unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

19
Journal of Pathology J Pathol 2010; 220: 198–216 Published online 30 October 2009 in Wiley InterScience (www.interscience.wiley.com) DOI: 10.1002/path.2639 Invited Review Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders Amy J McKnight, Diane Currie and Alexander P Maxwell* Nephrology Research Group, Queen’s University of Belfast, Northern Ireland, UK *Correspondence to: Alexander P Maxwell, Nephrology Research Group, Queen’s University of Belfast, c/o Regional Nephrology Unit, Level 11, Tower Block, Belfast City Hospital, Lisburn Road, Belfast BT9 7AB, Northern Ireland. E-mail: [email protected] No conflicts of interest were declared. Received: 21 September 2009 Revised: 25 September 2009 Accepted: 26 September 2009 Abstract Chronic kidney disease is common with up to 5% of the adult population reported to have an estimated glomerular filtration rate of <60 ml/min/1.73 m 2 . A large number of pathogenic mutations have been identified that are responsible for ‘single gene’ renal disorders, such as autosomal dominant polycystic kidney disease and X-linked Alport syndrome. These single gene disorders account for <15% of the burden of end-stage renal disease that requires dialysis or kidney transplantation. It has proved more difficult to identify the genetic susceptibility underlying common, complex, multifactorial kidney conditions, such as diabetic nephropathy and hypertensive nephrosclerosis. This review describes success to date and explores strategies currently employed in defining the genetic basis for a number of renal disorders. The complementary use of linkage studies, candidate gene and genome-wide association analyses are described and a collation of renal genetic resources highlighted. Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd. Keywords: angiotensin-converting enzyme; degeneration; polycystic kidney disease; copy number variation; kidney; glomerular filtration rate; genome-wide association; microRNA; mitochondrial DNA; polycystic kidney disease; systemic lupus erythematosis; single nucleotide polymorphisms; diabetes Introduction Chronic kidney disease (CKD), defined by an esti- mated glomerular filtration rate (GFR) <60 ml/min/ 1.73 m 2 , is common with a reported prevalence of almost 5% in most populations studied [1–3]. CKD is also associated with significantly increased rates of hospitalization and cardiovascular events and there is a strong inverse correlation between mor- tality and estimated GFR at levels of renal function <45 ml/min/1.73 m 2 [4,5]. These recent observations have increased awareness of the public health perspec- tive of renal disease and the escalating economic costs of managing end-stage renal disease (ESRD) with dial- ysis and/or renal transplantation [6]. The underlying pathological basis for ESRD has shifted dramatically over the last four decades with the emergence of diabetes and hypertension as the primary aetiologies for kidney failure, which together account for up to 75% of the ESRD recorded on some national renal registries [7,8]. This reflects improved survival of hypertensive and diabetic patients and more liberal access to renal replacement therapies [9]. The more ‘classical’ pathological kidney disorders, such as chronic glomerulonephritis, polycystic kidney disease and chronic pyelonephritis remain important aetiologies for ESRD [7,8]. Whilst there have been spectacular successes in identifying causative mutations for a wide variety of less common Mendelian renal disorders there has been much less progress defining the genetic contribution to common complex renal diseases, such as diabetic nephropathy and hypertensive nephrosclerosis [10,11]. Reviews of published genetic studies for acute kidney injury [12] and kidney transplantation [13], respec- tively, do not provide definitive evidence identifying genetic risk factors for either of these clinical con- ditions. The small sample sizes, complex phenotypes and various clinical outcomes make interpretation of currently available genetic data difficult. The genetics of renal tumours have also been recently evaluated in detail [14,15]. This review explores both the methodological approaches and current challenges faced by researchers trying to establish the genetic basis for chronic kidney disease. Early progress from genetic linkage studies Almost 25 years ago, autosomal dominant polycys- tic kidney disease (ADPKD) was first localized to the short arm of chromosome 16 by linkage analysis using highly polymorphic DNA markers [16]. A long period of sustained research using positional cloning techniques refined the locus and led to cloning of the PKD1 gene [17] encoding polycystin-1 [18]. The identification of PKD1 was complicated by its long Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk

Upload: amy-j-mcknight

Post on 06-Jul-2016

223 views

Category:

Documents


2 download

TRANSCRIPT

Page 1: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

Journal of PathologyJ Pathol 2010; 220: 198–216Published online 30 October 2009 in Wiley InterScience(www.interscience.wiley.com) DOI: 10.1002/path.2639

Invited Review

Unravelling the genetic basis of renal diseases; from singlegene to multifactorial disordersAmy J McKnight, Diane Currie and Alexander P Maxwell*Nephrology Research Group, Queen’s University of Belfast, Northern Ireland, UK

*Correspondence to:Alexander P Maxwell,Nephrology Research Group,Queen’s University of Belfast, c/oRegional Nephrology Unit, Level11, Tower Block, Belfast CityHospital, Lisburn Road, BelfastBT9 7AB, Northern Ireland.E-mail: [email protected]

No conflicts of interest weredeclared.

Received: 21 September 2009Revised: 25 September 2009Accepted: 26 September 2009

AbstractChronic kidney disease is common with up to 5% of the adult population reported to have anestimated glomerular filtration rate of <60 ml/min/1.73 m2. A large number of pathogenicmutations have been identified that are responsible for ‘single gene’ renal disorders, suchas autosomal dominant polycystic kidney disease and X-linked Alport syndrome. Thesesingle gene disorders account for <15% of the burden of end-stage renal disease thatrequires dialysis or kidney transplantation. It has proved more difficult to identify thegenetic susceptibility underlying common, complex, multifactorial kidney conditions, suchas diabetic nephropathy and hypertensive nephrosclerosis. This review describes success todate and explores strategies currently employed in defining the genetic basis for a number ofrenal disorders. The complementary use of linkage studies, candidate gene and genome-wideassociation analyses are described and a collation of renal genetic resources highlighted.Copyright 2009 Pathological Society of Great Britain and Ireland. Published by JohnWiley & Sons, Ltd.

Keywords: angiotensin-converting enzyme; degeneration; polycystic kidney disease; copynumber variation; kidney; glomerular filtration rate; genome-wide association; microRNA;mitochondrial DNA; polycystic kidney disease; systemic lupus erythematosis; singlenucleotide polymorphisms; diabetes

Introduction

Chronic kidney disease (CKD), defined by an esti-mated glomerular filtration rate (GFR) <60 ml/min/1.73 m2, is common with a reported prevalenceof almost 5% in most populations studied [1–3].CKD is also associated with significantly increasedrates of hospitalization and cardiovascular events andthere is a strong inverse correlation between mor-tality and estimated GFR at levels of renal function<45 ml/min/1.73 m2 [4,5]. These recent observationshave increased awareness of the public health perspec-tive of renal disease and the escalating economic costsof managing end-stage renal disease (ESRD) with dial-ysis and/or renal transplantation [6].

The underlying pathological basis for ESRD hasshifted dramatically over the last four decades withthe emergence of diabetes and hypertension as theprimary aetiologies for kidney failure, which togetheraccount for up to 75% of the ESRD recorded on somenational renal registries [7,8]. This reflects improvedsurvival of hypertensive and diabetic patients andmore liberal access to renal replacement therapies [9].The more ‘classical’ pathological kidney disorders,such as chronic glomerulonephritis, polycystic kidneydisease and chronic pyelonephritis remain importantaetiologies for ESRD [7,8].

Whilst there have been spectacular successes inidentifying causative mutations for a wide variety of

less common Mendelian renal disorders there has beenmuch less progress defining the genetic contributionto common complex renal diseases, such as diabeticnephropathy and hypertensive nephrosclerosis [10,11].Reviews of published genetic studies for acute kidneyinjury [12] and kidney transplantation [13], respec-tively, do not provide definitive evidence identifyinggenetic risk factors for either of these clinical con-ditions. The small sample sizes, complex phenotypesand various clinical outcomes make interpretation ofcurrently available genetic data difficult. The geneticsof renal tumours have also been recently evaluated indetail [14,15].

This review explores both the methodologicalapproaches and current challenges faced by researcherstrying to establish the genetic basis for chronic kidneydisease.

Early progress from genetic linkage studies

Almost 25 years ago, autosomal dominant polycys-tic kidney disease (ADPKD) was first localized tothe short arm of chromosome 16 by linkage analysisusing highly polymorphic DNA markers [16]. A longperiod of sustained research using positional cloningtechniques refined the locus and led to cloning ofthe PKD1 gene [17] encoding polycystin-1 [18]. Theidentification of PKD1 was complicated by its long

Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.www.pathsoc.org.uk

Page 2: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

Genetics of renal disease 199

DNA sequence length, the presence of six pseudo-genes in a duplicated region adjacent to the original16p13.3 PKD1 locus, and the presence of multiple(>300) unique PKD1 pathogenic mutations in differ-ent families with the clinical and pathological diag-nosis of ADPKD ( [19]; database accessed 3 Septem-ber 2009). This early success has been followed bydetailed assessment of the biochemical function ofpolycystins encoded by PKD1 and PKD2 (chromo-some 4q21-q23) [20–23] that has helped elucidate thepathophysiology of cyst growth. An inherited autoso-mal dominant germline mutation is not sufficient forcyst growth and a second somatic mutation in the cellis necessary for cell cycle regulation to be disrupted,associated with alteration in the function of primarycilia on renal tubular epithelial cells [24]. These mech-anistic studies, both in vitro and in animal modelsof polycystic kidney disease, have led to the devel-opment of potential therapeutic options for patientswith ADPKD for whom clinical trials of tolvaptan,sirolimus and somatostatin are now in progress [25,26](Figure 1).

The diversity of genetic mechanismscontributing to renal disease

Differences in DNA range from a single nucleotidebase change to large-scale chromosome rearrange-ments. Pathogenic gene mutations have been identifiedfor many kidney disorders that tend to be inherited ina Mendelian manner (Table 1); however, these typ-ically account for <15% of individuals with ESRDin national registries [7,8]. More than 300 inheriteddisorders have been described with kidney involve-ment and further studies of common renal diseases

Figure 1. The route from linkage studies of autosomaldominant polycystic kidney disease (ADPKD) to clinical trials ofnovel therapies for this renal disorder

have shown that patterns of inheritance are complex[11]. In addition to traditional Mendelian inheritancepatterns (autosomal dominant, autosomal recessiveand X-linked), renal diseases may also be influencedby epistasis [27], variable gene expression or pene-trance [28] and mosaicism [29]. Differential allelicexpression, due to epigenetic mechanisms modifyinggene function or the presence of polymorphisms inregulatory elements, affects the expression of morethan 20% of genes [30]. Allelic heterogeneity mayalso be evident; for example, different mutations ofthe collagen COL4A5 gene reflect phenotypic differ-ences in X-linked Alport syndrome [31] and indeedsome types of mutation are associated with a higherrisk of ESRD [32]. Deletions, duplications and sin-gle nucleotide polymorphisms (SNPs) in the comple-ment factor H (CFH ) gene have been associated withatypical haemolytic uraemic syndrome and glomeru-lonephritis [33]. Common genetic variants increasesusceptibility to chronic kidney disease and progres-sion to ESRD [34,35]. A coding variant in the nitricoxide synthase (NOS3 ) gene [36] and a haplotype inthe vascular endothelial growth factor (VEGF ) gene[37] are reported to act as genetic modifiers of therate of progression to ESRD in ADPKD patients.A combination of genetic and environmental modi-fiers partly explain why there is clinical heterogeneityin a ‘single-gene’ disorder such as ADPKD, wherethere can be wide variation in the age of onset ofESRD in individuals with the same pathogenic PKD1gene mutation [38]. Renal disease phenotypes are theresult of dynamic interactions between genetic factorsand environmental influences modifying the ‘risk’ ofdeveloping kidney disease (Figure 2).

To date there has been more limited success inidentifying definitive genetic risks for renal disorderswith a complex, multifactorial aetiology. The rateof progression of a ‘genetic’ chronic kidney diseasemay be influenced by environmental factors, suchas whether the individual is a smoker or receivestreatment for hypertension. These external factorswill have an impact on whether a particular renalphenotype, such as ESRD, is reached. Furthermore,even an individual’s response to medication, such asangiotensin-converting enzyme (ACE) inhibitors, maybe partly genetically determined [39,40].

