touchENDOCRINOLOGY touchENDOCRINOLOGY
Diabetes
Read Time: 7 mins

Understanding Diabetic Nephropathy – Is There a Genetic Susceptibility?

Copy Link
Published Online: Jun 6th 2011 European Endocrinology, 2008; 4(2):66-9; DOI: http://doi.org/10.17925/EE.2008.04.02.66
Authors: Savage David, Maxwell Peter
Quick Links:
Abstract
Article
Article Information
Abstract:
Overview

Diabetes is rapidly increasing in prevalence,1 resulting in profound socioeconomic impacts in both developed and developing countries. Furthermore, the parallel increase in the prevalence of complications of diabetes, particularly nephropathy, retinopathy and associated cardiovascular disease, is placing enormous demands on healthcare budgets.2

Article:

Improved understanding of the aetiology and pathogenesis of these complications is urgently needed. Effective screening strategies to identify individuals with diabetes most likely to develop complications could improve outcomes by focusing resources on those at highest risk.

What Is Diabetic Nephropathy?

Improved understanding of the aetiology and pathogenesis of these complications is urgently needed. Effective screening strategies to identify individuals with diabetes most likely to develop complications could improve outcomes by focusing resources on those at highest risk.

What Is Diabetic Nephropathy?
Diabetic nephropathy (DN) is a microvascular complication affecting patients with both type 1 and type 2 diabetes. It has become the leading cause of end-stage renal disease (ESRD) in Europe and the US managed by renal replacement therapy (kidney dialysis and/or renal transplantation).3,4 The clinical phenotype includes persistent proteinuria, a decreasing glomerular filtration rate (GFR) and hypertension.5 A sizeable minority of patients with diabetes eventually develop definite DN.6 The earliest clinical manifestation is microalbuminuria (incipient nephropathy), which may progress to overt proteinuria (dipstick urinalysis positive for proteinuria) followed by the emergence of hypertension, a declining GFR and, later, development of ESRD. Nevertheless, microalbuminura is not an absolutely reliable predictor of progression to DN as some patients remain microalbuminuric while others show regression of albuminuria to normal albumin excretion rates.7 Patients with DN have much higher mortality rates than individuals with diabetes with normal albumin excretion rates. The excess mortality is largely attributed to higher rates of coronary artery disease, stroke and amputation,8 and the five-year survival rate for ESRD patients with diabetes is worse than for most cancers.

DN in type 1 and 2 diabetes differs in a number of ways. For example, compared with type 1 diabetes the rate of progression of renal failure in patients with type 2 diabetes is more variable. In patients with type 2 diabetes, microalbuminuria progression to advanced renal disease is less frequent and increases in blood pressure generally occur before onset of microalbuminuria.9 Older patients with type 2 diabetes may also have concurrent hypertensive renal vascular disease, and unlike patients with type 1 diabetes the age at diabetes onset is more difficult to establish. Therefore, phenotypic definition of DN is simpler in patients with type 1 diabetes, which is one reason why efforts to discover genetic variants associated with risk of DN might be more successful in these patients.

Pathophysiological features of DN include glomerular capillary hypertension, glomerular hyperfiltration, mesangial matrix expansion and glomerulosclerosis. Prolonged hyperglycaemia leads to chronic metabolic and haemodynamic changes that modify the activity of various intracellular signalling pathways and transcription factors.10 There is the subsequent induction of cytokines, chemokines and growth factors, particularly transforming growth factor beta (TGFβ). These effects promote structural abnormalities in the kidney such as glomerular basement membrane thickening, podocyte injury and mesangial matrix expansion with the later development of irreversible glomerular sclerosis and tubulointerstitial fibrosis associated with declining GFR. The clinical management of DN includes optimal glycaemic control, treatment of dyslipidaemia and aggressive lowering of blood pressure ideally with angiotensin-converting enzyme (ACE) inhibitors and/or angiotensin II type 1 receptor (ARB) blockers.11–14

