Genetic epidemiology of diabetic retinopathy
Review Article

Genetic epidemiology of diabetic retinopathy

Lisa Tom, Samaneh Davoudi, Lucia Sobrin

Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA

Contributions: (I) Conception and design: All authors; (II) Administrative support: All authors; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Lucia Sobrin, MD, MPH. Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 243 Charles Street, 12th floor, 1252E, Boston, MA 02114, USA. Email: Lucia_Sobrin@meei.harvard.edu.

Abstract: The disease burden of diabetic retinopathy (DR) is tremendous around the world. While DR is correlated with hemoglobin A1c (HbA1c) and duration of diabetes, genetic differences likely account for variation in susceptibility to DR. DR is a polygenic disorder with demonstrated heritability. However, linkage and admixture analyses, candidate gene association studies, and genome-wide association studies (GWAS) have not identified many loci for DR that can be consistently replicated. Larger, collaborative, multi-ethnic GWAS are needed to identify common variants with small effects. Rigorous defining of controls groups as patients with a long duration of diabetes without DR, and case groups as patients with severe DR will also aid in finding genes associated with DR. Replication in independent cohorts will be key to establishing associated loci for DR. Investigations of mitochondrial DNA and epigenetics in DR are ongoing. Whole exome sequencing presents new opportunities to identify rare variants that might be implicated in DR development. Continued research in the genetic epidemiology of DR is needed, with the potential to elucidate pathogenesis and treatment of an important disease.

Keywords: Diabetic retinopathy (DR); genetics; genome-wide association studies; genetic association studies; diabetes complications


Received: 28 May 2017; Accepted: 12 July 2017; Published: 03 August 2017.

doi: 10.21037/aes.2017.07.04


Introduction

There are an estimated 93 million people with diabetic retinopathy (DR) and 17 million with proliferative diabetic retinopathy (PDR) worldwide (1). Duration of type 1 diabetes (T1D) and type 2 diabetes (T2D) and hyperglycemia, as measured by hemoglobin A1c (HbA1c), are strongly associated risk factors for retinopathy (2,3). However, as little as approximately 11% of variation in retinopathy risk is attributed to HbA1c and duration of disease (4). Transient hyperglycemia, hypertension, hyperlipidemia, and other environmental and genetic factors have been proposed as additional determinants of the development of DR (4-6). Genetic susceptibility may explain much of the heterogeneity in DR among patients with similar glycemic exposure.

Linkage disequilibrium (LD) studies, candidate gene association studies, admixture analysis, and genome wide association studies have been conducted to try to elucidate the genetic factors that influence progression of DR. These studies have been limited by insufficient sample size, lack of replication, and variation in how case/control groups are defined (7,8). Mitochondrial DNA and epigenetic changes, including miRNA, in DR have also begun to be examined (9-12). This review will summarize the current understanding of the genetics of DR, highlighting key findings and future directions of investigation.


Heritability and LD

Studies of twins and families first established a genetic basis for DR. In the 1970’s and 80’s, studies of monozygotic twins showed high rates of concordance for severity of DR in T1D and T2D (13,14). Relatives of patients with DR have a 2–4 times higher risk of developing DR compared to relatives of diabetic patients without retinopathy (15-21). This familial clustering has been shown in South Indian, Mexican American, Chinese, and multi-ethnic populations (15-21). Heritabilities of PDR and DR are estimated at 25–52% and 18–27% respectively (16,17). More extreme phenotypes—advanced PDR despite relatively low glycemic exposure, or absence of DR after decades of uncontrolled diabetes—likely have stronger genetic underpinnings than more common phenotypes. However, finding sufficient numbers of these patients with rarer phenotypes is challenging.

Linkage analyses identified possible loci with DR genes in the 1990’s and 2000’s. A significant logarithm of odds (LOD) score depends in part on the density of the genome sampled, and a score of 3.3 is the threshold for significance in the least strict definition of genome-wide significance (22). A genome-wide linkage analysis in the Pima Indian population showed evidence of linkage on chromosome 1p (LOD 3.1) (17). Unconditional linkage analysis found suggestive linkage at chromosomes 3 and 12 in a Mexican American population (23). Linkage analyses have not led to the identification of definitive DR genes, however, as linkage studies classically identify loci for monogenic diseases that exhibit Mendelian inheritance in large multigenerational pedigrees, and DR is likely a complex polygenic disease with environmental components (8,24).