Genome-wide linkage studies for kidneydiseases and traits

Historically, familial aggregation of a particular phe-notype was observed and linkage mapping, usinghighly polymorphic microsatellite markers, conductedto investigate correlations between phenotype andpatterns of allele segregation within families. Con-siderable efforts have been expended conductinggenome-wide linkage scans for common renal disease;however, few studies have achieved the levels ofstatistical evidence proposed for significance [41]

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 3: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

200 AJ McKnight et al

Table 1. Genes identified for glomerular, tubular and ciliary disorders

Disease Locus Gene symbol Gene name Reference

Alport syndrome 2q35-37 COL4A3 COL4A4 Collagen, type IV, α3 (Goodpasture antigen);Collagen, type IV, α4

[193,194]

Xq22 COL4A5 Collagen, type IV, α5 (Alport syndrome) [195]Thin basement membranenephropathy

2q35-37 COL4A3 COL4A4 Collagen, type IV, α3 (Goodpasture antigen);Collagen, type IV, α4

[196,197]

1q25-q31 NPHS2 Nephrosis 2, idiopathic, steroid-resistant (podocin) [198]Xq22 COL4A5 Collagen, type IV, α5 ( [199], sourced

from abstract)Benign familial haematuria 2q35-37 COL4A3 COL4A4 Collagen, type IV, α3 (Goodpasture antigen);

Collagen, type IV, α4[200]

Congenital nephroticsyndrome of Finnish type

19q12-q13.1 NPHS1 Nephrosis 1, congenital, Finnish type (nephrin) [201]

Pierson’s syndrome 3p21.3-p21.2 LAMB2 Laminin, β2 (laminin S) [202]Bartter’s syndrome 15q15-q21 SLC12A1 Solute carrier family 12 (sodium/potassium/chloride

transporters), member 1[203]

11q24 KCNJ1 Potassium inwardly-rectifying channel, subfamily J,member 1

[204]

1p32.3 1p36 BSND, CLCNKA,CLCNKB

Bartter syndrome, infantile, with sensorineuraldeafness (Barttin); chloride channels Ka and Kb

[205–207]

Distal renal tubular acidosis 17q12-21 SLC4A1 Solute carrier family 4, anion exchanger, member 1(erythrocyte membrane protein band 3, Diegoblood group)

[208]

8q23 SLC26A7 Solute carrier family 26, member 7 [209]7q34 ATP6V0A4 ATPase, H+ transporting, lysosomal V0 subunit a4 [210]2p13 ATP6V1B1 ATPase, H+ transporting, lysosomal 56/58 kDa, V1

subunit B1[211]

Gitelman’s syndrome 16q13 SLC12A3 Solute carrier family 12 (sodium/chloridetransporters), member 3

[212]

Liddle’s syndrome 16p12 16p12 SCNN1B SCNN1G Sodium channel, non-voltage-gated 1, β (Liddle’ssyndrome); sodium channel, non-voltage-gated 1, γ

[213,214]

Nephrogenic diabetesinsipidus

Xq28 AVPR2 Arginine vasopressin receptor 2 (nephrogenicdiabetes insipidus)

[215]

12q12-13 AQP2 Aquaporin 2 (collecting duct) [216]Autosomal dominantpolycystic kidney disease

16p13.3 PKD1 Polycystic kidney disease 1 (autosomal dominant) [17]

4q21-23 PKD2 Polycystic kidney disease 2 (autosomal dominant) [217]2p PKD3 Polycystic kidney disease 3 (autosomal dominant) [218,219]

Autosomal recessivepolycystic kidney disease

6p21.1-p12 PKDH1 Polyductin [220]

Meckel–Gruber syndrome 17q21-q24 MKS1 Meckel syndrome, type 1 [221]11q13 MKS2 Meckel syndrome, type 2 [222]8q22.1 TMEM67 Transmembrane protein 67 [223]

Medullary cystic kidneydisease

1q21 MCKD1 Medullary cystic kidney disease 1 (autosomaldominant)

[224]

16p12.3 UMOD Uromodulin (uromucoid, Tamm–Horsfallglycoprotein)

[97]

Nephronophthisis 2q13 NPHP1 Nephronophthisis 1 (juvenile) [225]9q31 INVS Inversin [226]3q22 NPHP3 Nephronophthisis 3 (adolescent) [183]1p36 NPHP4 Nephronophthisis 4 [227]

3q21.1 IQCB1 IQ motif containing B1 [182]12q21.33 CEP290 Centrosomal protein 290 kDa [228]16p13.3 GLIS2 GLIS family zinc finger 2 [229]

Xp11.23-23 CLCN5 Chloride channel 5 (nephrolithiasis 2, X-linked,Dent’s disease)

[230]

17q11.1 NEK8 NIMA (never in mitosis gene a)-related kinase 8 [231]16q12.2 RPGRIP1L RPGRIP1-like [232]8q22.1 TMEM67 Transmembrane protein 67 [233]

6q23.2 AHI1 Abelson helper integration site 1 [234]

and identifying disease-causing mutations by posi-tional cloning methods has proved difficult [42].Genetic linkage to renal diseases and quantitativetraits such as GFR [43] has been reported for all

chromosomes [11] (Table 2; see also Supporting infor-mation, Table S1). Many studies have demonstratedlinkage to chromosome 3q with further associationstudies refining loci and identifying several biological

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 4: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

Genetics of renal disease 201

Figure 2. The interactions between genes and environment governing the development of renal disease phenotypes

and positional candidate genes for renal phenotypes(Figure 3).

Relatively few single nucleotide polymorphism(SNP)-based genome-wide linkage scans have beenconducted for renal disorders. The first identified anovel, disease-causing mutation in PAX2 for autoso-mal dominant renal dysplasia [44]. Examining 100discordant sib pairs using >5000 SNPs highlightedlinkage to chromosome 19 (maximum likelihood score= 3.1) for nephropathy in type 1 diabetes [45]. Mostrecently, SNP-based linkage for renal function (mea-sured by serum creatinine) was established with chro-mosomes 7p14, 9p21, 11p15, 15q15-21, 16p13 and18p11 and a novel locus on chromosome 10p11 [46].

Several methods for the meta-analysis of linkagedata have been proposed. The multiple scan probabil-ity approach combines reported p values and appearsrobust to heterogeneity [47]. A modified genome scanmeta-analysis method has been proposed to adjustfor heterogeneity between studies (HEGESMA) [48].A novel, kernel-based estimation procedure has beenapplied to rheumatoid arthritis datasets [49] and col-laborative analyses on raw data would be ideal for thepurposes of meta-analysis [41,50,51]. Linkage analysishas proved successful for many ‘single-gene’ disor-ders, however association studies are believed to bemore powerful in detecting susceptibility genes forcomplex diseases such as CKD [52,53].

Candidate gene associations for renal diseases

Robust replication of initial candidate gene associa-tions has proven difficult, largely due to differences inbiological exposures or study designs [54]. The powerto identify statistically significant genetic associationswith disease is crucially important in designing suchstudies. Genetic power calculations typically consider

model parameters such as disease prevalence, diseaseallele frequency, disease genotype relative risks, miss-ing or erroneous data, marker allele frequencies, corre-lations between markers and disease, significance lev-els and case : control ratios before estimating requiredsample sizes [55,56]. Despite the apparent lack of con-sistent replication in the majority of candidate genestudies, collaborative efforts have led to the robustreplication of several loci. For example, convincingevidence was reported for linkage to 18q22.3–23 (log-arithm of odds = 6.1) in Turkish individuals withdiabetic nephropathy [57] and subsequently supportedby data from African-American individuals [58] andin the Family Investigation of Nephropathy and Dia-betes (FIND) collection [59]. Carnosine (CNDP1 ) isa candidate gene located in this region that has beenassociated with diabetic nephropathy [60–62]. Withinthe last year, four papers have described an associ-ation between variants in the MYH9 gene and CKDin African-American individuals [63–66]. A linkagestudy for serum creatinine has also recently reportedassociation with MYH9 in European populations [46].

Frequently studied genes for common, multifactorialrenal disease include ACE, AGT, AGTIR, AKR1B1,APOE, FCGR, GNB3, NOS3 and MTHFR. Combiningpublished results by meta-analysis is now practicalfor several genes (Table 3). The majority of studiesfor renal disease, which is inherited in a complexmanner, are based on the common variant–commondisease hypothesis [67]. This hypothesis suggests thatpredisposition to chronic kidney disease is based onnumerous genetic variants that are relatively commonin the population. It is possible that rare alleles with aminor allele frequency <5%, or multiple variants withsmall individual effects, contribute to chronic kidneydisease, but many thousands of individuals would berequired to identify these markers.

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 5: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

202 AJ McKnight et al

Table 2. Genes for common forms of renal disease identified from genome-wide studies

Phenotype Study Chromosomal region Gene(s) identified References

CKD Association. 70 987 SNPs 15q24.3, p = 0.001, rs6495446 MTHFS [235]CKD Association. Up to 900 000 SNPs 16p12.3, p = 2.8 × 10−09, rs12917707 UMOD [85]T1DN Linkage. 5382 SNPs in DSPs 20p12.1, max LOD = 2.8, rs466243 MACROD2 [45]

19q1, max LOD = 3.1, rs306450 RGS1010q25, max LOD = 2.4, rs14678135q, max LOD = 2.7, rs27342

T1DN Association. Affymetrix 10 K inCC and trios

2p22, p = 2.12 × 10−5, rs1368086;p = 8.96 × 10−4, rs725238

PLEKHH2 [236]

2p21, p = 1.86 × 10−4, rs11886047DN Linkage. 404 markers, sibs 18q22-23, max LOD = 2.1,

D18S1371–D18S1390CNDP1, CNDP2 [57,60–62,237]

DN Association. 81 315 SNPs in CC 7p14.1, p = 0.000008, Intron18A9170G

ELMO1 [114,117,118,238]

DN Association. 360 000 SNPs in CC 9q21.33, p = 5.0 × 10−7, rs1888747 FRMD3 [82]11p15.5, p = 3.1 × 10−6, rs451041 CARS

DN Association. 6000 markers in CC 10p15, pcorrected = 0.005, D10S558 andD10S1435

GTPBP4 [77,239]

DN Association. ∼80 000 SNPs inCC

1615 loci where p < 0.01. ACE [113,115,238,240]

p = 0.00005, (ACE I/D)/M (AGTM235T)/A (AGTR1

AGT

A1166C) AGTR116q13, rs2289116 ELMO17p14.1, rs741301 SLC12A3

ESRD Association. 1536 SNPs in CC 22q13.1, max LOD = 4.55 (non DNmax LOD = 8.56)

MYH9 [63,63,66,241]

ESRD Association. 115 352 SNPs in CC 8q24, rs2648875, p = 2 × 10−6 PVT1 [83]FSGS Association. 1272 SNPs in CC 22q13.1, max LOD = 12.4,

p = 1 × 10−7, rs5756152MYH9 [64]

IgA nephropathy Association. 80 000 SNPs in CC 1q31-41, p = 0.0003, A580V PIGR [81]Lupus nephritis Association. HumanHap550 2q32.2-q32.3, p < 10−11, rs7574865 STAT4 [79]Renal dysplasia Linkage. 10 000 markers in MF 10q23.31-q25.1, 55 SNPs, G874A PAX2 [44]GFR Association. 100 000 SNPs 21q22.3, p = 1.6 × 10−05, rs2839235 PCNT [242]eGFR—creatinine Linkage. 387 markers in sibs 3q27, LOD = 3.61 [243,244]

6q15-q21, LOD = 2.97eGFR—creatinine Association. Up to 900 000 SNPs 16p12.3, p = 3.0 × 10−11, rs12917707 UMOD [85]

4q21.1, p = 9.7 × 10−08, rs17319721 SHROOM315q15.1, p = 3.4 × 10−07, rs2467853 GATM–SPATA5L1

eGFR—creatinine Linkage. 747–6008 markers 22q13.1, p = 0.0089, rs11089788 MYH9 [46]eGFR—cystatin C Association. Up to 900 000 SNPs 16p12.3, rs12917707, p = 2.0E–07 UMOD [85]

8p22-p12, rs1731274, p = 4.6E–08 STC120p11.2, rs13038305, p = 2.2E–88 CST3–CST9

eGFR—cystatin C Association. 70 897 SNPs 20p11.21, p = 8.5 × 10−9, rs1158167 CST9L, CST9, CST3 [242]UAE Association. 70 897 SNPs 11q23.2, p = 1.9 × 10−6, rs1712790 FAM55B [242]Serum uric acidconcentration andgout

Association. 503 551 SNPs 4p16.1, p = 7.0 × 10−168, rs16890979 SLC2A9 [102,245]

4q22.1, p = 2.5 × 10−60, rs2231142 ABCG2 [246,247]6p22.2, p = 3.3 × 10−26, rs1165205 SLC17A3 [248,249]

ACR, urinary albumin excretion; MF, multigenerational family; ASPs, affected sib pairs; DSPs, discordant sib pairs; CC, cases vs. controls; UAE,urinary albumin excretion.

MicroRNAs (miRNAs) are small, non-protein cod-ing RNAs that function by post-transcriptional repres-sion of target coding RNAs. The role of miRNAsin the kidney is not yet clearly defined; however,miRNA function appears critical for maintenance ofthe glomerular filtration barrier [68]. Recent studieshave revealed genetic variants affecting miRNAs asso-ciated with renal disease. Horikawa and colleagues[69] observed SNPs involved with miRNA machinery(Asn929Asp, Cys1033Arg in the GEMIN4 gene) asso-ciated with the risk of renal cell carcinoma. The PVT1locus encodes several miRNAs [70] and variants in

this gene have been associated with diabetic nephropa-thy [71]. A1166C in the Ang II type 1 receptor gene(AGTR1 ) has been associated with renal disease; thisvariant attenuates binding of a specific miRNA (miR-155) [72].