Table 1: Key Risk Factors for Diabetic Nephropathy

Does Genetic Variation Contribute to Risk of Diabetic Nephropathy?
A number of factors can alter the risk of nephropathy in patients with diabetes (see Table 1). Intervention trials have demonstrated that excellent glycaemic control can reduce the risk of progression to nephropathy.11–13 However, hyperglycaemia alone is not responsible for development of DN, since some patients with diabetes do not develop nephropathy despite poor glycaemic control. Epidemiological studies have suggested that some individuals are ‘protected’ from DN because it generally develops within 15–20 years after diagnosis of diabetes or not at all.6 Family studies have demonstrated significant differences in recurrence risk for nephropathy in siblings with diabetes in which the proband had nephropathy compared with probands without nephropathy.15 The large differential in cumulative risk between these groups cannot be explained in terms of environmental factors alone. Certain ethnic groups (e.g. African-Americans, Hispanics and American Indians) are also at greater risk of DN, and there is clustering of hypertension, cardiovascular disease and dyslipidaemia among parents of patients with DN.16,17Many of these characteristics also have a genetic basis. Segregation analysis in families with type 2 diabetes also suggests that genetic factors determine urinary albumin excretion levels.18 Collectively there is compelling evidence for a genetic predisposition to DN. Therefore, identifying susceptibility genes and causal variants is a major goal, as it should lead to prediction of those individuals with diabetes at low and high risk of nephropathy. It may also help identify new targets for therapy within molecular pathways involved in this serious complication of diabetes.

Candidate Gene-based Studies in Diabetic Nephropathy
A number of genetic association (case-control, and to a lesser extent family-based) and linkage studies have been conducted in DN, but replication of positive findings has proved inconsistent. Case-control association studies in DN generally assess the frequencies of single nucleotide polymorphisms (SNPs) in patients with diabetes with (cases) and without (controls) nephropathy for correlations with disease. Finding causal variants in these studies relies on the fact that there is non-random association of alleles (linkage disequilibrium) within the human genome. Thus, a positive association could mean that the finding is true and causal, or true because of linkage disequilibrium or indeed false by chance. Genotyping redundancy due to linkage disequilibrium has been exploited, notably in genome-wide association studies (GWAS), by the use of so-called tag SNPs (proxies) in order to have good genome coverage while minimising genotyping effort and cost. Despite the lack of robust replication studies it is noteworthy that a large meta-analysis would tend to support involvement of the ACE insertion/deletion polymorphism in DN.19 The strategies employed and findings of many of these studies have been reviewed elsewhere.20–23

Many of the problems in identifying disease-causing genes in DN may be related to inappropriate phenotypic selection criteria and study design, especially statistically under-powered studies (both initial and replication) where there were small sample sizes or artefacts generated by population substructure. However, other factors have contributed, notably difficulties in obtaining genetic material from family members for linkage and family-based association studies because the disease is relatively late-onset. Furthermore, it has been suggested that there is likely to be heterogeneity for susceptibility genes in patients with proteinuria alone compared with those who progress to ESRD.24

Genome-wide Association Studies
Recently, a wealth of GWAS have been published for a range of diseases (largely in populations of European ancestry)25–28 by the Wellcome Trust Case Control initiative. Studies employed approximately 2,000 cases and 3,000 controls, and identified known and unknown gene variant associations for several diseases, notably metabolic and cardiovascular disease,26 autoimmune disease27 and cancer.28 The success of GWAS is due to improvements in the statistical power of studies, the availability of good SNP maps, advances in high-throughput genotyping platforms and computing power for bioinformatics and statistical genetics.29A recent analysis of published GWAS has (unsurprisingly) demonstrated that most common variants (minor allele frequency ≥5.0%) associated with disease have odds ratios of 1.2–1.5, whereas more penetrant rare variants have odds ratios ≥2.0.30 In many respects, GWAS have been successful, but for each disease the gene variants identified still explain only a proportion of the risk due to genetic susceptibility. Some of these limitations can be explained by the fact that GWAS are essentially based on the hypothesis that common disease is due to common variants,31 and that to detect common variants with small effects or rare variants (<5%) with large effects will require much larger sample sizes of cases and controls. However, there are ways to assist in finding new gene variants that include meta-analyses of GWAS, and also analysis of data from GWAS in populations of non-European ancestry.32,33

Figure 1: Strategies for Identifying Therapeutic Targets

Genome-wide association studies (GWAS) and next-generation sequencing studies will improve knowledge of the molecular pathways involved in diabetic nephropathy, which should lead to the development of novel therapies.

