Genetics of Diabetes and Its Complications

Stephen S. Rich Department of Public Health Sciences, The Wake Forest University School of Medicine, Winston-Salem, North Carolina J Am Soc Nephrol 17: 353–360, 2006. doi: 10.1681/ASN.2005070770

iabetes constitutes a major public health problem. The genetic basis for type 2 diabetes has been difficult to Although substantial progress has been made in de- resolve. Unlike type 1 diabetes, in which there seems to be an D fining the genetic risk for specific subtypes of diabe- autoimmune process, type 2 diabetes is a disease of relative tes (e.g., maturity-onset diabetes of the young), the majority of rather than absolute insulin deficiency. In type 2 diabetes, the genetic risk of diabetes (for type 1 and type 2) remain unre- pancreatic ␤ cells become progressively less able to secrete solved. This review focuses on the current knowledge of the sufficient insulin to maintain normal carbohydrate and lipid genetic basis of diabetes and its complications, specifically di- homeostasis (7). Furthermore, there is insulin resistance cou- abetic nephropathy (DN). Ultimately, identification of pled with effects of aging, obesity, and reduced exercise (8). In that contribute to risk (or protection) of diabetes and its com- addition, the strength of the genetic contribution to the cause of plications will allow identification of patients who have diabe- type 2 diabetes seems to be less than that for type 1 diabetes, tes and are at risk and targeted treatment/interventional strat- with overall genetic risk ratio ranging from 2 to 4 (9). Despite egies. the relatively low genetic risk ratio for type 2 diabetes, results of several genomewide scans have reported a number of chro- Genetics of Diabetes mosomal regions that may harbor genes that are involved in Type 1 diabetes is the third most prevalent chronic disease of type 2 diabetes, with the most promising, replicated findings on childhood, affecting up to 0.4% of children in some populations 1q21-q24, 2q37, 12q24 and 20 (reviewed in ref. by age 30 yr, with an overall lifetime risk of nearly 1% (1,2). It [10]). The finding of linkage to type 2 diabetes in Mexican is believed that a large proportion of cases of type 1 diabetes Americans to 2q37 (11) led to the characterization result from the autoimmune destruction of the pancreatic ␤ of susceptibility variants within the CAPN10 (12). Other cells, leading to complete dependence on exogenous insulin to type 2 diabetes linkage regions are being searched by develop- regulate blood glucose levels (3). Type 1 diabetes is strongly ment of consortia (the International 1q Consortium), and novel clustered in families with an overall genetic risk ratio (the type 2 diabetes susceptibility genes may be identified in the prevalence in siblings of a proband relative to the population ␭ future using the same approach. Several type 2 diabetes genes prevalence, S) of approximately 15 (4). (This compares with ␭ have been identified using investigations of candidate genes. the less familial but more prevalent type 2 diabetes with S of approximately 2). At least one that contributes strongly to Common variants in peroxisome proliferator–activated recep- ␥ this familial clustering resides within the MHC on chromosome tor- (13) and KCNJ11 (14) may predispose to type 2 diabetes 6p21, which accounts for nearly 40% of the observed familial with relatively low odds ratios (approximately 1.2). Variants clustering of type 1 diabetes, with a locus-specific genetic risk that affect insulin signaling through IRS-1 (15,16) and glucose ␭ homeostasis PTPN1 (17,18) seem to be less common in fre- ratio ( S) of approximately 3 (5). In a recent analysis of data from three previous genomewide scans (United States, United quency, suggesting that combinations of rare and common Kingdom, and Scandinavia) as well as new families collected variants contribute to risk for type 2 diabetes in different path- for the Type 1 Diabetes Genetics Consortium (http:// ways in different populations. www.t1dgc.org), 1435 multiplex families provided evidence for In summary, there is ample evidence that type 1 and type 2 linkage of type 1 diabetes to the MHC (IDDM1), insulin (INS, diabetes are, in part, genetically determined. The magnitude of IDDM2), a region that contains several genes, including CTLA4 the genetic contribution to type 1 and type 2 diabetes differs ␭ (2q31-q33 [IDDM12 and IDDM7]) and seven other chromosome dramatically (as shown by the genetic risk ratio [ S] for type 1 ␭ regions (6). diabetes of 15, but S is approximately 2 for type 2 diabetes). Furthermore, there is evidence that the genes that may contrib- ute to risk for type 1 diabetes may differ from those that

Published online ahead of print. Publication date available at www.jasn.org. contribute to risk for type 2 diabetes. As we show below, the genetic risk for diabetic complications may be defined by yet Address correspondence to: Dr. Stephen S. Rich, Department of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC. Phone: another set of genes than those that modify risk for diabetes 336-716-5778; Fax: 336-716-5425; E-mail: [email protected] alone.