Genome-wide association studies for renalphenotypes

The nature of linkage analysis data means thatgenomic regions of interest are typically large and may

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 6: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

Genetics of renal disease 203

Figure 3. Chromosome 3, demonstrating the distribution of microsatellite and SNP markers related to kidney disease and thehighlighted genes that are associated with renal phenotypes

contain many genes. Genome-wide association (GWA)may be used as a complementary approach to linkagestudies or where robust linkage analysis is not feasible.GWA studies have been especially successful recentlyin identifying genes implicated in autoimmune dis-orders [73], including type 1 diabetes [74], rheuma-toid arthritis [75] and inflammatory bowel disease[76]. Since complex polygenic disorders have provedtractable to GWA analyses, it is hoped that employingsimilar strategies can determine genetic contributionsto autoimmune renal disease, such as glomerulonephri-tis. Technical innovations have reduced the cost ofSNP-based genotyping and, together with improvedbioinformatic and genetic statistical tools, have madelarge-scale GWA screens possible. Genome-wide case-control association studies for renal disease have beenconducted using microsatellites [77], gene-based SNPs[78], SNPs indirectly distributed across the genome[79] or putatively functional SNPs [80]. Genome-wide screens are advantageous in that they requireno a priori hypothesis, neither do they limit analy-sis to previously examined genes. Several renal dis-eases and traits have been, or are currently, the sub-ject of GWA studies, including IgA nephropathy [81],diabetic nephropathy [78,82], ESRD [83], gout [84],glomerular filtration rate [85] and blood pressure [86].

Genome-wide association studies are designed toestablish the relationships between genotype and phe-notype. These steps include:

1. Accurate phenotyping (clinical ascertainment) toincrease the likelihood of observing true differencesat the level of individual genomes.

2. Extraction and normalization of DNA from bloodor tissues.

3. Genotyping of DNA samples with appropriatequality controls.

4. Statistical tests of association with stringent pvalues to correct for multiple testing.

5. Replication of the results in another similar cohort.

Arguably the main objective of GWA studies is toidentify biological pathways implicated in the diseaseor trait of interest, rather than the identification ofa causative mutation per se that is then employedin population screening or risk prediction [87]. Suchnew insights will not translate quickly into changes inclinical practice until the gap between genotype andphenotype is bridged [88]. The necessary translationof genetic mapping data to clinical applications willrequire connections to be made with gene expressionprofiles in cells and tissues under resting and stimu-lated states [89,90], eg renal mesangial cells exposedto normal and high extracellular glucose to mimicdiabetic kidney injury [91,92]. Such clinical connec-tions are increasingly being made between genotypesand human physiology, as recently exemplified by thefinding that an interleukin-2 receptor α (IL2RA) locusis directly related to cell surface expression of theIL2RA protein (or CD25) on T cells [93]. In this study,higher CD25 expression was associated with IL2RAhaplotypes that confer protection from type 1 diabetes.There is now a real impetus to undertake the detailedclinical studies of biological variation in geneticallycharacterized populations to fully understand the linksbetween gene(s) and phenotype(s).

Chronic kidney disease and genome-wideassociation analyses

Chronic kidney disease (CKD) is one of the broadclinical phenotypes that can be easily ascertained in anumber of existing GWA datasets that included mea-surements of kidney function (serum creatinine ± cys-tatin C) when individuals were recruited. CKD is asso-ciated with a number of loci in a recent meta-analysisof GWA studies performed on >40 000 individuals ofEuropean ancestry [85]. The meta-analysis identifiedSNPs in the UMOD gene locus (UMOD ; chromosome16p12.3) to be associated with CKD in 2388 cases, andthese findings were then replicated in a further 1932

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 7: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

204 AJ McKnight et al

Table 3. Meta-analysis of candidate genes for kidney disease

Gene Investigation DNAs Outcome Ref

Lupus nephritisTNFAIP3 rs5029939 from two studies n = 6973 (cases, 1453;

controls, 3381; trios, 713)Associated with SLE, particularly inpatients with renal manifestations.(p = 1.67 × 10−14; OR = 2.09; 95% CI,1.68–2.60)

[250]

FCGR2A R/H131 from 29 studies n = 7960 (cases, 1685;controls, 1892; 4383non-SLE controls)

RR is a risk factor (OR = 1.24; 95% CI,1.04–1.48

[251]

FCGR2A R/H131 from 17 studies n = 5694 (cases, 1405;controls, 1709; 2580non-SLE controls)

No association [252]

FCGR3A V/F158 from 11 studies n = 3870 (cases, 1154;controls, 1261; 1455non-SLE controls)

No association [253]

Renal transplantation

TGFβ 1, TNFA, IL-10 TGFβ1, cd10, cd25; TNFA,−308; IL-10, −1082 G/A in amaximum of five studies

n = 1087 Possible association between TGFβ1cd10 and IL-10 with poor outcomes inrenal transplantation

[254]

ADPKD

ACE Indel in 13 studies n = 1940 (cases, 398;controls, 1022)

No association [255]

IgA nephropathy

SCGB1A1 G38A in six studies n = (cases, 930; controls,768)

No association [256]

ACE Indel in five studies n = 901 (cases, 346;controls, 555)

DD associated with progression(p < 0.05)

[257]

ACE Indel in 11 studies ? DD associated [258]ACE Indel n = 1838 (cases, 981;

controls, 857)No association [259]

Diabetic nephropathy

AKR1B1 (CA)n in eight T1Ds and nineT2Ds

n = 5302 (cases, 2751;controls, 2551)

Z−2 allele risk (OR 1.40, 1.07, 1.84).Z+2 protective with OR 0.77. Noassociation with T2D

[260]

AKR1B1 (CA)n and 106C > T in 11studies

n = 2391 (cases, 1165;controls, 1226)

No association with (CA)n Associationwith −106T

[261]

MTHFR C677T in 15 studies n = 4027 (cases, 1877;controls, 2150)

Pooled random effects: OR = 1.30; 95%CI, 1.03–1.64; no association whendHWE studies removed. More rigorousstudies are required

[262]

SLC2A1 Xbal in six studies n = 1460 (cases, 710;controls, 750)

Large studies failed to show associationwhile small studies claimed an association

[263]

ACE Indel in 53 studies n = 17 791 II protective (pooled OR = 0.78; 95%CI, 0.70–0.87; p < 0.001)

[264]

ACE Indel in 47 studies n = 14 727 (cases, 8663;controls, 6064)

II associated with lower risk of DN [265]

ACE Indel in 18 studies ? Homozygous deletion associated withpoorer renal outcome

[266]

ACE Indel in 21 studies n = 5016 (cases, 2579;controls, 2437)

Trend towards protective effect ofinsertion on renal function

[267]

ACE Indel in 18 studies n = 4773 (cases, 2495;controls, 2278)

Deletion allele associated with DN in adominant model

[268]

ACE Indel in 11 studies ? Deletion allele is not associated withhypertension, but is a marker for DN

[269]

ACE Indel in 17 studies n = 2960 (cases, 1365;controls, 1595)

D allele is associated with progression,not initiation of DN

[270]

dHWE, deviation from Hardy–Weinberg equilibrium; T1D, type 1 diabetes mellitus; T2D, type 1 diabetes mellitus; DN, diabetic nephropathy;SLE, systemic lupus erythematosis.

CKD cases. This is an intriguing finding, since UMODencodes uromodulin (Tamm–Horsfall protein) whichis the most abundant protein in normal urine [94].

Uromodulin has antimicrobial properties providingdefence against uropathogens responsible for urinarytract infection [95] and may also play a role in

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 8: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

Genetics of renal disease 205

preventing crystallization of calcium and uric acidin kidneys and urine [96]. Although defects in theuromodulin gene have been associated with rareMendelian kidney disorders, such as autosomal dom-inant medullary cystic disease-2 and familial hyper-uricemic nephropathy [97,98], it is possible that subtlerdefects in uromodulin function contribute to progres-sive CKD. It is known that immunization of animalmodels with Tamm–Horsfall protein is associated witha chronic tubulointerstitial nephritis [99,100] and uro-modulin gene-deficient mice have a >60% reductionin GFR (as measured by creatinine clearance) com-pared to wild-type mice [101].

Renal function and GWA studies

The GWA meta-analysis by Kottgen and co-workersalso highlighted an association between the UMODlocus and the continuous renal trait of GFR (mea-sured by estimated GFR incorporating serum creati-nine) [85]. In addition, estimated GFR was associ-ated with intronic SNPs in genes encoding Jagged 1(JAG1 ; chromosome 20p12.1-p11.23), shroom fam-ily member 3 (SHROOM3 ; chromosome 4q21.1and spermatogenesis-associated 5-like 1 (SPATA5L1/GATM ; chromosome 15q21.1) (Table 2; see also Sup-porting information, Table S1). Using an alternativemethod of estimating GFR, incorporating cystatin Cassays, the UMOD locus was again found to be asso-ciated with renal function [85].

Gout and GWA analyses

GWA analyses have proved to be spectacularly suc-cessful in determining the links between plasma uratelevels and kidney function, with multiple novel locireported [102,103] (Table 2; see also Supporting infor-mation, Table S1). These novel insights into renalphysiology have been extended recently with the iden-tification of a urate transporter, human ATP-bindingcassette, subfamily G, 2 (ABCG2 ), which harbours acommon functional polymorphism causing gout [84].The native ABCG2 protein is located in the brush bor-der membrane of kidney proximal tubule cells, whereit mediates renal urate secretion. The common SNPrs2231142 (NP 004 818.2:p.Gln141Lys) reduces uratetransport rates by 53% compared to the wild-typeABCG2. This finding is an excellent example of thecommon variant–common disease hypothesis, wherea genetic association with ABCG2 provides physio-logical insights to explain the mechanism for hyper-uricaemia and gout.

IgA nephropathy and GWA studies

A major challenge now for investigators is to estab-lish large enough collections of DNA from care-fully phenotyped individuals, with and without renaldisease, for GWA studies to be sufficiently pow-erful to detect significant associations. This usu-ally requires national or international collaboration

to agree both on the definition of phenotype andto recruit patients for study. IgA nephropathy [104]is the most common glomerulonephritis identifiedin renal biopsy registries [105–107]. Several largeconsortia have been assembled to try and identifythe genetic susceptibility to IgA nephropathy includ-ing the European IgA nephropathy Biobank [108],MRC Kidney Research UK National DNA Bankfor Glomerulonephritis (www.renal.org/Research/GN-DNAbank.html) and an alliance of researchers fromCanada, Finland and France [109]. A limited genome-wide analysis (in terms of SNP coverage) was under-taken in 2003 in Japanese patients with and withoutIgA nephropathy [81]. This study, employing approx-imately 80 000 SNPs, identified a significant associa-tion between IgA nephropathy and six SNPs locatedin the polymeric immunoglobulin receptor gene atchromosome 1q31-q41. The larger groups represent-ing the IgA nephropathy consortia are expected toreport more extensive GWA data within the nextyear.

Diabetic nephropathy and GWA approaches

Diabetic nephropathy is the commonest cause ofESRD and there is evidence for a genetic sus-ceptibility to diabetic kidney disease [110]. Sus-tained efforts have been made to assemble suf-ficiently large case-control collections to achieveadequate statistical power in GWA analyses anda number of multicentre collaborations have beenforged into consortia, such as Genetics of Kid-neys in Diabetes (GoKinD) groups in the USAand UK (www.niddkrepository.org/niddk/jsp/public/GOKIND/publications.jsp), the Warren 3 DNA Col-lection [111], the EURAGEDIC study [112], FinnDi-ane, (www.finndiane.fi) and the Family Investigation of Nephropa-thy and Diabetes (darwin.cwru.edu/FIND). The ben-efits of such collaboration are agreement on rigor-ous phenotypic criteria for recruitment of cases andcontrols, pooling of DNA resources, shared costsof large-scale GWA studies and combined expertisein analysis and replication. In 2009 a SNP-basedGWA study for nephropathy in type 1 diabetes wasreported by Pezzolesi and colleagues, highlightingfour loci (FERMD3, CARS, CHN2 and CPVL) withmoderately strong association with kidney disease[82].

Diabetic nephropathy is arguably a more diffi-cult renal phenotype to ascertain in type 2 diabetes,since the patients recruited for genetic studies tendto be older than individuals with nephropathy andtype 1 diabetes. The renal phenotype in older type2 diabetes individuals may be confounded by coex-isting hypertensive kidney injury and/or atheroma-tous renal vascular disease. Confirmation of diabeticnephropathy by renal biopsy is usually not under-taken. GWA studies of nephropathy in type 2 diabeteshave been reported in Japanese populations. These

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 9: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

206 AJ McKnight et al

studies reported the association of several candidategenes with nephropathy, including the solute carrierfamily 12 (sodium/chloride transporters) member 3(SLC12A3 ) [113] and the engulfment and cell motility1 gene (ELMO1 ) [114]. Variants in SLC12A3 weresubsequently found to be associated in a relativelysmall collection (n = 177 cases) [115]; however, thisfinding was not replicated in a Caucasian popula-tion with type 2 diabetes [116]. Different SNPs inELMO1 have been associated with diabetic nephropa-thy in American individuals with diabetic nephropathy[117,118].

Improvements in genotype technologies and dataanalyses have enabled the extension of GWA studiesto identify copy number variants that are associatedwith susceptibility to common disease [119].

Copy number variation

Copy number variation (CNV) represents an intrigu-ing field for renal geneticists. Resequencing of thehuman genome has shown that fragments of DNAare deleted, duplicated or inverted and that thesegenetic rearrangements may alter the number of copiesfor a particular gene [120]. Copy number variationrepresents a substantial source of variation withinthe human genome and there is significant over-representation in genes involved with differentiationand development [121]. Structural rearrangements ofthe genome increase the risk of developing complexdiseases [122–124] and almost 15% of genes collatedin the OMIM morbid map overlapped with a first-generation map of copy number variation [120]. Lowcopy number of the FCGR3B gene is associated withglomerulonephritis in systemic lupus erythematosis inboth rat models and human disease [124]. Willcocksand colleagues demonstrated the functional effect ofCNV at the FCGR3B locus on protein expressionand function [125]. Copy number variation may alsoplay a role in the development of nephronophthisis[120].