Genome-wide Association Studies in Diabetic Nephropathy
A GWAS for DN in type 1 diabetes has very recently been completed under the Genetic Association Information Network (GAIN) initiative34 employing US Genetics of Kidneys in Diabetes (GoKinD)35 study cases and controls and Affymetrix 5.0 500K SNP arrays. After exclusion criteria were applied, good-quality data were available for a total of almost 360,000 autosomal SNPs in 820 cases and 885 controls (AS Krolewski, personal communication). The two most promising SNP associations with DN to emerge from the primary analysis that approached genome-wide significance were rs10868025 (odds ratio [OR] 1.45; p=5.0×10-7), which is located near the 5‘ end of the FERM domain containing 3 (FRMD3) gene on chromosome 9q, and rs451041 (OR 1.36; p=3.1×10-6), located in an intronic region of the cysteinyl-tRNA synthetase (CARS) gene on chromosome 11p. Imputation analysis (methods for deducing missing genotypes for untyped SNPs) identified an association with rs1888747 (p=4.7×10-7) on chromosome 9q, which was confirmed by genotyping this SNP in the US GoKinD case-control collection (p=6.3×10-7). Further analyses showed the 9q SNP associations to be associated with both proteinuria and ESRD, whereas rs451041 on chromosome 11p was associated with ESRD alone. Replication studies in Diabetes Control and Complications Trial/ Epidemiology of Diabetes Interventions and Complications13 samples, which analysed time to nephropathy, supported the findings of the US GoKinD study; however, these replication data should be treated with caution as the study was statistically underpowered. In addition, expression analysis in human cell lines, including mesangial and renal proximal tubule cells, revealed high levels of expression for both FRMD3 and CARS genes.Furthermore, immunohistochemistry analysis showed that FRMD3 protein expression was increased in biopsy samples from patients with DN compared with controls. This GWAS represents a landmark in DN genetics, and is a starting point for the identification of causal gene variants in this complication (see Figure 1). Since the power of the US GoKinD GWAS is sufficient to identify common variants with large effects, the findings might therefore suggest that common variants with large effects are not involved in DN, but additional studies are required to confirm this. More GWAS in DN (both European and non-European ancestry) are urgently required to increase the statistical power to confirm initial findings and identify new variants. To assist in this effort, case control collections of similar size to that employed for the US GoKinD GWAS have been assembled including GoKinD UK/Warren36 and FinnDiane37 collections.

Transcriptomics and Pathway-based Approaches
Expression profiling studies using in vitro and in vivo models of disease, and also human renal biopsies, have furthered understanding of the molecular drivers involved in DN. For example, recent studies have identified approximately 200 genes to be differentially expressed in mesangial cells exposed to high levels of extracellular glucose.38-41 Novel genes and gene transcripts identified include connective tissue growth factor (CTGF), and induced in high glucose-2 (IHG-2; Gremlin [GREM1]).

CTGF is a downstream mediator of TGFβ1-directed matrix production, and regulates actin cytoskeleton disassembly. Gremlin is a bone morphogenetic protein and is regarded as a novel target gene in DN.42 Recently, studies using bioinformatic analyses to identify genes with similar sequence and structure to GREM1, revealed significant similarity in both promoter and predicted microRNA (miRNA)-binding elements to the Notch ligand Jagged1 and its downstream effector, hairy enhancer of split-1 (Hes1).43 Furthermore, TGFβ1 increased expression of these genes in vitro, and increased expression of Gremlin, Jagged1 and Hes1 was also found in renal biopsies from patients with DN. Upregulation of these genes was also found to co-localise to areas of tubulointerstitial fibrosis. These findings point to co-regulation of Gremlin and Notch signalling, and a possible new pathway implicated in DN.