Copyright © 2006 by the American Society of Nephrology ISSN: 1046-6673/1702-0353 354 Journal of the American Society of Nephrology J Am Soc Nephrol 17: 353–360, 2006

Indirect Evidence that Favors a Genetic Basis basis of disease as well as the available source of subjects of DN (Figure 1). Susceptibility genes that are prevalent with large The complications of diabetes involve several organ systems effect are unlikely to exist (single Mendelian genes with and are often categorized as microvascular (retinopathy, ne- large effect are usually rare). Similarly, genes that are rare with phropathy, and neuropathy) or macrovascular (cardiovascular small effect are unlikely to be detectable with current analytical and cerebrovascular). There is mounting evidence for the role strategies. In contrast, prevalent genes with small effect (com- of genetic factors in several diabetic complications, particularly mon variant) or rare with large effect (rare variant) may be DN, and cardiovascular and cerebrovascular complications of identified (32–34). Under a common variant hypothesis, asso- diabetes (19). DN remains the most common and rapidly in- ciation approaches (case-control, cohort studies) using a dense creasing cause of ESRD in developed nations. Approximately marker map have been proposed (whole genome association 35% of European Americans with diabetes are at risk for the scans). Under a rare variant hypothesis, linkage approaches development of overt proteinuria, chronic renal insufficiency, using extended families or sibling pairs (whole genome linkage and ESRD, with higher proportions observed among diabetic scans) often have been performed (Figure 2). African Americans, Hispanic Americans, and Native Ameri- Within either the common variant or rare variant hypotheses, cans (20). Genetic predisposition or protection may be the most both candidate gene evaluations and genome scans have been important DN risk determinants in type 1 diabetes. Only ap- conducted. The characterization of the contribution of variants proximately one half of patients with poor glycemic control within candidate genes has evolved into two (often overlap- develop DN, whereas some patients do so despite relatively ping) strategies. The first strategy involves the study of poly- good control (21). Familial clustering of DN has been demon- morphisms within individual genes that should (based on the strated by several investigators (22–24). underlying biologic knowledge) have an effect on risk. The The majority of individuals with diabetes will develop back- second is to explore a cluster of genes that may be involved in ground diabetic retinopathy; however, only one third of indi- a pathophysiologic pathway, recognizing that multiple genes in viduals with diabetes will develop overt kidney disease (25). the pathway may have recognized effects with each other and The factors that initiate overt nephropathy are unclear, but unrecognized effects on genes in the pathway and other genes consistent familial aggregation of kidney disease (albuminuria, (35–37). In each scenario, issues related to candidate gene se- creatinine clearance, and ESRD) and renal histologic changes lection, polymorphism evaluation within candidate genes, and have been observed frequently. The familial clustering of overt analysis of multiple genes in pathways remain a significant and DN and diabetic ESRD has been observed widely in multiple unresolved area of research. racial and ethnic groups, with the earliest reports of familial aggregation of diabetic kidney disease in patients with type 1 Pathways and Candidate Genes Involved in diabetes (22,24). Family members with diabetes, even in the DN absence of clinical nephropathy, demonstrate similar patterns Current knowledge of the pathophysiology of DN suggests a of glomerular involvement. Renal biopsy material from sibling complex interaction of multiple pathways associated with al- pairs with type 1 diabetes were evaluated, and evidence of terations in the balance between produc- familial concordance in the percentage of the glomerulus occu- tion and removal, with interactions between genetic and envi- pied by mesangial matrix and the patterns of injury (presence ronmental factors. These pathophysiologic concepts have been of thickened glomerular basement membrane) were observed reviewed previously (38,39). The genes that contribute to vari- (26). In addition, consistent evidence across multiple studies ation and expression in the pathways are just now being ex- also supports familial aggregation of albuminuria (27,28), GFR, plored in the context of complex metabolic networks. Complex- and creatinine clearance (29–31).

Approaches to Identifying DN Susceptibility Genes Multiple strategies exist for exploring the for evidence of genetic factors that contribute to risk for complex disease (e.g., not determined by a defect in a single gene, often termed “Mendelian”). DN is an example of a complex human disease, with many genes and environmental risk factors con- tributing to susceptibility. These risk factors may be of differing magnitude, and not all may be required for initiation or pro- gression of disease. Furthermore, the constellation of factors may vary from one population to another. All approaches to detect disease susceptibility genes are based on demonstrating segregation of a trait (DN) with a measure of genetic variation. These approaches are dependent Figure 1. Effect and frequency of risk alleles dictate mapping on the unobservable “true” genetic architecture of the genetic strategies. J Am Soc Nephrol 17: 353–360, 2006 Genetics of Diabetes and Its Complications 355

be due to many reasons, including small sample size, incom- plete genetic dissection of the polymorphisms in the candidate gene, and extensive genetic and phenotypic heterogeneity. A partial list of candidate genes for DN is provided in Table 1.