SNPs in complement factor H (CFH ) genes areassociated with atypical haemolytic uraemic syn-drome, membranoproliferative glomerulonephritis,chronic kidney disease and age-related macular degen-eration (AMD) [126–128]. The relationship betweenrenal function and AMD [127,129] is not clearlydefined; however, complement genes are subject toCNV and complete duplication of complement factorH-related genes (CFHR1–CFHR4 ) has been observed[130]. A recent study suggests that CNV contributesto the development of AMD and may provide furtherinsights to renal disease mechanisms [131].

The mitochondrial genome

There is increasing awareness of the critical role ofmitochondrial defects in a diverse range of clinical

disorders. Mitochondrial DNA (mtDNA) content incells is lower in those individuals with renal fail-ure compared to healthy controls and, of interest,higher mtDNA copy number in patients with ESRD onhaemodialysis has been associated with improved sur-vival [132]. Mitochondrial mutations have been impli-cated in a range of complex diseases including dia-betes mellitus [133], age-related macular degeneration[134] and neurodegenerative disorders [135]. Alteredrenal phenotypes have been observed in several com-plex mitochondrial disorders, including Kearns–Sayre,Pearson and Leigh syndromes [136]. Increased preva-lence of mitochondrial deletions [137,138] and singlebase variants [139] have been reported in individu-als with ESRD compared to healthy controls. A 5778bp mitochondrial deletion [140] and the 3243A >G single base mutation [141–143] in the mitochon-drial genome have been suggested to contribute to thepathogenesis of diabetic nephropathy. Renal functionwas impaired in more than half of individuals withmaternally inherited diabetes and deafness associatedwith 3243A > G [144]. Renal tubular dysfunction isassociated with multiple overlapping deletions, dupli-cations and rearrangements [145–147]. Tubulointer-stitial nephropathy is associated with several geneticabnormalities, including two large-scale deletions andthe 608A > G SNP at the distal end of the tRNA(Phe)molecule [148]. Both the 7374 bp ( [149] deletionand 3243A > G variant have been investigated forglomerulosclerosis [150]. The 3243A > G mutationhas been observed in patients with focal segmen-tal glomerulosclerosis [151], with further individualswith this renal disease reported to have the 5843A> G mutation [152]. Several reports of progressiverenal disease are described with deletions of mito-chondrial DNA, along with 3243A > G [153] and11 778G > A [154]; three cases with 3243A > Ghave presented with a phenotype similar to Alportsyndrome [155–157]. Multiple SNPs have been inves-tigated for essential hypertension with the 10 086A >G variant found to be significantly associated withhypertension-associated ESRD in African-Americans[158].

Due to complex inheritance patterns, heteroplas-mic differences, copy number variance, existence ofnuclear–mitochondrial pseudogenes and high muta-tional rate of mitochondrial DNA, the importanceof accurate sequencing and efficient quality con-trol mechanisms cannot be underestimated for mito-chondrial DNA projects [159,160]. Determining therole of mitochondrial DNA variants and imple-menting effective therapeutic strategies for com-mon disease remain a major challenge for renalgeneticists.

Towards a systems biology approach

Technological advances now facilitate the generationof large-scale datasets. The rapid accumulation of

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 10: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

Genetics of renal disease 207

genomic, transcriptomic and proteomic informationprovides renal researchers with the opportunity tointegrate these datasets to highlight biological path-ways for investigation. Systems biology employs amultidisciplinary approach, combining clinical insightwith basic research to investigate how molecules,cells and tissues interact in both healthy and diseasedstates. Several online resources have been created tostore biological information [161], including those thatdistribute results from GWA projects. For example,the Database of Genotypes and Phenotypes (dbGaP)presently contains information on 109 studies ( [162];accessed 3 September 2009) linking clinical charac-teristics with genetic profiles. A unique catalogue hasbeen developed to store the results from genome-wide association studies; analysis of > 150 studiesand 500 SNP-trait associations revealed that hundredsof common genetic variations have been reproduciblyidentified for more than 80 disorders [163]. An inte-grated genomics approach has recently suggested that> 35% of cis-acting SNPs associated with expressiontraits are located more than 100 kb distant to transcrip-tion start/stop sites, so that genes physically closestto SNPs highlighted in GWA studies may not be theappropriate gene(s) of interest [164]. GWA studieshave also revealed several genes in linkage disequi-librium; in silico network modelling relating genesto relevant pathways may further resolve causal loci[165].

Exploring integrated datasets has the potential toreveal novel pathophysiological mechanisms con-tributing to disease states. The integration of genomicand transcriptomic data from human and mousedatasets identified SORT1 and CELSR2 genes and

candidates at chromosome 1p13.3, influencing suscep-tibility to coronary artery disease and hyperlipidaemia[166]. An international effort led to the development ofa unique, coordinated systems biology-based approachfor type 1 diabetes (T1DBase [167]). T1DBase is aweb-based resource that presents a simplified, inte-grated view of complex datasets including annotatedgene data for linkage and association results, geneexpression data, visualization of combined molecularinteraction networks and results from animal models.Systems biology is being employed to provide novelinsights to understanding diverse disorders from pan-creatic β cell death in type 1 diabetes [164,168], tocardiovascular disease [169] and adverse drug reac-tions [170]. Such comprehensive projects are not yetdeveloped for kidney diseases; however, several onlineresources have been specifically developed to read-ily facilitate public access to renal-related informa-tion (Table 4). Stand-alone resources also exist suchas the catalogue of predicted protein–protein inter-actions that was developed from meta-analysis ofglomerular transcriptional profiles to improve under-standing of glomerular signalling networks (GlomNet,[171]).

Online resources are often individualized with theirown content, layout and dynamic capabilities [172],so that automated integration of diverse datasets andeffortless retrieval of relevant information will beessential to maximize our understanding of complexdisease processes [173]. Systems biology provides amechanism of combining knowledge from biologicalpathways and large-scale ‘omics’ data to inform per-sonalized clinical medicine. Computational systemsmedicine has been proposed to enable changes from

Table 4. Online resources for renal genetics

Autosomal dominant polycystic kidney disease http://pkdb.mayo.edu/cgi-bin/mutations.cgi[mutation database (PKD1/PKD2)]Autosomal recessive polycystic kidney disease http://www.humgen.rwth-aachen.de(mutation database (ARPKD/PKHD1)]CDDB http://cddb.nhlbi.nih.gov/cddb(Collecting Duct DataBase)CORGI http://www.qub.ac.uk/neph-res/CORGI(Centralized Online Renal Genetics Initiative)EuReGene http://www.euregene.org(European Renal Genome Project)GENECURE http://www.genecure.eu(GENomic stratEgies for treatment and prevention of Cardiovasculardeath in Uraemia and end-stage REnal disease)GUDMAP http://www.gudmap.org(GenitoUrinary Development Molecular Anatomy Project)KGDB http://www.urogene.org/kgdb(human Kidney Gene DataBase)Kidney development database http://golgi.ana.ed.ac.uk/kidhome.html(collation of studies relating to kidney development)Predictions http://www.rzuser.uni-heidelberg.de/∼jb5/aboutproject.htm(EU-funded PREvention of DIabetic ComplicaTIONS)ReGeNet http://www.regenet.eu(the REnal GEnome NETwork project)Renal genes http://www.renalgenes.org(aims to provide information on nephronophthisis, nephrotic syndromeand urinary tract malformations)

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 11: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

208 AJ McKnight et al

population-based to personal health care strategies[174].

Conclusions

Genetic studies reported now are very different fromthose published > 5–10 years ago, with standardsimproving as we learn more about the confound-ing factors for genetic analyses [11,175]. Guidelineshave been published for STrengthening the REport-ing of Genetic Association studies (STREGA) [176].Important study design issues for genetic investiga-tions include careful phenotyping, appropriate samplesizes, tests for cryptic relatedness, a sound rationalefor genetic marker selection, minimizing genotypingerrors, managing population stratification, avoidanceof bias, rigorous statistical methods, replication ofpositive results and the public availability of non-identifiable data.

Commercial organizations now offer to ‘explain’how an individual European’s genetic data will affecttheir odds of developing kidney disease (https://www.23andme.com/health/pre kidney disease). This is apremature application of the burgeoning area of‘renal genetics’ research. Nonetheless, there are real-istic prospects of enhancing our understanding ofmultiple genes and biological pathways involved inthe pathogenesis of CKD. With continued effortsto bridge the many knowledge gaps between geno-types and phenotypes, it should prove possible toenhance our ability to predict and identify individ-uals ‘at risk’ of renal disease and improve the effi-cacy of treatment by developing novel therapies orutilizing pharmacogenetic insights for existing medi-cations.

Acknowledgements

DC is supported by a Northern Ireland Kidney Research FundPhD studentship.

Teaching Materials

PowerPoint slides of the figures from this revieware supplied as supporting information in the onlineversion of this article.

References

1. Jones CA, McQuillan GM, Kusek JW, Eberhardt MS, Her-man WH, Coresh J, et al. Serum creatinine levels in the USpopulation: third National Health and Nutrition Examination Sur-vey. Am J Kidney Dis 1998;32:992–999.

2. Hallan SI, Coresh J, Astor BC, Asberg A, Powe NR, Romund-stad S, et al. International comparison of the relationship ofchronic kidney disease prevalence and ESRD risk. J Am SocNephrol 2006;17:2275–2284.

3. Quinn MP, Rainey A, Cairns KJ, Marshall AH, Savage G,Kee F, et al. The practical implications of using standardized

estimation equations in calculating the prevalence of chronickidney disease. Nephrol Dial Transpl 2008;23:542–548.

4. Go AS, Chertow GM, Fan D, McCulloch CE, Hsu CY. Chronickidney disease and the risks of death, cardiovascular events, andhospitalization. N Engl J Med 2004;351:1296–1305.

5. Johnson ES, Thorp ML, Yang X, Charansonney OL, Smith DH.Predicting renal replacement therapy and mortality in CKD. AmJ Kidney Dis 2007;50:559–565.

6. Foley RN, Collins AJ. End-stage renal disease in the UnitedStates: an update from the United States Renal Data System.J Am Soc Nephrol 2007;18:2644–2648.

7. US Renal Data System: USRDS 2008 Annual data report: atlas ofchronic kidney disease and end-stage renal disease in the UnitedStates. National Institutes of Health, National Institute of Diabetesand Digestive and Kidney Diseases: Bethesda, MD. US CensusData 2008: www.usrds.org.

8. Ansell D, Feehally J, Fogarty D, Ford D, Hodson A, Tom-son C, et al. UK Renal Registry 11th Annual Report (Decem-ber 2008). Nephron Clin Pract 2009;111(suppl 1):c43–68;www.renalreg.com/Reports/2008.html.

9. Courtney AE, McNamee PT, Maxwell AP. The evolution ofrenal transplantation in clinical practice: for better, for worse?Qu J Med 2008;101:967–978.

10. Mogensen CE. Genetics and diabetic renal disease: still a bigblack hole. Diabetes Care 2003;26:1631–1632.

11. McKnight AJ, O’Donoghue D, Maxwell AP. Annotated chromo-some maps for renal disease. Hum Mutat 2009;30:314–320.

12. Lu JC, Coca SG, Patel UD, Cantley L, Parikh CR. Searching forgenes that matter in acute kidney injury: a systematic review.Clin J Am Soc Nephrol 2009;4:1020–1031.

13. Goldfarb-Rumyantzev AS, Naiman N. Genetic prediction ofrenal transplant outcome. Curr Opin Nephrol Hypertens2008;17:573–579.

14. Brunelli M, Gobbo S, Cossu-Rocca P, Cheng L, Ficarra V, et al.Fluorescent cytogenetics of renal cell neoplasms. Pathologica2008;100:454–460.

15. Yan BC, Mackinnon AC, Al Ahmadie HA. Recent developmentsin the pathology of renal tumors: morphology and molecularcharacteristics of select entities. Arch Pathol Lab Med2009;133:1026–1032.

16. Reeders ST, Breuning MH, Davies KE, Nicholls RD, Jar-man AP, Higgs DR, et al. A highly polymorphic DNA markerlinked to adult polycystic kidney disease on chromosome 16.Nature 1985;317:542–544.

17. The European Polycystic Kidney Disease Consortium: Thepolycystic kidney disease 1 gene encodes a 14 kb transcriptand lies within a duplicated region on chromosome 16. Cell1994;77:881–894.

18. Geng L, Segal Y, Peissel B, Deng N, Pei Y, Carone F, et al.Identification and localization of polycystin, the PKD1 geneproduct. J Clin Invest 1996;98:2674–2682.

19. Gout AM, Martin NC, Brown AF, Ravine D. PKDB: PolycysticKidney Disease Mutation Database — a gene variant databasefor autosomal dominant polycystic kidney disease. Hum Mutat2007;8:654–659.

20. Ong AC, Harris PC. Molecular basis of renal cyst forma-tion — one hit or two? Lancet 1997;349:1039–1040.

21. Brasier JL, Henske EP. Loss of the polycystic kidney disease(PKD1 ) region of chromosome 16p13 in renal cyst cells supportsa loss-of-function model for cyst pathogenesis. J Clin Invest1997;99:194–199.

