1
|
Moldovan RA, Hidalgo MR, Castañé H, Jiménez-Franco A, Joven J, Burks DJ, Galán A, García-García F. Landscape of sex differences in obesity and type 2 diabetes in subcutaneous adipose tissue: a systematic review and meta-analysis of transcriptomics studies. Metabolism 2025; 168:156241. [PMID: 40157598 DOI: 10.1016/j.metabol.2025.156241] [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: 12/04/2024] [Revised: 03/13/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
Obesity represents a significant risk factor in the development of type 2 diabetes (T2D), a chronic metabolic disorder characterized by elevated blood glucose levels, and a previous step for its development. Significant sex differences have been identified in the prevalence, development, and pathophysiology of obesity and T2D; however, the underlying molecular mechanisms remain unclear. This study aims to identify sex-specific signatures in obesity and T2D and enhance our understanding of the underlying mechanisms associated with sex differences by integrating expression data. We performed a systematic review and individual transcriptomic analysis of eight selected studies which included 302 subcutaneous adipose tissue samples. Then, we conducted different gene-level meta-analyses and functional characterizations for obesity and T2D separately, identifying common and sex-specific transcriptional profiles, many of which were previously associated with obesity or T2D. The obesity meta-analysis yielded nineteen differentially-expressed genes from a sex-specific perspective (e.g., SPATA18, KREMEN1, NPY4R, and PRM3), while a comparison of the expression profiles between sexes in T2D prompted the identification and validation of specific transcriptomic signatures in males (SAMD9, NBPF3, LDHD, and EHD3) and females (RETN, HEY1, PLPP2, and PM20D2). At the functional level, we highlighted the fundamental role of the Wnt pathway in the development of obesity and T2D in females, and the roles of mitochondrial damage and free fatty acids in males. Overall, our sex-specific meta-analyses supported the detection of differentially expressed genes in males and females associated with the development of obesity and further T2D development, emphasizing the relevance of sex-based information in biomedical data and opening new avenues for research.
Collapse
Affiliation(s)
- Roxana Andreea Moldovan
- Computational Biomedicine Laboratory, Príncipe Felipe Research Center (CIPF), 46012, Valencia, Spain; Department of Applied Statistics and Operations Research and Quality, Universitat Politècnica de València, Valencia 46022, Spain
| | - Marta R Hidalgo
- Computational Biomedicine Laboratory, Príncipe Felipe Research Center (CIPF), 46012, Valencia, Spain; Departament de Matemàtiques, Facultat de Matemàtiques, Universitat de València, 46010, Valencia, Spain
| | - Helena Castañé
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Andrea Jiménez-Franco
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Deborah J Burks
- Molecular Neuroendocrinology Laboratory, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Amparo Galán
- Molecular Neuroendocrinology Laboratory, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de València, 46010, Valencia, Spain.
| | - Francisco García-García
- Computational Biomedicine Laboratory, Príncipe Felipe Research Center (CIPF), 46012, Valencia, Spain.
| |
Collapse
|
2
|
Zhu H, Li B, Huang T, Wang B, Li S, Yu K, Cai L, Ye Y, Chen S, Zhu H, Xu J, Lu Q, Ji L. Update in the molecular mechanism and biomarkers of diabetic retinopathy. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167758. [PMID: 40048937 DOI: 10.1016/j.bbadis.2025.167758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 01/27/2025] [Accepted: 02/25/2025] [Indexed: 04/15/2025]
Abstract
Diabetic retinopathy (DR) is a serious complication of diabetes caused by long-term hyperglycemia that leads to microvascular and neuronal damage in the retina. The molecular mechanisms of DR involve oxidative stress, inflammatory responses, neurodegenerative changes, and vascular dysfunction triggered by hyperglycemia. Oxidative stress activates multiple metabolic pathways, such as the polyol, hexosamine, and protein kinase C (PKC) pathways, resulting in the production of, which in turn promote the formation of advanced glycation end products (AGEs). These pathways exacerbate vascular endothelial damage and the release of inflammatory factors, activating inflammatory signaling pathways such as the NF-κB pathway, leading to retinal cell damage and apoptosis. Additionally, DR involves neurodegenerative changes, including the activation of glial cells, neuronal dysfunction, and cell death. Research on the multiomics molecular markers of DR has revealed complex mechanisms at the genetic, epigenetic, and transcriptional levels. Genome-wide association studies (GWASs) have identified multiple genetic loci associated with DR that are involved in metabolic and inflammatory pathways. Noncoding RNAs, such as miRNAs, circRNAs, and lncRNAs, participate in the development of DR by regulating gene expression. Proteomic, metabolomic and lipidomic analyses have revealed specific proteins, metabolites and lipid changes associated with DR, providing potential biomarkers for the early diagnosis and treatment of this disease. This review provides a comprehensive perspective for understanding the molecular network of DR and facilitates the exploration of innovative therapeutic approaches.
Collapse
Affiliation(s)
- Hui Zhu
- Department of Ophthalmology, the Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, China
| | - Bingqi Li
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Tao Huang
- Department of Clinical Medicine, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Bin Wang
- Department of Clinical Medicine, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Shuoyu Li
- Department of Clinical Medicine, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Kuai Yu
- Department of Clinical Medicine, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Liwei Cai
- Department of Clinical Medicine, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Yuxin Ye
- Department of Clinical Medicine, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Siyuan Chen
- Department of Clinical Medicine, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Haotian Zhu
- Department of Clinical Medicine, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Jin Xu
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China; Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Qinkang Lu
- Department of Ophthalmology, the Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, China.
| | - Lindan Ji
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China.
| |
Collapse
|
3
|
Imamura M, Kadowaki T, Maeda S. Genetic studies on metabolic disorder-associated kidney diseases. Kidney Int 2025:S0085-2538(25)00321-7. [PMID: 40252921 DOI: 10.1016/j.kint.2025.01.042] [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: 09/08/2024] [Revised: 12/17/2024] [Accepted: 01/03/2025] [Indexed: 04/21/2025]
Abstract
Diabetic kidney disease (DKD) and obesity-related kidney diseases are the representative chronic kidney diseases related to metabolic disorders. Genome-wide association studies have been extensively performed, and a substantial number of confirmed loci have been identified to be associated with many common diseases or quantitative traits, including type 2 diabetes, obesity, and chronic kidney diseases. By contrast, genome-wide association studies for DKD have identified a limited number of susceptible loci, and the robust replication of these loci in independent studies has not yet been accomplished. As of 2024, no genome-wide association study has been reported on obesity-related kidney diseases. Therefore, the genetic studies on DKD or obesity-related kidney diseases have not provided satisfiable results. However, genetic correlation studies and Mendelian randomization studies, that were performed using multitrait genome-wide association study data, suggested that DKD, obesity-related kidney diseases, and obesity share common genetic mechanisms. Because obesity or overweight is a reversible condition, the effective interventions to reduce body weights might contribute to the prevention of the development of not only obesity-related kidney diseases, but also DKD or other types of chronic kidney diseases. Further genetic studies are necessary to understand the genetic architecture of DKD and obesity-related kidney diseases and should be expanded.