Genes identified by these approaches can provide interesting new candidates for genetic association studies in DN, particularly if they converge with findings from other studies such as GWAS. Also, expression profiling studies should assist in the identification of new therapeutic targets for intervention trials; this is particularly relevant since TGFβ itself is excluded as a suitable target due to its range of important actions.

Epigenetics and Diabetic Nephropathy
In addition to genetic variation, dysregulation of the epigenome can also lead to disease, notably in cancer,44,45 but also in other common diseases such as diabetes.46 Epigenetic changes include DNA methylation (covalent attachment of methyl groups at CpG dinucleotides), histone modifications (acetylation, methylation, phosphorylation and ubiquitination) and RNA-based silencing. DNA Methylation
DNA methylation influences gene regulation largely as a result of transcriptional silencing, and indirect evidence points to the involvement of DNA methylation in DN. For instance, alterations in DNA methylation may be implicated in vascular disease,47,48 and characteristics known to be linked to DN such as hyperhomocysteinaemia, dyslipidaemia, inflammation and oxidative stress can promote aberrant DNA methylation.49–51 The identification of aberrant DNA methylation in DN will provide new insights into the causes of this complication, and importantly could provide the basis for the development of novel treatments (e.g. DNA methylation inhibitors).

MicroRNAs
miRNAs are involved in post-transcriptional regulation of gene expression, and there is a growing awareness that altered (miRNA) expression is involved in disease.52-54 These non-coding RNAs (~21 nucleotides long) bind to the 3’ untranslated regions (UTRs) of target genes and affect stability or translation of messenger RNA (mRNA), and may be implicated in DN.55–58 Of note, it has been shown that the ‘C’ allele of a SNP (1166A>C) in the 3’ UTR of the AGTR1 gene is functional in that it results in increased levels of AGTR1 by abrogating the regulation of hsa-miR-155.56 Since the AGTR1 1166C allele has been reported to be associated with hypertension in many (but not all) studies, these findings
could provide new targets for therapeutic treatments. Furthermore, a recent study has also demonstrated that miR-377 is elevated in experimental models of disease and that this indirectly leads to increased production of fibronectin, an important matrix protein in DN.57

Therefore, comprehensive studies are required to assess altered expression of miRNA in both experimental models of disease and human biopsy material from affected and unaffected control individuals. Target genes for differentially expressed miRNAs can then be identified from established databases, and studies performed to assess if SNPs in the 3’UTR of target genes are associated with DN.59 These new avenues of research may reveal more about the aetiology of DN.

Next-generation Sequencing and Future Approaches
The feasibility of cost-effectively and efficiently re-sequencing entire genomes of humans to identify causal gene variants has improved considerably with the recent development of next-generation sequencing technologies.60,61 Although the chemistries, throughput and cost per nucleotide base vary between these platforms, they all involve massive parallel sequencing and miniaturisation that provides ultra-high throughput (up to billions of bases per run) at a hugely reduced cost compared with standard dideoxy sequencing methods.62,63 Furthermore, with the ability to bar-code DNA samples (multiplexing)64 and enrich for specific genomic region (e.g. exons) by array-based capture methods,65 this could result in still further reductions in cost and improvements in throughput.

Next-generation sequencing methods will be exceptionally useful in studying the genetic aetiology of DN (see Figure 1). For example, targeted re-sequencing of association signals within the genomic regions identified by GWAS will no longer be as cost prohibitive or inefficient to conduct. In addition, this technology has many other applications including transcriptome sequencing and the analysis of DNA methylation.Concluding Remarks
It is abundantly clear that the identification of genetic and epigenetic risk factors for DN requires multidisciplinary approaches involving international collaborations. The need to increase sample sizes in case-control studies, to perform more GWAS informed by the results from expression profiling to assist SNP selection and conduct meta-analyses of available genotype data is now recognised. Additional case-control collections for replication of provisional positive associations from these scans will also be required to confirm the reliability of the data. The challenge will be moving from association signals to causal variants and subsequent identification of new therapeutic targets in molecular pathways (see Figure 1). The application of ultra-high-throughput and cost-effective next-generation sequencing technologies will almost certainly revolutionise the ability of investigators to compare the genomes, transcriptomes and epigenomes of patients with diabetes with and without nephropathy. Armed with these new technologies and improved international collaboration to increase sample sizes, there is a real sense that we have opportunities (hitherto unavailable) to ultimately identify the aetiology of DN and develop treatments for patients with this devastating condition.