22. Igarashi P, Somlo S. Genetics and pathogenesis of polycystickidney disease. J Am Soc Nephrol 2002;13:2384–2398.

23. Qian Q, Hunter LW, Li M, Marin-Padilla M, Prakash YS,Somlo S, et al. Pkd2 haploinsufficiency alters intracellularcalcium regulation in vascular smooth muscle cells. Hum MolGenet 2003;12:1875–1880.

24. Yoder BK. Role of primary cilia in the pathogenesis of polycystickidney disease. J Am Soc Nephrol 2007;18:1381–1388.

25. Patel V, Chowdhury R, Igarashi P. Advances in the pathogenesisand treatment of polycystic kidney disease. Curr Opin NephrolHypertens 2009;18:99–106.

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 12: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

Genetics of renal disease 209

26. Torres VE, Harris PC. Autosomal dominant polycystic kidneydisease: the last 3 years. Kidney Int 2009;76:149–168.

27. Fain PR, McFann KK, Taylor MR, Tison M, Johnson AM,Reed B, et al. Modifier genes play a significant role in thephenotypic expression of PKD1 . Kidney Int 2005;67:1256–1267.

28. Sampson JR, Scahill SJ, Stephenson JB, Mann L, Connor JM.Genetic aspects of tuberous sclerosis in the west of Scotland.J Med Genet 1989;26:28–31.

29. Krol RP, Nozu K, Nakanishi K, Iijima K, Takeshima Y, Fu XJ,et al. Somatic mosaicism for a mutation of the COL4A5 gene isa cause of mild phenotype male Alport syndrome. Nephrol DialTranspl 2008;23:2525–2530.

30. Serre D, Gurd S, Ge B, Sladek R, Sinnett D, Harmsen E,et al. Differential allelic expression in the human genome:a robust approach to identify genetic and epigenetic cis-acting mechanisms regulating gene expression. PLoS Genet2008;4:e1000006.

31. Jais JP, Knebelmann B, Giatras I, De Marchi M, Rizzoni G,Renieri A, et al. X-linked Alport syndrome: natural history in195 families and genotype–phenotype correlations in males.J Am Soc Nephrol 2000;11:649–657.

32. Albert I, Jais JP. The use of frailty models in genetic studies:application to the relationship between end-stage renal failure andmutation type in Alport syndrome. European Community AlportSyndrome Concerted Action Group (ECASCA). J EpidemiolBiostat 2000;5:169–175.

33. Boon CJ, van de Kar NC, Klevering BJ, Keunen JE, Cre-mers FP, Klaver CC, et al. The spectrum of phenotypes causedby variants in the CFH gene. Mol Immunol 2009;46:1573–1594.

34. Yoshida T, Kato K, Fujimaki T, Yokoi K, Oguri M, Watanabe S,et al. Association of genetic variants with chronic kidney diseasein Japanese individuals. Clin J Am Soc Nephrol 2009;4:883–890.

35. Manchanda PK, Singh R, Mittal RD. Cytokine (IL-10–1082and -819) and chemokine receptor (CCR2 and CCR5 ) genepolymorphism in North Indian patients with end-stage renaldisease. DNA Cell Biol 2009;28:177–183.

36. Stefanakis N, Ziroyiannis P, Trygonis S, Lamnissou K. Modifiereffect of the Glu298Asp polymorphism of endothelial nitric oxidesynthase gene in autosomal-dominant polycystic kidney disease.Nephron Clin Pract 2008;110:c101–106.

37. Reiterova J, Obeidova H, Lenicek M, Stekrova J, Merta M,Maixnerova D, et al. Influence of VEGF polymorphism onprogression of autosomal dominant polycystic kidney disease.Kidney Blood Press Res 2008;31:398–403.

38. Rossetti S, Harris PC. Genotype-phenotype correlations inautosomal dominant and autosomal recessive polycystic kidneydisease. J Am Soc Nephrol 2007;18:1374–1380.

39. Jacobsen PK, Tarnow L, Parving HH. Time to consider ACEinsertion/deletion genotypes and individual renoprotective treat-ment in diabetic nephropathy? Kidney Int 2006;69:1293–1295.

40. Costa-Scharplatz M, van Asselt AD, Bachmann LM, KesselsAG, Severens JL. Cost-effectiveness of pharmacogenetic testingto predict treatment response to angiotensin-converting enzymeinhibitor. Pharmacogenet Genom 2007;17:359–368.

41. Lander E, Kruglyak L. Genetic dissection of complex traits:guidelines for interpreting and reporting linkage results. NatGenet 1995;11:241–247.

42. McCarthy MI. Susceptibility gene discovery for commonmetabolic and endocrine traits. J Mol Endocrinol 2002;28:1–17.

43. Kiryluk K. Quantitative genetics of renal function: tacklingcomplexities of the eGFR phenotype in gene mapping studies.Kidney Int 2008;74:1109–1112.

44. Sellick GS, Longman C, Tolmie J, Newbury-Ecob R, Geen-halgh L, Hughes S, et al. Genomewide linkage searches forMendelian disease loci can be efficiently conducted using high-density SNP genotyping arrays. Nucleic Acids Res 2004;32:e164.

45. Rogus JJ, Poznik GD, Pezzolesi MG, Smiles AM, Dunn J,Walker W, et al. High-density single nucleotide polymorphismgenome-wide linkage scan for susceptibility genes for diabeticnephropathy in type 1 diabetes: discordant sibpair approach.Diabetes 2008;57:2519–2526.

46. Pattaro C, Aulchenko YS, Isaacs A, Vitart V, Hayward C,Franklin CS, et al. Genome-wide linkage analysis of serumcreatinine in three isolated European populations. Kidney Int2009;[Epub ahead of print].

47. Badner JA, Gershon ES. Regional meta-analysis of publisheddata supports linkage of autism with markers on chromosome7. Mol Psychiat 2002;7:56–66.

48. Zintzaras E, Ioannidis JP. HEGESMA: genome search meta-analysis and heterogeneity testing. Bioinformatics 2005;21:3672–3673.

49. Briollais L, Durrieu G, Upathilake R. Novel approach forgenome scan meta-analysis of rheumatoid arthritis: a kernel-basedestimation procedure. BMC Proc 2007;1(suppl 1):S96.

50. Goldstein DR, Sain SR, Guerra R, Etzel CJ. Meta-analysis bycombining parameter estimates: simulated linkage studies. GenetEpidemiol 1999;17(Suppl 1):S581–586.

51. Guerra R, Etzel CJ, Goldstein DR, Sain Sr. Meta-analysis bycombining p values: simulated linkage studies. Genet Epidemiol1999;17:(suppl 1): S605–609.

52. Risch N, Merikangas K. The future of genetic studies of complexhuman diseases. Science 1996;273:1516–1517.

53. Wang WY, Barratt BJ, Clayton DG, Todd JA. Genome-wideassociation studies: theoretical and practical concerns. Nat RevGenet 2005;6:109–118.

54. Hattersley AT, McCarthy MI. What makes a good geneticassociation study? Lancet 2005;366:1315–1323.

55. Purcell S, Cherny SS, Sham PC. Genetic power calculator:design of linkage and association genetic mapping studies ofcomplex traits. Bioinformatics 2003;19:149–150.

56. Gordon D, Finch SJ. Factors affecting statistical power inthe detection of genetic association. J Clin Invest 2005;115:1408–1418.

57. Vardarli I, Baier LJ, Hanson RL, Akkoyun I, Fischer C, RohmeissP, et al. Gene for susceptibility to diabetic nephropathy in type 2diabetes maps to 18q22.3–23. Kidney Int 2002;62:2176–2183.

58. Bowden DW, Colicigno CJ, Langefeld CD, Sale MM, WilliamsA, Anderson PJ, et al. A genome scan for diabetic nephropathyin African Americans. Kidney Int 2004;66:1517–1526.

59. Iyengar SK, Abboud HE, Goddard KA, Saad MF, Adler SG,Arar NH, et al. Genome-wide scans for diabetic nephropathyand albuminuria in multiethnic populations: the familyinvestigation of nephropathy and diabetes (FIND). Diabetes2007;56:1577–1585.

60. Freedman BI, Hicks PJ, Sale MM, Pierson ED, Langefeld CD,Rich SS, et al. A leucine repeat in the carnosinase gene CNDP1is associated with diabetic end-stage renal disease in EuropeanAmericans. Nephrol Dial Transpl 2007;22:1131–1135.

61. Janssen B, Hohenadel D, Brinkkoetter P, Peters V, Rind N,Fischer C, et al. Carnosine as a protective factor in diabeticnephropathy: association with a leucine repeat of the carnosinasegene CNDP1 . Diabetes 2005;54:2320–2327.

62. McDonough CW, Hicks PJ, Lu L, Langefeld CD, Freedman BI,Bowden DW. The influence of carnosinase gene polymorphismson diabetic nephropathy risk in African-Americans. Hum Genet2009;126:265–275.

63. Kao WH, Klag MJ, Meoni LA, Reich D, Berthier-Schaad Y,Li M, et al. MYH9 is associated with nondiabetic end-stage renaldisease in African Americans. Nat Genet 2008;40:1185–1192.

64. Kopp JB, Smith MW, Nelson GW, Johnson RC, Freedman BI,Bowden DW, et al. MYH9 is a major-effect risk gene for focalsegmental glomerulosclerosis. Nat Genet 2008;40:1175–1184.

65. Freedman BI, Hicks PJ, Bostrom MA, Cunningham ME, Liu Y,Divers J, et al. Polymorphisms in the non-muscle myosin heavychain 9 gene (MYH9 ) are strongly associated with end-stagerenal disease historically attributed to hypertension in AfricanAmericans. Kidney Int 2009;75:736–745.

66. Freedman BI, Hicks PJ, Bostrom MA, Comeau ME, Divers J,Bleyer AJ, et al. Non-muscle myosin heavy chain 9 gene MYH9associations in African Americans with clinically diagnosedtype 2 diabetes mellitus-associated ESRD. Nephrol Dial Transpl2009;[Epub ahead of print].

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 13: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

210 AJ McKnight et al

67. Reich DE, Lander ES. On the allelic spectrum of human disease.Trends Genet 2001;17:502–510.

68. Ho J, Ng KH, Rosen S, Dostal A, Gregory RI, Kreidberg JA.Podocyte-specific loss of functional microRNAs leads torapid glomerular and tubular injury. J Am Soc Nephrol2008;19:2069–2075.

69. Horikawa Y, Wood CG, Yang H, Zhao H, Ye Y, Gu J, et al.Single nucleotide polymorphisms of microRNA machinery genesmodify the risk of renal cell carcinoma. Clin Cancer Res2008;14:7956–7962.

70. Huppi K, Volfovsky N, Runfola T, Jones TL, Mackiewicz M,Martin SE, et al. The identification of microRNAs in agenomically unstable region of human chromosome 8q24. MolCancer Res 2008;6:212–221.

71. Millis MP, Bowen D, Kingsley C, Watanabe RM, Wolford JK.Variants in the plasmacytoma variant translocation gene (PVT1 )are associated with end-stage renal disease attributed to type 1diabetes. Diabetes 2007;56:3027–3032.

72. Martin MM, Buckenberger JA, Jiang J, Malana GE, Nuovo GJ,Chotani M, et al. The human angiotensin II type 1 receptor+ 1166 A/C polymorphism attenuates microRNA-155 binding.J Biol Chem 2007;282:24262–24269.

73. Lettre G, Rioux JD. Autoimmune diseases: insights fromgenome-wide association studies. Hum Mol Genet 2008;17:R116–121.

74. Todd JA, Walker NM, Cooper JD, Smyth DJ, Downes K,Plagnol V, et al. Robust associations of four new chromosomeregions from genome-wide analyses of type 1 diabetes. Nat Genet2007;39:857–864.

75. Hinks A, Eyre S, Ke X, Barton A, Martin P, Flynn E, et al.Overlap of disease susceptibility loci for rheumatoid arthritis(RA) and juvenile idiopathic arthritis (JIA). Ann Rheum Dis2009;[Epub ahead of print].

76. Franke A, Balschun T, Karlsen TH, Hedderich J, May S, Lu T,et al. Replication of signals from recent studies of Crohn’sdisease identifies previously unknown disease loci for ulcerativecolitis. Nat Genet 2008;40:713–715.

77. McKnight AJ, Maxwell AP, Sawcer S, Compston A, Setakis E,Patterson CC, et al. A genome-wide DNA microsatelliteassociation screen to identify chromosomal regions harboringcandidate genes in diabetic nephropathy. J Am Soc Nephrol2006;17:831–836.

78. Maeda S. Genome-wide search for susceptibility gene to diabeticnephropathy by gene-based SNP. Diabetes Res Clin Pract2004;66(suppl 1):S45–47.

79. Taylor KE, Remmers EF, Lee AT, Ortmann WA, Plenge RM,Tian C, et al. Specificity of the STAT4 genetic association forsevere disease manifestations of systemic lupus erythematosus.PLoS Genet 2008;4:e1000084.

80. Savage DA, Patterson CC, Deloukas P, Whittaker P, McK-night AJ, Morrison J, et al. Genetic association analyses ofnon-synonymous single nucleotide polymorphisms in diabeticnephropathy. Diabetologia 2008;51:1998–2002.

