:232-248. [PMID: 36670309 PMCID: PMC9857923 DOI: 10.1038/s41574-022-00797-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 01/22/2023]
Abstract
Our understanding of diabetes mellitus has benefited from a combination of clinical investigations and work in model organisms and cell lines. Organoid models for a wide range of tissues are emerging as an additional tool enabling the study of diabetes mellitus. The applications for organoid models include studying human pancreatic cell development, pancreatic physiology, the response of target organs to pancreatic hormones and how glucose toxicity can affect tissues such as the blood vessels, retina, kidney and nerves. Organoids can be derived from human tissue cells or pluripotent stem cells and enable the production of human cell assemblies mimicking human organs. Many organ mimics relevant to diabetes mellitus are already available, but only a few relevant studies have been performed. We discuss the models that have been developed for the pancreas, liver, kidney, nerves and vasculature, how they complement other models, and their limitations. In addition, as diabetes mellitus is a multi-organ disease, we highlight how a merger between the organoid and bioengineering fields will provide integrative models.
Collapse
Affiliation(s)
- Belin Selcen Beydag-Tasöz
- The Novo Nordisk Foundation Center for Stem Cell Biology, Copenhagen, Denmark
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Siham Yennek
- The Novo Nordisk Foundation Center for Stem Cell Biology, Copenhagen, Denmark
| | - Anne Grapin-Botton
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
- Paul Langerhans Institute Dresden, Dresden, Germany.
| |
Collapse
|
37
|
Lowe WL. Genetics and Epigenetics: Implications for the Life Course of Gestational Diabetes. Int J Mol Sci 2023; 24:6047. [PMID: 37047019 PMCID: PMC10094577 DOI: 10.3390/ijms24076047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Gestational diabetes (GDM) is one of the most common complications of pregnancy, affecting as many as one in six pregnancies. It is associated with both short- and long-term adverse outcomes for the mother and fetus and has important implications for the life course of affected women. Advances in genetics and epigenetics have not only provided new insight into the pathophysiology of GDM but have also provided new approaches to identify women at high risk for progression to postpartum cardiometabolic disease. GDM and type 2 diabetes share similarities in their pathophysiology, suggesting that they also share similarities in their genetic architecture. Candidate gene and genome-wide association studies have identified susceptibility genes that are shared between GDM and type 2 diabetes. Despite these similarities, a much greater effect size for MTNR1B in GDM compared to type 2 diabetes and association of HKDC1, which encodes a hexokinase, with GDM but not type 2 diabetes suggest some differences in the genetic architecture of GDM. Genetic risk scores have shown some efficacy in identifying women with a history of GDM who will progress to type 2 diabetes. The association of epigenetic changes, including DNA methylation and circulating microRNAs, with GDM has also been examined. Targeted and epigenome-wide approaches have been used to identify DNA methylation in circulating blood cells collected during early, mid-, and late pregnancy that is associated with GDM. DNA methylation in early pregnancy had some ability to identify women who progressed to GDM, while DNA methylation in blood collected at 26-30 weeks gestation improved upon the ability of clinical factors alone to identify women at risk for progression to abnormal glucose tolerance post-partum. Finally, circulating microRNAs and long non-coding RNAs that are present in early or mid-pregnancy and associated with GDM have been identified. MicroRNAs have also proven efficacious in predicting both the development of GDM as well as its long-term cardiometabolic complications. Studies performed to date have demonstrated the potential for genetic and epigenetic technologies to impact clinical care, although much remains to be done.
Collapse
Affiliation(s)
- William L Lowe
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Rubloff 12, 420 E. Superior Street, Chicago, IL 60611, USA
| |
Collapse
|
38
|
Azarova I, Polonikov A, Klyosova E. Molecular Genetics of Abnormal Redox Homeostasis in Type 2 Diabetes Mellitus. Int J Mol Sci 2023; 24:4738. [PMID: 36902173 PMCID: PMC10003739 DOI: 10.3390/ijms24054738] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
Numerous studies have shown that oxidative stress resulting from an imbalance between the production of free radicals and their neutralization by antioxidant enzymes is one of the major pathological disorders underlying the development and progression of type 2 diabetes (T2D). The present review summarizes the current state of the art advances in understanding the role of abnormal redox homeostasis in the molecular mechanisms of T2D and provides comprehensive information on the characteristics and biological functions of antioxidant and oxidative enzymes, as well as discusses genetic studies conducted so far in order to investigate the contribution of polymorphisms in genes encoding redox state-regulating enzymes to the disease pathogenesis.