Search for DN Susceptibility Genes The search for genes that influence risk for DN can take place at many levels. The two classical approaches are the case- control and family-based designs. Within the case-control de- sign, the vast majority of studies have used a candidate gene approach, whether interrogating individual candidate genes or genes related to specific biologic pathways. The case-control Figure 2. Methods for genetic dissection of complex human design determines the evidence of association between a ge- traits. Solid symbols represent patients (diabetic nephropathy); netic variant and the outcome of DN or an intermediate, quan- open symbols represent control subjects (individuals with dia- titative phenotype (quantitative trait locus [QTL]). Within the betes of long duration without nephropathy). family-based approach, the unit of analysis is often a pair of siblings, as parents are often unavailable for study (except in the case of type 1 DN). Several general analyses of family data ity is also observed in the variation in structural features (e.g., have been used: The genome scan for linkage of DN, the the thickness in the capillary wall, mesangial matrix volume, candidate gene evaluation of linkage, and the family-based and width of the GBM), which have been shown to be deter- association approach. Recently, interest in mapping QTL for mined, in part, by genetic factors (26). Furthermore, the diabetic complex disease has been increasing. For example, the clinical state itself can modify the observed genetically determined phenotype (DN) is the result of multiple genetic and environ- structures. mental factors; however, the underlying physiologic (quantita- The major hypotheses as to how the diabetic state leads to tive) trait (e.g., urinary albumin excretion) may have stronger DN include (1) increased activity of a variety of growth factors, genetic effects (in the absence of those environmental factors including TGF-␤, growth hormone, IGF-1, vascular endothelial that lead to overt disease). Thus, the genetic predisposition to growth factor, and EGF; (2) activation of protein kinase C nephropathy may be reflected in the level of urinary albumin isoforms; (3) increased release of hormones, including renin, excretion as a quantitative trait, and, with exposure to diabetes, angiotensin, endothelin, and bradykinin; (4) formation of reac- high-normal levels of urinary albumin excretion may worsen to tive oxygen species; (5) increased formation of advanced gly- microalbuminuria and overt proteinuria. An implication of this cation end-products (AGE); (6) increased activity of the aldose hypothesis is that the genetic variation in the quantity of albu- reductase pathway; and (7) abnormalities in glucose transport min excreted in the urine is determined by the same QTL as mechanisms. The various hypotheses are interrelated, and the those genes that predispose to nephropathy but may not be key components of the pathways are genetically determined. dependent on the diabetic state. Results of mapping genes for Recently, a unifying hypothesis that links several important DN or urinary albumin excretion, under this model, would lead pathways in the pathogenesis of DN was developed (40). This to the same subset of genes in a common pathway. concept theorizes that hyperglycemia-induced mitochondrial In a genome screen, no a priori hypothesis is made as to the superoxide overproduction results in an increased activation of nature or the location of the specific genetic susceptibility genes protein kinase C isoforms, increased formation of AGE, accel- for DN or its intermediate phenotypes. Instead, the genetic eration of glucose flux through the aldose reductase pathway, determinants of these phenotypes are tested on a large number and an increased glucose flux into the hexosamine pathway. of polymorphic DNA markers scattered at roughly equal inter- These perturbations, with variation determined in part by vals throughout the human genome. This approach has been genes in the specific pathways that interact with environmental applied successfully in a number of Mendelian disorders, with factors, stimulate growth factors that result in extracellular investigations proceeding from positive linkage results to link- matrix accumulation, leading to DN. age disequilibrium mapping to gene identification. Once link- Many studies of candidate genes have examined functional age has identified a region in the genome that may contain a polymorphisms that affect levels (or activity) in many candi- susceptibility gene, use of markers in the narrow region in cases date pathways, including the renin-angiotensin, nitric oxide, and controls (for example) could find markers that are associ- and bradykinin systems. Other pathways that are involved ated (rather than linked) to the disease. Current strategy uses with insulin resistance, lipid production, and metabolism and haplotype-tagging single-nucleotide polymorphisms (SNP) de- glucose homeostasis, including aldose reductase, TGF-␤, and rived from HapMap Project, which allows the definition of glucose transporter-1, have been explored because of common causative “haplotypes” that segregate with disease in the fam- mechanisms that may be involved in several disease processes. ilies. There has been no consistent and replicated evidence for the It is, however, a much greater challenge to apply this ap- contributions by a set of candidate gene(s) or surrogate markers proach to genetically complex disorders. For most complex for DN risk (or protection). Failure to identify risk genes could diseases, it is likely that multiple genes influence disease ex- 356 Journal of the American Society of Nephrology J Am Soc Nephrol 17: 353–360, 2006

Table 1. A subset of candidate genes for diabetic nephropathya

Symbol Name Position Rationale (References)

NHE-1 (SLC9A1) Naϩ/H ϩ antiport-1 1p36.1-p35 NHE-1 activity is increased in type 1 diabetes with nephropathy (51,52) TGF-␤ (TGF-␤1, Transforming growth factor- 19q12-q13.31, 1q41, 14q24 TGF-␤ mRNA, proteins (and TGF-␤2, TGF-␤3) ␤ (1, 2, 3) TGF-␤-receptor mRNA) identified in rodent glomerular cells (53–55) GH1 Growth hormone 17q24.2 Diabetes causes decreased hepatic production of IGF- IGF-1, IGF-1R Insulin-like growth factor-1, 12q22-q23, 15q26.3 1; this decrease leads to an insulin-like growth factor- increased in growth 1 receptor hormone secretion (56–60) VEGF Vascular endothelial growth 6p12 VEGF expression increased factor in glomeruli of diabetic animals; diabetic rats that are treated with anti- VEGF antibody do not have hyperfiltration and reduced AER (61–63) RAAS ACE, AGT, AT1 (AGTR1), 17q23, 1q42-q43, 3q21-q25, BP regulation and sodium AT2 (AGTR2) Xq22-q23 homeostasis are crucial to progression of nephropathy (64–66)

aAER, albumin excretion ratio, RAAS, renin-angiotensin-aldosterone system; ACE, angiotensin-converting inhibitor; AGT, angiotensin-1; AT1, angiotensin II receptor type 1; AT2, angiotensin II receptor type 2.

