1
|
Tariq Z, Abusnana S, Mussa BM, Zakaria H. New insights on genetic background of major diabetic vascular complications. Diabetol Metab Syndr 2024; 16:243. [PMID: 39375805 PMCID: PMC11457557 DOI: 10.1186/s13098-024-01473-y] [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: 08/26/2024] [Accepted: 09/21/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND By 2045, it is expected that 693 million individuals worldwide will have diabetes and with greater risk of morbidity, mortality, loss of vision, renal failure, and a decreased quality of life due to the devastating effects of macro- and microvascular complications. As such, clinical variables and glycemic control alone cannot predict the onset of vascular problems. An increasing body of research points to the importance of genetic predisposition in the onset of both diabetes and diabetic vascular complications. OBJECTIVES Purpose of this article is to review these approaches and narrow down genetic findings for Diabetic Mellitus and its consequences, highlighting the gaps in the literature necessary to further genomic discovery. MATERIAL AND METHODS In the past, studies looking for genetic risk factors for diabetes complications relied on methods such as candidate gene studies, which were rife with false positives, and underpowered genome-wide association studies, which were constrained by small sample sizes. RESULTS The number of genetic findings for diabetes and diabetic complications has over doubled due to the discovery of novel genomics data, including bioinformatics and the aggregation of global cohort studies. Using genetic analysis to determine whether diabetes individuals are at the most risk for developing diabetic vascular complications (DVC) might lead to the development of more accurate early diagnostic biomarkers and the customization of care plans. CONCLUSIONS A newer method that uses extensive evaluation of single nucleotide polymorphisms (SNP) in big datasets is Genome-Wide Association Studies (GWAS).
Collapse
Affiliation(s)
- Zuira Tariq
- Diabetes and Endocrinology Department, University Hospital Sharjah, P.O. Box: 27272, Sharjah, United Arab Emirates
| | - Salah Abusnana
- Diabetes and Endocrinology Department, University Hospital Sharjah, P.O. Box: 27272, Sharjah, United Arab Emirates.
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
| | - Bashair M Mussa
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Hala Zakaria
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
2
|
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
|
3
|
Tang S, An X, Sun W, Zhang Y, Yang C, Kang X, Sun Y, Jiang L, Zhao X, Gao Q, Ji H, Lian F. Parallelism and non-parallelism in diabetic nephropathy and diabetic retinopathy. Front Endocrinol (Lausanne) 2024; 15:1336123. [PMID: 38419958 PMCID: PMC10899692 DOI: 10.3389/fendo.2024.1336123] [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: 11/10/2023] [Accepted: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
Diabetic nephropathy (DN) and diabetic retinopathy (DR), as microvascular complications of diabetes mellitus, are currently the leading causes of end-stage renal disease (ESRD) and blindness, respectively, in the adult working population, and they are major public health problems with social and economic burdens. The parallelism between the two in the process of occurrence and development manifests in the high overlap of disease-causing risk factors and pathogenesis, high rates of comorbidity, mutually predictive effects, and partial concordance in the clinical use of medications. However, since the two organs, the eye and the kidney, have their unique internal environment and physiological processes, each with specific influencing molecules, and the target organs have non-parallelism due to different pathological changes and responses to various influencing factors, this article provides an overview of the parallelism and non-parallelism between DN and DR to further recognize the commonalities and differences between the two diseases and provide references for early diagnosis, clinical guidance on the use of medication, and the development of new drugs.
Collapse
Affiliation(s)
- Shanshan Tang
- College of Traditional Chinese Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Xuedong An
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenjie Sun
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuehong Zhang
- Fangshan Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Cunqing Yang
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaomin Kang
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuting Sun
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Linlin Jiang
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuefei Zhao
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Qing Gao
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Hangyu Ji
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengmei Lian
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Sikka R, Raina P, Soni R, Gupta H, Bhanwer AJS. Genomic profile of diabetic retinopathy in a north indian cohort. Mol Biol Rep 2023; 50:9769-9778. [PMID: 37700140 DOI: 10.1007/s11033-023-08772-z] [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: 04/20/2023] [Accepted: 08/18/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND Diabetic Retinopathy (DR) is one of the major microvascular complications of diabetes. Being a complex disease, it is important to delineate the genetic and environmental factors that influence the susceptibility to DR in a population. Therefore, the present study was designed to investigate the role of genetic and lifestyle risk factors associated with DR susceptibility in a North-Indian population. METHODS A total of 848 subjects were enrolled, comprising of DR cases (n = 414) and healthy controls (n = 434). The Sequenom MassARRAY technology was used to perform target genome analysis of 111 SNPs across 57 candidate genes and 14 intergenic region SNPs that are involved in the metabolic pathways associated with type 2 diabetes (T2D) and DR. Allele, genotype and haplotype frequencies were determined and compared among cases and controls. Logistic regression models were used to determine genotype-phenotype and phenotype-phenotype correlations. RESULTS The strongest association was observed with TCF7L2 rs12255372 T allele [p < 0.0001; odds ratio (OR) = 1.81 (1.44-2.27)] and rs11196205 C allele [p < 0.0008; OR = 1.62 (1.32-1.99)]. Genotype-phenotype and phenotype-phenotype correlations were found in the present study. CONCLUSION Our study provides strong evidence of association between the TCF7L2 variants and DR susceptibility.
Collapse
Affiliation(s)
- Ruhi Sikka
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India.
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, UP, India.
| | - Priyanka Raina
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
- Mosaic Therapeutics, Wellcome Genome Campus, Cambridge, UK
| | | | - Himanshu Gupta
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, UP, India
| | - A J S Bhanwer
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
- Department of Genetics, Guru Ram Das University of Health Sciences, Amritsar, Punjab, India
| |
Collapse
|
5
|
Berrabeh S, Elmehraoui O, Benouda S, Assarrar I, Rouf S, Latrech H. Prevalence and Risk Factors of Retinopathy in Type 1 Diabetes: A Cross-Sectional Study. Cureus 2023; 15:e47993. [PMID: 38034238 PMCID: PMC10686625 DOI: 10.7759/cureus.47993] [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] [Accepted: 10/13/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction Diabetic retinopathy (DR) is a severe complication of diabetes. It remains a major cause of visual impairment and blindness, especially in young people. It is a silent affection that only becomes symptomatic at the onset of complications. Our study aimed to estimate the prevalence of retinopathy in patients with type 1 diabetes mellitus (T1DM) and evaluate the associated risk factors in our population. Materials and methods A descriptive and analytical study, with a cross-sectional study involving 359 patients with type 1 diabetes, was followed up in the Department of Endocrinology, Diabetology, and Nutrition of the University Hospital Center Mohammed VI Oujda, Morocco. Data were collected from medical records and analyzed by binary logistic regression using IBM Corp. Released 2012. IBM SPSS Statistics for Windows, Version 21.0. Armonk, NY: IBM Corp. Results The average age of our patients was 24.2 ± 11.4 years. The mean duration of diabetes was 11.8±4.4 years. The average glycated hemoglobin (HbA1c) at admission was 10.1 ± 2.4%. DR was found in 30% of patients, including 28.6% with minimal non-proliferative diabetic retinopathy (NPDR), 19.1% with moderate NPDR, 19.1% with severe NPDR, and 33.3% with proliferative DR. Patients with diabetic retinopathy appear to have a longer duration of diabetes (13.05±9.05 vs. 10.6±8.07 years). The longer duration of diabetes, neuropathy, and nephropathy was significantly associated with diabetic retinopathy (p=0.02, p=0.002, and p=0.0001, respectively). Conclusion The frequency of diabetic retinopathy increases with age, poor glycemic control, and the duration of diabetes. Therefore, cooperation between diabetologists and ophthalmologists is essential for making an early diagnosis and providing early treatment.
Collapse
Affiliation(s)
- Soumiya Berrabeh
- Department of Endocrinology-Diabetology and Nutrition, Mohammed VI University Hospital Center, Oujda, MAR
- Department of Endocrinology-Diabetology and Nutrition, Faculty of Medicine and Pharmacy, Mohamed First University, Oujda, MAR
| | - Ouafae Elmehraoui
- Department of Endocrinology-Diabetology and Nutrition, Mohammed VI University Hospital Center, Oujda, MAR
- Department of Endocrinology-Diabetology and Nutrition, Faculty of Medicine and Pharmacy, Mohamed First University, Oujda, MAR
| | - Siham Benouda
- Department of Endocrinology-Diabetology and Nutrition, Mohammed VI University Hospital Center, Oujda, MAR
- Department of Endocrinology-Diabetology and Nutrition, Faculty of Medicine and Pharmacy, Mohamed First University, Oujda, MAR
| | - Imane Assarrar
- Department of Endocrinology-Diabetology and Nutrition, Mohammed VI University Hospital Center, Oujda, MAR
- Department of Endocrinology-Diabetology and Nutrition, Faculty of Medicine and Pharmacy, Mohamed First University, Oujda, MAR
| | - Siham Rouf
- Department of Endocrinology-Diabetology and Nutrition, Mohammed VI University Hospital Center, Oujda, MAR
- Department of Endocrinology-Diabetology and Nutrition, Faculty of Medicine and Pharmacy, Mohamed First University, Oujda, MAR
- Laboratory of Epidemiology, Clinical Research and Public Health, Faculty of Medicine and Pharmacy, Mohammed First University, Oujda, MAR
| | - Hanane Latrech
- Department of Endocrinology-Diabetology and Nutrition, Mohammed VI University Hospital Center, Oujda, MAR
- Department of Endocrinology-Diabetology and Nutrition, Faculty of Medicine and Pharmacy, Mohamed First University, Oujda, MAR
- Laboratory of Epidemiology, Clinical Research and Public Health, Faculty of Medicine and Pharmacy, Mohammed First University, Oujda, MAR
| |
Collapse
|
6
|
Tasdika TE, Choudhury N, Hossain QMI, Kabir Y. Association of glutathione S-transferase M1 and T1 polymorphisms on the susceptibility of diabetic retinopathy in the Bangladeshi population. J Diabetes Metab Disord 2023; 22:325-332. [PMID: 37255812 PMCID: PMC10225443 DOI: 10.1007/s40200-022-01142-9] [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: 07/01/2022] [Accepted: 10/08/2022] [Indexed: 06/01/2023]
Abstract
Objectives This study investigated the role of glutathione-S-transferase gene (GSTM1 and GSTT1) polymorphisms in the predisposition of type 2 diabetes mellitus (T2DM) with or without diabetic retinopathy (DR). Methods The case-control study included 188 subjects: 50 T2DM with DR, 63 T2DM without DR, and 75 healthy individuals' presenting no clinical signs or evidence of diabetes mellitus. Zinc and magnesium levels were measured using a flame atomic absorption spectrophotometer, and the lipid profile was evaluated using standard methods. The gene polymorphism of GSTs was performed by the multiplex-PCR method. Results Compared to the control, DR and T2DM had considerably greater total cholesterol, LDL-C, and decreased HDL-C levels. Magnesium levels were significantly lower in DR and T2DM than in control. Total cholesterol, LDL, TG, and magnesium levels didn't differ significantly between DR and T2DM groups. In DR, the GSTT1-null genotype was more prevalent than in T2DM subjects and controls (26.0%, 12.7%, and 10.7%, respectively). GSTT1-null genotype was considerably more common in DR than in controls and associated with 2.94-folds enhancing the chance of developing DR (OR = 2.94; 95% CI = 1.12-7.75; p = 0.02). However, the recurrence of GSTM1-null genotype was not clearly distinguishable among these three populations (28.0%, 38.1% and 29.3%, respectively) and not particularly prone to the risk of DR compared to T2DM subjects and controls (OR = 0.63; 95% CI = 0.28-1.41; p = 0.26; OR = 0.94; 95% CI = 0.42-2.07; p = 0.87, respectively). Conclusions Taken together, these findings suggest the potential role of GSTT1 deletion mutation as a risk factor for the vulnerability of DR among T2DM patients in the Bangladeshi population.
Collapse
Affiliation(s)
- Tafriha E Tasdika
- Department of Biochemistry and Molecular Biology, Primeasia University, Dhaka, Bangladesh
| | - Nuzhat Choudhury
- Department of Ophthalmology, Bangabandhu Sheikh Mujib Medical University (BSMMU), Dhaka, Bangladesh
| | - Q. M. Iqbal Hossain
- Department of Ophthalmology, Bangladesh Institute of Health Sciences (BIHS), Dhaka, Bangladesh
| | - Yearul Kabir
- Department of Biochemistry and Molecular Biology, Primeasia University, Dhaka, Bangladesh
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
7
|
Gouliopoulos N, Gazouli M, Karathanou K, Moschos MM. The association of AGER and ALDH2 gene polymorphisms with diabetic retinopathy. Eur J Ophthalmol 2022; 33:11206721221126287. [PMID: 36113108 DOI: 10.1177/11206721221126287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2023]
Abstract
PURPOSE To evaluate the possible associations between AGER (rs1051993, rs2070600) and ALDH2 (rs671) gene polymorphisms with nonproliferative (NPDR) and proliferative (PDR) diabetic retinopathy, in a well-defined Greek population. MATERIALS 66 NPDR patients and 57 PDR patients participated in our study, along with 156 age- and gender-matched healthy-control subjects (CL). All the participants underwent a complete ophthalmological examination, while clinical and demographic data were collected. Furthermore, they were genotyped for the studied polymorphisms. RESULTS No significant differences were detected among the studied groups regarding the participants' age and gender status. We found that the ALDH2 AA genotype was significantly more frequent in PDR patients than in CL (p = 0.014). Furthermore, between NPDR and PDR groups, the AGER rs1051993 GT and TT genotype frequencies were significantly elevated in PDR patients (p < 0.0001 and 0.04, respectively). Moreover, we demonstrated that the heterozygous GT genotype in DR patients is accompanied by 71.11 times higher risk of developing PDR (OR = 71.11: 95% CI- 4.14-1215.2), while the homozygous TT genotype is associated with 12.71 times elevated risk for PDR development (OR = 12.71: 95% CI- 0.63-254.1). CONCLUSIONS We documented that the ALDH2 AA and AGER rs1051993 GT and TT genotypes were observed significantly more frequently in PDR Greek diabetic patients. Our findings also support the genetic theory, suggesting that heritability is significantly implicated in the development of DR, providing additional evidence in the understanding of DR pathogenesis.