Collapse
Affiliation(s)
- Iuliia Azarova
- Department of Biological Chemistry, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
| | - Alexey Polonikov
- Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia
| | - Elena Klyosova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
| |
Collapse
|
39
|
Kupai K, Várkonyi T, Török S, Gáti V, Czimmerer Z, Puskás LG, Szebeni GJ. Recent Progress in the Diagnosis and Management of Type 2 Diabetes Mellitus in the Era of COVID-19 and Single Cell Multi-Omics Technologies. Life (Basel) 2022; 12:1205. [PMID: 36013384 PMCID: PMC9409806 DOI: 10.3390/life12081205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/29/2022] [Accepted: 08/05/2022] [Indexed: 11/17/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is one of the world's leading causes of death and life-threatening conditions. Therefore, we review the complex vicious circle of causes responsible for T2DM and risk factors such as the western diet, obesity, genetic predisposition, environmental factors, and SARS-CoV-2 infection. The prevalence and economic burden of T2DM on societal and healthcare systems are dissected. Recent progress on the diagnosis and clinical management of T2DM, including both non-pharmacological and latest pharmacological treatment regimens, are summarized. The treatment of T2DM is becoming more complex as new medications are approved. This review is focused on the non-insulin treatments of T2DM to reach optimal therapy beyond glycemic management. We review experimental and clinical findings of SARS-CoV-2 risks that are attributable to T2DM patients. Finally, we shed light on the recent single-cell-based technologies and multi-omics approaches that have reached breakthroughs in the understanding of the pathomechanism of T2DM.
Collapse
Affiliation(s)
- Krisztina Kupai
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, 6726 Szeged, Hungary
- Department of Internal Medicine, University of Szeged, Korányi fasor 8, 6720 Szeged, Hungary
| | - Tamás Várkonyi
- Department of Internal Medicine, University of Szeged, Korányi fasor 8, 6720 Szeged, Hungary
| | - Szilvia Török
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, 6726 Szeged, Hungary
| | - Viktória Gáti
- Laboratory of Functional Genomics, Biological Research Centre, Temesvári krt. 62, 6726 Szeged, Hungary
| | - Zsolt Czimmerer
- Laboratory of Functional Genomics, Biological Research Centre, Temesvári krt. 62, 6726 Szeged, Hungary
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Life Science Building, Egyetem tér 1, 4032 Debrecen, Hungary
| | - László G. Puskás
- Laboratory of Functional Genomics, Biological Research Centre, Temesvári krt. 62, 6726 Szeged, Hungary
- Avidin Ltd., Alsó kikötő sor 11/D, 6726 Szeged, Hungary
| | - Gábor J. Szebeni
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, 6726 Szeged, Hungary
- Laboratory of Functional Genomics, Biological Research Centre, Temesvári krt. 62, 6726 Szeged, Hungary
- CS-Smartlab Devices Ltd., Ady E. u. 14, 7761 Kozármisleny, Hungary
| |
Collapse
|
40
|
Laakso M, Fernandes Silva L. Genetics of Type 2 Diabetes: Past, Present, and Future. Nutrients 2022; 14:nu14153201. [PMID: 35956377 PMCID: PMC9370092 DOI: 10.3390/nu14153201] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/30/2022] [Accepted: 08/03/2022] [Indexed: 02/01/2023] Open
Abstract
Diabetes has reached epidemic proportions worldwide. Currently, approximately 537 million adults (20–79 years) have diabetes, and the total number of people with diabetes is continuously increasing. Diabetes includes several subtypes. About 80% of all cases of diabetes are type 2 diabetes (T2D). T2D is a polygenic disease with an inheritance ranging from 30 to 70%. Genetic and environment/lifestyle factors, especially obesity and sedentary lifestyle, increase the risk of T2D. In this review, we discuss how studies on the genetics of diabetes started, how they expanded when genome-wide association studies and exome and whole-genome sequencing became available, and the current challenges in genetic studies of diabetes. T2D is heterogeneous with respect to clinical presentation, disease course, and response to treatment, and has several subgroups which differ in pathophysiology and risk of micro- and macrovascular complications. Currently, genetic studies of T2D focus on these subgroups to find the best diagnoses and treatments for these patients according to the principles of precision medicine.