81. Obara W, Iida A, Suzuki Y, Tanaka T, Akiyama F, Maeda S,et al. Association of single-nucleotide polymorphisms in thepolymeric immunoglobulin receptor gene with immunoglobulinA nephropathy (IgAN) in Japanese patients. J Hum Genet2003;48:293–299.

82. Pezzolesi MG, Poznik GD, Mychaleckyj JC, Paterson AD, BaratiMT, Klein JB, et al. Genome-wide association scan for diabeticnephropathy susceptibility genes in type 1 diabetes. Diabetes2009;58:1403–1410.

83. Hanson RL, Craig DW, Millis MP, Yeatts KA, Kobes S, Pear-son JV, et al. Identification of PVT1 as a candidate gene forend-stage renal disease in type 2 diabetes using a pooling-basedgenome-wide single nucleotide polymorphism association study.Diabetes 2007;56:975–983.

84. Woodward OM, Kottgen A, Coresh J, Boerwinkle E, Gug-gino WB, Kottgen M. Identification of a urate transporter,ABCG2, with a common functional polymorphism causing gout.Proc Natl Acad Sci USA 2009;106:10338–10342.

85. Kottgen A, Glazer NL, Dehghan A, Hwang SJ, Katz R, Li M,et al. Multiple loci associated with indices of renal function andchronic kidney disease. Nat Genet 2009;[Epub ahead of print].

86. Newton-Cheh C, Johnson T, Gateva V, Tobin MD, Bochud M,Coin L, et al. Genome-wide association study identifies eight lociassociated with blood pressure. Nat Genet 2009;[Epub ahead ofprint].

87. Hirschhorn JN. Genomewide association studies — illuminatingbiologic pathways. N Engl J Med 2009;360:1699–1701.

88. Gregersen PK. Closing the gap between genotype and phenotype.Nat Genet 2009;41:958–959.

89. Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ,et al. The Connectivity Map: using gene-expression signaturesto connect small molecules, genes, and disease. Science2006;313:1929–1935.

90. Altshuler D, Daly MJ, Lander ES. Genetic mapping in humandisease. Science 2008;322:881–888.

91. McMahon R, Murphy M, Clarkson M, Taal M, Mackenzie HS,Godson C, et al. IHG-2, a mesangial cell gene induced by highglucose, is human gremlin. Regulation by extracellular glucoseconcentration, cyclic mechanical strain, and transforming growthfactor-β1. J Biol Chem 2000;275:9901–9904.

92. Dolan V, Murphy M, Sadlier D, Lappin D, Doran P, Godson C,et al. Expression of gremlin, a bone morphogenetic proteinantagonist, in human diabetic nephropathy. Am J Kidney Dis2005;45:1034–1039.

93. Dendrou CA, Plagnol V, Fung E, Yang JH, Downes K, CooperJD, et al. Cell-specific protein phenotypes for the autoimmunelocus IL2RA using a genotype-selectable human bioresource. NatGenet 2009;41:1011–1015.

94. Devuyst O, Dahan K, Pirson Y. Tamm–Horsfall protein oruromodulin: new ideas about an old molecule. Nephrol DialTranspl 2005;20:1290–1294.

95. Saemann MD, Weichhart T, Horl WH, Zlabinger GJ.Tamm–Horsfall protein: a multilayered defence molecule againsturinary tract infection. Eur J Clin Invest 2005;35:227–235.

96. Schlieper G, Westenfeld R, Brandenburg V, Ketteler M. Inhibitorsof calcification in blood and urine. Semin Dial 2007;20:113–121.

97. Hart TC, Gorry MC, Hart PS, Woodard AS, Shihabi Z, Sandhu J,et al. Mutations of the UMOD gene are responsible for medullarycystic kidney disease 2 and familial juvenile hyperuricaemicnephropathy. J Med Genet 2002;39:882–892.

98. Williams SE, Reed AA, Galvanovskis J, Antignac C, Good-ship T, Karet FE, et al. Uromodulin mutations causing familialjuvenile hyperuricaemic nephropathy lead to protein maturationdefects and retention in the endoplasmic reticulum. Hum MolGenet 2009;18:2963–2974.

99. Hoyer Jr. Tubulointerstitial immune complex nephritis inrats immunized with Tamm–Horsfall protein. Kidney Int1980;17:284–292.

100. Mayrer AR, Kashgarian M, Ruddle NH, Marier R, Hodson CJ,Richards FF, et al. Tubulointerstitial nephritis and immunologicresponses to Tamm–Horsfall protein in rabbits challengedwith homologous urine or Tamm–Horsfall protein. J Immunol1982;128:2634–2642.

101. Bachmann S, Mutig K, Bates J, Welker P, Geist B, Gross V,et al. Renal effects of Tamm–Horsfall protein (uromodulin) defi-ciency in mice. Am J Physiol Renal Physiol 2005;288:F559–567.

102. Dehghan A, Kottgen A, Yang Q, Hwang SJ, Kao WL,Rivadeneira F, et al. Association of three genetic loci with uricacid concentration and risk of gout: a genome-wide associationstudy. Lancet 2008;372:1953–1961.

103. Kolz M, Johnson T, Sanna S, Teumer A, Vitart V, Perola M,et al. Meta-analysis of 28 141 individuals identifies commonvariants within five new loci that influence uric acidconcentrations. PLoS Genet 2009;5:e1000504.

104. Working Group of the International IgA Nephropathy Networkand the Renal Pathology Society, Cook HT, Troyanov S,Alpers CE, Amore A, Barratt J, et al. The Oxford classificationof IgA nephropathy: pathology definitions, correlations, andreproducibility. Kidney Int 2009;76:546–556.

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 14: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

Genetics of renal disease 211

105. Swaminathan S, Leung N, Lager DJ, Melton LJ, III, BergstralhEJ, Rohlinger A, et al. Changing incidence of glomerular diseasein Olmsted County, Minnesota: a 30-year renal biopsy study. ClinJ Am Soc Nephrol 2006;1:483–487.

106. Zhou FD, Zhao MH, Zou WZ, Liu G, Wang H. The changingspectrum of primary glomerular diseases within 15 years: asurvey of 3331 patients in a single Chinese centre. Nephrol DialTranspl 2009;24:870–876.

107. Hanko JB, Mullan RN, O’Rourke DM, McNamee PT, MaxwellAP, Courtney AE. The changing pattern of adult primaryglomerular disease. Nephrol Dial Transpl 2009;24:3050–3054.

108. Schena FP, Cerullo G, Torres DD, Scolari F, Foramitti M,Amoroso A, et al. The IgA nephropathy biobank. An importantstarting point for the genetic dissection of a complex trait. BMCNephrol 2005;6:14.

109. Liu XQ, Paterson AD, He N, George-Hyslop P, Rauta V,Gronhagen-Riska C, et al. IL5RA and TNFRSF6B gene variantsare associated with sporadic IgA nephropathy. J Am Soc Nephrol2008;19:1025–1033.

110. Conway BR, Maxwell AP. Genetics of diabetic nephropathy: arethere clues to the understanding of common kidney diseases?Nephron Clin Pract 2009;112:c213–221.

111. McKnight AJ, Woodman AM, Parkkonen M, Patterson CC, Sav-age DA, Forsblom C, et al. Investigation of DNA polymorphismsin SMAD genes for genetic predisposition to diabetic nephropa-thy in patients with type 1 diabetes mellitus. Diabetologia2009;52:844–849.

112. Tarnow L, Groop PH, Hadjadj S, Kazeem G, Cambien F,Marre M, et al. European rational approach for the genetics ofdiabetic complications — EURAGEDIC: patient populations andstrategy. Nephrol Dial Transpl 2008;23:161–168.

113. Tanaka N, Babazono T, Saito S, Sekine A, Tsunoda T, Haneda M,et al. Association of solute carrier family 12 (sodium/chloride)member 3 with diabetic nephropathy, identified by genome-wide analyses of single nucleotide polymorphisms. Diabetes2003;52:2848–2853.

114. Shimazaki A, Kawamura Y, Kanazawa A, Sekine A, Saito S,Tsunoda T, et al. Genetic variations in the gene encoding ELMO1are associated with susceptibility to diabetic nephropathy.Diabetes 2005;54:1171–1178.

115. Kim JH, Shin HD, Park BL, Moon MK, Cho YM,Hwang YH, et al. SLC12A3 [solute carrier family 12member (sodium/chloride) 3] polymorphisms are associatedwith end-stage renal disease in diabetic nephropathy. Diabetes2006;55:843–848.

116. Ng DP, Nurbaya S, Choo S, Koh D, Chia KS, Krolewski AS.Genetic variation at the SLC12A3 locus is unlikely to explainrisk for advanced diabetic nephropathy in Caucasians with type2 diabetes. Nephrol Dial Transpl 2008;23:2260–2264.

117. Pezzolesi MG, Katavetin P, Kure M, Poznik GD, Skupien J,Mychaleckyj JC, et al. Confirmation of genetic associationsat ELMO1 in the GoKinD collection support its roleas a susceptibility gene in diabetic nephropathy. Diabetes2009;58:1403–1410.

118. Leak TS, Perlegas PS, Smith SG, Keene KL, Hicks PJ, Lange-feld CD, et al. Variants in intron 13 of the ELMO1 gene areassociated with diabetic nephropathy in African Americans. AnnHum Genet 2009;73:152–159.

119. McCarroll SA. Extending genome-wide association studies tocopy-number variation. Hum Mol Genet 2008;17:R135–142.

120. Redon R, Ishikawa S, Fitch KR, Feuk L, Perry GH, AndrewsTD, et al. Global variation in copy number in the human genome.Nature 2006;444:444–454.

121. de Smith AJ, Tsalenko A, Sampas N, Scheffer A, Yamada NA,Tsang P, et al. Array CGH analysis of copy number variationidentifies 1284 new gene variants in healthy white males:implications for association studies of complex diseases. HumMol Genet 2007;16:2783–2794.

122. Lupski JR, Stankiewicz P. Genomic disorders: molecular mecha-nisms for rearrangements and conveyed phenotypes. PLoS Genet2005;1:e49.

123. Hollox EJ, Huffmeier U, Zeeuwen PL, Palla R, Lascorz J,Rodijk-Olthuis D, et al. Psoriasis is associated with increasedβ-defensin genomic copy number. Nat Genet 2008;40:23–25.

124. Aitman TJ, Dong R, Vyse TJ, Norsworthy PJ, Johnson MD,Smith J, et al. Copy number polymorphism in Fcgr3 pre-disposes to glomerulonephritis in rats and humans. Nature2006;439:851–855.

125. Willcocks LC, Lyons PA, Clatworthy MR, Robinson JI, Yang W,Newland SA, et al. Copy number of FCGR3B, which isassociated with systemic lupus erythematosus, correlates withprotein expression and immune complex uptake. J Exp Med2008;205:1573–1582.

126. Goodship TH. Factor H genotype–phenotype correlations:lessons from aHUS, MPGN II, and AMD. Kidney Int2006;70:12–13.

127. Liew G, Mitchell P, Wong TY, Iyengar SK, Wang JJ. CKDincreases the risk of age-related macular degeneration. J Am SocNephrol 2008;19:806–811.

128. Xing C, Sivakumaran TA, Wang JJ, Rochtchina E, Joshi T,Smith W, et al. Complement factor H polymorphisms, renalphenotypes and age-related macular degeneration: the BlueMountains Eye Study. Genes Immunol 2008;9:231–239.

129. Klein R, Knudtson MD, Lee KE, Klein BE. Serum cystatin Clevel, kidney disease markers, and incidence of age-relatedmacular degeneration: the Beaver Dam Eye Study. ArchOphthalmol 2009;127:193–199.

130. Alkan C, Kidd JM, Marques-Bonet T, Aksay G, Antonacci F,Hormozdiari F, et al. Personalized copy number and segmentalduplication maps using next-generation sequencing. Nat Genet2009;41:1061–1067.

131. Schmid-Kubista KE, Tosakulwong N, Wu Y, Ryu E, Hecker LA,Baratz KH, et al. Contribution of copy number variation inthe regulation of complement activation locus to developmentof age-related macular degeneration. Invest Ophthalmol Vis Sci2009;[Epub ahead of print].

132. Rao M, Li L, Demello C, Guo D, Jaber BL, Pereira BJ, et al.Mitochondrial DNA injury and mortality in hemodialysis patients.J Am Soc Nephrol 2009;20:189–196.

133. de Andrade PB, Rubi B, Frigerio F, van den Ouweland JM,Maassen JA, Maechler P. Diabetes-associated mitochondrialDNA mutation A3243G impairs cellular metabolic path-ways necessary for β cell function. Diabetologia 2006;49:1816–1826.

134. Canter JA, Olson LM, Spencer K, Schnetz-Boutaud N, Ander-son B, Hauser MA, et al. Mitochondrial DNA polymorphismA4917G is independently associated with age-related maculardegeneration. PLoS One 2008;3:e2091.

135. Lynch D, Wanglund C, Spathis R, Chan CW, Reiff DM,Lum JK, et al. The contribution of mitochondrial dysfunctionto a gene–environment model of Guamanian ALS and PD.Mitochondrion 2008;8:109–116.

136. Finsterer J. Overview on visceral manifestations of mitochondrialdisorders. Neth J Med 2006;64:61–71.

137. Lim PS, Cheng YM, Wei YH. Large-scale mitochondrial DNAdeletions in skeletal muscle of patients with end-stage renaldisease. Free Radic Biol Med 2000;29:454–463.