pression, making it difficult to isolate and characterize the genes). The advantage of the genome screen strategy is the effects of each disease-determining locus. Genes may interact comprehensive evaluation of the entire genome. (epistasis) or induce susceptibility independently (heterogene- To date, only a few full descriptions of genome scans for DN ity). In addition, complex diseases such as DN are influenced have been published. The first genome scan that searched for by environmental factors. nephropathy and retinopathy loci (microvascular complica- Despite these complicating factors, the genome screen strat- tions) was performed in the Pima Indians (44). The strongest egy has been successful for type 1 diabetes (6), type 2 diabetes evidence for linkage was observed on chromosome 7q. Addi- (12), Crohn’s disease (41), asthma (42), and schizophrenia (43). tional evidence for linkage was found on chromosomes 3 and 9 The likelihood of success has increased with advances in high- (DN and retinopathy) and 20 (DN only). A similar approach throughput genotyping, development of semiparametric ge- was used in families with type 1 diabetes (45), in which linkage netic analysis methods, the potential to accumulate sufficient was found to a region that contains the angiotensin II type 1 patient and family materials, and more refined definition and receptor gene (AGTR1). Other genome scans were completed measurement of phenotypes. Although the most complex to recently. Members of a large Turkish kindred that contains execute, genome scans of appropriate family collections with multiple individuals who have type 2 DN (with multiple cous- significant prevalence of DN offer hope for identifying the in–cousin marriages and significant inbreeding) were subjected primary genetic components of DN. For complex genetic enti- to a genome scan, and a strong linkage peak was observed on ties such as DN, analysis of 1000 DNA (or more) from many chromosome 18 (46). Evaluation of these loci in the Pima Indi- different families is necessary. This corresponds to Ͼ400,000 ans also showed evidence of linkage, although this region was genotypes. The microsatellite or SNP markers are not specifi- not linked in the original Pima genome scan. The first genome cally associated with genes but are markers for the inheritance scan of DN in African American families has only now been of each small segment of the human genome within which they completed (47). Evidence for a nephropathy locus on chromo- lie. When the genome scan is completed, the result is a data set some 3 was detected in approximately 30% of the families, on that follows the inheritance of every part of every chromosome chromosome 7p in approximately 40% of the families (those in each individual in the study. The resulting statistical analy- with longer duration of diabetes before diagnosis of ESRD), and ses would identify those chromosomal regions with the stron- on chromosome 18q in approximately 65% of the families gest evidence of linkage (assumed to contain the susceptibility (those with early age of diabetes diagnosis). Although these J Am Soc Nephrol 17: 353–360, 2006 Genetics of Diabetes and Its Complications 357 genome scan studies have been performed in diverse popula- complications for targeted intervention and to continue to ex- tions with different study designs and analytical methods, in- plore underlying mechanisms that contribute to their greater vestigators can take some encouragement from the results. risk for disease. Genetic characterization is one important facet Several of these linkage locations are consistent between stud- of an individual’s risk profile that has yet to be proved as an ies. These complementary data increase the likelihood that efficacious strategy, yet it has the potential to allow a targeted several chromosomal locations mapped by linkage analysis and effective approach to identify a smaller subset of patients reflect the presence of true DN genes. for whom specific interventions/treatments should be most A major limitation in gene identification has been adequate effective. sample size with carefully and consistently phenotyped indi- It is apparent from genetic studies of DN and studies of other viduals. Several major multicenter studies are examining sub- diseases such as type 1 diabetes and type 2 diabetes that con- jects and establishing renewable resources for genetic studies, firmation of results in different populations are of great value in particularly in the area of DN. These initiatives include GoKinD trying to assess the validity of individual findings. Evidence of (Genetics of Kidney in Diabetes), FIND (Family Investigation of confirmation from multiple studies is the most widely used Nephropathy and Diabetes), and EDIC (Epidemiology of Dia- metric for accepting evidence of the importance of a gene in betes In Complications) Genetics. The purpose of the GoKinD contribution to variation to risk for diabetes and its complica- Study is to establish a repository of DNA and clinical informa- tions. Although this outcome would be comforting, there are (at tion from adults with long-term type 1 diabetes, with or with- least) two situations that limit the utility of population replica- out kidney disease, along with their parents (trios). The Go- tion as a means for determining success of susceptibility gene KinD study has screened Ͼ5500 participants and obtained identification. One limitation is if the “truth” in the genetics of DNA to generate lymphoblast cell lines that are available for diabetes or complications is in a series of population-specific researchers. The FIND study also has established immortalized private mutations within a common susceptibility framework. cell lines from approximately 6000 individuals in families that In this case, each population would be consistent with evidence are enriched for the presence of overt DN (mainly from type 2 for linkage at multiple sites, but the specific pattern of muta- diabetes). A data from a sibling without diabetes has also been tions could be different. A second limitation is that the same collected for mapping of type 2 diabetes genes. African Amer- constellation of genetic factors contributes to susceptibility in ican, European American, Mexican American, and American all populations, but the subset of factors and effect sizes may Indian families are being recruited in nearly equal numbers. differ. Thus, simple comparison of results across populations The EDIC Genetics study has recruited nearly 1500 individuals may provide erroneous “failure to replicate.” with type 1 diabetes (including measures of nephropathy and The technology for carrying out large-scale molecular genetic retinopathy) and 3000 relatives. Thus, in the near future, a studies is rapidly improving. In the near future, it will be significant set of resources will become available for investiga- possible to combine the power and the ease of association tors of the genetic basis of type 1 and 2 diabetes and DN. studies with comprehensive examination of the genome (whole genome association). One intriguing study that used elements Summary of this approach has been reported (50) with 55,000 gene-based For effective reduction of the effect of diabetes and its com- SNP loci. A single gene encoding solute carrier family 12 mem- plications and numerous public and personal health conse- ber3(SLC12A3), the gene associated with Gitelman’s syn- quences, every effort must be made to minimize the develop- drome, exhibited consistent evidence for association with DN ment of diabetes and early progression to diabetic susceptibility with a single SNP in intron 24 having statistically complications. A current limitation of progress in this regard is significant association. The results suggested that the uncertainty on who is “at risk.” Although there are no recog- Arg913Gln substitution in the SLC12A3 gene provided protec- nized preventive measures for type 1 diabetes, numerous stud- tion from development of DN. With increasing coverage of the ies have demonstrated the relative efficacy of weight loss, diet, genome (approximately 500,000 SNP), these studies may be- and exercise on normalization of glucose homeostasis, even in come a valuable tool in identification of common variants that individuals with no clinical manifestations of type 2 diabetes enhance the search for diabetes and complication susceptibility (48,49). Unfortunately, despite significant relative risk reduc- genes. tion of these procedures, the absolute risk reduction remains low, resulting in the need to treat several hundred “healthy” (without diabetes) individuals to prevent a single person from References progressing to overt disease event. The low absolute risk re- 1. Rewers M, LaPorte RE, King H, Tuomilehto J: Trends in the duction in any single individual may also contribute to the prevalence and incidence of diabetes: Insulin-dependent diabetes mellitus in childhood. World Health Stat Q 41: underutilization of these procedures by patients and would 179–189, 1988 require substantial increases in health care cost if it were ad- 2. Rewers MJ: The changing face of the epidemiology of ministered to all patients who are at risk. It is also clear that insulin-dependent diabetes mellitus (IDDM): Research de- many people develop diabetes and (ultimately) diabetic com- signs and models of disease causation. Ann Med 23: 419– plications despite the absence of these risk factors. These facts 426, 1991 underscore the need to develop more effective means to iden- 3. Genuth S, Alberti KG, Bennett P, Buse J, Defronzo R, Kahn tify individuals who are most likely to develop diabetes and R, Kitzmiller J, Knowler WC, Lebovitz H, Lernmark A, 358 Journal of the American Society of Nephrology J Am Soc Nephrol 17: 353–360, 2006