Collapse
Affiliation(s)
- Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland
- Department of Medicine, Kuopio University Hospital, 70210 Kuopio, Finland
- Correspondence: ; Tel.: +358-40-672-3338
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland
| |
Collapse
|
41
|
Pipal KV, Mamtani M, Patel AA, Jaiswal SG, Jaisinghani MT, Kulkarni H. Susceptibility Loci for Type 2 Diabetes in the Ethnically Endogamous Indian Sindhi Population: A Pooled Blood Genome-Wide Association Study. Genes (Basel) 2022; 13:1298. [PMID: 35893037 PMCID: PMC9331904 DOI: 10.3390/genes13081298] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 12/10/2022] Open
Abstract
Type 2 diabetes (T2D) is a complex metabolic derangement that has a strong genetic basis. There is substantial population-specificity in the association of genetic variants with T2D. The Indian urban Sindhi population is at a high risk of T2D. The genetic basis of T2D in this population is unknown. We interrogated 28 pooled whole blood genomes of 1402 participants from the Diabetes In Sindhi Families In Nagpur (DISFIN) study using Illumina's Global Screening Array. From a total of 608,550 biallelic variants, 140 were significantly associated with T2D after adjusting for comorbidities, batch effects, pooling error, kinship status and pooling variation in a random effects multivariable logistic regression framework. Of the 102 well-characterized genes that these variants mapped onto, 70 genes have been previously reported to be associated with T2D to varying degrees with known functional relevance. Excluding open reading frames, intergenic non-coding elements and pseudogenes, our study identified 22 novel candidate genes in the Sindhi population studied. Our study thus points to the potential, interesting candidate genes associated with T2D in an ethnically endogamous population. These candidate genes need to be fully investigated in future studies.
Collapse
Affiliation(s)
- Kanchan V. Pipal
- Lata Medical Research Foundation, Nagpur 440002, India; (K.V.P.); (M.M.); (A.A.P.); (S.G.J.); (M.T.J.)
| | - Manju Mamtani
- Lata Medical Research Foundation, Nagpur 440002, India; (K.V.P.); (M.M.); (A.A.P.); (S.G.J.); (M.T.J.)
- M&H Research, LLC, San Antonio, TX 78249, USA
| | - Ashwini A. Patel
- Lata Medical Research Foundation, Nagpur 440002, India; (K.V.P.); (M.M.); (A.A.P.); (S.G.J.); (M.T.J.)
| | - Sujeet G. Jaiswal
- Lata Medical Research Foundation, Nagpur 440002, India; (K.V.P.); (M.M.); (A.A.P.); (S.G.J.); (M.T.J.)
| | - Manisha T. Jaisinghani
- Lata Medical Research Foundation, Nagpur 440002, India; (K.V.P.); (M.M.); (A.A.P.); (S.G.J.); (M.T.J.)
| | - Hemant Kulkarni
- Lata Medical Research Foundation, Nagpur 440002, India; (K.V.P.); (M.M.); (A.A.P.); (S.G.J.); (M.T.J.)
- M&H Research, LLC, San Antonio, TX 78249, USA
| |
Collapse
|
42
|
Overview of Transcriptomic Research on Type 2 Diabetes: Challenges and Perspectives. Genes (Basel) 2022; 13:genes13071176. [PMID: 35885959 PMCID: PMC9319211 DOI: 10.3390/genes13071176] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
Type 2 diabetes (T2D) is a common chronic disease whose etiology is known to have a strong genetic component. Standard genetic approaches, although allowing for the detection of a number of gene variants associated with the disease as well as differentially expressed genes, cannot fully explain the hereditary factor in T2D. The explosive growth in the genomic sequencing technologies over the last decades provided an exceptional impetus for transcriptomic studies and new approaches to gene expression measurement, such as RNA-sequencing (RNA-seq) and single-cell technologies. The transcriptomic analysis has the potential to find new biomarkers to identify risk groups for developing T2D and its microvascular and macrovascular complications, which will significantly affect the strategies for early diagnosis, treatment, and preventing the development of complications. In this article, we focused on transcriptomic studies conducted using expression arrays, RNA-seq, and single-cell sequencing to highlight recent findings related to T2D and challenges associated with transcriptome experiments.
Collapse
|
43
|
Jyothish UK, Pradeep L, Fernando R, Ravichandran K, Das S. Comparison of forearm muscle fatigue among apparently healthy young adults with and without diabetogenic genes. JOURNAL OF CURRENT RESEARCH IN SCIENTIFIC MEDICINE 2022. [DOI: 10.4103/jcrsm.jcrsm_67_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|