Collapse
Affiliation(s)
- Minako Imamura
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan; Division of Clinical Laboratory and Blood Transfusion, University of the Ryukyus Hospital, Okinawa, Japan.
| | | | - Shiro Maeda
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan; Division of Clinical Laboratory and Blood Transfusion, University of the Ryukyus Hospital, Okinawa, Japan.
| |
Collapse
|
4
|
Hashiba T, Sugawara Y, Hirakawa Y, Sato D, Inagi R, Nangaku M. Pathogenic variants prevalence patients with diabetic kidney disease in Japan: A descriptive study. J Diabetes Investig 2025. [PMID: 40197820 DOI: 10.1111/jdi.70041] [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: 11/07/2024] [Revised: 03/26/2025] [Accepted: 03/30/2025] [Indexed: 04/10/2025] Open
Abstract
AIMS/INTRODUCTION The impact of rare pathogenic variants on diabetic kidney disease (DKD) has not been investigated in detail. Previous studies have detected pathogenic variants in 22% of Caucasian patients with DKD; however, this proportion may vary depending on ethnicity and updates to the database. Therefore, we performed a whole-genome analysis of patients with DKD in type 2 diabetes mellitus in Japan, utilizing a recent database to investigate the prevalence of kidney-related pathogenic variants and describe the characteristics of these patients. MATERIALS AND METHODS Whole-genome sequencing was performed, and variants were analyzed following the GATK Best Practices. We extracted data on 790 genes associated with Mendelian kidney and genitourinary diseases. Pathogenic variants were defined based on the American College of Medical Genetics criteria, including both heterozygous and homozygous variants classified as pathogenic or likely pathogenic. RESULTS Among 79 participants, heterozygous pathogenic variants were identified in 27 (34.1%), a higher prevalence than previously reported. No homozygous pathogenic variants were detected. The identified heterozygous pathogenic variants were roughly divided into 23.7% related to glomerulopathy, 36.8% related to tubulointerstitial disease, 10.5% related to cystic disease/ciliopathy, and 28.9% related to others. Diagnostic variants were found in 10 patients (12.7%) in seven genes (ABCC6, ALPL, ASXL1, BMPR2, GCM2, PAX2, and WT1), all associated with autosomal dominant congenital disease. CONCLUSIONS This study identified a considerable number of patients with DKD in Japan who carried kidney-related heterozygous pathogenic variants. These findings suggest potential ethnic differences and highlight the impact of database updates on variant detection.
Collapse
Affiliation(s)
- Toyohiro Hashiba
- Division of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuka Sugawara
- Division of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yosuke Hirakawa
- Division of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Dai Sato
- Division of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Reiko Inagi
- Division of Chronic Kidney Disease Pathophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
5
|
Huang Y, Rao S, Sun X, Liu J. Advances in molecular epidemiology of diabetic retinopathy: from genomics to gut microbiomics. Mol Biol Rep 2025; 52:304. [PMID: 40080283 DOI: 10.1007/s11033-025-10383-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/25/2025] [Indexed: 03/15/2025]
Abstract
Diabetic retinopathy (DR) remains a prevalent complication of diabetes mellitus and a leading cause of blindness worldwide. The growing global diabetic population underscores the urgency to deepen our understanding of DR pathogenesis and develop effective prevention strategies. This review synthesizes recent advancements in molecular epidemiology, spanning genomics, epigenomics, transcriptomics, proteomics, metabolomics, and gut microbiomics, elucidating genetic underpinnings, epigenetic modifications, transcriptional alterations, protein biomarkers, metabolic disruptions, and gut microbiota dysbiosis associated with DR. Highlighted are key findings from genome-wide association studies (GWAS), Mendelian randomization (MR) studies, candidate gene association studies, and advancements in epigenetic mechanisms, revealing intricate disease pathways and potential therapeutic targets. Additionally, insights into altered metabolic profiles and gut microbiota compositions in DR underscore their emerging roles in disease progression and complications. Challenges and future directions in molecular epidemiological research are discussed to accelerate the translation of these findings into clinical applications for personalized DR management. The integration of multi-omics research findings may provide novel perspectives for facilitating rapid and accurate disease diagnosis, enabling dynamic disease monitoring, and advancing targeted therapeutic strategies.
Collapse
Affiliation(s)
- Yida Huang
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Suyun Rao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xufang Sun
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jun Liu
- Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| |
Collapse
|
6
|
Liu M, Luo T, Li R, Yin W, Yang F, Ge D, Liu N. Ent-pimarane and ent-kaurane diterpenoids from Siegesbeckiapubescens and their anti-endothelial damage effect in diabetic retinopathy. Chin J Nat Med 2025; 23:234-244. [PMID: 39986699 DOI: 10.1016/s1875-5364(25)60827-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/28/2024] [Accepted: 06/17/2024] [Indexed: 02/24/2025]
Abstract
Diabetic retinopathy, a prevalent and vision-threatening microvascular complication of diabetes mellitus, is the leading cause of blindness among middle-aged and elderly individuals. Natural diterpenoids isolated from Siegesbeckia pubescens demonstrate potent anti-inflammatory properties. This study aimed to identify novel bioactive diterpenoids from S. pubescens and investigate their effects on oxidative stress and inflammatory responses in diabetic retinopathy, both in vitro and in vivo. Three new ent-pimarane-type diterpenoids (1-3) and six known compounds (4-9) were isolated from the aerial parts of S. pubescens. Their structures were elucidated through spectroscopic data interpretation, and absolute configurations were determined by comparing calculated and experimental electronic circular dichroism (ECD) spectra. Among these compounds, 14β,16-epoxy-ent-3β,15α,19-trihydroxypimar-7-ene (5) exhibited the most potent protective effect against high glucose and interleukin-1β (IL-1β)-stimulated human retinal endothelial cells. Mechanistically, compound 5 promoted endothelial cell survival while ameliorating oxidative stress and inflammatory response in diabetic retinopathy, both in vivo and in vitro. These findings not only suggest that diterpenoids such as compound 5 are important anti-inflammatory constituents in S. pubescens, but also indicate that compound 5 may serve as a lead compound for preventing or treating vascular complications associated with diabetic retinopathy.