Candidate gene association studies

Population-based studies that seek to identify common genetic variants are more promising for polygenic diseases like DR. Candidate gene association studies investigate whether a variant of a gene with a hypothesized role in the disease is significantly more common in a group of cases vs. controls. The ideal association study for DR has several characteristics. First, the presence of DR should be determined with fundus photographs and grading with a standard scale such as the Early Treatment Diabetic Retinopathy Study (ETDRS) criteria. Cases should be advanced (PDR or diabetic macular edema) because these are likely the more heritable forms of the disease. Control groups consisting of patients without diabetes can result in the identification of genes that contribute to diabetes in general rather than DR specifically. Controls should ideally be patients who have had diabetes but no or minimal DR. It is best if the controls also have a long duration of diabetes, at least 10–15 years. This minimizes misclassification of cases as controls, as some patients with no DR and short duration of diabetes will go on to develop severe DR with longer diabetes duration. It is not yet clear if DR risk variants will differ between patients with T2D and T1D. DR has more similarities than differences clinically between T1D and T2D patients, so it is likely that there are some shared variants. However, to limit heterogeneity, many genetic studies restrict the discovery phase to one of the diabetes types. Finally, large sample sizes and independent replication cohorts are key to reliably identifying the common variants of modest effect that we hypothesize are involved in DR.

Previous reviews have summarized findings for a number of candidate DR genes (7,8,12,25,26). Genes in the renin-angiotensin system as well as vascular endothelial growth factor (VEGF), erythropoietin (EPO), transcription factor 7-like 2 (TCF7L2), aldose reductase (AKR1B1), receptor for advanced glycation end products (RAGE), nitrous oxide synthase (NOS3), methylenetetrahydrofolate reductase (MTHFR), solute carrier family 19 member 3 (SLC19A3), nuclear factor erythroid 2-like 2 (NFE2L2), CDK5 regulatory subunit associated protein 1 like 1 (CDKAL1), and complement pathway genes have been investigated (7,8,12,25-31). No consistent, rigorously replicated gene has emerged for DR (12,32), likely due to insufficient sample size, lack of comprehensive coverage of genetic variants, or incorrect hypotheses of the candidate genes involved.

The Candidate Gene Association Resource (CARe) conducted one of the best powered candidate gene studies for DR with a large sample size (n=8,040) including discovery and replication cohorts (33). This study was performed with comprehensive coverage of 2,000 genes in inflammatory, metabolic and cardiovascular pathways, with correction for multiple hypothesis testing. This strategy increased the likelihood that the correct variant would be chosen by choosing multiple genes in relevant pathways and maximizing coverage of all the variants in these genes (33). P selectin (SELP) and iduronidase (IDUA) were associated with DR in the discovery sample, but not in replication cohorts. An EPO association was consistent with initial report (34), but was not significant when corrected for multiple hypothesis testing (33).

The P selectin association was further followed up in a study of DR in the Jackson Heart Study (JHS). Among 629 African Americans with T2D in the JHS, Penman et al. showed that higher plasma P selectin levels were associated with DR [odds ratio (OR) =1.11, 95% confidence interval (CI) =1.02‒1.21, P=0.02] and PDR (OR =1.23, 95% CI =0.03–1.46, P=0.02) (35). Subjects with T2D without retinopathy were more likely to be minor allele homozygotes (TT) for the single nucleotide polymorphism (SNP) rs6128 in the P selectin gene than those with retinopathy (P=0.03) (35). This same variant in P selectin was one of those found to be associated with DR in Caucasians with the same direction of effect as in the CARe discovery sample (33,35).

Recently, Porta et al. found that variants of SLC19A3, which encodes a thiamine transporter, were associated with a decreased risk of severe DR. Thiamine regulates intracellular glucose metabolism, supporting an a priori hypothesis for involvement in DR. The study population was comprised of T1D patients from the FinnDiane Study: 1,566 cases of severe DR (defined as ETDRS score ≥53 or any retinal laser treatment) and 218 control subjects with no/mild DR (ETDRS score <35, no laser treatment, and diabetes duration >20 years). Two SNPs in SLC19A3 in LD with each other (FinnDiane r2=0.93) were associated with a reduced risk of severe DR and the combined phenotype of end-stage renal disease and severe DR: rs12694743 [P=3.81×10−6, OR 0.51 (95% CI: 0.38–0.68)] and rs6713116 [P=3.15×10−6, OR 0.41 (0.28–0.60)]. However, the negative association of these two SNPs with DR could not be replicated in two independent cohorts (28), the Diabetes Control and Complications Trial (DCCT)/Epidemiology of Diabetes Interventions and Complications (EDIC) (3) and the Wisconsin Epidemiologic Study of Diabetic Retinopathy (WESDR) (36). However, rs12694743 was found to be associated with a decreased risk of the combined phenotype of severe DR and end-stage renal disease in the study’s meta-analysis of the FinnDiane and WESDR cohorts, even after adjustment for body mass index and HbA1c (P=2.30×10−8) (28).


Admixture mapping

Admixed individuals inherit chromosomal segments from two distinct continental populations that mixed relatively recently in evolutionary history, within the last 20 generations (37). African Americans are an example of an admixed population with European and African ancestry. Admixed individuals allow for detection of a variant associated with a disease if the variant differs in frequency between the ancestral populations.