Collapse
Affiliation(s)
- Nikolaos Gouliopoulos
- 1st Department of Ophthalmology, Medical School, 393206National and Kapodistrian University of Athens, Athens, Greece
- 2nd Department of Ophthalmology, Medical School, 393206National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Gazouli
- Department of Basic Medical Sciences, Laboratory of Biology Medical School, 393206National and Kapodistrian University of Athens, Athens, Greece
| | | | - Marilita M Moschos
- 1st Department of Ophthalmology, Medical School, 393206National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
8
|
Gouliopoulos N, Siasos G, Oikonomou D, Oikonomou E, Konsola T, Kollia C, Athanasiou D, Dimitropoulos S, Rouvas A, Kassi E, Tousoulis D, Moschos MM. The association of T786C and G894T polymorphisms of eNOS gene with diabetic retinopathy in Greece. Eur J Ophthalmol 2022; 32:2582-2588. [PMID: 34779284 DOI: 10.1177/11206721211054724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE To investigate whether eNOS T786C (rs2070744) and G894T (rs1799983) gene polymorphisms are associated with diabetic retinopathy in Greek diabetic patients. MATERIALS 271 patients with type-2 diabetes mellitus participated in our study; 130 suffered from diabetic retinopathy and 141 not. All the patients underwent a complete ophthalmological examination, while clinical and demographic data were assessed. Furthermore, they were genotyped for rs2070744 and rs1799983 single nucleotide polymorphisms of eNOS gene. RESULTS Regarding the clinical and demographic data, no significant differences were detected between the studied groups, except for hemoglobin A1c levels and the frequency of insulin treatment (higher in patients with diabetic retinopathy). The frequency of rs1799983 GT genotype was significantly elevated in patients with diabetic retinopathy (55% vs. 40%, P = 0.011) and was associated with a 2-fold increased risk of developing retinopathy (OR 1.92, 95% CI 1.16-3.17). Furthermore, we demonstrated that the aforementioned genotype was significantly and independently associated with increased odds for retinopathy onset in diabetic subjects (OR 2.23, 95% CI 1.28-3.90, P = 0.005), regardless of the impact of other confounders. CONCLUSIONS We documented that rs1799983 GT genotype could be recognized as an independent risk factor of retinopathy in Greek patients with type-2 diabetes mellitus, while no role for rs2070744 polymorphism was identified. Further research in different ethnic groups will clarify the exact association of these polymorphisms with the risk for diabetic retinopathy development.
Collapse
Affiliation(s)
- Nikolaos Gouliopoulos
- 2nd Department of Ophthalmology, University of Athens Medical School, 69038Attikon Hospital, Athens, Greece
- 1st Department of Ophthalmology, University of Athens Medical School, G. Gennimatas Hospital, Athens, Greece
| | - Gerasimos Siasos
- 1st Department of Cardiology, 393206University of Athens Medical School, Hippokration Hospital, Athens, Greece
| | - Dimitra Oikonomou
- 2nd Department of Ophthalmology, University of Athens Medical School, 69038Attikon Hospital, Athens, Greece
| | - Evangelos Oikonomou
- 1st Department of Cardiology, 393206University of Athens Medical School, Hippokration Hospital, Athens, Greece
| | - Theodosia Konsola
- 1st Department of Cardiology, 393206University of Athens Medical School, Hippokration Hospital, Athens, Greece
| | - Christina Kollia
- 1st Department of Cardiology, 393206University of Athens Medical School, Hippokration Hospital, Athens, Greece
| | - Dimitris Athanasiou
- 1st Department of Cardiology, 393206University of Athens Medical School, Hippokration Hospital, Athens, Greece
| | - Stathis Dimitropoulos
- 1st Department of Cardiology, 393206University of Athens Medical School, Hippokration Hospital, Athens, Greece
| | - Alexandros Rouvas
- 2nd Department of Ophthalmology, University of Athens Medical School, 69038Attikon Hospital, Athens, Greece
| | - Eva Kassi
- 1st Department of Propaedeutic and Internal Medicine, Division of Diabetes, 68989University of Athens Medical School, Laiko Hospital, Athens, Greece
| | - Dimitrios Tousoulis
- 1st Department of Cardiology, 393206University of Athens Medical School, Hippokration Hospital, Athens, Greece
| | - Marilita M Moschos
- 1st Department of Ophthalmology, University of Athens Medical School, G. Gennimatas Hospital, Athens, Greece
| |
Collapse
|
9
|
Gouliopoulos N, Siasos G, Bouratzis N, Oikonomou E, Kollia C, Konsola T, Oikonomou D, Rouvas A, Kassi E, Tousoulis D, Moschos MM. Polymorphism analysis of ADIPOQ gene in Greek patients with diabetic retinopathy. Ophthalmic Genet 2022; 43:326-331. [PMID: 34895017 DOI: 10.1080/13816810.2021.2015787] [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: 05/13/2021] [Revised: 11/13/2021] [Accepted: 11/30/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Several genetic polymorphisms have been identified as risk factors for diabetic retinopathy (DR) onset. The purpose of our study was to determine whether ADIPOQ rs1501299 and rs2241766 gene polymorphisms are associated with DR in a cohort of Greek diabetic patients. MATERIALS AND METHODS 218 patients with type-2 diabetes mellitus (T2DM) were included in the study; 109 suffered from DR and 109 not. All the participants underwent a complete ophthalmological examination, while clinical and demographic data were assessed. Furthermore, they were genotyped for G276T (rs1501299) and T45G (rs2241766) single nucleotide polymorphisms of ADIPOQ gene. RESULTS Between the studied groups, no significant differences were detected regarding the demographic and clinical data (p > .05 for all), except for hemoglobin A1c levels and frequency of insulin treatment (higher in DR patients). We detected that the frequency of rs1501299 GT genotype was significantly elevated in DR patients (53% vs. 34%, p = .004) and was associated with a higher risk of developing retinopathy (OR 2.31, 95% CI 1.30-4.11). Furthermore, we demonstrated that the rs1501299 GT genotype was significantly and independently associated with increased odds for DR development in diabetic subjects (OR 2.68, 95% CI 1.38-5.21, p = .004), regardless of the impact of other known risk factors. CONCLUSIONS We documented that rs1501299 GT genotype could be recognized as an independent risk factor of retinopathy in T2DM Greek patients, while no role for rs2241766 polymorphism was identified. Further research in different ethnic groups will clarify the exact association of these polymorphisms with the risk of DR development.
Collapse
Affiliation(s)
- Nikolaos Gouliopoulos
- 2nd Department of Ophthalmology, University of Athens Medical School, Athens, Greece
- 1st Department of Ophthalmology, University of Athens Medical School, Athens, Greece
| | - Gerasimos Siasos
- 1st Department of Cardiology, University of Athens Medical School, Athens, Greece
| | - Nikolaos Bouratzis
- 2 Department of Ophthalmology, University of Athens Medical School, Athens, Greece
| | - Evangelos Oikonomou
- 1 Department of Cardiology, University of Athens Medical School, Athens, Greece
| | - Christina Kollia
- 1 Department of Cardiology, University of Athens Medical School, Athens, Greece
| | - Theodosia Konsola
- 1 Department of Cardiology, University of Athens Medical School, Athens, Greece
| | - Dimitra Oikonomou
- 2 Department of Ophthalmology, University of Athens Medical School, Athens, Greece
| | - Alexandros Rouvas
- 2 Department of Ophthalmology, University of Athens Medical School, Athens, Greece
| | - Eva Kassi
- 1st Department of Propaedeutic and Internal Medicine, Division of Diabetes, University of Athens Medical School, Athens, Greece
| | - Dimitrios Tousoulis
- 1 Department of Cardiology, University of Athens Medical School, Athens, Greece
| | - Marilita M Moschos
- 1 Department of Ophthalmology, University of Athens Medical School, Athens, Greece
| |
Collapse
|
10
|
Kim J, Jensen A, Ko S, Raghavan S, Phillips LS, Hung A, Sun Y, Zhou H, Reaven P, Zhou JJ. Systematic Heritability and Heritability Enrichment Analysis for Diabetes Complications in UK Biobank and ACCORD Studies. Diabetes 2022; 71:1137-1148. [PMID: 35133398 PMCID: PMC9044130 DOI: 10.2337/db21-0839] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 02/02/2022] [Indexed: 11/13/2022]
Abstract
Diabetes-related complications reflect longstanding damage to small and large vessels throughout the body. In addition to the duration of diabetes and poor glycemic control, genetic factors are important contributors to the variability in the development of vascular complications. Early heritability studies found strong familial clustering of both macrovascular and microvascular complications. However, they were limited by small sample sizes and large phenotypic heterogeneity, leading to less accurate estimates. We take advantage of two independent studies-UK Biobank and the Action to Control Cardiovascular Risk in Diabetes trial-to survey the single nucleotide polymorphism heritability for diabetes microvascular (diabetic kidney disease and diabetic retinopathy) and macrovascular (cardiovascular events) complications. Heritability for diabetic kidney disease was estimated at 29%. The heritability estimate for microalbuminuria ranged from 24 to 60% and was 41% for macroalbuminuria. Heritability estimates of diabetic retinopathy ranged from 6 to 33%, depending on the phenotype definition. More severe diabetes retinopathy possessed higher genetic contributions. We show, for the first time, that rare variants account for much of the heritability of diabetic retinopathy. This study suggests that a large portion of the genetic risk of diabetes complications is yet to be discovered and emphasizes the need for additional genetic studies of diabetes complications.
Collapse
Affiliation(s)
- Juhyun Kim
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA
- Department of Biostatistics, University of Michigan, Ann Arbor, MI
| | - Aubrey Jensen
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA
| | - Seyoon Ko
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA
| | - Sridharan Raghavan
- University of Colorado School of Medicine, Aurora, CO
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | - Lawrence S. Phillips
- Division of Endocrinology, Emory University School of Medicine, Atlanta, GA
- Atlanta Veterans Affairs Medical Center, Decatur, GA
| | - Adriana Hung
- Tennessee Valley Healthcare System and Vanderbilt University, Nashville, TN
| | - Yan Sun
- Department of Epidemiology, Emory University, Atlanta, GA
| | - Hua Zhou
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA
| | - Peter Reaven
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ
| | - Jin J. Zhou
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ
- Department of Epidemiology and Biostatistics, University of Arizona, Tucson, AZ
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
11
|
Scanlon PH. Improving the screening of risk factors in diabetic retinopathy. Expert Rev Endocrinol Metab 2022; 17:235-243. [PMID: 35730170 DOI: 10.1080/17446651.2022.2078305] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 05/12/2022] [Indexed: 10/17/2022]
Abstract
INTRODUCTION In 2002, Diabetic Retinopathy was reported as the leading cause of blindness in the working age group. The introduction of systematic screening programs in the UK has reduced visual loss and blindness due to diabetic retinopathy, but it does still occur with catastrophic consequences for the individual. AREAS COVERED The author conducted an ongoing search for articles relating to diabetic retinopathy since 2000 utilizing Zetoc Alert with keywords and contents page lists from relevant journals. This review covers the risk factors for loss of vision due to diabetic retinopathy and discusses ways in which the awareness of these risk factors can be used to further reduce visual loss. Some risk factors such as glycemic and B/P control are well known from landmark trials. This review has included these factors but concentrated more on the evidence behind those risk factors that are not so clearly defined or so well known. EXPERT OPINION The major risk factors are well known, but one continues to find that people with diabetes lose vision in situations in which a better awareness of the risks by both the individual with diabetes and the health workers involved may have prevented the visual loss.
Collapse
Affiliation(s)
- Peter H Scanlon
- Consultant Ophthalmologist, Department of Ophthalmologist, Gloucestershire Hospitals NHS Foundation Trust Cheltenham, UK
- National Clinical Lead, NHS Diabetic Eye Screening Programme (Ophthalmology), Public Health Commissioning and Operations, England
- Associate Professor, Nuffield Department of Clinical Neuroscience, University of Oxford, UK
- Visiting Professor, School of Health and Social Care, University of Gloucestershire, UK
| |
Collapse
|
12
|
Kowluru RA. Long Noncoding RNAs and Mitochondrial Homeostasis in the Development of Diabetic Retinopathy. Front Endocrinol (Lausanne) 2022; 13:915031. [PMID: 35733767 PMCID: PMC9207305 DOI: 10.3389/fendo.2022.915031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Retinopathy is one of the most devastating complications of diabetes, which a patient fears the most. Hyperglycemic environment results in many structural, functional, molecular and biochemical abnormalities in the retina, and overproduction of mitochondrial superoxide, induced by hyperglycemic milieu, is considered to play a central role in the development of diabetic retinopathy. Expression of many genes associated with maintaining mitochondrial homeostasis is also altered. Recent research has shown that several long noncoding RNAs, RNAs with more than 200 nucleotides but without any reading frames, are aberrantly expressed in diabetes, and altered expression of these long noncoding RNAs is now being implicated in the development of diabetes and its complications including retinopathy. This review focuses the role of long noncoding RNAs in the development of diabetic retinopathy, with a special emphasis on the maintenance of mitochondrial homeostasis.