Collapse
Affiliation(s)
- Mengjia Liu
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Tingting Luo
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Rongxian Li
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Wenying Yin
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Fengying Yang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Di Ge
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China.
| | - Na Liu
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China.
| |
Collapse
|
7
|
Breeyear JH, Mitchell SL, Nealon CL, Hellwege JN, Charest B, Khakharia A, Halladay CW, Yang J, Garriga GA, Wilson OD, Basnet TB, Hung AM, Reaven PD, Meigs JB, Rhee MK, Sun Y, Lynch MG, Sobrin L, Brantley MA, Sun YV, Wilson PW, Iyengar SK, Peachey NS, Phillips LS, Edwards TL, Giri A. Development of electronic health record based algorithms to identify individuals with diabetic retinopathy. J Am Med Inform Assoc 2024; 31:2560-2570. [PMID: 39158361 PMCID: PMC11491608 DOI: 10.1093/jamia/ocae213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024] Open
Abstract
OBJECTIVES To develop, validate, and implement algorithms to identify diabetic retinopathy (DR) cases and controls from electronic health care records (EHRs). MATERIALS AND METHODS We developed and validated electronic health record (EHR)-based algorithms to identify DR cases and individuals with type I or II diabetes without DR (controls) in 3 independent EHR systems: Vanderbilt University Medical Center Synthetic Derivative (VUMC), the VA Northeast Ohio Healthcare System (VANEOHS), and Massachusetts General Brigham (MGB). Cases were required to meet 1 of the following 3 criteria: (1) 2 or more dates with any DR ICD-9/10 code documented in the EHR, (2) at least one affirmative health-factor or EPIC code for DR along with an ICD9/10 code for DR on a different day, or (3) at least one ICD-9/10 code for any DR occurring within 24 hours of an ophthalmology examination. Criteria for controls included affirmative evidence for diabetes as well as an ophthalmology examination. RESULTS The algorithms, developed and evaluated in VUMC through manual chart review, resulted in a positive predictive value (PPV) of 0.93 for cases and negative predictive value (NPV) of 0.91 for controls. Implementation of algorithms yielded similar metrics in VANEOHS (PPV = 0.94; NPV = 0.86) and lower in MGB (PPV = 0.84; NPV = 0.76). In comparison, the algorithm for DR implemented in Phenome-wide association study (PheWAS) in VUMC yielded similar PPV (0.92) but substantially reduced NPV (0.48). Implementation of the algorithms to the Million Veteran Program identified over 62 000 DR cases with genetic data including 14 549 African Americans and 6209 Hispanics with DR. CONCLUSIONS/DISCUSSION We demonstrate the robustness of the algorithms at 3 separate healthcare centers, with a minimum PPV of 0.84 and substantially improved NPV than existing automated methods. We strongly encourage independent validation and incorporation of features unique to each EHR to enhance algorithm performance for DR cases and controls.
Collapse
Affiliation(s)
- Joseph H Breeyear
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- VA Tennessee Valley Healthcare System (626), Nashville, TN 37212, United States
| | - Sabrina L Mitchell
- VA Tennessee Valley Healthcare System (626), Nashville, TN 37212, United States
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Cari L Nealon
- Eye Clinic, VA Northeast Ohio Healthcare System, Cleveland, OH 44106, United States
| | - Jacklyn N Hellwege
- VA Tennessee Valley Healthcare System (626), Nashville, TN 37212, United States
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN 37232, United States
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Brian Charest
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA 02111, United States
| | - Anjali Khakharia
- VA Atlanta Healthcare System, Decatur, GA 30033, United States
- Department of Medicine and Geriatrics, Emory University School of Medicine, Atlanta, GA 30307, United States
| | | | - Janine Yang
- Department of Ophthalmology, Mass Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, United States
| | - Gustavo A Garriga
- Division of Quantitative and Clinical Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Otis D Wilson
- VA Tennessee Valley Healthcare System (626), Nashville, TN 37212, United States
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Til B Basnet
- VA Tennessee Valley Healthcare System (626), Nashville, TN 37212, United States
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN 37232, United States
- Division of Quantitative and Clinical Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Adriana M Hung
- VA Tennessee Valley Healthcare System (626), Nashville, TN 37212, United States
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Peter D Reaven
- Phoenix VA Health Care System, Phoenix, AZ 85012, United States
- College of Medicine, University of Arizona, Phoenix, AZ 85721, United States
| | - James B Meigs
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, United States
- Department of Medicine, Harvard Medical School, Boston, MA 02115, United States
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA 02114, United States
| | - Mary K Rhee
- VA Atlanta Healthcare System, Decatur, GA 30033, United States
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30307, United States
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94305, United States
| | - Mary G Lynch
- VA Atlanta Healthcare System, Decatur, GA 30033, United States
| | - Lucia Sobrin
- Department of Ophthalmology, Mass Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, United States
| | - Milam A Brantley
- VA Tennessee Valley Healthcare System (626), Nashville, TN 37212, United States
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN 37232, United States
| | - Yan V Sun
- VA Atlanta Healthcare System, Decatur, GA 30033, United States
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA 30307, United States
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA 30307, United States
| | - Peter W Wilson
- VA Atlanta Healthcare System, Decatur, GA 30033, United States
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30307, United States
| | - Sudha K Iyengar
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH 44106, United States
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States
| | - Neal S Peachey
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH 44106, United States
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH 44106, United States
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, United States
| | - Lawrence S Phillips
- VA Atlanta Healthcare System, Decatur, GA 30033, United States
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30307, United States
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- VA Tennessee Valley Healthcare System (626), Nashville, TN 37212, United States
| | - Ayush Giri
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- VA Tennessee Valley Healthcare System (626), Nashville, TN 37212, United States
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN 37232, United States
- Division of Quantitative and Clinical Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| |
Collapse
|
8
|
Imamura M, Maeda S. Genetic studies of type 2 diabetes, and microvascular complications of diabetes. Diabetol Int 2024; 15:699-706. [PMID: 39469559 PMCID: PMC11512959 DOI: 10.1007/s13340-024-00727-4] [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: 01/16/2024] [Accepted: 04/24/2024] [Indexed: 10/30/2024]
Abstract
Genome-wide association studies (GWAS) have significantly advanced the identification of genetic susceptibility variants associated with complex diseases. As of 2023, approximately 800 variants predisposing individuals to the risk of type 2 diabetes (T2D) were identified through GWAS, and the majority of studies were predominantly conducted in European populations. Despite the shared nature of the majority of genetic susceptibility loci across diverse ethnic populations, GWAS in non-European populations, including Japanese and East Asian populations, have revealed population-specific T2D loci. Currently, polygenic risk scores (PRSs), encompassing millions of associated variants, can identify individuals with a higher T2D risk than the general population. However, GWAS focusing on microvascular complications of diabetes have identified a limited number of disease-susceptibility loci. Ongoing efforts are crucial to enhance the applicability of PRS for all ethnic groups and unravel the genetic architecture of microvascular complications of diabetes.