Tandon et al. studied participants who self-identified as African American with T2D: 305 DR cases (ETDRS score >60) and 1,135 controls (ETDRS score <60) (38). The proportion of African ancestry was not associated with PDR after adjustment for clinical (duration of diabetes, HbA1c, systolic blood pressure), demographic, and socioeconomic (income, education) factors. Admixture analysis failed to identify a genome-wide significant locus (38). Larger sample sizes may be needed for admixture analysis to reveal potential loci for DR genes.


Genome-wide association studies

Rather than investigate hypothesized candidate genes, genome-wide association studies (GWAS) agnostically analyze SNPs across the genome that commonly vary between humans. A higher threshold of significance P<5×10−8 is the accepted standard in the field, and associations should be replicated in independent cohorts (8). In many complex diseases, including age-related macular degeneration (AMD), GWAS have successfully identified genes, providing novel opportunities for understanding pathogenesis and treatment (39-42).

GWAS for DR are summarized in Table 1. The results from these GWAS have been well-summarized in previous reviews (8,12). Two of these GWAS studies found variants that achieved genome-wide significance of P<5×10−8 in their discovery samples, but neither study corrected for the multiple genetic models tested or had independent replication of their results (12,47). One GWAS has found a genome-wide significant finding including replication in independent cohorts. Burdon et al. found an association of genome-wide significance between rs9896052 and sight-threatening DR (P=4.15×10−8) in a meta-analysis of three cohorts in the study: Caucasian patients with T2D, Indian patients with T2D, and Caucasian patients with T1D (50). The growth factor receptor bound protein 2 (GRB2) gene downstream of this locus binds phosphorylated insulin receptor substrate 1 to activate the MAPK pathway via Ras in response to insulin (51,52). This gene is also involved in VEGF signaling (52). Burdon et al. showed expression of GRB2 in the human retina and increased expression in a mouse model of retinopathy, supporting a possible association between this locus and DR (50).

Table 1

Published GWAS in diabetic retinopathy

Study Diabetes type Number of cases, case definition Number of controls, control definition Top findings OR 95% CI P value
Fu et al. 2010 (43) 2 103; severe NPDR, PDR 183; no DR or early NPDR CAMK4 2.6 1.6–4.3 6.0×10−5
FMN1 0.3 0.2–0.5 6.2×10−5
Grassi et al. 2011 (44) 1 973; laser treatment 1,856; no laser treatment rs476141 1.3 NR 1.2×10−7
CCDC101 0.6 NR 3.4×10−6
No loci replicated
Huang et al. 2011 (45) 2 174;NPDR, PDR 575; no DR and non-diabetics ARHGAP22 1.6 1.0–2.5 1.9×10−9
PLXDC2 1.7 1.1–2.7 3.5×10−7
Sheu et al. 2013 (46) 2 437; PDR 570; no DR, ≥8 years of T2D TBC1D4 1.7 NR 1.3×10−7
LRP2-BBS5 1.5 NR 2.0×10−6
Lin et al. 2013 (47) 2 174; NPDR and PDR 574; no DR rs10499299 1.7 1.0–1.3 8.5×10−21
rs17827966 1.7 1.0–3.0 8.7×10−21
Awata et al. 2014 (48,49) 2 837; any DR 1,149; no DR rs9362054 1.6 NR 1.4×10−7
Burdon et al. 2015* (50) 1 and 2 336; sight-threatening DR 508; no DR rs3805931 0.5 0.4–0.7 2.7×10−7
Did not replicate
GRB2 1.7 1.3–2.2 6.6×10−5
With replication 1.5 1.1–2.0 4.2×10−8

*, study pursued replication. NR, not reported; CAMK4, calcium/calmodulin dependent protein kinase IV; FMN1, formin 1; CCDC101, coiled-coil domain-containing protein 101; ARHGAP22, rho GTPase activating protein 22; PLXDC2, plexin domain containing 2; TBC1D4, TBC1 domain family member 4; LRP2-BBS5, LDL receptor related protein 2-Bardet-Biedl syndrome 5; DR, diabetic retinopathy; GWAS, genome-wide association studies; NPDR, non-proliferative diabetic retinopathy; PDR, proliferative diabetic retinopathy; GRB2, growth factor receptor bound protein 2; T2D, type 2 diabetes.

Four studies have attempted to replicate reported loci associations with DR in independent cohorts (32,53-55). Grassi et al. first attempted replication of 389 putatively associated SNPs identified in the Genetics of Kidneys in Diabetes (GoKinD) and EDIC discovery cohort (44) in the WESDR cohort in 2012. The study compared 208 cases (prior laser treatment for either PDR or diabetic macular edema) and 261 controls (all other patients in WESDR). No associations reached genome-wide significance (53).