138. Liu CS, Ko LY, Lim PS, Kao SH, Wei YH. Biomarkers ofDNA damage in patients with end-stage renal disease:mitochondrial DNA mutation in hair follicles. Nephrol DialTranspl 2001;16:561–565.

139. Watson B Jr, Khan MA, Desmond RA, Bergman S. Mitochon-drial DNA mutations in black Americans with hypertension-associated end-stage renal disease. Am J Kidney Dis 2001;38:529–536.

140. Hinokio Y, Suzuki S, Komatu K, Ohtomo M, Onoda M, Mat-sumoto M, et al. A new mitochondrial DNA deletion associatedwith diabetic amyotrophy, diabetic myoatrophy and diabetic fattyliver. Muscle Nerve 1995;3:S142–149.

141. Yamagata K, Tomida C, Umeyama K, Urakami K, Ishizu T,Hirayama K, et al. Prevalence of Japanese dialysis patients withan A-to-G mutation at nucleotide 3243 of the mitochondrialtRNA[Leu(UUR)] gene. Nephrol Dial Transpl 2000;15:385–388.

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 15: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

212 AJ McKnight et al

142. Nakamura S, Yoshinari M, Doi Y, Yoshizumi H, Katafuchi R,Yokomizo Y, et al. Renal complications in patients with diabetesmellitus associated with an A to G mutation of mitochondrialDNA at the 3243 position of leucine tRNA. Diabetes Res ClinPract 1999;44:183–189.

143. Iwasaki N, Babazono T, Tsuchiya K, Tomonaga O, Suzuki A,Togashi M, et al. Prevalence of A-to-G mutation at nucleotide3243 of the mitochondrial tRNA[Leu(UUR)] gene in Japanesepatients with diabetes mellitus and end stage renal disease.J Hum Genet 2001;46:330–334.

144. Massin P, Dubois-Laforgue D, Meas T, Laloi-Michelin M,Gin H, Bauduceau B, et al. Retinal and renal complicationsin patients with a mutation of mitochondrial DNA at position3243 (maternally inherited diabetes and deafness). A case-controlstudy. Diabetologia 2008;51:1664–1670.

145. Martin-Hernandez E, Garcia-Silva MT, Vara J, Campos Y,Cabello A, Muley R, et al. Renal pathology in children withmitochondrial diseases. Pediatr Nephrol 2005;20:1299–1305.

146. Tulinius MH, Oldfors A, Holme E, Larsson NG, Houshmand M,Fahleson P, et al. Atypical presentation of multisystem disordersin two girls with mitochondrial DNA deletions. Eur J Pediatr1995;154:35–42.

147. Majander A, Suomalainen A, Vettenranta K, Sariola H,Perkkio M, Holmberg C, et al. Congenital hypoplastic anemia,diabetes, and severe renal tubular dysfunction associated with amitochondrial DNA deletion. Pediatr Res 1991;30:327–330.

148. Tzen CY, Tsai JD, Wu TY, Chen BF, Chen ML, Lin SP, et al.Tubulointerstitial nephritis associated with a novel mitochondrialpoint mutation. Kidney Int 2001;59:846–854.

149. Morikawa Y, Matsuura N, Kakudo K, Higuchi R, Koike M,Kobayashi Y. Pearson’s marrow/pancreas syndrome: a histolog-ical and genetic study. Virchows Arch A Pathol Anat Histopathol1993;423:227–231.

150. Hirano M, Konishi K, Arata N, Iyori M, Saruta T, Kuramochi S,et al. Renal complications in a patient with A-to-G mutation ofmitochondrial DNA at the 3243 position of leucine tRNA. InternMed 2002;41:113–118.

151. Hotta O, Inoue CN, Miyabayashi S, Furuta T, Takeuchi A,Taguma Y. Clinical and pathologic features of focal segmentalglomerulosclerosis with mitochondrial tRNA Leu(UUR) genemutation. Kidney Int 2001;59:1236–1243.

152. Scaglia F, Vogel H, Hawkins EP, Vladutiu GD, Liu LL, WongLJ. Novel homoplasmic mutation in the mitochondrial tRNA Tyrgene associated with atypical mitochondrial cytopathy presentingwith focal segmental glomerulosclerosis. Am J Med Genet A2003;123A:172–178.

153. Yorifuji T, Kawai M, Momoi T, Sasaki H, Furusho K, Muroi J,et al. Nephropathy and growth hormone deficiency in a patientwith mitochondrial tRNA[Leu(UUR)] mutation. J Med Genet1996;33:621–622.

154. Souied E, Pisella PJ, Ossareh B, Brezin A, Junes P, Wild-Decrette C, et al. [Positive diagnosis of Leber’s hereditaryoptic neuropathy using molecular genetics]. J Fr Ophthalmol1997;20:65–70.

155. Fujii H, Mori Y, Kayamori K, Igari T, Ito E, Akashi T, et al.A familial case of mitochondrial disease resembling Alportsyndrome. Clin Exp Nephrol 2008;12:159–163.

156. Jansen JJ, Maassen JA, van der Woude FJ, Lemmink HA, vanden Ouweland JM, t’Hart LM, et al. Mutation in mitochondrialtRNA [Leu(UUR)] gene associated with progressive kidneydisease. J Am Soc Nephrol 1997;8:1118–1124.

157. Cheong HI, Chae JH, Kim JS, Park HW, Ha IS, Hwang YS,et al. Hereditary glomerulopathy associated with a mitochondrialtRNA (Leu) gene mutation. Pediatr Nephrol 1999;13:477–480.

158. Schwartz F, Duka A, Sun F, Cui J, Manolis A, Gavras H.Mitochondrial genome mutations in hypertensive individuals. AmJ Hypertens 2004;17:629–635.

159. Salas A, Carracedo A, Macaulay V, Richards M, Bandelt HJ. Apractical guide to mitochondrial DNA error prevention in clinical,forensic, and population genetics. Biochem Biophys Res Commun2005;335:891–899.

160. Bandelt HJ, Achilli A, Kong QP, Salas A, Lutz-Bonengel S,Sun C, et al. Low ‘penetrance’ of phylogenetic knowledge inmitochondrial disease studies. Biochem Biophys Res Commun2005;333:122–130.

161. Galperin MY, Cochrane GR. Nucleic Acids Research annualDatabase Issue and the NAR online Molecular Biology DatabaseCollection in 2009. Nucleic Acids Res 2009;37:D1–4.

162. Mailman MD, Feolo M, Jin Y, Kimura M, Tryka K, Bagoutdi-nov R, et al. The NCBI dbGaP database of genotypes and phe-notypes. Nat Genet 2007;39:1181–1186.

163. Hindorff LA, Sethupathy P, Junkins HA, Ramos EM, Mehta JP,Collins FS, et al. Potential etiologic and functional implicationsof genome-wide association loci for human diseases and traits.Proc Natl Acad Sci USA 2009;106:9362–9367.

164. Schadt EE, Zhang B, Zhu J. Advances in systems biology areenhancing our understanding of disease and moving us closer tonovel disease treatments. Genetica 2009;136:259–269.

165. Lusis AJ, Attie AD, Reue K. Metabolic syndrome: from epidemi-ology to systems biology. Nat Rev Genet 2008;9:819–830.

166. Schadt EE, Molony C, Chudin E, Hao K, Yang X, Lum PY,et al. Mapping the genetic architecture of gene expression inhuman liver. PLoS Biol 2008;6:e107.

167. Hulbert EM, Smink LJ, Adlem EC, Allen JE, Burdick DB,Burren OS, et al. T1DBase: integration and presentation ofcomplex data for type 1 diabetes research. Nucleic Acids Res2007;35:D742–746.

168. Eizirik DL, Moore F, Flamez D, Ortis F. Use of a systemsbiology approach to understand pancreatic β-cell death in type 1diabetes. Biochem Soc Trans 2008;36:321–327.

169. Wheelock CE, Wheelock AM, Kawashima S, Diez D, Kane-hisa M, van Erk M, et al. Systems biology approaches and path-way tools for investigating cardiovascular disease. Mol Biosyst2009;5:588–602.

170. Xie L, Li J, Xie L, Bourne PE. Drug discovery using chemicalsystems biology: identification of the protein–ligand bindingnetwork to explain the side effects of CETP inhibitors. PLoSComput Biol 2009;5:e1000387.

171. He L, Sun Y, Takemoto M, Norlin J, Tryggvason K, Samuels-son T, et al. The glomerular transcriptome and a pre-dicted protein–protein interaction network. J Am Soc Nephrol2008;19:260–268.

172. Adriaens ME, Jaillard M, Waagmeester A, Coort SL, Pico AR,Evelo CT. The public road to high-quality curated biologicalpathways. Drug Discov Today 2008;13:856–862.

173. Bauer-Mehren A, Furlong LI, Sanz F. Pathway databases andtools for their exploitation: benefits, current limitations andchallenges. Mol Syst Biol 2009;5:290.

174. Deisboeck TS. Personalizing medicine: a systems biologyperspective. Mol Syst Biol 2009;5:249.

175. Mondry A, Loh M, Laurence KB, Low N. DNA polymorphismsand renal disease: a critical appraisal of studies presented at theannual ERA/EDTA and ASN conferences in 2004 and 2005.Nephrol Dial Transpl 2006;21:2775–2779.

176. Little J, Higgins JP, Ioannidis JP, Moher D, Gagnon F, vonElm E, et al. Strengthening the Reporting of Genetic AssociationStudies (STREGA) — an extension of the STROBE statement.Genet Epidemiol 2009;33:581–598.

177. Vionnet N, Tregouet D, Kazeem G, Gut I, Groop PH, Tarnow L,et al. Analysis of 14 candidate genes for diabetic nephropathyon chromosome 3q in European populations: strongest evidencefor association with a variant in the promoter region of theadiponectin gene. Diabetes 2006;55:3166–3174.

178. Bostrom MA, Freedman BI, Langefeld CD, Liu L, Hicks PJ,Bowden DW. Association of adiponectin gene polymorphismswith type 2 diabetes in an African American population enrichedfor nephropathy. Diabetes 2009;58:499–504.

179. Zhang D, Ma J, Brismar K, Efendic S, Gu HF. A singlenucleotide polymorphism alters the sequence of SP1 bindingsite in the adiponectin promoter region and is associated withdiabetic nephropathy among type 1 diabetic patients in theGenetics of Kidneys in Diabetes Study. J Diabetes Complic2009;23:265–272.

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 16: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

Genetics of renal disease 213

180. Antonellis A, Rogus JJ, Canani LH, Makita Y, Pezzolesi MG,Nam M, et al. A method for developing high-density SNP mapsand its application at the type 1 angiotensin II receptor (AGTR1 )locus. Genomics 2002;79:326–332.

181. Osawa N, Koya D, Araki S, Uzu T, Tsunoda T, Kashiwagi A,et al. Combinational effect of genes for the renin–angiotensinsystem in conferring susceptibility to diabetic nephropathy.J Hum Genet 2007;52:143–151.

182. Otto EA, Loeys B, Khanna H, Hellemans J, Sudbrak R, Fan S,et al. Nephrocystin-5, a ciliary IQ domain protein, is mutatedin Senior–Loken syndrome and interacts with RPGR andcalmodulin. Nat Genet 2005;37:282–288.

183. Olbrich H, Fliegauf M, Hoefele J, Kispert A, Otto E, Volz A,et al. Mutations in a novel gene, NPHP3, cause adolescentnephronophthisis, tapeto-retinal degeneration and hepatic fibrosis.Nat Genet 2003;34:455–459.

184. Wilson GN, Dasouki M, Barr M Jr. Further delineation of thedup(3q) syndrome. Am J Med Genet 1985;22:117–123.

185. Toncheva D, Dimitrov T, Tzoneva M. Cytogenetic studies inBalkan endemic nephropathy. Nephron 1988;48:18–21.

186. Osterholm AM, He B, Pitkaniemi J, Albinsson L, Berg T,Sarti C, et al. Genome-wide scan for type 1 diabetic nephropathyin the Finnish population reveals suggestive linkage to a singlelocus on chromosome 3q. Kidney Int 2007;71:140–145.

187. Freedman BI, Langefeld CD, Rich SS, Valis CJ, Sale MM,Williams AH, et al. A genome scan for ESRD in blackfamilies enriched for nondiabetic nephropathy. J Am Soc Nephrol2004;15:2719–2727.

188. Chistiakov DA, Savost’anov KV, Shestakova MV, ChugunovaLA, Samkhalova MS, Dedov II, et al. Confirmation of asusceptibility locus for diabetic nephropathy on chromosome3q23-q24 by association study in Russian type 1 diabetic patients.Diabetes Res Clin Pract 2004;66:79–86.

189. Moczulski DK, Rogus JJ, Antonellis A, Warram JH, KrolewskiAS. Major susceptibility locus for nephropathy in type 1 diabeteson chromosome 3q: results of novel discordant sib-pair analysis.Diabetes 1998;47:1164–1169.

190. Chen G, Adeyemo AA, Zhou J, Chen Y, Doumatey A, Lash-ley K, et al. A genome-wide search for linkage to renal functionphenotypes in West Africans with type 2 diabetes. Am J KidneyDis 2007;49:394–400.

191. Placha G, Poznik GD, Dunn J, Smiles A, Krolewski B, Glew T,et al. A genome-wide linkage scan for genes controlling variationin renal function estimated by serum cystatin C levels in extendedfamilies with type 2 diabetes. Diabetes 2006;55:3358–3365.