Collapse
Affiliation(s)
- Minako Imamura
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara-Cho, Okinawa 903-0215 Japan
- Division of Clinical Laboratory and Blood Transfusion, University of the Ryukyus Hospital, Nishihara-Cho, Okinawa 930-0215 Japan
| | - Shiro Maeda
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara-Cho, Okinawa 903-0215 Japan
- Division of Clinical Laboratory and Blood Transfusion, University of the Ryukyus Hospital, Nishihara-Cho, Okinawa 930-0215 Japan
| |
Collapse
|
9
|
Breeyear JH, Hellwege JN, Schroeder PH, House JS, Poisner HM, Mitchell SL, Charest B, Khakharia A, Basnet TB, Halladay CW, Reaven PD, Meigs JB, Rhee MK, Sun Y, Lynch MG, Bick AG, Wilson OD, Hung AM, Nealon CL, Iyengar SK, Rotroff DM, Buse JB, Leong A, Mercader JM, Sobrin L, Brantley MA, Peachey NS, Motsinger-Reif AA, Wilson PW, Sun YV, Giri A, Phillips LS, Edwards TL. Adaptive selection at G6PD and disparities in diabetes complications. Nat Med 2024; 30:2480-2488. [PMID: 38918629 PMCID: PMC11555759 DOI: 10.1038/s41591-024-03089-1] [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: 02/16/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024]
Abstract
Diabetes complications occur at higher rates in individuals of African ancestry. Glucose-6-phosphate dehydrogenase deficiency (G6PDdef), common in some African populations, confers malaria resistance, and reduces hemoglobin A1c (HbA1c) levels by shortening erythrocyte lifespan. In a combined-ancestry genome-wide association study of diabetic retinopathy, we identified nine loci including a G6PDdef causal variant, rs1050828 -T (Val98Met), which was also associated with increased risk of other diabetes complications. The effect of rs1050828 -T on retinopathy was fully mediated by glucose levels. In the years preceding diabetes diagnosis and insulin prescription, glucose levels were significantly higher and HbA1c significantly lower in those with versus without G6PDdef. In the Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial, participants with G6PDdef had significantly higher hazards of incident retinopathy and neuropathy. At the same HbA1c levels, G6PDdef participants in both ACCORD and the Million Veteran Program had significantly increased risk of retinopathy. We estimate that 12% and 9% of diabetic retinopathy and neuropathy cases, respectively, in participants of African ancestry are due to this exposure. Across continentally defined ancestral populations, the differences in frequency of rs1050828 -T and other G6PDdef alleles contribute to disparities in diabetes complications. Diabetes management guided by glucose or potentially genotype-adjusted HbA1c levels could lead to more timely diagnoses and appropriate intensification of therapy, decreasing the risk of diabetes complications in patients with G6PDdef alleles.
Collapse
Affiliation(s)
- Joseph H Breeyear
- Biostatistics and Computational Biology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Durham, NC, USA
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- VA Tennessee Valley Healthcare System (626), Nashville, TN, USA
| | - Jacklyn N Hellwege
- VA Tennessee Valley Healthcare System (626), Nashville, TN, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Philip H Schroeder
- Program in Metabolism, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - John S House
- Biostatistics and Computational Biology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Hannah M Poisner
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Sabrina L Mitchell
- VA Tennessee Valley Healthcare System (626), Nashville, TN, USA
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Brian Charest
- Massachusetts Veterans Epidemiology Research and Information Center, Boston, MA, USA
| | - Anjali Khakharia
- Atlanta VA Medical Center, Decatur, GA, USA
- Department of Medicine and Geriatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Til B Basnet
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Peter D Reaven
- Phoenix VA Health Care System, Phoenix, AZ, USA
- College of Medicine, University of Arizona, Phoenix, AZ, USA
| | - James B Meigs
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mary K Rhee
- Atlanta VA Medical Center, Decatur, GA, USA
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
- Veterans Administration Palo Alto Health Care System, Palo Alto, California, USA
| | | | - Alexander G Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Otis D Wilson
- VA Tennessee Valley Healthcare System (626), Nashville, TN, USA
| | - Adriana M Hung
- VA Tennessee Valley Healthcare System (626), Nashville, TN, USA
| | - Cari L Nealon
- Eye Clinic, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Department of Ophthalmology & Visual Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sudha K Iyengar
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Daniel M Rotroff
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Endocrinology and Metabolism Institute, Cleveland Clinic, Cleveland, OH, USA
- Center for Quantitative Metabolic Research, Cleveland Clinic, Cleveland, OH, USA
| | - John B Buse
- Division of Endocrinology & Metabolism, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Aaron Leong
- Program in Metabolism, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Josep M Mercader
- Program in Metabolism, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Lucia Sobrin
- Department of Ophthalmology, Mass Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Milam A Brantley
- VA Tennessee Valley Healthcare System (626), Nashville, TN, USA
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Neal S Peachey
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Alison A Motsinger-Reif
- Biostatistics and Computational Biology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Peter W Wilson
- Atlanta VA Medical Center, Decatur, GA, USA
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Yan V Sun
- Atlanta VA Medical Center, Decatur, GA, USA
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ayush Giri
- VA Tennessee Valley Healthcare System (626), Nashville, TN, USA.