Nathan D, Palmer J, Rizza R, Saudek C, Shaw J, Steffes M, E, Tuncman G, Gorgun C, Glimcher LH, Hotamisligil GS: Stern M, Tuomilehto J, Zimmet P: Expert Committee on the Endoplasmic reticulum stress links obesity, insulin action, Diagnosis and Classification of Diabetes Mellitus. Fol- and type 2 diabetes. Science 306: 457–461, 2004 low-up report on the diagnosis of diabetes mellitus. Dia- 16. Kaneto H, Nakatani Y, Miyatsuka T, Kawamori D, Mat- betes Care 31: 3160–3167, 2003 suoka TA, Matsuhisa M, Kajimoto Y, Ichijo H, Yamasaki Y, 4. Pociot F, McDermott MF: Genetics of type 1 diabetes mel- Hori M: Possible novel therapy for diabetes with cell- litus. Genes Immun 3: 235–249, 2002 permeable JNK-inhibitory peptide. Nat Med 10: 1128–1132, 5. Rich SS: Mapping genes in diabetes: A genetic epidemio- 2004 logical perspective. Diabetes 39: 1315–1319, 1990 17. Palmer ND, Bento JL, Mychaleckyj JC, Langefeld CD, 6. Concannon P, Erlich HA, Julier C, Morahan G, Pociot F, Campbell JK, Norris JM, Haffner SM, Bergman RN, Bow- Nerup J, Todd JA, Rich SS: Type 1 diabetes: Evidence for den DW: Association of protein tyrosine phosphatase 1B susceptibility loci from four genome-wide scans in 1435 gene polymorphisms with measures of glucose homeosta- multiplex families. Diabetes 54, 2995–3001, 2005 sis in Hispanic Americans: The Insulin Resistance Athero- 7. Bell GI, Polonsky KS: Diabetes mellitus and genetically sclerosis Study (IRAS) Family Study. Diabetes 53: 3013– programmed defects in beta-cell function. Nature 414: 788– 3019, 2004 791, 2001 18. Bento JL, Palmer ND, Mychaleckyj JC, Lange LA, Lange- 8. Saltiel AR, Kahn CR: Insulin signalling and the regulation feld CD, Rich SS, Freedman BI, Bowden DW: Association of glucose and lipid metabolism. Nature 414: 799–806, 2002 of protein tyrosine phosphatase 1B polymorphisms with 9. Weijnen CF, Rich SS, Meigs JB, Krolewski AS, Warram JS: type 2 diabetes. Diabetes 53: 3007–3012, 2004 Risk of diabetes in siblings of index cases with type 2 19. Bowden DW: Genetics of diabetes complications. Curr Diab diabetes: Implications for genetic studies. Diabet Med 19: Rep 2: 191–200, 2002 41–50, 2002 20. US Renal Date System: USRDS 2003 Annual Report: Atlas of 10. Demenais F, Kanninen T, Lindgren CM, Wiltshire S, Gaget End-Stage Renal Disease in the United States, Bethesda, Na- S, Dandrieux C, Almgren P, Sjogren M, Hattersley A, Dina tional Institutes of Health, National Institute of Diabetes C, Tuomi T, McCarthy MI, Froguel P, Groop LC: A meta- and Digestive and Kidney Diseases, 2003 analysis of four European genome screens (GIFT Consor- 21. Krolewski M, Eggers PW, Warram JH: Magnitude of end- tium) shows evidence for a novel region on chromosome stage renal disease in IDDM: A 35-year follow-up study. 