Collapse
|
13
|
Sobrin L, Susarla G, Stanwyck L, Rouhana JM, Li A, Pollack S, Igo RP, Jensen RA, Li X, Ng MCY, Smith AV, Kuo JZ, Taylor KD, Freedman BI, Bowden DW, Penman A, Chen CJ, Craig JE, Adler SG, Chew EY, Cotch MF, Yaspan B, Mitchell P, Wang JJ, Klein BEK, Wong TY, Rotter JI, Burdon KP, Iyengar SK, Segrè AV. Gene Set Enrichment Analsyes Identify Pathways Involved in Genetic Risk for Diabetic Retinopathy. Am J Ophthalmol 2022; 233:111-123. [PMID: 34166655 PMCID: PMC8678352 DOI: 10.1016/j.ajo.2021.06.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 04/19/2021] [Accepted: 06/12/2021] [Indexed: 01/03/2023]
Abstract
To identify functionally related genes associated with diabetic retinopathy (DR) risk using gene set enrichment analyses applied to genome-wide association study meta-analyses. METHODS We analyzed DR GWAS meta-analyses performed on 3246 Europeans and 2611 African Americans with type 2 diabetes. Gene sets relevant to 5 key DR pathophysiology processes were investigated: tissue injury, vascular events, metabolic events and glial dysregulation, neuronal dysfunction, and inflammation. Keywords relevant to these processes were queried in 4 pathway and ontology databases. Two GSEA methods, Meta-Analysis Gene set Enrichment of variaNT Associations (MAGENTA) and Multi-marker Analysis of GenoMic Annotation (MAGMA), were used. Gene sets were defined to be enriched for gene associations with DR if the P value corrected for multiple testing (Pcorr) was <.05. RESULTS Five gene sets were significantly enriched for numerous modest genetic associations with DR in one method (MAGENTA or MAGMA) and also at least nominally significant (uncorrected P < .05) in the other method. These pathways were regulation of the lipid catabolic process (2-fold enrichment, Pcorr = .014); nitric oxide biosynthesis (1.92-fold enrichment, Pcorr = .022); lipid digestion, mobilization, and transport (1.6-fold enrichment, P = .032); apoptosis (1.53-fold enrichment, P = .041); and retinal ganglion cell degeneration (2-fold enrichment, Pcorr = .049). The interferon gamma (IFNG) gene, previously implicated in DR by protein-protein interactions in our GWAS, was among the top ranked genes in the nitric oxide pathway (best variant P = .0001). CONCLUSIONS These GSEA indicate that variants in genes involved in oxidative stress, lipid transport and catabolism, and cell degeneration are enriched for genes associated with DR risk. NOTE: Publication of this article is sponsored by the American Ophthalmological Society.
Collapse
Affiliation(s)
- Lucia Sobrin
- From the Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary.
| | - Gayatri Susarla
- From the Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary
| | - Lynn Stanwyck
- From the Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary
| | - John M Rouhana
- From the Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary
| | - Ashley Li
- From the Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary
| | - Samuela Pollack
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Robert P Igo
- Department of Population and Quantitative Health Sciences, Case Western University, Cleveland, Ohio
| | - Richard A Jensen
- Cardiovascular Health Research Unit, Department of Medicine, Epidemiology and Health Services, University of Washington, Seattle, Washington
| | - Xiaohui Li
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, the Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California
| | - Maggie C Y Ng
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine; Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA; Vanderbilt Genetics Institute and Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Albert V Smith
- Department of Medicine, University of Iceland, Reykjavík, Iceland
| | - Jane Z Kuo
- Medical Affairs, Ophthalmology, Sun Pharmaceutical Industries, Inc, Princeton, New Jersey
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, the Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California
| | - Barry I Freedman
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine; Department of Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Donald W Bowden
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine; Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Alan Penman
- Department of Preventive Medicine, John D. Bower School of Population Health (A.P.), Department of Ophthalmology
| | - Ching J Chen
- Department of Preventive Medicine, John D. Bower School of Population Health (A.P.), Department of Ophthalmology
| | - Jamie E Craig
- University of Mississippi Medical Center, Jackson, Mississippi, USA, FHMRI Eye & Vision, Flinders University, Bedford Park, SA, Australia
| | - Sharon G Adler
- Department of Nephrology and Hypertension, Los Angeles Biomedical Research Institute at Harbor-University of California, Torrance, California
| | - Emily Y Chew
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Mary Frances Cotch
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Brian Yaspan
- Genentech Inc, South San Francisco, California, USA
| | - Paul Mitchell
- Department of Ophthalmology, Centre for Vision Research, Westmead Institute for Medical Research, University of Sydney, Sydney, Australia
| | - Jie Jin Wang
- Department of Ophthalmology, Centre for Vision Research, Westmead Institute for Medical Research, University of Sydney, Sydney, Australia; Center of Clinician-Scientist Development, Duke-NUS Medical School, Singapore
| | - Barbara E K Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Tien Y Wong
- Center of Clinician-Scientist Development, Duke-NUS Medical School, Singapore; Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, the Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California
| | - Kathyrn P Burdon
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Sudha K Iyengar
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Population and Quantitative Health Sciences, Case Western University, Cleveland, Ohio
| | - Ayellet V Segrè
- From the Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary; Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| |
Collapse
|
14
|
VEGF and eNOS genes polymorphism features in patients with diabetes mellitus with and without initial non-proliferative diabetic retinopathy. ACTA BIOMEDICA SCIENTIFICA 2021. [DOI: 10.29413/abs.2021-6.6-1.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The endothelial NO synthase (eNOS) and vascular endothelial growth factor (VEGF) imbalance and the polymorphism of these genes may be the predisposition for diabetic retinopathy (DR) development and progression.The aim: to analyze VEGF (rs699947 and rs3025039) and eNOS (rs2070744) genes polymorphism and their combinations in patients with type 2 diabetes mellitus (DM2) with and without initial non-proliferative DR.Materials and methods. The study included 200 patients with type 2 diabetes (155 women and 45 men, age – 43–70 years): 111 people without and 89 people with DR. The polymorphism of the regulatory regions of VEGF (rs699947 and rs3025039) and eNOS (rs2070744) genes was studied using restriction fragment length polymorphism analysis and TaqMan Real-Time PCR by. Statistical processing was carried out using the software packages Statistica 10.0, SPSS Statistics 23 and the package of original programs for volumetric processing of bioinformation.Results. The VEGF-2578 heterozygosity and two complex genotypes – VEGF-2578CA:VEGF+936CC and NOS3-786CT:VEGF-2578CA:VEGF+936CC – signifi cantly decreased in patients with DR. The predisposition to early DR development to minor genotype of eNOS gene in the NOS3-786CC:VEGF+936CT complex and signifi cantly decreased the homozygous wild-type eNOS genotype in DM2 patients with ophthalmopathology were shown. NOS3-86TT:VEGF2578AA genotype signifi cantly decreased in group with retinopathy developing and the glycated hemoglobin high level.Conclusion. Along with the clinical risk factors for the development of DR in DM2, the genetic polymorphism of the regulatory regions of the genes analyzed by us has a signifi cant weight. When analyzing potential genetic markers, it is important to consider possible joint epistatic/hypostatic effects. The complex analysis of polymorphic gene can help early prognosis of the DR development.
Collapse
|
15
|
|
16
|
Behl T, Kumar K, Singh S, Sehgal A, Sachdeva M, Bhatia S, Al-Harrasi A, Buhas C, Teodora Judea-Pusta C, Negrut N, Alexandru Munteanu M, Brisc C, Bungau S. Unveiling the role of polyphenols in diabetic retinopathy. J Funct Foods 2021. [DOI: https://doi.org/10.1016/j.jff.2021.104608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
17
|
Ustinova M, Peculis R, Rescenko R, Rovite V, Zaharenko L, Elbere I, Silamikele L, Konrade I, Sokolovska J, Pirags V, Klovins J. Novel susceptibility loci identified in a genome-wide association study of type 2 diabetes complications in population of Latvia. BMC Med Genomics 2021; 14:18. [PMID: 33430853 PMCID: PMC7802349 DOI: 10.1186/s12920-020-00860-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/20/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Type 2 diabetes complications cause a serious emotional and economical burden to patients and healthcare systems globally. Management of both acute and chronic complications of diabetes, which dramatically impair the quality of patients' life, is still an unsolved issue in diabetes care, suggesting a need for early identification of individuals with high risk for developing diabetes complications. METHODS We performed a genome-wide association study in 601 type 2 diabetes patients after stratifying them according to the presence or absence of four types of diabetes complications: diabetic neuropathy, diabetic nephropathy, macrovascular complications, and ophthalmic complications. RESULTS The analysis revealed ten novel associations showing genome-wide significance, including rs1132787 (GYPA, OR = 2.71; 95% CI = 2.02-3.64) and diabetic neuropathy, rs2477088 (PDE4DIP, OR = 2.50; 95% CI = 1.87-3.34), rs4852954 (NAT8, OR = 2.27; 95% CI = 2.71-3.01), rs6032 (F5, OR = 2.12; 95% CI = 1.63-2.77), rs6935464 (RPS6KA2, OR = 2.25; 95% CI = 6.69-3.01) and macrovascular complications, rs3095447 (CCDC146, OR = 2.18; 95% CI = 1.66-2.87) and ophthalmic complications. By applying the targeted approach of previously reported susceptibility loci we managed to replicate three associations: MAPK14 (rs3761980, rs80028505) and diabetic neuropathy, APOL1 (rs136161) and diabetic nephropathy. CONCLUSIONS Together these results provide further evidence for the implication of genetic factors in the development of type 2 diabetes complications and highlight several potential key loci, able to modify the risk of developing these conditions. Moreover, the candidate variant approach proves a strong and consistent effect for multiple variants across different populations.
Collapse
Affiliation(s)
- Monta Ustinova
- Latvian Biomedical Research and Study Centre, Ratsupites iela 1, Riga, 1067, Latvia
| | - Raitis Peculis
- Latvian Biomedical Research and Study Centre, Ratsupites iela 1, Riga, 1067, Latvia
| | - Raimonds Rescenko
- Latvian Biomedical Research and Study Centre, Ratsupites iela 1, Riga, 1067, Latvia
| | - Vita Rovite
- Latvian Biomedical Research and Study Centre, Ratsupites iela 1, Riga, 1067, Latvia
| | - Linda Zaharenko
- Latvian Biomedical Research and Study Centre, Ratsupites iela 1, Riga, 1067, Latvia
| | - Ilze Elbere
- Latvian Biomedical Research and Study Centre, Ratsupites iela 1, Riga, 1067, Latvia
| | - Laila Silamikele
- Latvian Biomedical Research and Study Centre, Ratsupites iela 1, Riga, 1067, Latvia
| | - Ilze Konrade
- Latvian Biomedical Research and Study Centre, Ratsupites iela 1, Riga, 1067, Latvia
- Faculty of Medicine, Riga Stradins University, Dzirciema iela 16, Riga, 1007, Latvia
| | | | - Valdis Pirags
- Latvian Biomedical Research and Study Centre, Ratsupites iela 1, Riga, 1067, Latvia
- Faculty of Medicine, University of Latvia, Jelgavas iela 3, Riga, 1004, Latvia
| | - Janis Klovins
- Latvian Biomedical Research and Study Centre, Ratsupites iela 1, Riga, 1067, Latvia.
| |
Collapse
|
18
|
Abstract
Diabetes is one of the fastest growing diseases worldwide, projected to affect 693 million adults by 2045. Devastating macrovascular complications (cardiovascular disease) and microvascular complications (such as diabetic kidney disease, diabetic retinopathy and neuropathy) lead to increased mortality, blindness, kidney failure and an overall decreased quality of life in individuals with diabetes. Clinical risk factors and glycaemic control alone cannot predict the development of vascular complications; numerous genetic studies have demonstrated a clear genetic component to both diabetes and its complications. Early research aimed at identifying genetic determinants of diabetes complications relied on familial linkage analysis suited to strong-effect loci, candidate gene studies prone to false positives, and underpowered genome-wide association studies limited by sample size. The explosion of new genomic datasets, both in terms of biobanks and aggregation of worldwide cohorts, has more than doubled the number of genetic discoveries for both diabetes and diabetes complications. We focus herein on genetic discoveries for diabetes and diabetes complications, empowered primarily through genome-wide association studies, and emphasize the gaps in research for taking genomic discovery to the next level.
Collapse
Affiliation(s)
- Joanne B Cole
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, MA, USA
| | - Jose C Florez
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
Cabrera AP, Mankad RN, Marek L, Das R, Rangasamy S, Monickaraj F, Das A. Genotypes and Phenotypes: A Search for Influential Genes in Diabetic Retinopathy. Int J Mol Sci 2020; 21:E2712. [PMID: 32295293 PMCID: PMC7215289 DOI: 10.3390/ijms21082712] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 12/13/2022] Open
Abstract
Although gene-environment interactions are known to play an important role in the inheritance of complex traits, it is still unknown how a genotype and the environmental factors result in an observable phenotype. Understanding this complex interaction in the pathogenesis of diabetic retinopathy (DR) remains a big challenge as DR appears to be a disease with heterogenous phenotypes with multifactorial influence. In this review, we examine the natural history and risk factors related to DR, emphasizing distinct clinical phenotypes and their natural course in retinopathy. Although there is strong evidence that duration of diabetes and metabolic factors play a key role in the pathogenesis of DR, accumulating new clinical studies reveal that this disease can develop independently of duration of diabetes and metabolic dysfunction. More recently, studies have emphasized the role of genetic factors in DR. However, linkage analyses, candidate gene studies, and genome-wide association studies (GWAS) have not produced any statistically significant results. Our recently initiated genomics study, the Diabetic Retinopathy Genomics (DRGen) Study, aims to examine the contribution of rare and common variants in the development DR, and how they can contribute to clinical phenotype, rate of progression, and response to available therapies. Our preliminary findings reveal a novel set of genetic variants associated with proangiogenic and inflammatory pathways that may contribute to DR pathogenesis. Further investigation of these variants is necessary and may lead to development of novel biomarkers and new therapeutic targets in DR.