In 2014, McAuley et al. examined 24 of the most significant SNPs previously reported by Grassi et al. (44) and Huang et al. (45) in a predominantly Caucasian population with T1D and T2D in Australia: 163 cases with severe non-proliferative diabetic retinopathy (NPDR) or PDR and 300 controls with T2D for 5 years or more with no or mild DR (54). McAuley et al. found that rs1073203 was associated in a dominant model (P=0.005). The SNP rs1073203 was first reported to be associated with severe DR (diabetic macular edema or PDR) in the GoKinD and EDIC T1D cohort (P=8.5×10−6) (44). McAuley et al. also found rs4838605 to be significant in an additive model (P=0.047). A Taiwanese GWAS for T2D had reported an association for rs4838605 with DR (P=1.87×10−9) (45), raising the possibility that this locus may affect DR in different ethnicities (12,47,54). The multiple models tested for these variants make it difficult to determine if they truly are significant after correction for multiple hypothesis testing.

In 2015, Hosseini et al. (32) attempted replication for 90 SNPs at 34 independent loci for DR. Top reported signals (P<10−5, 54 SNPs in 11 loci) came from four previous GWAS studies (43-46) and two large candidate gene association studies with broad coverage (33,56). Hosseini et al. also used top reported signals (P<10−5, 22 SNPs in 11 loci) (32) from a GWAS of retinopathy in subjects without diabetes (57). SNPs with evidence of association with DR (P<0.05, 18 SNPs in 16 loci) from previous candidate gene association studies were also included (25,58-74). Of these, 87 SNPs (32 loci) were genotyped or imputed in Hosseini et al.’s data and suitable proxies (r2>0.9) were identified for three more SNPs. Hosseini et al. did not find any genome-wide significant associations among the 90 tested SNPs at 34 independent loci after accounting for multiple hypothesis testing (32). Peng et al. similarly did not find any genome-wide significant associations for 40 SNPs previously reported in three GWAS [Mexican-Americans (43), GoKinD and EDIC (44), and WESDR (43,44,53)] in a large study of Chinese patients with T2D (819 with DR and 1,153 without) (55).

Replication of associations with genome-wide significance has been challenging for DR. One contributing factor is that variants for DR likely have small effects, requiring larger sample sizes for detection. In many other complex diseases, sample sizes upwards of 20,000 participants have been necessary to identify replicable variants (75). Thus far, sample sizes this large have not yet been collected for DR. With larger collaborative efforts, GWAS with larger sample sizes for DR will be possible. To increase the likelihood of success of such efforts, precise phenotyping and strict definitions for cases and controls are needed.


Mitochondrial DNA and epigenetics

Exploration of the role of mitochondrial DNA (mtDNA) and epigenetics in DR is beginning. Mishra et al. used extended-length PCR to measure mtDNA damage in peripheral blood mtDNA in rat and mouse models of diabetes (9). Diabetic rats had a significantly reduced ratio of long to short amplicons of mtDNA and decreased mtDNA copy numbers, compared with age-matched normal rats, suggesting higher mtDNA damage in diabetes (P<0.05) (9). Lipoic acid, which prevents retinopathy in diabetic rats, decreased mtDNA damage in this study (P<0.05). Overexpression of superoxide dismutase 2 (SOD2) or suppression of matrix metallopeptidase 9 (MMP-9) prevented retinopathy in diabetic mice, and these populations of mice also did not show the increase in mtDNA seen in diabetic mice (9). Mishra et al. also measured mitochondrial DNA damage as significantly increased in diabetic patients with retinopathy (n=6) vs. without retinopathy (n=5) (P<0.05) (9).

Estopinal et al. showed that mitochondrial haplotypes are associated with severity of DR in Caucasian patients (153 with NPDR and 138 with PDR) (76). The study found that the frequency of PDR differed significantly by mitochondrial haplogroup (P=0.027). In the study, an independent cohort of Caucasian patients with DR (44 NPDR; 57 PDR) confirmed this association (P=0.0064) (76). In the combined cohort, patients from the common haplogroup H were more likely to have PDR [OR =2.0 (95% CI =1.3–3.0), P=0.0012]. Patients from haplogroup UK had a decreased risk of having PDR [OR =0.5 (95% CI =0.3–0.8, P=0.0049)]. These associations with PDR were independent of HbA1c, diabetes duration, and hypertension in multivariate logistic regression analyses haplogroup H [OR =2.1 (95% CI =1.3–3.4)]; haplogroup UK [OR =0.41 (95% CI =0.23–0.73)] (76).

Investigation into epigenetic mechanisms in DR has been underway (77). Diabetic eyes have been shown to have miRNA changes including upregulation of miR200b, a VEGF-regulating miRNA (10). An increase in miR-29b is thought to be protective against apoptosis of retinal ganglion cells in streptozotocin-induced diabetic rats (11). Agardh et al. analyzed DNA methylation genome-wide in 485,577 sites in T1D patients. False discovery rate analysis was used to account for multiple hypothesis testing. Cases had PDR (n=28) and controls (n=30) were defined as having at least 10 years of diabetes with no or mild DR. The study identified differential DNA methylation at 349 CpG sites, representing 233 genes, the majority of which (79%) had decreased methylation in the PDR group. The Natural Killer cell-mediated cytotoxicity pathway was significantly (P=0.006) enriched among these differentially methylated genes (78).