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA.
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Lawrence S Phillips
- Atlanta VA Medical Center, Decatur, GA, USA
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- VA Tennessee Valley Healthcare System (626), Nashville, TN, USA.
| |
Collapse
|
10
|
Pei X, Huang D, Li Z. Genetic insights and emerging therapeutics in diabetic retinopathy: from molecular pathways to personalized medicine. Front Genet 2024; 15:1416924. [PMID: 39246572 PMCID: PMC11378321 DOI: 10.3389/fgene.2024.1416924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 08/12/2024] [Indexed: 09/10/2024] Open
Abstract
Diabetic retinopathy (DR) is a major complication of diabetes worldwide, significantly causing vision loss and blindness in working-age adults, and imposing a substantial socioeconomic burden globally. This review examines the crucial role of genetic factors in the development of DR and highlights the shift toward personalized treatment approaches. Advances in genetic research have identified specific genes and variations involved in angiogenesis, inflammation, and oxidative stress that increase DR susceptibility. Understanding these genetic markers enables early identification of at-risk individuals and the creation of personalized treatment plans. Incorporating these genetic insights, healthcare providers can develop early intervention strategies and tailored treatment plans to improve patient outcomes and minimize side effects. This review emphasizes the transformative potential of integrating genetic information into clinical practice, marking a paradigm shift in DR management and advancing toward a more personalized and effective healthcare model.
Collapse
Affiliation(s)
- Xiaoting Pei
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Duliurui Huang
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhijie Li
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Yu J, Brelen ME, Chan CKM, Chen ZJ, Mohamed S, Yam JC, Lam DSC, Pang CP, Tham CC, Chen LJ. Genetic association of TIE2 with diabetic retinopathy and diabetic macular edema. Asia Pac J Ophthalmol (Phila) 2024; 13:100068. [PMID: 38750959 DOI: 10.1016/j.apjo.2024.100068] [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: 04/13/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 09/18/2024] Open
Abstract
PURPOSE To evaluate the associations of the TIE2 gene with diabetic retinopathy (DR) and diabetic macular edema (DME). METHODS This study included a Chinese cohort of 285 non-proliferative DR patients and 433 healthy controls. The DR patients were classified further into those with or without DME. Thirty haplotype-tagging single-nucleotide polymorphisms (SNPs) in TIE2 were genotyped using TaqMan technology. Associations of DR and subtypes were analyzed by logistic regression adjusted for age and sex. Stratification association analysis by sex was performed. RESULTS TIE2 rs625767 showed a nominal but consistent association with DR [odds ratio (OR) = 0.71, P = 0.005] and subtypes (DR without DME: OR = 0.69, P = 0.016; DME: OR = 0.73, P = 0.045). SNP rs652010 was consistently associated with overall DR (OR = 0.74, P = 0.011) and DR without DME (OR = 0.70, P = 0.016), but not with DME. Moreover, SNPs rs669441, rs10967760, rs549099 and rs639225 showed associations with overall DR, whilst rs17761403, rs664461 and rs1413825 with DR without DME. In stratification analysis, three SNPs, rs625767 (OR = 0.62, P = 0.005), rs669441 (OR = 0.63, P = 0.006) and rs652010 (OR = 0.64, P = 0.007), were associated with DR in females, but not in males. Moreover, one haplotype T-T defined by rs625767 and rs669441 was significantly associated with DR in females only. CONCLUSIONS This study revealed TIE2 as a susceptibility gene for DR and DME in Chinese, with a sex-specific association in females. Further validation should be warranted.
Collapse
Affiliation(s)
- Jun Yu
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Marten E Brelen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong; Hong Kong Eye Hospital, Hong Kong; Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong
| | - Carmen K M Chan
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong; Hong Kong Eye Hospital, Hong Kong
| | - Zhen Ji Chen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Shaheeda Mohamed
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong; Hong Kong Eye Hospital, Hong Kong
| | - Jason C Yam
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong; Hong Kong Eye Hospital, Hong Kong
| | - Dennis S C Lam
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong; C-MER Dennis Lam & Partners Eye Center, C-MER International Eye Care Group, Hong Kong
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Clement C Tham
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong; Hong Kong Eye Hospital, Hong Kong
| | - Li Jia Chen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong; Hong Kong Eye Hospital, Hong Kong; Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong.
| |
Collapse
|
12
|
Yang WQ, Ge JY, Zhang X, Zhu WY, Lin L, Shi Y, Xu B, Liu RJ. THUMPD2 catalyzes the N2-methylation of U6 snRNA of the spliceosome catalytic center and regulates pre-mRNA splicing and retinal degeneration. Nucleic Acids Res 2024; 52:3291-3309. [PMID: 38165050 PMCID: PMC11014329 DOI: 10.1093/nar/gkad1243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024] Open
Abstract
The mechanisms by which the relatively conserved spliceosome manages the enormously large number of splicing events that occur in humans (∼200 000 versus ∼300 in yeast) are poorly understood. Here, we show deposition of one RNA modification-N2-methylguanosine (m2G) on the G72 of U6 snRNA (the catalytic center of the spliceosome) promotes efficient pre-mRNA splicing activity in human cells. This modification was identified to be conserved among vertebrates. Further, THUMPD2 was demonstrated as the methyltransferase responsible for U6 m2G72 by explicitly recognizing the U6-specific sequences and structural elements. The knock-out of THUMPD2 eliminated U6 m2G72 and impaired the pre-mRNA splicing activity, resulting in thousands of changed alternative splicing events of endogenous pre-mRNAs in human cells. Notably, the aberrantly spliced pre-mRNA population elicited the nonsense-mediated mRNA decay pathway. We further show that THUMPD2 was associated with age-related macular degeneration and retinal function. Our study thus demonstrates how an RNA epigenetic modification of the major spliceosome regulates global pre-mRNA splicing and impacts physiology and disease.