Collapse
Affiliation(s)
- Andrea P. Cabrera
- Department of Surgery, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (A.P.C.); (R.N.M.); (L.M.); (R.D.); (F.M.)
| | - Rushi N. Mankad
- Department of Surgery, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (A.P.C.); (R.N.M.); (L.M.); (R.D.); (F.M.)
| | - Lauren Marek
- Department of Surgery, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (A.P.C.); (R.N.M.); (L.M.); (R.D.); (F.M.)
| | - Ryan Das
- Department of Surgery, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (A.P.C.); (R.N.M.); (L.M.); (R.D.); (F.M.)
| | - Sampath Rangasamy
- Translational & Genomics Research Institute, Phoenix, AZ 85004, USA;
| | - Finny Monickaraj
- Department of Surgery, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (A.P.C.); (R.N.M.); (L.M.); (R.D.); (F.M.)
- New Mexico VA Health Care System, Albuquerque, NM 87108, USA
| | - Arup Das
- Department of Surgery, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (A.P.C.); (R.N.M.); (L.M.); (R.D.); (F.M.)
- New Mexico VA Health Care System, Albuquerque, NM 87108, USA
| |
Collapse
|
20
|
Han M, Dong Z, Duan H, Sun X, Zhang T, Ying M. Associations of rs2300782CAMK4, rs2292239ERBB3and rs10491034ARHGAP22with Diabetic Retinopathy Among Chinese Hui Population. DNA Cell Biol 2020; 39:398-403. [PMID: 31976761 DOI: 10.1089/dna.2019.5027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Mei Han
- The Third Department of Vitreoretinal, Tianjin Eye Hospital, Tianjin, P.R. China
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, P.R. China
| | - Zuyan Dong
- Department of Ophthalmology, Linxia State People Hospital of Gansu Province, Linxia, P.R. China
| | - Hongtao Duan
- The Third Department of Vitreoretinal, Tianjin Eye Hospital, Tianjin, P.R. China
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, P.R. China
| | - Xiaoli Sun
- The Third Department of Vitreoretinal, Tianjin Eye Hospital, Tianjin, P.R. China
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, P.R. China
| | - Tongmei Zhang
- The Third Department of Vitreoretinal, Tianjin Eye Hospital, Tianjin, P.R. China
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, P.R. China
| | - Ming Ying
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, P.R. China
| |
Collapse
|
21
|
Cabrera AP, Monickaraj F, Rangasamy S, Hobbs S, McGuire P, Das A. Do Genomic Factors Play a Role in Diabetic Retinopathy? J Clin Med 2020; 9:E216. [PMID: 31947513 PMCID: PMC7019561 DOI: 10.3390/jcm9010216] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 02/07/2023] Open
Abstract
Although there is strong clinical evidence that the control of blood glucose, blood pressure, and lipid level can prevent and slow down the progression of diabetic retinopathy (DR) as shown by landmark clinical trials, it has been shown that these factors only account for 10% of the risk for developing this disease. This suggests that other factors, such as genetics, may play a role in the development and progression of DR. Clinical evidence shows that some diabetics, despite the long duration of their diabetes (25 years or more) do not show any sign of DR or show minimal non-proliferative diabetic retinopathy (NPDR). Similarly, not all diabetics develop proliferative diabetic retinopathy (PDR). So far, linkage analysis, candidate gene studies, and genome-wide association studies (GWAS) have not produced any statistically significant results. We recently initiated a genomics study, the Diabetic Retinopathy Genetics (DRGen) Study, to examine the contribution of rare and common variants in the development of different phenotypes of DR, as well as their responsiveness to anti-VEGF treatment in diabetic macular edema (DME). Our preliminary findings reveal a novel set of genetic variants involved in the angiogenesis and inflammatory pathways that contribute to DR progression or protection. Further investigation of variants can help to develop novel biomarkers and lead to new therapeutic targets in DR.
Collapse
Affiliation(s)
- Andrea P. Cabrera
- Department of Surgery/Ophthalmology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (A.P.C.); (F.M.); (S.H.)
| | - Finny Monickaraj
- Department of Surgery/Ophthalmology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (A.P.C.); (F.M.); (S.H.)
- New Mexico VA Health Care System, Albuquerque, NM 87131, USA
| | | | - Sam Hobbs
- Department of Surgery/Ophthalmology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (A.P.C.); (F.M.); (S.H.)
| | - Paul McGuire
- Department of Cell Biology & Physiology, UNM, Albuquerque, NM 87131, USA;
| | - Arup Das
- Department of Surgery/Ophthalmology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (A.P.C.); (F.M.); (S.H.)
- New Mexico VA Health Care System, Albuquerque, NM 87131, USA
- Department of Cell Biology & Physiology, UNM, Albuquerque, NM 87131, USA;
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW The goal of this paper is to review the latest findings in understanding the genetics of diabetic retinopathy. We highlight recent literature using a variety of molecular genetic techniques to identify variants which contribute to genetic susceptibility for diabetic retinopathy. RECENT FINDINGS New genome-wide association study (GWAS) and whole-exome sequencing approaches have been utilized to identify both common and rare variants associated with diabetic retinopathy. While variants have been identified in isolated studies, no variants have been replicated across multiple studies. The identification of genetic factors associated with diabetic retinopathy remains elusive. This is due to the multifactorial nature of the disease, small sample sizes for GWAS, and difficulty in controlling covariates of the disease. Larger populations as well as utilization of new sequencing and data analysis techniques may lead to new insights into genetic factors associated with diabetic retinopathy in the future.
Collapse
Affiliation(s)
- Jonathan Han
- School of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Leonardo Lando
- Shiley Eye Institute, Andrew Viterbi Department of Ophthalmology, University of California San Diego, 9415 Campus Point Dr, La Jolla, San Diego, CA, 92093, USA
| | - Dorota Skowronska-Krawczyk
- Shiley Eye Institute, Andrew Viterbi Department of Ophthalmology, University of California San Diego, 9415 Campus Point Dr, La Jolla, San Diego, CA, 92093, USA
| | - Daniel L Chao
- Shiley Eye Institute, Andrew Viterbi Department of Ophthalmology, University of California San Diego, 9415 Campus Point Dr, La Jolla, San Diego, CA, 92093, USA.
- Shiley Eye Institute; Andrew Viterbi Department of Ophthalmology, University of California San Diego, 9500 Gilman Dr MC 0946, La Jolla, San Diego, CA, 93094, USA.
| |
Collapse
|
23
|
Pollack S, Igo RP, Jensen RA, Christiansen M, Li X, Cheng CY, Ng MCY, Smith AV, Rossin EJ, Segrè AV, Davoudi S, Tan GS, Chen YDI, Kuo JZ, Dimitrov LM, Stanwyck LK, Meng W, Hosseini SM, Imamura M, Nousome D, Kim J, Hai Y, Jia Y, Ahn J, Leong A, Shah K, Park KH, Guo X, Ipp E, Taylor KD, Adler SG, Sedor JR, Freedman BI, Lee IT, Sheu WHH, Kubo M, Takahashi A, Hadjadj S, Marre M, Tregouet DA, Mckean-Cowdin R, Varma R, McCarthy MI, Groop L, Ahlqvist E, Lyssenko V, Agardh E, Morris A, Doney ASF, Colhoun HM, Toppila I, Sandholm N, Groop PH, Maeda S, Hanis CL, Penman A, Chen CJ, Hancock H, Mitchell P, Craig JE, Chew EY, Paterson AD, Grassi MA, Palmer C, Bowden DW, Yaspan BL, Siscovick D, Cotch MF, Wang JJ, Burdon KP, Wong TY, Klein BEK, Klein R, Rotter JI, Iyengar SK, Price AL, Sobrin L. Multiethnic Genome-Wide Association Study of Diabetic Retinopathy Using Liability Threshold Modeling of Duration of Diabetes and Glycemic Control. Diabetes 2019; 68:441-456. [PMID: 30487263 PMCID: PMC6341299 DOI: 10.2337/db18-0567] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 11/12/2018] [Indexed: 12/18/2022]
Abstract
To identify genetic variants associated with diabetic retinopathy (DR), we performed a large multiethnic genome-wide association study. Discovery included eight European cohorts (n = 3,246) and seven African American cohorts (n = 2,611). We meta-analyzed across cohorts using inverse-variance weighting, with and without liability threshold modeling of glycemic control and duration of diabetes. Variants with a P value <1 × 10-5 were investigated in replication cohorts that included 18,545 European, 16,453 Asian, and 2,710 Hispanic subjects. After correction for multiple testing, the C allele of rs142293996 in an intron of nuclear VCP-like (NVL) was associated with DR in European discovery cohorts (P = 2.1 × 10-9), but did not reach genome-wide significance after meta-analysis with replication cohorts. We applied the Disease Association Protein-Protein Link Evaluator (DAPPLE) to our discovery results to test for evidence of risk being spread across underlying molecular pathways. One protein-protein interaction network built from genes in regions associated with proliferative DR was found to have significant connectivity (P = 0.0009) and corroborated with gene set enrichment analyses. These findings suggest that genetic variation in NVL, as well as variation within a protein-protein interaction network that includes genes implicated in inflammation, may influence risk for DR.