Conclusions

Current understanding of the genetics of DR is incomplete. Linkage studies, candidate gene association studies, admixture analysis, and GWAS have not yet achieved consistent replication of many loci or genes associated with DR.

GWAS will require larger international collaborative efforts to assemble multi-ethnic cohorts and increase sample sizes. To maximize the chances of identifying true variants, cases should be defined as those with PDR or diabetic macular edema determined from a standard grading scale of imaging, while control groups should be defined rigorously as patients without DR despite a long duration of diabetes (15–20 years). Patients with mild DR are sometimes classified as cases (48) and sometimes as controls (43,79), biasing the field towards null findings. Future studies also need to correctly account for glycemic control, which is strongly correlated with DR. Replication in independent data sets will also be key to strengthening future GWAS findings (8,12). In addition, examination of rare variation and DR risk has not yet been attempted. Whole exome sequencing may reveal associated rare variants, particularly if the extremes of phenotype are examined. Epigenetics and mitochondrial DNA also deserve further investigation. Ongoing research in these areas and large collaborations for GWAS have the potential to illuminate the genetic foundations of DR.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the Guest Editors (Chi-Chao Chan and Mingguang He) for the series “Medical Retinal Diseases and Epidemiology” published in Annals of Eye Science. The article has undergone external peer review.