Collapse
Affiliation(s)
- Wen-Qing Yang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jian-Yang Ge
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaofeng Zhang
- Division of Reproduction and Genetics, The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, China
| | - Wen-Yu Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Lin Lin
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yigong Shi
- Institute of Biology, Westlake Institute for Advanced Study, Westlake University, Hangzhou 310064,Zhejiang Province, China
| | - Beisi Xu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ru-Juan Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
13
|
Breeyear JH, Mitchell SL, Nealon CL, Hellwege JN, Charest B, Khakharia A, Halladay CW, Yang J, Garriga GA, Wilson OD, Basnet TB, Hung AM, Reaven PD, Meigs JB, Rhee MK, Sun Y, Lynch MG, Sobrin L, Brantley MA, Sun YV, Wilson PW, Iyengar SK, Peachey NS, Phillips LS, Edwards TL, Giri A. Development of Portable Electronic Health Record Based Algorithms to Identify Individuals with Diabetic Retinopathy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.11.10.23298311. [PMID: 38014167 PMCID: PMC10680882 DOI: 10.1101/2023.11.10.23298311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Objectives To develop, validate and implement algorithms to identify diabetic retinopathy (DR) cases and controls from electronic health care records (EHR)s. Methods : We developed and validated EHR-based algorithms to identify DR cases and individuals with type I or II diabetes without DR (controls) in three independent EHR systems: Vanderbilt University Medical Center Synthetic Derivative (VUMC), the VA Northeast Ohio Healthcare System (VANEOHS), and Massachusetts General Brigham (MGB). Cases were required to meet one of three criteria: 1) two or more dates with any DR ICD-9/10 code documented in the EHR, or 2) at least one affirmative health-factor or EPIC code for DR along with an ICD9/10 code for DR on a different day, or 3) at least one ICD-9/10 code for any DR occurring within 24 hours of an ophthalmology exam. Criteria for controls included affirmative evidence for diabetes as well as an ophthalmology exam. Results The algorithms, developed and evaluated in VUMC through manual chart review, resulted in a positive predictive value (PPV) of 0.93 for cases and negative predictive value (NPV) of 0.97 for controls. Implementation of algorithms yielded similar metrics in VANEOHS (PPV=0.94; NPV=0.86) and lower in MGB (PPV=0.84; NPV=0.76). In comparison, use of DR definition as implemented in Phenome-wide association study (PheWAS) in VUMC, yielded similar PPV (0.92) but substantially reduced NPV (0.48). Implementation of the algorithms to the Million Veteran Program identified over 62,000 DR cases with genetic data including 14,549 African Americans and 6,209 Hispanics with DR. Conclusions/Discussion We demonstrate the robustness of the algorithms at three separate health-care centers, with a minimum PPV of 0.84 and substantially improved NPV than existing high-throughput methods. We strongly encourage independent validation and incorporation of features unique to each EHR to enhance algorithm performance for DR cases and controls.
Collapse
|
14
|
Sienkiewicz-Szłapka E, Fiedorowicz E, Król-Grzymała A, Kordulewska N, Rozmus D, Cieślińska A, Grzybowski A. The Role of Genetic Polymorphisms in Diabetic Retinopathy: Narrative Review. Int J Mol Sci 2023; 24:15865. [PMID: 37958858 PMCID: PMC10650381 DOI: 10.3390/ijms242115865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Diabetic retinopathy (DR) is renowned as a leading cause of visual loss in working-age populations with its etiopathology influenced by the disturbance of biochemical metabolic pathways and genetic factors, including gene polymorphism. Metabolic pathways considered to have an impact on the development of the disease, as well as genes and polymorphisms that can affect the gene expression, modify the quantity and quality of the encoded product (protein), and significantly alter the metabolic pathway and its control, and thus cause changes in the functioning of metabolic pathways. In this article, the screening of chromosomes and the most important genes involved in the etiology of diabetic retinopathy is presented. The common databases with manuscripts published from January 2000 to June 2023 have been taken into consideration and chosen. This article indicates the role of specific genes in the development of diabetic retinopathy, as well as polymorphic changes within the indicated genes that may have an impact on exacerbating the symptoms of the disease. The collected data will allow for a broader look at the disease and help to select candidate genes that can become markers of the disease.
Collapse
Affiliation(s)
- Edyta Sienkiewicz-Szłapka
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury, 10-719 Olsztyn, Poland; (E.S.-S.); (E.F.); (A.K.-G.); (N.K.); (D.R.)
| | - Ewa Fiedorowicz
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury, 10-719 Olsztyn, Poland; (E.S.-S.); (E.F.); (A.K.-G.); (N.K.); (D.R.)
| | - Angelika Król-Grzymała
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury, 10-719 Olsztyn, Poland; (E.S.-S.); (E.F.); (A.K.-G.); (N.K.); (D.R.)
| | - Natalia Kordulewska
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury, 10-719 Olsztyn, Poland; (E.S.-S.); (E.F.); (A.K.-G.); (N.K.); (D.R.)
| | - Dominika Rozmus
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury, 10-719 Olsztyn, Poland; (E.S.-S.); (E.F.); (A.K.-G.); (N.K.); (D.R.)
| | - Anna Cieślińska
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury, 10-719 Olsztyn, Poland; (E.S.-S.); (E.F.); (A.K.-G.); (N.K.); (D.R.)