Collapse
Affiliation(s)
- Samuela Pollack
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Robert P Igo
- Department of Population and Quantitative Health Sciences, Case Western University, Cleveland, OH
| | - Richard A Jensen
- Cardiovascular Health Research Unit, Department of Medicine, Epidemiology and Health Services, University of Washington, Seattle, WA
| | - Mark Christiansen
- Cardiovascular Health Research Unit, Department of Medicine, Epidemiology and Health Services, University of Washington, Seattle, WA
| | - Xiaohui Li
- Institute for Translational Genomics and Population Sciences, LA BioMed and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
| | - Ching-Yu Cheng
- Duke-NUS Medical School, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Maggie C Y Ng
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC
| | - Albert V Smith
- Department of Medicine, University of Iceland, Reykjavík, Iceland
| | - Elizabeth J Rossin
- Massachusetts Eye and Ear Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Ayellet V Segrè
- Massachusetts Eye and Ear Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Samaneh Davoudi
- Massachusetts Eye and Ear Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Gavin S Tan
- Duke-NUS Medical School, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, LA BioMed and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
| | - Jane Z Kuo
- Institute for Translational Genomics and Population Sciences, LA BioMed and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
- Medical Affairs, Ophthalmology, Sun Pharmaceutical Industries, Inc., Princeton, NJ
| | - Latchezar M Dimitrov
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC
| | - Lynn K Stanwyck
- Massachusetts Eye and Ear Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Weihua Meng
- Division of Population Health Sciences, Ninewells Hospital and Medical School, University of Dundee School of Medicine, Scotland, U.K
| | - S Mohsen Hosseini
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Minako Imamura
- Laboratory for Endocrinology, Metabolism and Kidney Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
- Division of Clinical Laboratory and Blood Transfusion, University of the Ryukyus Hospital, Nishihara, Japan
| | - Darryl Nousome
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Jihye Kim
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Yang Hai
- Institute for Translational Genomics and Population Sciences, LA BioMed and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
| | - Yucheng Jia
- Institute for Translational Genomics and Population Sciences, LA BioMed and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
| | - Jeeyun Ahn
- Department of Ophthalmology, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Aaron Leong
- Endocrine Unit and Diabetes Unit, Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA
| | - Kaanan Shah
- Section of Genetic Medicine, University of Chicago, Chicago, IL
| | - Kyu Hyung Park
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, LA BioMed and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
| | - Eli Ipp
- Section of Diabetes and Metabolism, Harbor-UCLA Medical Center, University of California, Los Angeles, Los Angeles, CA
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, LA BioMed and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
| | - Sharon G Adler
- Department of Nephrology and Hypertension, Los Angeles Biomedical Research Institute at Harbor-University of California, Torrance, CA
| | - John R Sedor
- Department of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH
- Division of Nephrology, MetroHealth System, Cleveland, OH
| | - Barry I Freedman
- Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - I-Te Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wayne H-H Sheu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Atsushi Takahashi
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
- Department of Genomic Medicine, Research Institute, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Samy Hadjadj
- CHU de Poitiers, Centre d'Investigation Clinique, Poitiers, France
- Université de Poitiers, UFR Médecine Pharmacie, Centre d'Investigation Clinique 1402, Poitiers, France
- INSERM, Centre d'Investigation Clinique 1402, Poitiers, France
- L'Institut du Thorax, INSERM, CNRS, CHU Nantes, Nantes, France
| | - Michel Marre
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- Department of Diabetology, Endocrinology and Nutrition, Assistance Publique-Hôpitaux de Paris, Bichat Hospital, DHU FIRE, Paris, France
- INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
| | - David-Alexandre Tregouet
- Team Genomics & Pathophysiology of Cardiovascular Diseases, UPMC, Sorbonne Universités, INSERM, UMR_S 1166, Paris, France
- Institute of Cardiometabolism and Nutrition, Paris, France
| | - Roberta Mckean-Cowdin
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Department of Ophthalmology, USC Roski Eye Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Rohit Varma
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Department of Ophthalmology, USC Roski Eye Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, U.K
- NIHR Oxford Biomedical Research Centre, Churchill Hospital, Oxford, U.K
| | - Leif Groop
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Emma Ahlqvist
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Valeriya Lyssenko
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Malmö, Sweden
- Department of Clinical Science, KG Jebsen Center for Diabetes Research, University of Bergen, Bergen, Norway
| | - Elisabet Agardh
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Andrew Morris
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, U.K
| | - Alex S F Doney
- Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, U.K
| | - Helen M Colhoun
- Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, U.K
| | - Iiro Toppila
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Niina Sandholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Shiro Maeda
- Laboratory for Endocrinology, Metabolism and Kidney Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
- Division of Clinical Laboratory and Blood Transfusion, University of the Ryukyus Hospital, Nishihara, Japan
| | - Craig L Hanis
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Alan Penman
- Department of Preventive Medicine, John D. Bower School of Population Health, University of Mississippi Medical Center, Jackson, MS
| | - Ching J Chen
- Department of Ophthalmology, University of Mississippi Medical Center, Jackson, MS
| | - Heather Hancock
- Department of Ophthalmology, University of Mississippi Medical Center, Jackson, MS
| | - Paul Mitchell
- Centre for Vision Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Jamie E Craig
- Department of Ophthalmology, Flinders University, Bedford Park, South Australia, Australia
| | - Emily Y Chew
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Andrew D Paterson
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Program in Genetics & Genome Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Epidemiology and Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Michael A Grassi
- Grassi Retina, Naperville, IL
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL
| | - Colin Palmer
- Pat MacPherson Centre for Pharmacogenetics and Pharmacogenomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, U.K
| | - Donald W Bowden
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC
| | | | - David Siscovick
- Institute for Urban Health, New York Academy of Medicine, New York, NY
| | - Mary Frances Cotch
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Jie Jin Wang
- Duke-NUS Medical School, Singapore
- Centre for Vision Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Kathryn P Burdon
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Tien Y Wong
- Duke-NUS Medical School, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Barbara E K Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI
| | - Ronald Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, LA BioMed and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
| | - Sudha K Iyengar
- Department of Population and Quantitative Health Sciences, Case Western University, Cleveland, OH
| | - Alkes L Price
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Lucia Sobrin
- Massachusetts Eye and Ear Department of Ophthalmology, Harvard Medical School, Boston, MA
| |
Collapse
|
24
|
Meng W, Shah KP, Pollack S, Toppila I, Hebert HL, McCarthy MI, Groop L, Ahlqvist E, Lyssenko V, Agardh E, Daniell M, Kaidonis G, Craig JE, Mitchell P, Liew G, Kifley A, Wang JJ, Christiansen MW, Jensen RA, Penman A, Hancock HA, Chen CJ, Correa A, Kuo JZ, Li X, Chen YDI, Rotter JI, Klein R, Klein B, Wong TY, Morris AD, Doney AS, Colhoun HM, Price AL, Burdon KP, Groop PH, Sandholm N, Grassi MA, Sobrin L, Palmer CN. A genome-wide association study suggests new evidence for an association of the NADPH Oxidase 4 (NOX4) gene with severe diabetic retinopathy in type 2 diabetes. Acta Ophthalmol 2018; 96:e811-e819. [PMID: 30178632 PMCID: PMC6263819 DOI: 10.1111/aos.13769] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 03/01/2018] [Indexed: 12/29/2022]
Abstract
Purpose Diabetic retinopathy is the most common eye complication in patients with diabetes. The purpose of this study is to identify genetic factors contributing to severe diabetic retinopathy. Methods A genome‐wide association approach was applied. In the Genetics of Diabetes Audit and Research in Tayside Scotland (GoDARTS) datasets, cases of severe diabetic retinopathy were defined as type 2 diabetic patients who were ever graded as having severe background retinopathy (Level R3) or proliferative retinopathy (Level R4) in at least one eye according to the Scottish Diabetic Retinopathy Grading Scheme or who were once treated by laser photocoagulation. Controls were diabetic individuals whose longitudinal retinopathy screening records were either normal (Level R0) or only with mild background retinopathy (Level R1) in both eyes. Significant Single Nucleotide Polymorphisms (SNPs) were taken forward for meta‐analysis using multiple Caucasian cohorts. Results Five hundred and sixty cases of type 2 diabetes with severe diabetic retinopathy and 4,106 controls were identified in the GoDARTS cohort. We revealed that rs3913535 in the NADPH Oxidase 4 (NOX4) gene reached a p value of 4.05 × 10−9. Two nearby SNPs, rs10765219 and rs11018670 also showed promising p values (p values = 7.41 × 10−8 and 1.23 × 10−8, respectively). In the meta‐analysis using multiple Caucasian cohorts (excluding GoDARTS), rs10765219 and rs11018670 showed associations for diabetic retinopathy (p = 0.003 and 0.007, respectively), while the p value of rs3913535 was not significant (p = 0.429). Conclusion This genome‐wide association study of severe diabetic retinopathy suggests new evidence for the involvement of the NOX4 gene.
Collapse
|
25
|
Kowluru RA, Mishra M. Therapeutic targets for altering mitochondrial dysfunction associated with diabetic retinopathy. Expert Opin Ther Targets 2018; 22:233-245. [PMID: 29436254 PMCID: PMC6088375 DOI: 10.1080/14728222.2018.1439921] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Retinopathy remains as one of the most feared blinding complications of diabetes, and with the prevalence of this life-long disease escalating at an alarming rate, the incidence of retinopathy is also climbing. Although the cutting edge research has identified many molecular mechanisms associated with its development, the exact mechanism how diabetes damages the retina remains obscure, limiting therapeutic options for this devastating disease. Areas covered: This review focuses on the central role of mitochondrial dysfunction/damage in the pathogenesis of diabetic retinopathy, and how damaged mitochondria initiates a self-perpetuating vicious cycles of free radicals. We have also reviewed how mitochondria could serve as a therapeutic target, and the challenges associated with the complex double mitochondrial membranes and a well-defined blood-retinal barrier for optimal pharmacologic/molecular approach to improve mitochondrial function. Expert opinion: Mitochondrial dysfunction provides many therapeutic targets for ameliorating the development of diabetic retinopathy including their biogenesis, DNA damage and epigenetic modifications. New technology to enhance pharmaceuticals uptake inside the mitochondria, nanotechnology to deliver drugs to the retina, and maintenance of mitochondrial homeostasis via lifestyle changes and novel therapeutics to prevent epigenetic modifications, could serve as some of the welcoming avenues for a diabetic patient to target this sight-threatening disease.
Collapse
Affiliation(s)
- Renu A Kowluru
- a Department of Ophthalmology, Kresge Eye Institute , Wayne State University , Detroit , MI , USA
| | - Manish Mishra
- a Department of Ophthalmology, Kresge Eye Institute , Wayne State University , Detroit , MI , USA
| |
Collapse
|
26
|
Mukhopadhyay N, Noble JA, Govil M, Marazita ML, Greenberg DA. Identifying genetic risk loci for diabetic complications and showing evidence for heterogeneity of type 1 diabetes based on complications risk. PLoS One 2018; 13:e0192696. [PMID: 29444168 PMCID: PMC5812614 DOI: 10.1371/journal.pone.0192696] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 01/29/2018] [Indexed: 12/13/2022] Open
Abstract
There is a growing body of evidence suggesting that type 1 diabetes (T1D) is a genetically heterogeneous disease. However, the extent of this heterogeneity, and what observations may distinguish different forms, is unclear. One indicator may be T1D-related microvascular complications (MVCs), which are familial, but occur in some families, and not others. We tested the hypothesis that T1D plus MVC is genetically distinct from T1D without MCV. We studied 415 families (2,462 individuals, 896 with T1D) using genome-wide linkage analysis, comparing families with and without MVC. We also tested for interaction between identified loci and alleles at the HLA-DRB1 locus. We found significant linkage scores at 1p36.12, 1q32.1, 8q21.3, 12p11.21 and 22q11.21. In all regions except 1p36.12, linkage scores differed between MVC-based phenotype groups, suggesting that families with MVCs express different genetic influences than those without. Our linkage results also suggested gene-gene interaction between the above putative loci and the HLA region; HLA-based strata produced significantly increased linkage scores in some strata, but not others within a phenotype group. We conclude that families with type 1 diabetes plus MVCs are genetically different from those with diabetes alone.
Collapse
Affiliation(s)
- Nandita Mukhopadhyay
- Center for Craniofacial and Dental Genetics, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| | - Janelle A. Noble
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
| | - Manika Govil
- Center for Craniofacial and Dental Genetics, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mary L. Marazita
- Center for Craniofacial and Dental Genetics, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Clinical and Translational Science Institute, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - David A. Greenberg
- Battelle Center for Mathematical Medicine, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, Wexner Medical Center, Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
27
|
Eshaq RS, Aldalati AMZ, Alexander JS, Harris NR. Diabetic retinopathy: Breaking the barrier. PATHOPHYSIOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY FOR PATHOPHYSIOLOGY 2017; 24:229-241. [PMID: 28732591 PMCID: PMC5711541 DOI: 10.1016/j.pathophys.2017.07.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 06/26/2017] [Accepted: 07/04/2017] [Indexed: 12/28/2022]
Abstract
Diabetic retinopathy (DR) remains a major complication of diabetes and a leading cause of blindness among adults worldwide. DR is a progressive disease affecting both type I and type II diabetic patients at any stage of the disease, and targets the retinal microvasculature. DR results from multiple biochemical, molecular and pathophysiological changes to the retinal vasculature, which affect both microcirculatory functions and ultimately photoreceptor function. Several neural, endothelial, and support cell (e.g., pericyte) mechanisms are altered in a pathological fashion in the hyperglycemic environment during diabetes that can disturb important cell surface components in the vasculature producing the features of progressive DR pathophysiology. These include loss of the glycocalyx, blood-retinal barrier dysfunction, increased expression of inflammatory cell markers and adhesion of blood leukocytes and platelets. Included in this review is a discussion of modifications that occur at or near the surface of the retinal vascular endothelial cells, and the consequences of these alterations on the integrity of the retina.
Collapse
Affiliation(s)
- Randa S Eshaq
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center -Shreveport, 1501 Kings Highway, Shreveport, LA 71130, United States
| | - Alaa M Z Aldalati
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center -Shreveport, 1501 Kings Highway, Shreveport, LA 71130, United States
| | - J Steven Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center -Shreveport, 1501 Kings Highway, Shreveport, LA 71130, United States
| | - Norman R Harris
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center -Shreveport, 1501 Kings Highway, Shreveport, LA 71130, United States.
| |
Collapse
|
28
|
Ren J, He T, Li Y, Liu S, Du Y, Jiang Y, Wu C. Network-based regularization for high dimensional SNP data in the case-control study of Type 2 diabetes. BMC Genet 2017; 18:44. [PMID: 28511641 PMCID: PMC5434559 DOI: 10.1186/s12863-017-0495-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 03/25/2017] [Indexed: 12/02/2022] Open
Abstract
Background Over the past decades, the prevalence of type 2 diabetes mellitus (T2D) has been steadily increasing around the world. Despite large efforts devoted to better understand the genetic basis of the disease, the identified susceptibility loci can only account for a small portion of the T2D heritability. Some of the existing approaches proposed for the high dimensional genetic data from the T2D case–control study are limited by analyzing a few number of SNPs at a time from a large pool of SNPs, by ignoring the correlations among SNPs and by adopting inefficient selection techniques. Methods We propose a network constrained regularization method to select important SNPs by taking the linkage disequilibrium into account. To accomodate the case control study, an iteratively reweighted least square algorithm has been developed within the coordinate descent framework where optimization of the regularized logistic loss function is performed with respect to one parameter at a time and iteratively cycle through all the parameters until convergence. Results In this article, a novel approach is developed to identify important SNPs more effectively through incorporating the interconnections among them in the regularized selection. A coordinate descent based iteratively reweighed least squares (IRLS) algorithm has been proposed. Conclusions Both the simulation study and the analysis of the Nurses’s Health Study, a case–control study of type 2 diabetes data with high dimensional SNP measurements, demonstrate the advantage of the network based approach over the competing alternatives.