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/aes.2017.07.04). The series “Medical Retinal Diseases and Epidemiology” was commissioned by the editorial office without any funding or sponsorship. The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Yau JW, Rogers SL, Kawasaki R, et al. Global Prevalence and Major Risk Factors of Diabetic Retinopathy. Diabetes Care 2012;35:556-64. [Crossref] [PubMed]
  2. Klein R, Klein BE, Moss SE, et al. Relationship of hyperglycemia to the long-term incidence and progression of diabetic retinopathy. Arch Intern Med 1994;154:2169-78. [Crossref] [PubMed]
  3. Group DCaCTR. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N Engl J Med 1993;329:977-86. [Crossref] [PubMed]
  4. Lachin JM, Genuth S, Nathan DM, et al. Effect of glycemic exposure on the risk of microvascular complications in the diabetes control and complications trial--revisited. Diabetes 2008;57:995-1001. [Crossref] [PubMed]
  5. Stolar M. Glycemic control and complications in type 2 diabetes mellitus. Am J Med 2010;123:S3-S11. [Crossref] [PubMed]
  6. Hirsch IB, Brownlee M. Beyond hemoglobin A1c—need for additional markers of risk for diabetic microvascular complications. JAMA 2010;303:2291-2. [Crossref] [PubMed]
  7. Kuo JZ, Wong TY, Rotter JI. Challenges in elucidating the genetics of diabetic retinopathy. JAMA Ophthalmol 2014;132:96-107. [Crossref] [PubMed]
  8. Cho H, Sobrin L. Genetics of diabetic retinopathy. Curr Diab Rep 2014;14:515. [Crossref] [PubMed]
  9. Mishra M, Lillvis J, Seyoum B, Kowluru RA. Peripheral Blood Mitochondrial DNA Damage as a Potential Noninvasive Biomarker of Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2016;57:4035-44. [Crossref] [PubMed]
  10. McArthur K, Feng B, Wu Y, et al. MicroRNA-200b regulates vascular endothelial growth factor–mediated alterations in diabetic retinopathy. Diabetes 2011;60:1314-23. [Crossref] [PubMed]
  11. Silva VA, Polesskaya A, Sousa TA, et al. Expression and cellular localization of microRNA-29b and RAX, an activator of the RNA-dependent protein kinase (PKR), in the retina of streptozotocin-induced diabetic rats. Mol Vis 2011;17:2228. [PubMed]
  12. Davoudi S, Sobrin L. Novel Genetic Actors of Diabetes-Associated Microvascular Complications: Retinopathy, Kidney Disease and Neuropathy. Rev Diabet Stud 2015;12:243-59. [Crossref] [PubMed]
  13. Leslie RD, Pyke DA. Diabetic retinopathy in identical twins. Diabetes 1982;31:19-21. [Crossref] [PubMed]
  14. Pyke DA, Tattersall RB. Diabetic Retinopathy in Identical Twins. Diabetes 1973;22:613-8. [Crossref] [PubMed]
  15. Group DCaCTR. Clustering of long-term complications in families with diabetes in the diabetes control and complications trial. Diabetes 1997;46:1829. [Crossref] [PubMed]
  16. Hietala K, Forsblom C, Summanen P, et al. Heritability of proliferative diabetic retinopathy. Diabetes 2008;57:2176-80. [Crossref] [PubMed]
  17. Looker HC, Nelson RG, Chew E, et al. Genome-wide linkage analyses to identify loci for diabetic retinopathy. Diabetes 2007;56:1160-6. [Crossref] [PubMed]
  18. Rema M, Saravanan G, Deepa R, et al. Familial clustering of diabetic retinopathy in South Indian type 2 diabetic patients. Diabet Med 2002;19:910-6. [Crossref] [PubMed]
  19. Zhang X, Gao Y, Zhou Z, et al. Familial clustering of diabetic retinopathy in Chongqing, China, type 2 diabetic patients. Eur J Ophthalmol 2010;20:911-8. [PubMed]
  20. Hallman DM, Huber JC, Gonzalez VH, et al. Familial aggregation of severity of diabetic retinopathy in Mexican Americans from Starr County, Texas. Diabetes Care 2005;28:1163-8. [Crossref] [PubMed]
  21. Arar NH, Freedman BI, Adler SG, et al. Heritability of the severity of diabetic retinopathy: the FIND-Eye study. Invest Ophthalmol Vis Sci 2008;49:3839-45. [Crossref] [PubMed]
  22. Lander E, Kruglyak L. Genetic dissection of complex traits: guidelines for interpreting and reporting linkage results. Nat Genet 1995;11:241-7. [Crossref] [PubMed]
  23. Hallman DM, Boerwinkle E, Gonzalez VH, et al. A genome-wide linkage scan for diabetic retinopathy susceptibility genes in Mexican Americans with type 2 diabetes from Starr County, Texas. Diabetes 2007;56:1167-73. [Crossref] [PubMed]
  24. Mishra B, Swaroop A, Kandpal RP. Genetic components in diabetic retinopathy. Indian J Ophthalmol 2016;64:55. [Crossref] [PubMed]
  25. Abhary S, Hewitt AW, Burdon KP, et al. A systematic meta-analysis of genetic association studies for diabetic retinopathy. Diabetes 2009;58:2137-47. [Crossref] [PubMed]
  26. Liew G, Klein R, Wong TY. The role of genetics in susceptibility to diabetic retinopathy. Int Ophthalmol Clin 2009;49:35-52. [Crossref] [PubMed]
  27. Luo S, Wang F, Shi C, et al. A Meta-Analysis of Association between Methylenetetrahydrofolate Reductase Gene (MTHFR) 677C/T Polymorphism and Diabetic Retinopathy. Int J Environ Res Public Health 2016;13:E806 [Crossref] [PubMed]
  28. Porta M, Toppila I, Sandholm N, et al. Variation in SLC19A3 and Protection From Microvascular Damage in Type 1 Diabetes. Diabetes 2016;65:1022-30. [Crossref] [PubMed]