References

  1. Wild S, Roglic G, Green A, et al., Global prevalence of diabetes: estimates for the year 2000 and projections for 2030, Diabetes Care, 2004;27:1047–53.
  2. Ray JA, Valentine WJ, Secnik K, et al., Review of the cost of diabetes complications in Australia, Canada, France, Germany, Italy and Spain, Curr Med Res Opin, 2005;21:1617–29.
  3. UK Renal Registry Report, UK Renal Registry, Bristol, 2007. Available at: www.renalreg.com
  4. US Renal Data System, USRDS 2007, Annual Data Report: Atlas of End-Stage Renal Disease in the United States, National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, 2007. Available at: www.usrds.org
  5. Ritz E, Pathogenesis, clinical manifestations and natural history of diabetic nephropathy, Comprehensive Clin Nephrol, 2007;13.
  6. Krolewski AS, Warram JH, Christlieb AR, et al., The changing natural history of nephropathy in type I diabetes, Am J Med, 1985;78:785–94.
  7. Perkins BA, Ficociello LH, Silva KH, et al., Regression of microalbuminuria in type 1 diabetes, N Engl J Med, 2003;348: 2285–93.
  8. Morgan CL, Currie CJ, Stott NC, et al., The prevalence of multiple diabetes-related complications, Diabet Med, 2000;17: 146–51.
  9. Remuzzi G, Schieppati A, Ruggenenti P, Clinical practice. Nephropathy in patients with type 2 diabetes, N Engl J Med, 2002;346:1145–51.
  10. Soldatos G, Cooper ME, Diabetic nephropathy: important pathophysiologic mechanisms, Diabetes Res Clin Pract, 2008;82(Suppl. 1):S75–9.
  11. The Diabetes Control and Complications Research Group, 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.
  12. UK Prospective Diabetes Study (UKPDS) Group, Intensive bloodglucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33), Lancet, 1998;352:837–53.
  13. Nathan DM, Cleary PA, Backlund JY, et al., Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) Study Research Group, Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes, N Engl J Med, 2005;353: 2643–53.
  14. Barnett AH, Treating to goal: challenges of current management, Eur J Endocrinol, 2004;151(Suppl. 2):T3–7.
  15. Quinn M, Angelico MC, Warram JH, et al., Familial factors determine the development of diabetic nephropathy in patients with IDDM, Diabetologia, 1996;39:940–45.
  16. Viberti GC, Keen H, Wiseman MJ, Raised arterial pressure in parents of proteinuric insulin dependent diabetics, Br Med J, 1987;295:515–17.
  17. De Cosmo S, Bacci S, Piras GP, et al., High prevalence of risk factors for cardiovascular disease in parents of IDDM patients with albuminuria, Diabetologia, 1997;40:1191–6.
  18. Fogarty DG, Hanna LS, Wantman M, et al., Segregation analysis of urinary albumin excretion in families with type 2 diabetes, Diabetes, 2000;49:1057–63.
  19. Ng DP, Tai BC, Koh D, et al., Angiotensin-I converting enzyme insertion/deletion polymorphism and its association with diabetic nephropathy, Diabetologia, 2005; 48:1008–16.
  20. Bain SC, Chowdhury TA, Genetics of diabetic nephropathy and microalbuminuria, J R Soc Med, 2000;93:62–6.
  21. Ng DP, Krolewski AS, Molecular genetic approaches for studying the etiology of diabetic nephropathy, Curr Mol Med, 2005;5:509–25.
  22. Savage DA, Bain SC, McKnight AJ, et al., Gene discovery in diabetic nephropathy, Curr Diabetes Report, 2007;7:139–45.