Collapse
Affiliation(s)
- Jie Ren
- Department of Statistics, Kansas State University, 1116 Mid-Campus Drive N., 66506, Manhattan, KS, USA
| | - Tao He
- Department of Mathematics, San Francisco State University, San Francisco, CA, USA
| | - Ye Li
- Department of Biostatistics, Yale University, New Haven, CT, USA
| | - Sai Liu
- Division of Nephrology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Yinhao Du
- Department of Statistics, Kansas State University, 1116 Mid-Campus Drive N., 66506, Manhattan, KS, USA
| | - Yu Jiang
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| | - Cen Wu
- Department of Statistics, Kansas State University, 1116 Mid-Campus Drive N., 66506, Manhattan, KS, USA.
| |
Collapse
|
29
|
Villegas-Ruiz V, Hendlmeier F, Buentello-Volante B, Rodríguez-Loaiza JL, Miranda-Duarte A, Zenteno JC. Genome-wide mRNA analysis reveals a TUBD1 isoform profile as a potential biomarker for diabetic retinopathy development. Exp Eye Res 2017; 155:99-106. [DOI: 10.1016/j.exer.2017.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 12/23/2016] [Accepted: 01/21/2017] [Indexed: 01/23/2023]
|
30
|
Wang JH, Ling D, Tu L, van Wijngaarden P, Dusting GJ, Liu GS. Gene therapy for diabetic retinopathy: Are we ready to make the leap from bench to bedside? Pharmacol Ther 2017; 173:1-18. [PMID: 28132907 DOI: 10.1016/j.pharmthera.2017.01.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Diabetic retinopathy (DR), a chronic and progressive complication of diabetes mellitus, is a sight-threatening disease characterized in the early stages by neuronal and vascular dysfunction in the retina, and later by neovascularization that further damages vision. A major contributor to the pathology is excess production of vascular endothelial growth factor (VEGF), a growth factor that induces formation of new blood vessels and increases permeability of existing vessels. Despite the recent availability of effective treatments for the disease, including laser photocoagulation and therapeutic VEGF antibodies, DR remains a significant cause of vision loss worldwide. Existing anti-VEGF agents, though generally effective, are limited by their short therapeutic half-lives, necessitating frequent intravitreal injections and the risk of attendant adverse events. Management of DR with gene therapies has been proposed for several years, and pre-clinical studies have yielded enticing findings. Gene therapy holds several advantages over conventional treatments for DR, such as a longer duration of therapeutic effect, simpler administration, the ability to intervene at an earlier stage of the disease, and potentially fewer side-effects. In this review, we summarize the current understanding of the pathophysiology of DR and provide an overview of research into DR gene therapies. We also examine current barriers to the clinical application of gene therapy for DR and evaluate future prospects for this approach.
Collapse
Affiliation(s)
- Jiang-Hui Wang
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia
| | - Damien Ling
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Discipline of Ophthalmology, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Leilei Tu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Peter van Wijngaarden
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia
| | - Gregory J Dusting
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia
| | - Guei-Sheung Liu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia; Menzies Institute for Medical Research, University of Tasmania, Tasmania, Australia.
| |
Collapse
|
31
|
Wu H, Wu H, Shi L, Yuan X, Yin Y, Yuan M, Zhou Y, Hu Q, Jiang K, Dong J. The Association of Haptoglobin Gene Variants and Retinopathy in Type 2 Diabetic Patients: A Meta-Analysis. J Diabetes Res 2017; 2017:2195059. [PMID: 28758129 PMCID: PMC5512055 DOI: 10.1155/2017/2195059] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 05/23/2017] [Indexed: 11/25/2022] Open
Abstract
AIMS/INTRODUCTION To collectively evaluate the association between haptoglobin (Hp) gene variants and diabetic retinopathy (DR) in patients with type 2 diabetes mellitus (T2DM). METHODS A comprehensive literature review was performed for eligible studies. After inclusion and exclusion selection as well as quality assessment, those studies meeting quality standards were included. In this study, diabetic patients with retinopathy were selected as the case group and those ones without DR were treated as the control group. The recessive model, allele model, additive model, heterozygote model, and homozygote model were utilized to investigate the association of three Hp gene variants and DR. Subgroup analysis on different severity of DR including nonproliferative diabetic retinopathy (NPDR) and proliferative diabetic retinopathy (PDR) was also conducted. RESULTS Six trials from different regions were finally included. A total of 1145 subjects containing 564 T2DM patients with retinopathy were included. The recessive model, allele model, additive model, and homozygote model results showed that Hp gene variants were not associated with DR, NPDR, and PDR. However, the heterozygote model indicated the association of Hp gene variants with DR. CONCLUSIONS No association was found between the Hp gene variants and PDR and NPDR. More studies are required to verify these findings.
Collapse
Affiliation(s)
- Huiqun Wu
- Department of Medical Informatics, Medical School of Nantong University, Nantong 226001, China
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Huan Wu
- Department of Medical Informatics, Medical School of Nantong University, Nantong 226001, China
| | - Lili Shi
- Department of Medical Informatics, Medical School of Nantong University, Nantong 226001, China
| | - Xinlu Yuan
- Department of Endocrinology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Ying Yin
- Department of Medical Informatics, Medical School of Nantong University, Nantong 226001, China
| | - Mingjie Yuan
- Department of Medical Informatics, Medical School of Nantong University, Nantong 226001, China
| | - Yushan Zhou
- Department of Medical Informatics, Medical School of Nantong University, Nantong 226001, China
| | - Qianwen Hu
- Department of Medical Informatics, Medical School of Nantong University, Nantong 226001, China
| | - Kui Jiang
- Department of Medical Informatics, Medical School of Nantong University, Nantong 226001, China
- *Kui Jiang:
| | - Jiancheng Dong
- Department of Medical Informatics, Medical School of Nantong University, Nantong 226001, China
| |
Collapse
|
32
|
Abstract
Diabetic retinopathy (DR) is a serious complication of diabetes, which is fast reaching epidemic proportions worldwide. While tight glycemic control remains the standard of care for preventing the progression of DR, better insights into DR etiology require understanding its genetic basis, which in turn may assist in the design of novel treatments. During the last decade, genomic medicine is increasingly being applied to common multifactorial diseases such as diabetes and age-related macular degeneration. The contribution of genetics to the initiation and progression of DR has been recognized for some time, but the involvement of specific genes and genetic variants remains elusive. Several investigations are currently underway for identifying DR susceptibility loci through linkage studies, candidate gene approaches, and genome-wide association studies. Advent of next generation sequencing and high throughput genomic technologies, development of novel bioinformatics tools and collaborations among research teams should facilitate such investigations. Here, we review the current state of genetic studies in DR and discuss reported findings in the context of biochemical, cell biological and therapeutic advances. We propose the development of a consortium in India for genetic studies with large cohorts of patients and controls from limited geographical areas to stratify the impact of the environment. Uniform guidelines should be established for clinical phenotyping and data collection. These studies would permit identification of genetic loci for DR susceptibility in the Indian population and should be valuable for better diagnosis and prognosis, and for clinical management of this blinding disease.
Collapse
Affiliation(s)
| | - Anand Swaroop
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
33
|
Abstract
Diabetic retinopathy (DR), a leading cause of acquired vision loss, is a microvascular complication of diabetes. While traditional risk factors for diabetic retinopathy including longer duration of diabetes, poor blood glucose control, and dyslipidemia are helpful in stratifying patient's risk for developing retinopathy, many patients without these traditional risk factors develop DR; furthermore, there are persons with long diabetes duration who do not develop DR. Thus, identifying biomarkers to predict DR or to determine therapeutic response is important. A biomarker can be defined as a characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention. Incorporation of biomarkers into risk stratification of persons with diabetes would likely aid in early diagnosis and guide treatment methods for those with DR or with worsening DR. Systemic biomarkers of DR include serum measures including genomic, proteomic, and metabolomics biomarkers. Ocular biomarkers including tears and vitreous and retinal vascular structural changes have also been studied extensively to prognosticate the risk of DR development. The current studies on biomarkers are limited by the need for larger sample sizes, cross-validation in different populations and ethnic groups, and time-efficient and cost-effective analytical techniques. Future research is important to explore novel DR biomarkers that are non-invasive, rapid, economical, and accurate to help reduce the incidence and progression of DR in people with diabetes.
Collapse
Affiliation(s)
- Daniel Shu Wei Ting
- Singapore National Eye Center, 11 Third Hospital Avenue, Singapore, 168751, Singapore
- Singapore Eye Research Institute, Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Kara-Anne Tan
- Singapore National Eye Center, 11 Third Hospital Avenue, Singapore, 168751, Singapore
| | - Val Phua
- Singapore National Eye Center, 11 Third Hospital Avenue, Singapore, 168751, Singapore
| | - Gavin Siew Wei Tan
- Singapore National Eye Center, 11 Third Hospital Avenue, Singapore, 168751, Singapore
- Singapore Eye Research Institute, Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Chee Wai Wong
- Singapore National Eye Center, 11 Third Hospital Avenue, Singapore, 168751, Singapore
- Singapore Eye Research Institute, Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Tien Yin Wong
- Singapore National Eye Center, 11 Third Hospital Avenue, Singapore, 168751, Singapore.
- Singapore Eye Research Institute, Singapore, Singapore.
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
34
|
Priščáková P, Minárik G, Repiská V. Candidate gene studies of diabetic retinopathy in human. Mol Biol Rep 2016; 43:1327-1345. [PMID: 27730450 PMCID: PMC5102952 DOI: 10.1007/s11033-016-4075-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 09/09/2016] [Indexed: 12/13/2022]
Abstract
Diabetic retinopathy (DR) is a multifactorial disease with complex pathophysiology. It is the main cause of blindness among the people in productive age. The purpose of this literature review is to highlight recent achievements in the genetics of diabetic retinopathy with particular focus on candidate gene studies. We summarized most of the available published data about candidate genes for diabetic retinopathy with the goal to identify main genetic aspects. We conclude that genetic studies reported contradictory findings and no genetic variants meet criteria of a diagnostic marker, or significantly elucidate the root of DR development. Based on these findings it is important to continue with the research in the field of DR genetics, mainly due to the fact that currently new possibilities and approaches associated with utilization of next-generation sequencing are available.
Collapse
Affiliation(s)
- Petra Priščáková
- Faculty of Medicine, Institute of Medical Biology, Genetics and Clinical Genetics, University Hospital Bratislava, Comenius University in Bratislava, Sasinkova 4, 81108, Bratislava, Slovakia
| | - Gabriel Minárik
- Medirex Group Academy n.o., Galvaniho 17/C, 82016, Bratislava, Slovakia
| | - Vanda Repiská
- Faculty of Medicine, Institute of Medical Biology, Genetics and Clinical Genetics, University Hospital Bratislava, Comenius University in Bratislava, Sasinkova 4, 81108, Bratislava, Slovakia.
| |
Collapse
|
35
|
Rajavi Z, Safi S, Javadi MA, Azarmina M, Moradian S, Entezari M, Nourinia R, Ahmadieh H, Shirvani A, Shahraz S, Ramezani A, Dehghan MH, Shahsavari M, Soheilian M, Nikkhah H, Ziaei H, Behboudi H, Farrahi F, Falavarjani KG, Parvaresh MM, Fesharaki H, Abrishami M, Shoeibi N, Rahimi M, Javadzadeh A, Karkhaneh R, Riazi-Esfahani M, Manaviat MR, Maleki A, Kheiri B, Golbafian F. Diabetic Retinopathy Clinical Practice Guidelines: Customized for Iranian Population. J Ophthalmic Vis Res 2016; 11:394-414. [PMID: 27994809 PMCID: PMC5139552 DOI: 10.4103/2008-322x.194131] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/24/2016] [Indexed: 12/12/2022] Open
Abstract
PURPOSE To customize clinical practice guidelines (CPGs) for management of diabetic retinopathy (DR) in the Iranian population. METHODS Three DR CPGs (The Royal College of Ophthalmologists 2013, American Academy of Ophthalmology [Preferred Practice Pattern 2012], and Australian Diabetes Society 2008) were selected from the literature using the AGREE tool. Clinical questions were designed and summarized into four tables by the customization team. The components of the clinical questions along with pertinent recommendations extracted from the above-mentioned CPGs; details of the supporting articles and their levels of evidence; clinical recommendations considering clinical benefits, cost and side effects; and revised recommendations based on customization capability (applicability, acceptability, external validity) were recorded in 4 tables, respectively. Customized recommendations were sent to the faculty members of all universities across the country to score the recommendations from 1 to 9. RESULTS Agreed recommendations were accepted as the final recommendations while the non-agreed ones were approved after revision. Eventually, 29 customized recommendations under three major categories consisting of screening, diagnosis and treatment of DR were developed along with their sources and levels of evidence. CONCLUSION This customized CPGs for management of DR can be used to standardize the referral pathway, diagnosis and treatment of patients with diabetic retinopathy.