17p11.2-q22 linked to type 2 diabetes. Hum Mol Genet 12: Kidney Int 50: 2041–2046, 1996 1865–1873, 2003 22. Seaquist ER, Goetz FC, Rich SS, Barbosa J: Familial clus- 11. Hanis CL, Boerwinkle E, Chakraborty R, Ellsworth DL, tering of diabetic kidney disease: Evidence for genetic sus- Concannon P, Stirling B, Morrison VA, Wapelhorst B, ceptibility to diabetic nephropathy. N Engl J Med 320: 1161– Spielman RS, Gogolin-Ewens KJ, Shepard JM, Williams SR, 1165, 1989 Risch N, Hinds D, Iwasaki N, Ogata M, Omori Y, Petzold 23. Borch-Johnsen K, Norgaard K, Hommel E, Mathiesen ER, C, Rietzch H, Schroder HE, Schulze J, Cox NJ, Menzel S, Jensen JS, Deckert T, Parving HH: Is diabetic nephropathy Boriraj VV, Chen X, Lim LR, Lindner T, Mereu LE, Wang an inherited complication? Kidney Int 41: 719–722, 1992 YQ, Xiang K, Yamagata K, Yang Y, Bell GI: A genome-wide 24. Quinn M, Angelico MC, Warram JH, Krolewski AS: Famil- search for human non-insulin-dependent (type 2) diabetes ial factors determine the development of diabetic nephrop- genes reveals a major susceptibility locus on chromosome athy in patients with IDDM. Diabetologia 39: 940–945, 1996 2. Nat Genet 13: 161–166, 1996 25. Microvascular and acute complication in IDDM patients: 12. Horikawa Y, Oda N, Cox NJ, Li X, Orho-Melander M, Hara The EURODIAB IDDM Complications Study. Diabetologia M, Hinokio Y, Lindner TH, Mashima H, Schwarz PE, del 37: 278–285, 1994 Bosque-Plata L, Horikawa Y, Oda Y, Yoshiuchi I, Colilla S, 26. Fioretto P, Steffes MW, Barboso J, Rich SS, Miller ME, Polonsky KS, Wei S, Concannon P, Iwasaki N, Schulze J, Mauer M: Is diabetic nephropathy inherited? Studies of Baier LJ, Bogardus C, Groop L, Boerwinkle E, Hanis CL, glomerular structure in type 1 diabetic sibling pairs. Dia- Bell GI: Genetic variation in the gene encoding calpain-10 betes 48: 865–869, 1999 is associated with type 2 diabetes mellitus. Nat Genet 26: 27. Langefeld CD, Beck SR, Bowden DW, Rich SS, Wagen- 163–175, 2000 knecht LE, Freedman BI: Heritability of GFR and albumin- 13. Altshuler D, Hirschhorn JN, Klannemark M, Lindgren CM, uria in Caucasians with type 2 diabetes mellitus. Am J Vohl MC, Nemesh J, Lane CR, Schaffner SF, Bolk S, Brewer Kidney Dis 5: 796–800, 2004 C, Tuomi T, Gaudet D, Hudson TJ, Daly M, Groop L, 28. Freedman BI, Beck SR, Rich SS, Heiss G, Lewis CE, Turner Lander ES: The common PPARg Pro12Ala polymorphism S, Province MA, Schwander KL, Arnett DK, Mellen BG: A is associated with decreased risk of type 2 diabetes. Nat genome-wide scan for urinary albumin excretion in hyper- Genet 26: 76–80, 2000 tensive families. Hypertension 42: 291–296, 2003 14. Gloyn AL, Weedon MN, Owen KR, Turner MJ, Knight BA, 29. Hunter DJ, Lange M, Snieder H, MacGregor AJ, Swami- Hitman GA, Walker M, Levy JC, Sampson MJ, Halford S, nathan R, Thakker RV, Spector TD: Genetic contribution to McCarthy MI, Hattersley AT, Frayling TM: Large scale renal function and electrolyte balance: A twin study. Clin association studies of variants in genes encoding pancre- Sci 103: 259–265, 2002 atic beta-cell K-ATP channel subunits Kir6.2 (KCNJ11) and 30. DeWan AT, Arnett DK, Atwood LD, Province MA, Lewis SUR1 (ABCC8) confirm that the KCNJ11 E23K variant is CE, Hunt SC, Eckfeldt J: A genome scan for renal function associated with increased risk for type 2 diabetes suscep- among hypertensives: The HyperGEN study. Am J Hum tibility. Am J Hum Genet 52: 568–572, 2003 Genet 68: 136–144, 2000 15. Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen 31. Hunt SC, Hasstedt SJ, Coon H, Camp NJ, Cawthon RM, J Am Soc Nephrol 17: 353–360, 2006 Genetics of Diabetes and Its Complications 359