| | - Andrzej Grzybowski
- Institute for Research in Ophthalmology, Foundation for Ophthalmology Development, Gorczyczewskiego 2/3, 61-553 Poznań, Poland;
| |
Collapse
|
15
|
Yu J, Yan Y, Hua C, Song H. EHD3 promotes gastric cancer progression via Wnt/β-catenin/EMT pathway and associates with clinical prognosis and immune infiltration. Am J Cancer Res 2023; 13:4401-4417. [PMID: 37818061 PMCID: PMC10560930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/21/2023] [Indexed: 10/12/2023] Open
Abstract
Gastric cancer (GC) shows high levels of heterogeneity and predicts a poor prognosis. The expressions of EHD3 are found to be misregulated in a number of tumors. However, the clinical significance and potential function of EHD3 expression in GC patients remain unknown. In this study, we found that EHD3 expression was distinctly increased in GC specimens and cell lines in both TCGA datasets and our cohort. High levels of EHD3 expression were linked to worse outcomes for patients with GC in clinical tests. Nomogram based on multivariate assays displayed good predictive accuracy for GC patients, as evidenced by C-indices and calibration graphs. Low levels of EHD3 mRNA were discovered in GC tissues due to EHD3 methylation's negative regulation of EHD3. In addition, EHD3 was observed to be related to several immune cells and might play a role in successful immunotherapy. Functionally, it was verified that knockdown of EHD3 remarkably suppressed the proliferation, migration and invasion of GC cells in vitro and in vivo. Results of Western blot confirmed that knockdown of EHD3 suppressed the expressions of β-catenin, MMP-9, and N-cadherin, while promoting the expression of E-cadherin. Overall, this research identified a novel GC-related gene EHD3 which might be a novel prognostic biomarker involved in tumor microenvironment. EHD3 promoted the proliferation and metastasis of GC cells through influencing the Wnt/β-catenin/EMT signaling pathway, suggesting it as a novel treatment target for GC patients.
Collapse
Affiliation(s)
- Jing Yu
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University Zhengzhou, Henan, China
| | - Yunmeng Yan
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University Zhengzhou, Henan, China
| | - Chunlan Hua
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University Zhengzhou, Henan, China
| | - Hairong Song
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University Zhengzhou, Henan, China
| |
Collapse
|
16
|
Shojima N, Yamauchi T. Progress in genetics of type 2 diabetes and diabetic complications. J Diabetes Investig 2023; 14:503-515. [PMID: 36639962 PMCID: PMC10034958 DOI: 10.1111/jdi.13970] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 01/15/2023] Open
Abstract
Type 2 diabetes results from a complex interaction between genetic and environmental factors. Precision medicine for type 2 diabetes using genetic data is expected to predict the risk of developing diabetes and complications and to predict the effects of medications and life-style intervention more accurately for individuals. Genome-wide association studies (GWAS) have been conducted in European and Asian populations and new genetic loci have been identified that modulate the risk of developing type 2 diabetes. Novel loci were discovered by GWAS in diabetic complications with increasing sample sizes. Large-scale genome-wide association analysis and polygenic risk scores using biobank information is making it possible to predict the development of type 2 diabetes. In the ADVANCE clinical trial of type 2 diabetes, a multi-polygenic risk score was useful to predict diabetic complications and their response to treatment. Proteomics and metabolomics studies have been conducted and have revealed the associations between type 2 diabetes and inflammatory signals and amino acid synthesis. Using multi-omics analysis, comprehensive molecular mechanisms have been elucidated to guide the development of targeted therapy for type 2 diabetes and diabetic complications.
Collapse
Affiliation(s)
- Nobuhiro Shojima
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
17
|
Yu X, Rong S. Genome-Wide Associations and Confirmatory Meta-Analyses in Diabetic Retinopathy. Genes (Basel) 2023; 14:653. [PMID: 36980925 PMCID: PMC10048213 DOI: 10.3390/genes14030653] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/21/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
The present study aimed to summarize and validate the genomic association signals for diabetic retinopathy (DR), proliferative DR, and diabetic macular edema/diabetic maculopathy. A systematic search of the genome-wide association study (GWAS) catalog and PubMed/MELINE databases was conducted to curate a comprehensive list of significant GWAS discoveries. The top signals were then subjected to meta-analysis using established protocols. The results indicate the need for improved consensus among DR GWASs, highlighting the importance of validation efforts. A subsequent meta-analysis confirmed the association of two SNPs, rs4462262 (ZWINT-MRPS35P3) (odds ratio = 1.38, p = 0.001) and rs7903146 (TCF7L2) (odd ratio = 1.30, p < 0.001), with DR in independent populations, strengthening the evidence of their true association. We also compiled a list of candidate SNPs for further validation. This study highlights the importance of consistent validation and replication efforts in the field of DR genetics. The two identified gene loci warrant further functional investigation to understand their role in DR pathogenesis.
Collapse
Affiliation(s)
- Xinting Yu
- Department of Medicine, Brigham and Women’s Hospital, Mass General Brigham, Harvard Medical School, Boston, MA 02445, USA
| | - Shisong Rong
- Department of Ophthalmology, Massachusetts Eye and Ear, Mass General Brigham, Harvard Medical School, Boston, MA 02445, USA
| |
Collapse
|
18
|
Fickweiler W, Mitzner M, Jacoba CMP, Sun JK. Circulatory Biomarkers and Diabetic Retinopathy in Racial and Ethnic Populations. Semin Ophthalmol 2023:1-11. [PMID: 36710371 DOI: 10.1080/08820538.2023.2168488] [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] [Indexed: 01/31/2023]
Abstract
Clinical staging systems for diagnosis and treatment of diabetic retinopathy (DR) must closely relate to endpoints that are both relevant for patients and feasible for physicians to implement. Current DR staging systems for clinical eye care and research provide detailed phenotypic characterization to predict patient outcomes in diabetes but have limitations. Biochemical biomarkers provide a rich pool of potential candidates for new DR staging systems that can be readily measured in accessible fluids. Circulating biomarkers that are specific to the retina and relate to angiogenesis and inflammation have been suggested as relevant for DR. Although there is a lack of multi-ethnic studies evaluating circulatory biomarkers in DR, variability in circulatory biomarkers have been reported in people from different ethnic and racial backgrounds. Therefore, there is a need for future studies to evaluate individual or combinations of biomarkers in diverse populations with DR from different ethnic and racial backgrounds.