Collapse
Affiliation(s)
- Zhale Rajavi
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Ophthalmic Epidemiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sare Safi
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Ophthalmic Epidemiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Javadi
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Azarmina
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siamak Moradian
- Ophthalmic Epidemiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Morteza Entezari
- Ophthalmic Epidemiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramin Nourinia
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Ahmadieh
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Armin Shirvani
- Standardization and CPG Development Office, Deputy of Curative Affairs, Ministry of Health and Medical Education, Tehran, Iran
| | | | - Alireza Ramezani
- Ophthalmic Epidemiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Ophthalmology, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Dehghan
- Ophthalmic Epidemiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Ophthalmology, Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Shahsavari
- Department of Ophthalmology, Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Soheilian
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Ophthalmology, Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homayoun Nikkhah
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Ophthalmology, Torfeh Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Ziaei
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Ophthalmic Epidemiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hasan Behboudi
- Department of Ophthalmology, Gilan University of Medical Sciences, Rasht, Iran
| | - Fereydoun Farrahi
- Department of Ophthalmology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Mohammad Mehdi Parvaresh
- Department of Ophthalmology, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Hamid Fesharaki
- Department of Ophthalmology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Majid Abrishami
- Department of Ophthalmology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nasser Shoeibi
- Department of Ophthalmology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mansour Rahimi
- Department of Ophthalmology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Javadzadeh
- Department of Ophthalmology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Karkhaneh
- Department of Ophthalmology, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Riazi-Esfahani
- Department of Ophthalmology, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Alireza Maleki
- Department of Ophthalmology, Al Zahra Eye Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Bahareh Kheiri
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
36
|
Fan Y, Fu YY, Chen Z, Hu YY, Shen J. Gene-gene interaction of erythropoietin gene polymorphisms and diabetic retinopathy in Chinese Han. Exp Biol Med (Maywood) 2016; 241:1524-30. [PMID: 27190272 DOI: 10.1177/1535370216645210] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 03/09/2016] [Indexed: 11/16/2022] Open
Abstract
The aim of this study was to investigate the association of three single nucleotide polymorphisms in the erythropoietin gene polymorphisms with diabetic retinopathy and additional role of gene-gene interaction on diabetic retinopathy risk. A total of 1193 patients (579 men, 614 women) with type 2 diabetes mellitus were selected, including 397 diabetic retinopathy patients and 796 controls (type 2 diabetes mellitus patients without diabetic retinopathy); the mean age of all participants was 56.7 ± 13.9 years. Three single nucleotide polymorphisms were selected: rs507392, rs1617640, and rs551238. The t-test was used for comparison of erythropoietin protein level erythropoietin levels in patients having different erythropoietin genotypes. Logistic regression model was used to examine the association between three single nucleotide polymorphisms and diabetic retinopathy. Odds ratio (OR) and 95% confident interval (95% CI) were calculated. Generalized multifactor dimensionality reduction was employed to analyze the impact of interaction among three single nucleotide polymorphisms on CVD risk. After covariates adjustment, the carriers of homozygous mutant of three single nucleotide polymorphisms have higher diabetic retinopathy risk than those with wild-type homozygotes, OR (95% CI) were 2.04 (1.12-2.35), 1.87 (1.10-2.41) and 1.15 (1.06-1.76), respectively. Generalized multifactor dimensionality reduction model indicated a significant three-locus model (p = 0.0010) involving rs507392, rs1617640, and rs551238. Overall, the three-locus models had a cross-validation consistency of 10 of 10, and had the testing accuracy of 60.72%. Subjects with TC or CC-TG or GG-AC or CC genotype have the highest diabetic retinopathy risk. In conclusion, our results support an important association of rs507392, rs1617640 and rs551238 minor allele of erythropoietin with increased diabetic retinopathy risk, and additional interaction among three single nucleotide polymorphisms.
Collapse
Affiliation(s)
- YanFei Fan
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Southern Medical University, No. 183, West Zhongshan Rd, Guangzhou 510500, China
| | - Yin-Yu Fu
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Southern Medical University, No. 183, West Zhongshan Rd, Guangzhou 510500, China
| | - Zhi Chen
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Southern Medical University, No. 183, West Zhongshan Rd, Guangzhou 510500, China
| | - Yuan-Yuan Hu
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Southern Medical University, No. 183, West Zhongshan Rd, Guangzhou 510500, China
| | - Jie Shen
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Southern Medical University, No. 183, West Zhongshan Rd, Guangzhou 510500, China
| |
Collapse
|
37
|
Ting DSW, Cheung GCM, Wong TY. Diabetic retinopathy: global prevalence, major risk factors, screening practices and public health challenges: a review. Clin Exp Ophthalmol 2016; 44:260-77. [DOI: 10.1111/ceo.12696] [Citation(s) in RCA: 444] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Revised: 12/07/2015] [Accepted: 12/21/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Daniel Shu Wei Ting
- Singapore National Eye Center, Singapore Health Service (SingHealth); Singapore Singapore
- Singapore Eye Research Institute; Singapore Singapore
| | - Gemmy Chui Ming Cheung
- Singapore National Eye Center, Singapore Health Service (SingHealth); Singapore Singapore
- Singapore Eye Research Institute; Singapore Singapore
- Duke-NUS Graduate Medical School; Singapore Singapore
| | - Tien Yin Wong
- Singapore National Eye Center, Singapore Health Service (SingHealth); Singapore Singapore
- Singapore Eye Research Institute; Singapore Singapore
- Duke-NUS Graduate Medical School; Singapore Singapore
| |
Collapse
|
38
|
Davoudi S, Sobrin L. Novel Genetic Actors of Diabetes-Associated Microvascular Complications: Retinopathy, Kidney Disease and Neuropathy. Rev Diabet Stud 2016; 12:243-59. [PMID: 26859656 DOI: 10.1900/rds.2015.12.243] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Both type 1 and type 2 diabetes mellitus can lead to the common microvascular complications of diabetic retinopathy, kidney disease, and neuropathy. Diabetic patients do not universally develop these complications. Long duration of diabetes and poor glycemic control explain a lot of the variability in the development of microvascular complications, but not all. Genetic factors account for some of the remaining variability because of the heritability and familial clustering of these complications. There have been a large number of investigations, including linkage studies, candidate gene studies, and genome-wide association studies, all of which have sought to identify the specific variants that increase susceptibility. For retinopathy, several genome-wide association studies have been performed in small or midsize samples, but no reproducible loci across the studies have been identified. For diabetic kidney disease, genome-wide association studies in larger samples have been performed, and loci for this complication are beginning to emerge. However, validation of the existing discoveries, and further novel discoveries in larger samples is ongoing. The amount of genetic research into diabetic neuropathy has been very limited, and much is dedicated to the understanding of genetic risk factors only. Collaborations that pool samples and aim to detect phenotype classifications more precisely are promising avenues for a better explanation of the genetics of diabetic microvascular complications.
Collapse
Affiliation(s)
- Samaneh Davoudi
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, 243 Charles Street, Boston, MA 02114, USA
| | - Lucia Sobrin
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, 243 Charles Street, Boston, MA 02114, USA
| |
Collapse
|
39
|
Hampton BM, Schwartz SG, Brantley MA, Flynn HW. Update on genetics and diabetic retinopathy. Clin Ophthalmol 2015; 9:2175-93. [PMID: 26648684 PMCID: PMC4664538 DOI: 10.2147/opth.s94508] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Clinical risk factors for diabetic retinopathy (DR), such as duration of disease and degree of glucose control, do not adequately predict disease progression in individual patients, suggesting the presence of a genetic component. Multiple smaller studies have investigated genotype–phenotype correlations in genes encoding vascular endothelial growth factor, aldose reductase, the receptor for advanced glycation end products, and many others. In general, reported results have been conflicting, due to factors including small sample sizes, variations in study design, differences in clinical end points, and underlying genetic differences between study groups. At this time, there is no confirmed association with any risk allele reported. As we continue to collect data from additional studies, the role of genetics in DR may become more apparent.
Collapse
Affiliation(s)
- Blake M Hampton
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Stephen G Schwartz
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Milam A Brantley
- Department of Ophthalmology, Vanderbilt Eye Institute, Nashville, TN, USA
| | - Harry W Flynn
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
40
|
Thorsen SU, Sandahl K, Nielsen LB, Broe R, Rasmussen ML, Peto T, Grauslund J, Andersen MLM, Mortensen HB, Pociot F, Olsen BS, Brorsson C. Polymorphisms in the CTSH gene may influence the progression of diabetic retinopathy: a candidate-gene study in the Danish Cohort of Pediatric Diabetes 1987 (DCPD1987). Graefes Arch Clin Exp Ophthalmol 2015; 253:1959-65. [PMID: 26245339 DOI: 10.1007/s00417-015-3118-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 06/16/2015] [Accepted: 07/18/2015] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The incidence of type 1 diabetes mellitus (T1DM) is increasing globally, and as a consequence, more patients are affected by microvascular complications such as diabetic retinopathy (DR). The aim of this study was to elucidate possible associations between diabetes-related single-nucleotide polymorphisms (SNP) and the development of DR. METHODS Three hundred and thirty-nine patients with T1DM from the Danish Cohort of Pediatric Diabetes 1987 (DCPD1987) went through an ophthalmic examination in 1995; 185 of these were reexamined in 2011. The development of DR was assessed by comparison of overall DR level between baseline and follow-up in the worst eye at baseline. Patients were graded on a modified version of the Early Treatment Diabetic Retinopathy Study (ETDRS) scale, and 20 SNPs were genotyped in 130 of the 185 patients. RESULTS We found the CTSH/rs3825932 variant (C > T) was associated with reduced risk of progression to proliferative diabetic retinopathy (PDR) (OR [95 % CI] = 0.20 [0.07-0.56], p = 2.4 × 10(-3), padjust = 0.048) and ERBB3/rs2292239 variant (G > T) associated with increased risk of two-step progression (OR [95 % CI] = 2.76 [1.31-5.80], p = 7.5 × 10(-3), padjust = 0.15). The associations were independent of other known risk factors, such as HbA1c, sex, and diastolic blood pressure. CONCLUSION In conclusion, CTSH/rs3825932 and ERBB3/rs2292239 SNPs were associated with reduced risk of progression to PDR and two-step progression of DR on the ETDRS scale accordingly. The variant CTSH remained statistically significant after adjusting for multiple testing. Our results suggest an overlap between genetic variants that confer risk of T1DM and progression of DR.
Collapse
Affiliation(s)
- Steffen U Thorsen
- Department of Peadiatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730, Herlev, Denmark
| | - Kristian Sandahl
- Department of Peadiatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730, Herlev, Denmark.
| | - Lotte B Nielsen
- Department of Peadiatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730, Herlev, Denmark
| | - Rebecca Broe
- Department of Ophthalmology, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense C, Denmark.,The Clinical Research Institute, University of Southern Denmark, Odense, Denmark.,OPEN Odense Patient data Explorative Network, Odense University Hospital, Odense, Denmark
| | - Malin L Rasmussen
- Department of Ophthalmology, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense C, Denmark.,The Clinical Research Institute, University of Southern Denmark, Odense, Denmark
| | - Tunde Peto
- The Clinical Research Institute, University of Southern Denmark, Odense, Denmark.,NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Jakob Grauslund
- Department of Ophthalmology, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense C, Denmark.,The Clinical Research Institute, University of Southern Denmark, Odense, Denmark
| | - Marie L M Andersen
- Department of Peadiatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730, Herlev, Denmark
| | - Henrik B Mortensen
- Department of Peadiatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730, Herlev, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Flemming Pociot
- Department of Peadiatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730, Herlev, Denmark
| | - Birthe S Olsen
- Department of Peadiatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730, Herlev, Denmark
| | - Caroline Brorsson
- Department of Peadiatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730, Herlev, Denmark
| |
Collapse
|
41
|
Peng D, Wang J, Zhang R, Jiang F, Tang S, Chen M, Yan J, Sun X, Wang S, Wang T, Yan D, Bao Y, Hu C, Jia W. Common variants in or near ZNRF1, COLEC12, SCYL1BP1 and API5 are associated with diabetic retinopathy in Chinese patients with type 2 diabetes. Diabetologia 2015; 58:1231-8. [PMID: 25819896 DOI: 10.1007/s00125-015-3569-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 03/05/2015] [Indexed: 10/23/2022]
Abstract
AIMS/HYPOTHESIS Three recent genome-wide association studies (GWAS) identified several single-nucleotide polymorphisms (SNPs) with modest effects on diabetic retinopathy in Mexican-American and white patients with diabetes. This study aimed to evaluate the effects of these variants on diabetic retinopathy in Chinese patients with type 2 diabetes. METHODS A total of 1,972 patients with type 2 diabetes were recruited to this study, including 819 patients with diabetic retinopathy and 1,153 patients with diabetes of ≥5 years duration but without retinopathy. Forty SNPs associated with diabetic retinopathy in three GWAS were genotyped. Fundus photography was performed to diagnose and classify diabetic retinopathy. RESULTS rs17684886 in ZNRF1 and rs599019 near COLEC12 were associated with diabetic retinopathy (OR 0.812, p = 0.0039 and OR 0.835, p = 0.0116, respectively) and with the severity of diabetic retinopathy (p = 0.0365 and p = 0.0252, respectively, for trend analysis). Sub-analysis in patients with diabetic retinopathy revealed that rs6427247 near SCYL1BP1 (also known as GORAB) and rs899036 near API5 were associated with severe diabetic retinopathy (OR 1.368, p = 0.0333 and OR 0.340, p = 0.0005, respectively). The associations between rs6427247 and rs899036 and severe diabetic retinopathy became more evident after a meta-analysis of published GWAS data (OR 1.577, p = 2.01 × 10(-4) for rs6427247; OR 0.330, p = 5.84 × 10(-7) for rs899036). CONCLUSIONS/INTERPRETATION We determined that rs17684886 and rs599019 are associated with diabetic retinopathy and that rs6427247 and rs899036 are associated with severe diabetic retinopathy in Chinese patients with type 2 diabetes.
Collapse
Affiliation(s)
- Danfeng Peng
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kwak SH, Park KS. Genetic Studies on Diabetic Microvascular Complications: Focusing on Genome-Wide Association Studies. Endocrinol Metab (Seoul) 2015; 30:147-58. [PMID: 26194074 PMCID: PMC4508258 DOI: 10.3803/enm.2015.30.2.147] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 05/26/2015] [Accepted: 05/26/2015] [Indexed: 01/13/2023] Open
Abstract
Diabetes is a common metabolic disorder with a worldwide prevalence of 8.3% and is the leading cause of visual loss, end-stage renal disease and amputation. Recently, genome-wide association studies (GWASs) have identified genetic risk factors for diabetic microvascular complications of retinopathy, nephropathy, and neuropathy. We summarized the recent findings of GWASs on diabetic microvascular complications and highlighted the challenges and our opinion on future directives. Five GWASs were conducted on diabetic retinopathy, nine on nephropathy, and one on neuropathic pain. The majority of recent GWASs were underpowered and heterogeneous in terms of study design, inclusion criteria and phenotype definition. Therefore, few reached the genome-wide significance threshold and the findings were inconsistent across the studies. Recent GWASs provided novel information on genetic risk factors and the possible pathophysiology of diabetic microvascular complications. However, further collaborative efforts to standardize phenotype definition and increase sample size are necessary for successful genetic studies on diabetic microvascular complications.