  29. Xu X, Sun J, Chang X, et al. Genetic variants of nuclear factor erythroid-derived 2-like 2 associated with the complications in Han descents with type 2 diabetes mellitus of Northeast China. J Cell Mol Med 2016;20:2078-88. [Crossref] [PubMed]
  30. Liu NJ, Xiong Q, Wu HH, et al. The association analysis polymorphism of CDKAL1 and diabetic retinopathy in Chinese Han population. Int J Ophthalmol 2016;9:707-12. [PubMed]
  31. Yang MM, Wang J, Ren H, et al. Genetic Investigation of Complement Pathway Genes in Type 2 Diabetic Retinopathy: An Inflammatory Perspective. Mediators Inflamm 2016;2016:1313027.
  32. Hosseini SM, Boright AP, Sun L, et al. The association of previously reported polymorphisms for microvascular complications in a meta-analysis of diabetic retinopathy. Hum Genet 2015;134:247-57. [Crossref] [PubMed]
  33. Sobrin L, Green T, Sim X, et al. Candidate gene association study for diabetic retinopathy in persons with type 2 diabetes: the Candidate gene Association Resource (CARe). Invest Ophthalmol Vis Sci 2011;52:7593-602. [Crossref] [PubMed]
  34. Tong Z, Yang Z, Patel S, et al. Promoter polymorphism of the erythropoietin gene in severe diabetic eye and kidney complications. Proc Natl Acad Sci 2008;105:6998-7003. [Crossref] [PubMed]
  35. Penman A, Hoadley S, Wilson JG, et al. P-selectin plasma levels and genetic variant associated with diabetic retinopathy in African Americans. Am J Ophthalmol 2015;159:1152-1160.e2. [Crossref] [PubMed]
  36. Klein R, Klein BE, Moss SE. Epidemiology of proliferative diabetic retinopathy. Diabetes Care 1992;15:1875-91. [Crossref] [PubMed]
  37. Shriner D. Overview of Admixture Mapping. Curr Protoc Hum Genet 2013;Chapter 1:Unit 1.23.
  38. Tandon A, Chen CJ, Penman A, et al. African Ancestry Analysis and Admixture Genetic Mapping for Proliferative Diabetic Retinopathy in African Americans. Invest Ophthalmol Vis Sci 2015;56:3999-4005. [Crossref] [PubMed]
  39. Visscher PM, Brown MA, McCarthy MI, et al. Five years of GWAS discovery. Am J Hum Genet 2012;90:7-24. [Crossref] [PubMed]
  40. Fritsche LG, Chen W, Schu M, et al. Seven new loci associated with age-related macular degeneration. Nat Genet 2013;45:433-9, 439e1-2.
  41. Neale BM, Fagerness J, Reynolds R, et al. Genome-wide association study of advanced age-related macular degeneration identifies a role of the hepatic lipase gene (LIPC). Proc Natl Acad Sci 2010;107:7395-400. [Crossref] [PubMed]
  42. Yu Y, Bhangale TR, Fagerness J, et al. Common variants near FRK/COL10A1 and VEGFA are associated with advanced age-related macular degeneration. Hum Mol Genet 2011;20:3699-709. [Crossref] [PubMed]
  43. Fu YP, Hallman DM, Gonzalez VH, et al. Identification of diabetic retinopathy genes through a genome-wide association study among Mexican-Americans from Starr County, Texas. J Ophthalmol 2010;2010. pii: 861291.
  44. Grassi MA, Tikhomirov A, Ramalingam S, et al. Genome-wide meta-analysis for severe diabetic retinopathy. Hum Mol Genet 2011;20:2472-81. [Crossref] [PubMed]
  45. Huang YC, Lin JM, Lin HJ, et al. Genome-wide association study of diabetic retinopathy in a Taiwanese population. Ophthalmology 2011;118:642-8. [Crossref] [PubMed]
  46. Sheu WH, Kuo JZ, Lee IT, et al. Genome-wide association study in a Chinese population with diabetic retinopathy. Hum Mol Genet 2013;22:3165-73. [Crossref] [PubMed]
  47. Lin HJ, Huang YC, Lin JM, et al. Association of genes on chromosome 6, GRIK2, TMEM217 and TMEM63B (linked to MRPL14) with diabetic retinopathy. Ophthalmologica 2013;229:54-60. [Crossref] [PubMed]
  48. Awata T, Yamashita H, Kurihara S, et al. 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 [Crossref] [PubMed]
  49. Awata T, Yamashita H, Kurihara S, et al. Correction: a genome-wide association study for diabetic retinopathy in a Japanese population: potential association with a long intergenic non-coding RNA. PLoS One 2015;10:e0126789 [Crossref] [PubMed]
  50. Burdon KP, Fogarty RD, Shen W, et al. Genome-wide association study for sight-threatening diabetic retinopathy reveals association with genetic variation near the GRB2 gene. Diabetologia 2015;58:2288-97. [Crossref] [PubMed]
  51. Skolnik EY, Lee CH, Batzer A, et al. The SH2/SH3 domain-containing protein GRB2 interacts with tyrosine-phosphorylated IRS1 and Shc: implications for insulin control of ras signaling. EMBO J 1993;12:1929-36. [PubMed]
  52. Anselmi F, Orlandini M, Rocchigiani M, et al. c-ABL modulates MAP kinases activation downstream of VEGFR-2 signaling by direct phosphorylation of the adaptor proteins GRB2 and NCK1. Angiogenesis 2012;15:187-97. [Crossref] [PubMed]
  53. Grassi MA, Tikhomirov A, Ramalingam S, et al. Replication Analysis for Severe Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2012;53:2377-81. [Crossref] [PubMed]
  54. McAuley AK, Wang JJ, Dirani M, et al. Replication of Genetic Loci Implicated in Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2014;55:1666-71. [Crossref] [PubMed]