Wu LL, Hopkins PN: Linkage of creatinine clearance to chromosome 3q: Results of novel discordant sib-pair anal- chromosome 10 in Utah pedigrees replicates a locus for ysis. Diabetes 47: 1164–1169, 1998 end-stage renal disease in humans and renal failure in the 46. Vardarli I, Baier L, Hanson JA, Akkoyun I, Fischer C, fawn-hooded rat. Kidney Int 62: 1143–1148, 2002 Rohmeiss P, Basci A, Bartram CR, Van Der Woude FJ, 32. Risch N, Merikangas K: The future of genetic studies of Janssen B: Gene for susceptibility to diabetic nephropathy complex human diseases. Science 273: 1516–1517, 1996 in type 2 diabetes maps to 18q22.3-23. Kidney Int 62: 2176– 33. Alshuler D, Daly M, Kruglyak L: Guilt by association. Nat 2183, 2002 Genet 26: 135–137, 2000 47. Bowden DW, Colicigno CJ, Langefeld CD, Sale MM, Wil- 34. Altmuller J, Palmer LJ, Fischer G, Scherb H, Wjst M: liams A, Anderson PJ, Rich SS, Freedman BI: A genome Genomewide scans of complex human diseases. True link- scan for diabetes associated end-stage renal disease in Af- age is hard to find. Am J Hum Genet 69: 936–950, 2001 rican Americans. Kidney Int 66: 1517–1526, 2004 35. Hill AB: The environment and disease: Association or cau- 48. Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, sation. Proc R Soc Med 58: 295–300, 1965 Lachin JM, Walker EA, Nathan DM: Diabetes Prevention 36. Clayton D, McKeigue PM: Epidemiological methods for Program Research Group: Reduction in the incidence of studying genes and environmental factors in complex dis- type 2 diabetes with lifestyle intervention or metformin. eases. Lancet 358: 1356–1360, 2001 N Engl J Med 346: 393–403, 2002 37. Chakravarti A: Nature, nurture and human disease. Nature 49. Kriska AM, Saremi A, Hanson RL, Bennett PH, Kobes S, 421: 413–414, 2003 Williams DE, Knowler WC: Physical activity, obesity, and 38. Caramori ML, Mauer M: Diabetes and nephropathy. Curr the incidence of type 2 diabetes in a high-risk population. Opin Nephrol Hypertens 12: 273–282, 2003 Am J Epidemiol 158: 669–675, 2003 39. Brownlee M, Livingston JN: Biochemical mechanisms of 50. Tanaka N, Babazono T, Saito S, Sekine A, Tsunoda T, microvascular disease. In: Ellenberg & Rifkin’s Diabetes Mel- Haneda M, Tanaka Y, Fujioka T, Kaku K, Kawamori R, litus, 6th Ed., edited by Porte D Jr, Sherwin RS, Baron A, Iwamoto Y, Nakamura Y, Maeda S: Association of solute New York, McGraw Hill, 2003, pp 181–196 carrier family 12 (sodium/chloride) member 3 with dia- 40. Du X-L, Edelstein D, Rossetti L, Fantus IG, Goldberg H, betic nephropathy, identified by genome-wide analyses of single nucleotide polymorphisms. Diabetes 52: 2848–2853, Ziyadeh F, Wu J, Brownlee M: Hyperglycemia-induced 2003 mitochondrial superoxide overproduction activates the 51. Trevisan R, Li LK, Messent J, Tariq T, Earle K, Walker JD, hexosamine pathway and induces plasminogen activator Viberti G: Naϩ/Hϩ antiport activity and cell growth in inhibitor-1 expression by increasing Sp1 glycosylation. cultured skin fibroblasts of IDDM patients with nephrop- Proc Natl Acad Sci U S A 97: 12222–12226, 2000 athy. Diabetes 41: 1239–1246, 1992 41. Hugot J, Chamaillard M, Zouali H, Lesage S, Cezard J, 52. Lurbe A, Fioretto P, Mauer M, LaPointe MS, Batlle D: Belaiche J, Almer S, Tysk C, O’Morain C, Gassull M, Binder Growth phenotype of cultured skin fibroblasts from IDDM V, Finkel Y, Cortot A, Modigliani R, Laurent-Puig P, patients with and without nephropathy and overactivity of Gower-Rousseau C, Macry J, Colombel J, Sahbatou M, the Naϩ/Hϩ antiporter. Kidney Int 50: 1684–1693, 1996 Thomas G: Association of Nod2 leucine-rich repeat vari- 53. Shankland SJ, Scholey JW, Ly H, Thai K: Expression of ants with susceptibility to Crohn’s disease. Nature 411: transforming growth factor-beta 1 during diabetic renal 599–602, 2001 hypertrophy. Kidney Int 46: 430–442, 1994 42. Van Eerdewegh P, Little RD, Dupuis J, Del Mastro RG, 54. Iwano M, Kubo A, Nishino T, Sato H, Nishoika H, Akai Y, Falls K, Simon J, Torrey D, Pandit S, McKenny J, Braun- Kurioka H, Fugii Y, Kanauchi M, Shiiki H, Doki K: Quan- schweiger K, Walsh A, Liu Z, Hayward B, Folz C, Manning tification of glomerular TGF-beta 1 mRNA in patients with SP, Bawa A, Saracino L, Thackston M, Benchekroun Y, diabetes mellitus. Kidney Int 49: 1120–1126, 1996 Capparell N, Wang M, Adair R, Feng Y, Dubois J, FitzGer- 55. Hill C, Flyvbjerg A, Gronbaek H, Petrik J, Hill DJ, Thomas ald MG, Huang H, Gibson R, Allen KM, Pedan A, Danzig CR, Sheppard MC, Logan A: The renal expression of trans- MR, Umland SP, Egan RW, Cuss FM, Rorke S, Clough JB, forming growth factor-beta isoforms and their receptors in Holloway JW, Holgate ST, Keith TP: Association of the acute and chronic experimental diabetes in rats. Endocri- ADAM33 gene with asthma and bronchial hyperrespon- nology 141: 1196–1208, 2000 siveness. Nature 418: 426–430, 2002 56. Janssen JA, Lamberts SW: Circulating IGF-I and its protec- 43. Straub RE, Jiang Y, MacLean CJ, Ma Y, Webb BT, Myaki- tive role in the pathogenesis of diabetic angiopathy. Clin shev MV, Harris-Kerr C, Wormley B, Sadek H, Kadambi B, Endocrinol (Oxf) 52: 1–9, 2000 Cesare AJ, Gibberman A, Wang X, O’Neill FA, Walsh D, 57. Elliot SJ, Striker LJ, Hattori M, Yang CW, He CJ, Peten EP, Kendler KS: Genetic variation in the 6p22.3 gene DTNBP1, Striker GE: Mesangial cells from diabetic NOD mice con- the human ortholog of the mouse dysbindin gene, is asso- stitutively secrete increased amounts of insulin-like ciated with schizophrenia. Am J Hum Genet 71: 337–348, growth factor-I. Endocrinology 133: 1783–1788, 1993 2002 58. Lupia E, Ellit SJ, Lenz O, Zheug F, Hattori M, Striker GE, 44. Imperatore G, Hanson RL, Pettitt DJ, Kobes S, Bennett PH, Striker J: IGF-1 decreases collagen degradation in diabetic Knowler WC: Sib-pair linkage analysis for susceptibility NOD mesangial cells: Implications for diabetic nephropa- genes for microvascular complications among Pima Indi- thy. Diabetes 48: 1638–1644, 1999 ans with type 2 diabetes. Pima Diabetes Genes Group. 59. Flyvbjerg A, Kessler U, Kiess W: Increased kidney and Diabetes 47: 821–830, 1998 liver insulin-like growth factor II/mannose-6-phosphate 45. Moczulski DK, Rogus JJ, Antonellis A, Warram JH: Major receptor concentration in experimental diabetes in rats. susceptibility locus for nephropathy in type 1 diabetes on Growth Regul 4: 188–193, 1994 360 Journal of the American Society of Nephrology J Am Soc Nephrol 17: 353–360, 2006