  23. Freedman BI, Bostrom M, Daeihagh P, et al., Genetic factors in diabetic nephropathy, Clin J Am Soc Nephrol, 2007;2:1306–16.
  24. Placha G, Canani LH, Warram JH, et al., Evidence for different susceptibility genes for proteinuria and ESRD in type 2 diabetes, Adv Chronic Kidney Dis, 2005;12:155–69.
  25. Wellcome Trust Case Control Consortium, Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls, Nature, 2007;447:661–78.
  26. Mohlke KL, Boehnke M, Abecasis GR, Metabolic and cardiovascular traits: an abundance of recently identified common genetic variants, Hum Mol Genet, 2008;17:R102–8.
  27. Lettre G, Rioux JD, Autoimmune diseases: insights from genome-wide association studies, Hum Mol Genet, 2008;17: R116–21.
  28. Easton DF, Eeles RA, Genome-wide association studies in cancer, Hum Mol Genet, 2008;17:R109–15.
  29. McCarthy MI, Abecasis GR, Cardon LR, et al., Genome-wide association studies for complex traits: consensus, uncertainty and challenges, Nat Rev Genet, 2008;9:356–69.
  30. Bodmer W, Bonilla C, Common and rare variants in multifactorial susceptibility to common diseases, Nat Genet, 2008;40:695–701.
  31. International HapMap Consortium, A haplotype map of the human genome, Nature, 2005;437:1299–1320.
  32. McCarthy MI, Hirschhorn JN, Genome-wide association studies: potential next steps on a genetic journey, Hum Mol Genet, 2008;17:R156–65.
  33. McCarthy MI, Casting a wider net for diabetes susceptibility genes, Nat Genet, 2008;40:1039–40.
  34. GAIN Collaborative Research Group, New models of collaboration in genome-wide association studies: the Genetic Association Information Network, Nat Genet, 2007;39: 1045–51.
  35. Mueller PW, Rogus JJ, Cleary PA, et al., Genetics of Kidneys in Diabetes (GoKinD) study: a genetics collection available for identifying genetic susceptibility factors for diabetic nephropathy in type 1 diabetes, J Am Soc Nephrol, 2006;17: 1782–90.
  36. Savage DA, Patterson CC, Deloukas P, et al., Genetic association analyses of non-synonymous single nucleotide polymorphisms in diabetic nephropathy, Diabetologia, 2008;51: 1998–2002.
  37. Thorn LM, Forsblom C, Fagerudd J, et al., Metabolic syndrome in type 1 diabetes: association with diabetic nephropathy and glycemic control (the FinnDiane study), Diabetes Care, 2005;28: 2019–24.
  38. Murphy M, Godson C, Cannon S, et al., Suppression subtractive hybridization identifies high glucose levels as a stimulus for expression of connective tissue growth factor and other genes in human mesangial cells, J Biol Chem, 1999;274:5830–34.
  39. McMahon R, Murphy M, Clarkson M, et al., IHG-2, a mesangial cell gene induced by high glucose, is human gremlin. Regulation by extracellular glucose concentration, cyclic mechanical strain, and transforming growth factor-beta1, J Biol Chem, 2000;275:9901–4.
  40. Dolan V, Murphy M, Sadlier D, et al., Expression of gremlin, a bone morphogenetic protein antagonist, in human diabetic nephropathy, Am J Kidney Dis, 2005;45:1034–9.
  41. Murphy M, Crean J, Brazil DP, et al., Regulation and consequences of differential gene expression in diabetic kidney disease, Biochem Soc Trans, 2008;36:941–5.
  42. Dolan V, Murphy M, Alarcon P, et al., Gremlin – a putative pathogenic player in progressive renal disease, Expert Opin Ther Targets, 2003;7:523–6.
  43. Walsh DW, Roxburgh SA, McGettigan P, et al., Co-regulation of Gremlin and Notch signalling in diabetic nephropathy, Biochim Biophys Acta, 2008;1782:10–21.