Collapse
Affiliation(s)
- Ward Fickweiler
- Research Division, Joslin Diabetes Center, Boston, MA, USA.,Beetham Eye Institute, Joslin Diabetes Center, Boston, MA, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Margalit Mitzner
- Research Division, Joslin Diabetes Center, Boston, MA, USA.,Beetham Eye Institute, Joslin Diabetes Center, Boston, MA, USA
| | - Cris Martin P Jacoba
- Beetham Eye Institute, Joslin Diabetes Center, Boston, MA, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Jennifer K Sun
- Research Division, Joslin Diabetes Center, Boston, MA, USA.,Beetham Eye Institute, Joslin Diabetes Center, Boston, MA, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Rodrigues Oliveira SM, Rebocho A, Ahmadpour E, Nissapatorn V, de Lourdes Pereira M. Type 1 Diabetes Mellitus: A Review on Advances and Challenges in Creating Insulin Producing Devices. MICROMACHINES 2023; 14:151. [PMID: 36677212 PMCID: PMC9867263 DOI: 10.3390/mi14010151] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/25/2022] [Accepted: 12/30/2022] [Indexed: 06/17/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is the most common autoimmune chronic disease in young patients. It is caused by the destruction of pancreatic endocrine β-cells that produce insulin in specific areas of the pancreas, known as islets of Langerhans. As a result, the body becomes insulin deficient and hyperglycemic. Complications associated with diabetes are life-threatening and the current standard of care for T1DM consists still of insulin injections. Lifesaving, exogenous insulin replacement is a chronic and costly burden of care for diabetic patients. Alternative therapeutic options have been the focus in these fields. Advances in molecular biology technologies and in microfabrication have enabled promising new therapeutic options. For example, islet transplantation has emerged as an effective treatment to restore the normal regulation of blood glucose in patients with T1DM. However, this technique has been hampered by obstacles, such as limited islet availability, extensive islet apoptosis, and poor islet vascular engraftment. Many of these unsolved issues need to be addressed before a potential cure for T1DM can be a possibility. New technologies like organ-on-a-chip platforms (OoC), multiplexed assessment tools and emergent stem cell approaches promise to enhance therapeutic outcomes. This review will introduce the disorder of type 1 diabetes mellitus, an overview of advances and challenges in the areas of microfluidic devices, monitoring tools, and prominent use of stem cells, and how they can be linked together to create a viable model for the T1DM treatment. Microfluidic devices like OoC platforms can establish a crucial platform for pathophysiological and pharmacological studies as they recreate the pancreatic environment. Stem cell use opens the possibility to hypothetically generate a limitless number of functional pancreatic cells. Additionally, the integration of stem cells into OoC models may allow personalized or patient-specific therapies.
Collapse
Affiliation(s)
- Sonia M. Rodrigues Oliveira
- HMRI-Hunter Medical Research Institute, New Lambton, NSW 2305, Australia
- CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - António Rebocho
- Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ehsan Ahmadpour
- Drug Applied Research Center, Department of Parasitology and Mycology, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
- Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| | - Veeranoot Nissapatorn
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
- School of Allied Health Sciences, Southeast Asia Water Team (SEAWater Team), World Union for Herbal Drug Discovery (WUHeDD), Research Excellence Center for Innovation and Health Products, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Maria de Lourdes Pereira
- CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
20
|
Gurung RL, Burdon KP, McComish BJ. A Guide to Genome-Wide Association Study Design for Diabetic Retinopathy. Methods Mol Biol 2023; 2678:49-89. [PMID: 37326705 DOI: 10.1007/978-1-0716-3255-0_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Diabetic retinopathy (DR) is the most common microvascular complication related to diabetes. There is evidence that genetics play an important role in DR pathogenesis, but the complexity of the disease makes genetic studies a challenge. This chapter is a practical overview of the basic steps for genome-wide association studies with respect to DR and its associated traits. Also described are approaches that can be adopted in future DR studies. This is intended to serve as a guide for beginners and to provide a framework for further in-depth analysis.
Collapse
Affiliation(s)
- Rajya L Gurung
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.
| | - Kathryn P Burdon
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.
| | - Bennet J McComish
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
21
|
Dissecting the Pathogenesis of Diabetic Retinopathy Based on the Biological ceRNA Network and Genome Variation Disturbance. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:9833142. [PMID: 34707685 PMCID: PMC8545528 DOI: 10.1155/2021/9833142] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/29/2021] [Indexed: 12/23/2022]
Abstract
Background Diabetic retinopathy (DR) is the most important manifestation of diabetic microangiopathy. It is essential to explore the gene regulatory relationship and genomic variation disturbance of biological networks in DR progression. Methods In this study, we constructed a comprehensive lncRNA-mRNA ceRNA network of DR procession (CLMN) and explored its topological characteristics. Results Modular and functional analysis indicated that the organization of CLMN performed fundamental and specific functions in diabetes and DR pathology. The differential expression of hub ceRNA nodes and positive correlation reveals the highly connected ceRNA regulation and important roles in the regulating of DR pathology. A large proportion of SNPs in the TFBS, DHS, and enhancer regions of lncRNAs will affect lncRNA transcription and further cause expression variation. Some SNPs were found to disrupt the lncRNA functional elements such as miRNA target binding sites. These results indicate the complex nature of genotypic effects in the disturbing of CLMN and further contribute to gene expression variation and different disease phenotypes. Conclusion The identification of individual genomic variations and analysis of biological network disturbance by these genomic variations will help provide more personalized treatment plans and promote the development of precision medicine for DR.
Collapse
|
22
|
Genetics of Diabetic Retinopathy, a Leading Cause of Irreversible Blindness in the Industrialized World. Genes (Basel) 2021; 12:genes12081200. [PMID: 34440374 PMCID: PMC8394456 DOI: 10.3390/genes12081200] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/29/2021] [Accepted: 07/29/2021] [Indexed: 12/14/2022] Open
Abstract
Diabetic retinopathy (DR) is a chronic complication of diabetes and a leading cause of blindness in the industrialized world. Traditional risk factors, such as glycemic control and duration of diabetes, are unable to explain why some individuals remain protected while others progress to a more severe form of the disease. Differences are also observed in DR heritability as well as the response to anti-vascular endothelial growth factor (VEGF) treatment. This review discusses various aspects of genetics in DR to shed light on DR pathogenesis and treatment. First, we discuss the global burden of DR followed by a discussion on disease pathogenesis as well as the role genetics plays in the prevalence and progression of DR. Subsequently, we provide a review of studies related to DR’s genetic contribution, such as candidate gene studies, linkage studies, and genome-wide association studies (GWAS) as well as other clinical and meta-analysis studies that have identified putative candidate genes. With the advent of newer cutting-edge technologies, identifying the genetic components in DR has played an important role in understanding DR incidence, progression, and response to treatment, thereby developing newer therapeutic targets and therapies.
Collapse
|