  55. Peng D, Wang J, Zhang R, et al. 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. [Crossref] [PubMed]
  56. Roy MS, Hallman D, Fu Y, et al. Assessment of 193 candidate genes for retinopathy in african americans with type 1 diabetes. Arch Ophthalmol 2009;127:605-12. [Crossref] [PubMed]
  57. Jensen RA, Sim X, Li X, et al. Genome-wide association study of retinopathy in individuals without diabetes. PLoS One 2013;8:e54232 [Crossref] [PubMed]
  58. Lu Y, Ge Y, Hu Q, et al. Association between angiotensin-converting enzyme gene polymorphism and diabetic retinopathy in the Chinese population. J Renin Angiotensin Aldosterone Syst 2012;13:289-95. [Crossref] [PubMed]
  59. Fujisawa T, Ikegami H, Kawaguchi Y, et al. Meta-analysis of association of insertion/deletion polymorphism of angiotensin I-converting enzyme gene with diabetic nephropathy and retinopathy. Diabetologia 1998;41:47-53. [Crossref] [PubMed]
  60. Zhou JB, Yang JK. Angiotensin-converting enzyme gene polymorphism is associated with proliferative diabetic retinopathy: a meta-analysis. Acta Diabetologica 2010;47:187-93. [Crossref] [PubMed]
  61. Niu W, Qi Y, Wu Z, et al. A meta-analysis of receptor for advanced glycation end products gene: four well-evaluated polymorphisms with diabetes mellitus. Mol Cell Endocrinol 2012;358:9-17. [Crossref] [PubMed]
  62. Yuan D, Liu Q. Association of the receptor for advanced glycation end products gene polymorphisms with diabetic retinopathy in type 2 diabetes: a meta-analysis. Ophthalmologica 2012;227:223-32. [Crossref] [PubMed]
  63. Hu C, Zhang R, Yu W, et al. CPVL/CHN2 genetic variant is associated with diabetic retinopathy in Chinese type 2 diabetic patients. Diabetes 2011;60:3085-9. [Crossref] [PubMed]
  64. Zintzaras E, Chatzoulis DZ, Karabatsas CH, et al. The relationship between C677T methylenetetrahydrofolate reductase gene polymorphism and retinopathy in type 2 diabetes: a meta-analysis. J Hum Genet 2005;50:267-75. [Crossref] [PubMed]
  65. Niu W, Qi Y. An updated meta-analysis of methylenetetrahydrofolate reductase gene 677C/T polymorphism with diabetic nephropathy and diabetic retinopathy. Diabetes Res Clin Pract 2012;95:110-8. [Crossref] [PubMed]
  66. Zhao S, Li T, Zheng B, et al. Nitric oxide synthase 3 (NOS3) 4b/a, T-786C and G894T polymorphisms in association with diabetic retinopathy susceptibility: a meta-analysis. Ophthalmic Genet 2012;33:200-7. [Crossref] [PubMed]
  67. Ma J, Li Y, Zhou F, et al. Meta-analysis of association between the Pro12Ala polymorphism of the peroxisome proliferator–activated receptor-γ2 gene and diabetic retinopathy in Caucasians and Asians. Mol Vis 2012;18:2352-60. [PubMed]
  68. Zhang T, Pang C, Li N, et al. Plasminogen activator inhibitor-1 4G/5G polymorphism and retinopathy risk in type 2 diabetes: a meta-analysis. BMC Med 2013;11:1. [Crossref] [PubMed]
  69. Tian C, Fang S, Du X, et al. Association of the C47T polymorphism in SOD2 with diabetes mellitus and diabetic microvascular complications: a meta-analysis. Diabetologia 2011;54:803-11. [Crossref] [PubMed]
  70. Syreeni A, El-Osta A, Forsblom C, et al. Genetic examination of SETD7 and SUV39H1/H2 methyltransferases and the risk of diabetes complications in patients with type 1 diabetes. Diabetes 2011;60:3073-80. [Crossref] [PubMed]
  71. Qiu M, Xiong W, Liao H, et al. VEGF− 634G> C polymorphism and diabetic retinopathy risk: a meta-analysis. Gene 2013;518:310-5. [Crossref] [PubMed]
  72. Zhao T, Zhao J. Association between the-634C/G polymorphisms of the vascular endothelial growth factor and retinopathy in type 2 diabetes: a meta-analysis. Diabetes Res Clin Pract 2010;90:45-53. [Crossref] [PubMed]
  73. Gong JY, Sun YH. Association of VEGF gene polymorphisms with diabetic retinopathy: a meta-analysis. PLoS One 2013;8:e84069 [Crossref] [PubMed]
  74. Lu Y, Ge Y, Shi Y, et al. Two polymorphisms (rs699947, rs2010963) in the VEGFA gene and diabetic retinopathy: an updated meta-analysis. BMC Ophthalmol 2013;13:56. [Crossref] [PubMed]
  75. Loos RJ. Genetic determinants of common obesity and their value in prediction. Best Pract Res Clin Endocrinol Metab 2012;26:211-26. [Crossref] [PubMed]
  76. Estopinal CB, Chocron IM, Parks MB, et al. Mitochondrial Haplogroups Are Associated With Severity of Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2014;55:5589-95. [Crossref] [PubMed]
  77. Zeng J, Chen B. Epigenetic Mechanisms in the Pathogenesis of Diabetic Retinopathy. Ophthalmologica 2014;232:1-9. [Crossref] [PubMed]
  78. Agardh E, Lundstig A, Perfilyev A, et al. Genome-wide analysis of DNA methylation in subjects with type 1 diabetes identifies epigenetic modifications associated with proliferative diabetic retinopathy. BMC Med 2015;13:182. [Crossref] [PubMed]
  79. Gao X, Gauderman WJ, Marjoram P, et al. Genetic variants associated with proliferative diabetic retinopathy in Latinos. Invest Ophthalmol Vis Sci 2014;55:2238.
doi: 10.21037/aes.2017.07.04
Cite this article as: Tom L, Davoudi S, Sobrin L. Genetic epidemiology of diabetic retinopathy. AME Med J 2017;2:56.

Download Citation