  44. Jones PA, Baylin SB, The epigenomics of cancer, Cell, 2007;128:683–92.
  45. Esteller M, Epigenetics in cancer, N Engl J Med, 2008;358: 1148–59.
  46. Gray SG, De Meyts P, Role of histone and transcription factor acetylation in diabetes pathogenesis, Diabetes Metab Res Rev, 2005;21:416–33.
  47. Dong C, Yoon W, Goldschmidt-Clermont PJ, DNA methylation and atherosclerosis, J Nutr, 2002;132:2406–9S.
  48. Sharma P, Kumar J, Garg G, et al., Detection of altered global DNA methylation in coronary artery disease patients, DNA Cell Biol, 2008;27:357–65.
  49. Ingrosso D, Cimmino A, Perna AF, et al., Folate treatment and unbalanced methylation and changes of allelic expression induced by hyperhomocysteinaemia in patients with uraemia, Lancet, 2003;361:1693–9.
  50. Zaina S, Lindholm MW, Lund G, Nutrition and aberrant DNA methylation patterns in atherosclerosis: more than just hyperhomocysteinemia?, J Nutr, 2005;135:5–8.
  51. Stenvinkel P, Karimi M, Johansson S, et al., Impact of inflammation on epigenetic DNA methylation - a novel risk factor for cardiovascular disease?, J Intern Med, 2007;261: 488–99.
  52. Soifer HS, Rossi JJ, Saetrom P, MicroRNAs in disease and potential therapeutic applications, Mol Ther, 2007;15: 2070–79.
  53. Mattes J, Collison A, Foster PS, Emerging role of microRNAs in disease pathogenesis and strategies for therapeutic modulation. Curr Opin Mol Ther, 2008;10:150–57.
  54. Couzin J, MicroRNAs make big impression in disease after disease, Science, 2008;319:1782–84.
  55. Kato M, Zhang J, Wang M, et al., MicroRNA-192 in diabetic kidney glomeruli and its function in TGF-beta-induced collagen expression via inhibition of E-box repressors, Proc Natl Acad Sci U S A, 2007;104:3432–7.
  56. Sethupathy P, Borel C, Gagnebin M, et al., Human microRNA-155 on chromosome 21 differentially interacts with its polymorphic target in the AGTR1 3’ untranslated region: a mechanism for functional single-nucleotide polymorphisms related to phenotypes, Am J Hum Genet, 2007;81:405–13.
  57. Wang Q, Wang Y, Minto AW, et al., MicroRNA-377 is up-regulated and can lead to increased fibronectin production in diabetic nephropathy, FASEB J, 2008. Epub ahead of print.
  58. Gregory PA, Bert AG, Paterson EL, et al., The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1, Nat Cell Biol, 2008;10:593–601.
  59. Saunders MA, Liang H, Li WH, Human polymorphism at microRNAs and microRNA target sites, Proc Natl Acad Sci U S A, 2007;104:3300–3305.
  60. Bentley DR, Whole-genome re-sequencing, Curr Opin Genet Dev, 2006;16:545–52.
  61. Wheeler DA, Srinivasan M, Egholm M, et al., The complete genome of an individual by massively parallel DNA sequencing, Nature, 2008;452:872–6.
  62. Mardis ER, The impact of next-generation sequencing technology on genetics, Trends Genet, 2008;24:133–41.
  63. Strausberg RL, Levy S, Rogers YH, Emerging DNA sequencing technologies for human genomic medicine, Drug Discov Today, 2008;13:569–77.
  64. Craig DW, Pearson JV, Szelinger S, et al., Identification of genetic variants using bar-coded multiplexed sequencing, Nat Methods, 2008;5:887–93.
  65. Albert TJ, Molla MN, Muzny DM, et al., Direct selection of human genomic loci by microarray hybridization, Nat Methods, 2007;4:903–5.