Collapse
Affiliation(s)
- Soo Heon Kwak
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kyong Soo Park
- Department of Internal Medicine, Seoul National University Hospital; Depatment of Internal Medicine, Seoul National University College of Medicine; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea.
| |
Collapse
|
43
|
Oxidative stress and epigenetic modifications in the pathogenesis of diabetic retinopathy. Prog Retin Eye Res 2015; 48:40-61. [PMID: 25975734 DOI: 10.1016/j.preteyeres.2015.05.001] [Citation(s) in RCA: 233] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/29/2015] [Accepted: 05/01/2015] [Indexed: 12/21/2022]
Abstract
Diabetic retinopathy remains the major cause of blindness among working age adults. Although a number of metabolic abnormalities have been associated with its development, due to complex nature of this multi-factorial disease, a link between any specific abnormality and diabetic retinopathy remains largely speculative. Diabetes increases oxidative stress in the retina and its capillary cells, and overwhelming evidence suggests a bidirectional relationship between oxidative stress and other major metabolic abnormalities implicated in the development of diabetic retinopathy. Due to increased production of cytosolic reactive oxygen species, mitochondrial membranes are damaged and their membrane potentials are impaired, and complex III of the electron transport system is compromised. Suboptimal enzymatic and nonenzymatic antioxidant defense system further aids in the accumulation of free radicals. As the duration of the disease progresses, mitochondrial DNA (mtDNA) is damaged and the DNA repair system is compromised, and due to impaired transcription of mtDNA-encoded proteins, the integrity of the electron transport system is encumbered. Due to decreased mtDNA biogenesis and impaired transcription, superoxide accumulation is further increased, and the vicious cycle of free radicals continues to self-propagate. Diabetic milieu also alters enzymes responsible for DNA and histone modifications, and various genes important for mitochondrial homeostasis, including mitochondrial biosynthesis, damage and antioxidant defense, undergo epigenetic modifications. Although antioxidant administration in animal models has yielded encouraging results in preventing diabetic retinopathy, controlled longitudinal human studies remain to be conducted. Furthermore, the role of epigenetic in mitochondrial homeostasis suggests that regulation of such modifications also has potential to inhibit/retard the development of diabetic retinopathy.
Collapse
|
44
|
Song Q, Zhang Y, Wu Y, Zhou F, Qu Y. Association of erythropoietin gene polymorphisms with retinopathy in a Chinese cohort with type 2 diabetes mellitus. Clin Exp Ophthalmol 2015; 43:544-9. [PMID: 25675872 DOI: 10.1111/ceo.12505] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 01/13/2015] [Indexed: 12/18/2022]
Affiliation(s)
- Qi Song
- Operating Room; Qilu Hospital of Shandong University; Jinan China
| | - Yue Zhang
- Department of Geriatrics; Qilu Hospital of Shandong University; Jinan China
| | - Yongzhong Wu
- State Key Lab of Crystal Materials; Shandong University; Jinan China
| | - Fang Zhou
- Department of Geriatrics; Qilu Hospital of Shandong University; Jinan China
| | - Yi Qu
- Department of Geriatrics; Qilu Hospital of Shandong University; Jinan China
| |
Collapse
|
45
|
Abstract
The rising global prevalence of diabetes mellitus is accompanied by an increasing burden of morbidity and mortality that is attributable to the complications of chronic hyperglycaemia. These complications include blindness, renal failure and cardiovascular disease. Current therapeutic options for chronic hyperglycaemia reduce, but do not eradicate, the risk of these complications. Success in defining new preventative and therapeutic strategies hinges on an improved understanding of the molecular processes involved in the development of these complications. This Review explores the role of human genetics in delivering such insights, and describes progress in characterizing the sequence variants that influence individual predisposition to diabetic kidney disease, retinopathy, neuropathy and accelerated cardiovascular disease. Numerous risk variants for microvascular complications of diabetes have been reported, but very few have shown robust replication. Furthermore, only limited evidence exists of a difference in the repertoire of risk variants influencing macrovascular disease between those with and those without diabetes. Here, we outline the challenges associated with the genetic analysis of diabetic complications and highlight ongoing efforts to deliver biological insights that can drive translational benefits.
Collapse
|
46
|
Maghbooli Z, Hossein-nezhad A, Larijani B, Amini M, Keshtkar A. Global DNA methylation as a possible biomarker for diabetic retinopathy. Diabetes Metab Res Rev 2015; 31:183-9. [PMID: 25069700 DOI: 10.1002/dmrr.2584] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 06/26/2014] [Accepted: 07/04/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND We evaluated whether global levels of DNA methylation status were associated with retinopathy as well as providing a predictive role of DNA methylation in developing retinopathy in a case-control study of 168 patients with type 2 diabetes. METHODS The 5-methylcytosine content was assessed by reversed-phase high-pressure liquid chromatography of peripheral blood leukocytes to determine an individual's global DNA methylation status in the two groups, either with or without retinopathy. RESULTS The global DNA methylation levels were significantly higher in diabetic retinopathy patients compared with those in non-retinopathy patients (4.90 ± 0.12 vs. 4.22 ± 0.13, respectively; p = 0.001). There was a significant increasing trend in global DNA methylation levels in terms of progressing retinopathy (without retinopathy, 4.22 ± 0.13; non-proliferative diabetic retinopathy, 4.62 ± 0.17; proliferative diabetic retinopathy, 5.07 ± 0.21) (p = 0.006). Additionally, global DNA methylation independent of retinopathy risk factors, which include dyslipidaemia, hypertension, hyperglycaemia and duration of diabetes, was a predictive factor for retinopathy (OR = 1.53, p = 0.015). CONCLUSIONS Global DNA methylation is modulated during or possibly before the primary stage of diabetes. This observation verifies the metabolic memory effect of hyperglycaemia in early stage of an aetiological process that leads to type 2 diabetes and its associated complications.
Collapse
Affiliation(s)
- Zhila Maghbooli
- Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | |
Collapse
|
47
|
Agarwal A, Soliman MK, Sepah YJ, Do DV, Nguyen QD. Diabetic retinopathy: variations in patient therapeutic outcomes and pharmacogenomics. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2014; 7:399-409. [PMID: 25548526 PMCID: PMC4271791 DOI: 10.2147/pgpm.s52821] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Diabetes and its microvascular complications in patients poses a significant challenge and constitutes a major health problem. When it comes to manifestations in the eye, each case of diabetic retinopathy (DR) is unique, in terms of the phenotype, genotype, and, more importantly, the therapeutic response. It is therefore important to identify factors that distinguish one patient from another. Personalized therapy in DR is a new trend aimed at achieving maximum therapeutic response in patients by identifying genotypic and phenotypic factors that may result in less than optimal response to conventional therapy, and consequently, lead to poorer outcome. With advances in the identification of these genetic markers, such as gene polymorphisms and human leucocyte antigen associations, as well as development of drugs that can target their effects, the future of personalized medicine in DR is promising. In this comprehensive review, data from various studies have been analyzed to present what has been achieved in the field of pharmacogenomics thus far. An insight into future research is also provided.
Collapse
Affiliation(s)
- Aniruddha Agarwal
- Ocular Imaging Research and Reading Center, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, USA
| | - Mohamed K Soliman
- Ocular Imaging Research and Reading Center, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, USA
| | - Yasir J Sepah
- Ocular Imaging Research and Reading Center, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, USA
| | - Diana V Do
- Ocular Imaging Research and Reading Center, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, USA
| | - Quan Dong Nguyen
- Ocular Imaging Research and Reading Center, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, USA
| |
Collapse
|
48
|
Tang ZH, Wang L, Zeng F, Zhang K. Human genetics of diabetic retinopathy. J Endocrinol Invest 2014; 37:1165-74. [PMID: 25201002 DOI: 10.1007/s40618-014-0172-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 08/25/2014] [Indexed: 01/03/2023]
Abstract
There is evidence demonstrating that genetic factors contribute to the risk of diabetic retinopathy (DR). Genetics variants, structural variants (copy number variation, CNV) and epigenetic changes play important roles in the development of DR. Genetic linkage and association studies have uncovered a number of genetic loci and common genetic variants susceptibility to DR. CNV and interactions of gene by environment have also been detected by association analysis. Apart from nucleus genome, mitochondrial DNA plays critical roles in regulation of development of DR. Epigenetic studies have indicated epigenetic changes in chromatin affecting gene transcription in response to environmental stimuli, which provided a large body of evidence of regulating development of diabetes mellitus. Identification of genetic variants and epigenetic changes contributed to risk or protection of DR will benefit uncovering the complex mechanism underlying DR. This review focused on the current knowledge of the genetic and epigenetic basis of DR.
Collapse
Affiliation(s)
- Z-H Tang
- Department of Endocrinology and Metabolism, Shanghai Tongji Hospital, Tongji University School of Medicine, Room 517 Building 2nd, NO. 389 Xincun Road, Shanghai, 200063, China,
| | | | | | | |
Collapse
|
49
|
Awata T, Yamashita H, Kurihara S, Morita-Ohkubo T, Miyashita Y, Katayama S, Mori K, Yoneya S, Kohda M, Okazaki Y, Maruyama T, Shimada A, Yasuda K, Nishida N, Tokunaga K, Koike A. A genome-wide association study for diabetic retinopathy in a Japanese population: potential association with a long intergenic non-coding RNA. PLoS One 2014; 9:e111715. [PMID: 25364816 PMCID: PMC4218806 DOI: 10.1371/journal.pone.0111715] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 09/30/2014] [Indexed: 12/31/2022] Open
Abstract
Elucidation of the genetic susceptibility factors for diabetic retinopathy (DR) is important to gain insight into the pathogenesis of DR, and may help to define genetic risk factors for this condition. In the present study, we conducted a three-stage genome-wide association study (GWAS) to identify DR susceptibility loci in Japanese patients, which comprised a total of 837 type 2 diabetes patients with DR (cases) and 1,149 without DR (controls). From the stage 1 genome-wide scan of 446 subjects (205 cases and 241 controls) on 614,216 SNPs, 249 SNPs were selected for the stage 2 replication in 623 subjects (335 cases and 288 controls). Eight SNPs were further followed up in a stage 3 study of 297 cases and 620 controls. The top signal from the present association analysis was rs9362054 in an intron of RP1-90L14.1 showing borderline genome-wide significance (Pmet = 1.4×10−7, meta-analysis of stage 1 and stage 2, allele model). RP1-90L14.1 is a long intergenic non-coding RNA (lincRNA) adjacent to KIAA1009/QN1/CEP162 gene; CEP162 plays a critical role in ciliary transition zone formation before ciliogenesis. The present study raises the possibility that the dysregulation of ciliary-associated genes plays a role in susceptibility to DR.
Collapse
Affiliation(s)
- Takuya Awata
- Department of Endocrinology and Diabetes, Faculty of Medicine, Saitama Medical University, Saitama, Japan
- * E-mail:
| | - Hisakuni Yamashita
- Department of Endocrinology and Diabetes, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Susumu Kurihara
- Department of Endocrinology and Diabetes, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Tomoko Morita-Ohkubo
- Department of Endocrinology and Diabetes, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Yumi Miyashita
- Division of RI Laboratory, Biomedical Research Center, Saitama Medical University, Saitama, Japan
| | - Shigehiro Katayama
- Department of Endocrinology and Diabetes, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Keisuke Mori
- Department of Ophthalmology, Faculty of Medicine, Saitama Medical University, Faculty of Medicine, Saitama, Japan
| | - Shin Yoneya
- Department of Ophthalmology, Faculty of Medicine, Saitama Medical University, Faculty of Medicine, Saitama, Japan
| | - Masakazu Kohda
- Division of Translational Research, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Yasushi Okazaki
- Division of Translational Research, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Taro Maruyama
- Department of Internal Medicine, Saitama Social Insurance Hospital, Saitama, Japan
| | - Akira Shimada
- Department of Internal Medicine, Saiseikai Central Hospital, Tokyo, Japan
| | - Kazuki Yasuda
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Nao Nishida
- Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Katsushi Tokunaga
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Asako Koike
- Central Research Laboratory, Hitachi Ltd, Tokyo, Japan
| |
Collapse
|
50
|
Alkayyali S, Lyssenko V. Genetics of diabetes complications. Mamm Genome 2014; 25:384-400. [PMID: 25169573 DOI: 10.1007/s00335-014-9543-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 08/13/2014] [Indexed: 12/11/2022]
Abstract
Chronic hyperglycemia and duration of diabetes are the major risk factors associated with development of micro- and macrovascular complications of diabetes. Although it is believed that hyperglycemia induces damage to the particular cell subtypes, e.g., mesangial cells in the renal glomerulus, capillary endothelial cells in the retina, and neurons and Schwann cells in peripheral nerves, the exact mechanisms underlying these damaging defects are not yet well understood. Clustering of micro- and macrovascular complications in families of patients with diabetes suggests a strong genetic susceptibility. However, until now only a handful number of genetic variants were reported to be associated with either nephropathy (ACE, ELMO1, FRMD3, and AKR1B1) or retinopathy (VEGF, AKR1B1, and EPO), and only a few studies were carried out for genetic susceptibility to cardiovascular diseases (ADIPOQ, GLUL) in patients with diabetes. It is, therefore, obvious that the accumulation of more data from larger studies and better phenotypically characterized cohorts is needed to facilitate genetic discoveries and unravel novel insights into the pathogenesis of diabetic complications.
Collapse
Affiliation(s)
- Sami Alkayyali
- Department of Clinical Sciences, Diabetes and Endocrinology, CRC, Lund University, Lund, Sweden,
| | | |
Collapse
|