60. Smith LE, Shen W, Perruzzi C, Soker S, Kinose F, Xu S, 63. Gruden G, Thomas S, Burt D, Zhou W, Chusney G, Gnudi Robinson G, Driver S, Bischoff J, Zhang B, Schaeffer JM, L, Viberti G: Interaction of angiotensin II and mechanical Senger DR: Regulation of vascular endothelial growth fac- stretch on vascular endothelial growth factor production tor-dependent retinal neovascularization by insulin-like by human mesangial cells. J Am Soc Nephrol 10: 730–737, growth factor-1 receptor. Nat Med 5: 1390–1395, 1999 1999 61. Simon M, Rockl W, Hornig C, Grone EF, Theis H, Weich HA, 64. Caramori MLA, Mauer M: Pathophysiology of renal com- Fuchs E, Vayon A, Grone HJ: Receptors of vascular endothe- plications. In: Ellenberg and Rifkin’s Diabetes Mellitus, 6th lial growth factor/vascular permeability factor (VEGF/VPF) Ed., edited by Porte D Jr, Sherwin RS, Baron A, New York, in fetal and adult human kidney: localization and [125I]VEGF McGraw-Hill, 2003, pp 697–722 binding sites. J Am Soc Nephrol 9: 1032–1044, 1998 65. Caramori MLA, Mauer M: Diabetes and nephropathy. Curr 62. Cooper ME, Vranes D, Youssef S, Stacker SA, Cox AJ, Opin Nephrol Hypertens 12: 273–282, 2003 Rizkalla B, Casley DJ, Bach LA, Kelly DJ, Gilbert RE: In- 66. Mauer M, Zinman B, Gardiner R, Drummond KN, Suissa creased renal expression of vascular endothelial growth S, Donnelly SM, Strand TD, Kramer MS, Klein R, Sinaiko factor (VEGF) and its receptor VEGFR-2 in experimental AR: ACE-1 and ARBs in early diabetic nephropathy. J diabetes. Diabetes 48: 2229–2239, 1999 Renin Angiotensin Aldosterone Syst 3: 262–269, 2002