Further Resources

Share this Article
Related Content In Diabetes
  • Copied to clipboard!
    accredited arrow-down-editablearrow-downarrow_leftarrow-right-bluearrow-right-dark-bluearrow-right-greenarrow-right-greyarrow-right-orangearrow-right-whitearrow-right-bluearrow-up-orangeavatarcalendarchevron-down consultant-pathologist-nurseconsultant-pathologistcrosscrossdownloademailexclaimationfeedbackfiltergraph-arrowinterviewslinkmdt_iconmenumore_dots nurse-consultantpadlock patient-advocate-pathologistpatient-consultantpatientperson pharmacist-nurseplay_buttonplay-colour-tmcplay-colourAsset 1podcastprinter scenerysearch share single-doctor social_facebooksocial_googleplussocial_instagramsocial_linkedin_altsocial_linkedin_altsocial_pinterestlogo-twitter-glyph-32social_youtubeshape-star (1)tick-bluetick-orangetick-red tick-whiteticktimetranscriptup-arrowwebinar Sponsored Department Location NEW TMM Corporate Services Icons-07NEW TMM Corporate Services Icons-08NEW TMM Corporate Services Icons-09NEW TMM Corporate Services Icons-10NEW TMM Corporate Services Icons-11NEW TMM Corporate Services Icons-12Salary £ TMM-Corp-Site-Icons-01TMM-Corp-Site-Icons-02TMM-Corp-Site-Icons-03TMM-Corp-Site-Icons-04TMM-Corp-Site-Icons-05TMM-Corp-Site-Icons-06TMM-Corp-Site-Icons-07TMM-Corp-Site-Icons-08TMM-Corp-Site-Icons-09TMM-Corp-Site-Icons-10TMM-Corp-Site-Icons-11TMM-Corp-Site-Icons-12TMM-Corp-Site-Icons-13TMM-Corp-Site-Icons-14TMM-Corp-Site-Icons-15TMM-Corp-Site-Icons-16TMM-Corp-Site-Icons-17TMM-Corp-Site-Icons-18TMM-Corp-Site-Icons-19TMM-Corp-Site-Icons-20TMM-Corp-Site-Icons-21TMM-Corp-Site-Icons-22TMM-Corp-Site-Icons-23TMM-Corp-Site-Icons-24TMM-Corp-Site-Icons-25TMM-Corp-Site-Icons-26TMM-Corp-Site-Icons-27TMM-Corp-Site-Icons-28TMM-Corp-Site-Icons-29TMM-Corp-Site-Icons-30TMM-Corp-Site-Icons-31TMM-Corp-Site-Icons-32TMM-Corp-Site-Icons-33TMM-Corp-Site-Icons-34TMM-Corp-Site-Icons-35TMM-Corp-Site-Icons-36TMM-Corp-Site-Icons-37TMM-Corp-Site-Icons-38TMM-Corp-Site-Icons-39TMM-Corp-Site-Icons-40TMM-Corp-Site-Icons-41TMM-Corp-Site-Icons-42TMM-Corp-Site-Icons-43TMM-Corp-Site-Icons-44TMM-Corp-Site-Icons-45TMM-Corp-Site-Icons-46TMM-Corp-Site-Icons-47TMM-Corp-Site-Icons-48TMM-Corp-Site-Icons-49TMM-Corp-Site-Icons-50TMM-Corp-Site-Icons-51TMM-Corp-Site-Icons-52TMM-Corp-Site-Icons-53TMM-Corp-Site-Icons-54TMM-Corp-Site-Icons-55TMM-Corp-Site-Icons-56TMM-Corp-Site-Icons-57TMM-Corp-Site-Icons-58TMM-Corp-Site-Icons-59TMM-Corp-Site-Icons-60TMM-Corp-Site-Icons-61TMM-Corp-Site-Icons-62TMM-Corp-Site-Icons-63TMM-Corp-Site-Icons-64TMM-Corp-Site-Icons-65TMM-Corp-Site-Icons-66TMM-Corp-Site-Icons-67TMM-Corp-Site-Icons-68TMM-Corp-Site-Icons-69TMM-Corp-Site-Icons-70TMM-Corp-Site-Icons-71TMM-Corp-Site-Icons-72