192. He B, Osterholm AM, Hoverfalt A, Forsblom C, Hjorleifsdot-tir EE, Nilsson AS, et al. Association of genetic variants at 3q22with nephropathy in patients with type 1 diabetes mellitus. Am JHum Genet 2009;84:5–13.

193. Lemmink HH, Mochizuki T, van den Heuvel LP, Schroder CH,Barrientos A, Monnens LA, et al. Mutations in the type IVcollagen α3 (COL4A3 ) gene in autosomal recessive Alportsyndrome. Hum Mol Genet 1994;3:1269–1273.

194. Jefferson JA, Lemmink HH, Hughes AE, Hill CM, Smeets HJ,Doherty CC, et al. Autosomal dominant Alport syndrome linkedto the type IV collage α3 and α4 genes (COL4A3 and COL4A4 ).Nephrol Dial Transpl 1997;12:1595–1599.

195. Barker DF, Hostikka SL, Zhou J, Chow LT, Oliphant AR,Gerken SC, et al. Identification of mutations in the COL4A5collagen gene in Alport syndrome. Science 1990;248:1224–1227.

196. Buzza M, Wang YY, Dagher H, Babon JJ, Cotton RG, Pow-ell H, et al. COL4A4 mutation in thin basement membranedisease previously described in Alport syndrome. Kidney Int2001;60:480–483.

197. Wang YY, Rana K, Tonna S, Lin T, Sin L, Savige J. COL4A3mutations and their clinical consequences in thin basementmembrane nephropathy (TBMN). Kidney Int 2004;65:786–790.

198. Tonna S, Wang YY, Wilson D, Rigby L, Tabone T, Cotton R,et al. The R229Q mutation in NPHS2 may predispose toproteinuria in thin-basement-membrane nephropathy. PediatrNephrol 2008;23:2201–2207.

199. Zhang C, Zhu S, Zhang Y. [X-linked thin basement membranenephropathy was linked with COL4A5 gene]. Zhonghua Yi XueZa Zhi 2000;80:582–584.

200. Longo I, Porcedda P, Mari F, Giachino D, Meloni I, Deplano C,et al. COL4A3 /COL4A4 mutations: from familial hematuria toautosomal-dominant or recessive Alport syndrome. Kidney Int2002;61:1947–1956.

201. Lenkkeri U, Mannikko M, McCready P, Lamerdin J, Gribou-val O, Niaudet PM, et al. Structure of the gene for congenitalnephrotic syndrome of the Finnish type (NPHS1 ) and character-ization of mutations. Am J Hum Genet 1999;64:51–61.

202. Matejas V, Al Gazali L, Amirlak I, Zenker M. A syndromecomprising childhood-onset glomerular kidney disease and ocularabnormalities with progressive loss of vision is caused by mutatedLAMB2 . Nephrol Dial Transpl 2006;21:3283–3286.

203. Adachi M, Asakura Y, Sato Y, Tajima T, Nakajima T, YamamotoT, et al. Novel SLC12A1 (NKCC2 ) mutations in two familieswith Bartter syndrome type 1. Endocr J 2007;54:1003–1007.

204. Kunzelmann K, Hubner M, Vollmer M, Ruf R, Hildebrandt F,Greger R, et al. A Bartter’s syndrome mutation of ROMK1 exertsdominant negative effects on K+ conductance. Cell PhysiolBiochem 2000;10:117–124.

205. Birkenhager R, Otto E, Schurmann MJ, Vollmer M, Ruf EM,Maier-Lutz I, et al. Mutation of BSND causes Bartter syndromewith sensorineural deafness and kidney failure. Nat Genet2001;29:310–314.

206. Schlingmann KP, Konrad M, Jeck N, Waldegger P, Reinal-ter SC, Holder M, et al. Salt wasting and deafness result-ing from mutations in two chloride channels. N Engl J Med2004;350:1314–1319.

207. Konrad M, Vollmer M, Lemmink HH, van den Heuvel LP,Jeck N, Vargas-Poussou R, et al. Mutations in the chloridechannel gene CLCNKB as a cause of classic Bartter syndrome.J Am Soc Nephrol 2000;11:1449–1459.

208. Bruce LJ, Cope DL, Jones GK, Schofield AE, Burley M, PoveyS, et al. Familial distal renal tubular acidosis is associated withmutations in the red cell anion exchanger (Band 3, AE1 ) gene.J Clin Invest 1997;100:1693–1707.

209. Xu J, Song P, Nakamura S, Miller M, Barone S, Alper SL, et al.Deletion of the chloride transporter Slc26a7 causes distal renaltubular acidosis and impairs gastric acid secretion. J Biol Chem2009;[Epub ahead of print].

210. Smith AN, Skaug J, Choate KA, Nayir A, Bakkaloglu A,Ozen S, et al. Mutations in ATP6N1B, encoding a new kidneyvacuolar proton pump 116 kD subunit, cause recessive distalrenal tubular acidosis with preserved hearing. Nat Genet2000;26:71–75.

211. Stover EH, Borthwick KJ, Bavalia C, Eady N, Fritz DM,Rungroj N, et al. Novel ATP6V1B1 and ATP6V0A4 mutationsin autosomal recessive distal renal tubular acidosis with newevidence for hearing loss. J Med Genet 2002;39:796–803.

212. Simon DB, Nelson-Williams C, Bia MJ, Ellison D, Karet FE,Molina AM, et al. Gitelman’s variant of Bartter’s syndrome,inherited hypokalaemic alkalosis, is caused by mutationsin the thiazide-sensitive Na-Cl cotransporter. Nat Genet1996;12:24–30.

213. Jeunemaitre X, Bassilana F, Persu A, Dumont C, Champigny G,Lazdunski M, et al. Genotype–phenotype analysis of a newlydiscovered family with Liddle’s syndrome. J Hypertens1997;15:1091–1100.

214. Hiltunen TP, Hannila-Handelberg T, Petajaniemi N, Kantola I,Tikkanen I, Virtamo J, et al. Liddle’s syndrome associated with apoint mutation in the extracellular domain of the epithelial sodiumchannel γ -subunit. J Hypertens 2002;20:2383–2390.

215. Wildin RS, Antush MJ, Bennett RL, Schoof JM, Scott CR.Heterogeneous AVPR2 gene mutations in congenital nephrogenicdiabetes insipidus. Am J Hum Genet 1994;55:266–277.

216. van Lieburg AF, Verdijk MA, Knoers VV, van Essen AJ,Proesmans W, Mallmann R, et al. Patients with autosomalnephrogenic diabetes insipidus homozygous for mutationsin the aquaporin 2 water-channel gene. Am J Hum Genet1994;55:648–652.

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 17: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

214 AJ McKnight et al

217. Mochizuki T, Wu G, Hayashi T, Xenophontos SL, Veldhuisen B,Saris JJ, et al. PKD2, a gene for polycystic kidney dis-ease that encodes an integral membrane protein. Science1996;272:1339–1342.

218. Norby S, Schwartz M. Possible locus for polycystic kidneydisease on chromosome 2. Lancet 1990;336:323–324.

219. Paterson AD, Pei Y. Is there a third gene for autosomal dominantpolycystic kidney disease? Kidney Int 1998;54:1759–1761.

220. Ward CJ, Hogan MC, Rossetti S, Walker D, Sneddon T, WangX, et al. The gene mutated in autosomal recessive polycystickidney disease encodes a large, receptor-like protein. Nat Genet2002;30:259–269.

221. Kyttala M, Tallila J, Salonen R, Kopra O, Kohlschmidt N,Paavola-Sakki P, et al. MKS1, encoding a component of theflagellar apparatus basal body proteome, is mutated in Meckelsyndrome. Nat Genet 2006;38:155–157.

222. Roume J, Genin E, Cormier-Daire V, Ma HW, Mehaye B,Attie T, et al. A gene for Meckel syndrome maps to chromosome11q13. Am J Hum Genet 1998;63:1095–1101.

223. Smith UM, Consugar M, Tee LJ, McKee BM, Maina EN,Whelan S, et al. The transmembrane protein meckelin (MKS3)is mutated in Meckel–Gruber syndrome and the wpk rat. NatGenet 2006;38:191–196.

224. Wolf MT, Mucha BE, Hennies HC, Attanasio M, Panther F,Zalewski I, et al. Medullary cystic kidney disease type 1:mutational analysis in 37 genes based on haplotype sharing. HumGenet 2006;119:649–658.

225. Konrad M, Saunier S, Calado J, Gubler MC, Broyer M,Antignac C. Familial juvenile nephronophthisis. J Mol Med1998;76:310–316.

226. Otto EA, Schermer B, Obara T, O’Toole JF, Hiller KS,Mueller AM, et al. Mutations in INVS encoding inversin causenephronophthisis type 2, linking renal cystic disease to thefunction of primary cilia and left–right axis determination. NatGenet 2003;34:413–420.

227. Hoefele J, Sudbrak R, Reinhardt R, Lehrack S, Hennig S,Imm A, et al. Mutational analysis of the NPHP4 gene in 250patients with nephronophthisis. Hum Mutat 2005;25:411.

228. Sayer JA, Otto EA, O’Toole JF, Nurnberg G, Kennedy MA,Becker C, et al. The centrosomal protein nephrocystin-6 ismutated in Joubert syndrome and activates transcription factorATF4. Nat Genet 2006;38:674–681.

229. Attanasio M, Uhlenhaut NH, Sousa VH, O’Toole JF, Otto E,Anlag K, et al. Loss of GLIS2 causes nephronophthisis inhumans and mice by increased apoptosis and fibrosis. Nat Genet2007;39:1018–1024.

230. Dinour D, Davidovitz M, Levin-Iaina N, Lotan D, Cleper R,Weissman I, et al. Truncating mutations in the chloride/protonClC-5 antiporter gene in seven Jewish Israeli families with Dent’s1 disease. Nephron Clin Pract 2009;112:c262–267.

231. Otto EA, Trapp ML, Schultheiss UT, Helou J, Quarmby LM,Hildebrandt F. NEK8 mutations affect ciliary and centrosomallocalization and may cause nephronophthisis. J Am Soc Nephrol2008;19:587–592.

232. Wolf MT, Saunier S, O’Toole JF, Wanner N, Groshong T,Attanasio M, et al. Mutational analysis of the RPGRIP1L genein patients with Joubert syndrome and nephronophthisis. KidneyInt 2007;72:1520–1526.

233. Otto EA, Tory K, Attanasio M, Zhou W, Chaki M, Paruchuri Y,et al. Hypomorphic mutations in meckelin (MKS3 /TMEM67 )cause nephronophthisis with liver fibrosis (NPHP11 ). J MedGenet 2009;[Epub ahead of print].

234. Utsch B, Sayer JA, Attanasio M, Pereira RR, Eccles M, Hen-nies HC, et al. Identification of the first AHI1 gene mutations innephronophthisis-associated Joubert syndrome. Pediatr Nephrol2006;21:32–35.

235. Kottgen A, Kao WH, Hwang SJ, Boerwinkle E, Yang Q, LevyD, et al. Genome-wide association study for renal traits in theFramingham Heart and Atherosclerosis Risk in CommunitiesStudies. BMC Med Genet 2008;9:49.

236. Greene CN, Keong LM, Cordovado SK, Mueller PW. Sequencevariants in the PLEKHH2 region are associated with diabetic

nephropathy in the GoKinD study population. Hum Genet2008;124:255–262.

237. Schelling JR, Abboud HE, Nicholas SB, Pahl MV, Sedor JR,Adler SG, et al. Genome-wide scan for estimated glomerularfiltration rate in multi-ethnic diabetic populations: the FamilyInvestigation of Nephropathy and Diabetes (FIND). Diabetes2008;57:235–243.

238. Maeda S, Osawa N, Hayashi T, Tsukada S, Kobayashi M,Kikkawa R. Genetic variations associated with diabetic nephropa-thy and type II diabetes in a Japanese population. Kidney Int Suppl2007;106:S43–48.

239. Yu H, Sale M, Rich SS, Spray BJ, Roh BH, Bowden DW, et al.Evaluation of markers on human chromosome 10, including thehomologue of the rodent Rf-1 gene, for linkage to ESRD in blackpatients. Am J Kidney Dis 1999;33:294–300.

240. Nishiyama K, Tanaka Y, Nakajima K, Mokubo A, Atsumi Y,Matsuoka K, et al. Polymorphism of the solute carrier family12 (sodium/chloride transporters) member 3, SLC12A3 geneat exon 23 (+78G/A: Arg913Gln) is associated with elevationof urinary albumin excretion in Japanese patients with type 2diabetes: a 10-year longitudinal study. Diabetologia 2005;48:1335–1338.

241. Capria M, Andreucci M, Fuiano L, Mancuso D, Cianfrone P,Comi N, et al. Double nucleotidic mutation of the MYH9 genein a young patient with end-stage renal disease. Nephrol DialTranspl 2004;19:249–251.

242. Hwang SJ, Yang Q, Meigs JB, Pearce EN, Fox CS. A genome-wide association for kidney function and endocrine-related traitsin the NHLBI’s Framingham Heart Study. BMC Med Genet2007;8(suppl 1):S10.

243. DeWan AT, Arnett DK, Atwood LD, Province MA, Lewis CE,et al. A genome scan for renal function among hypertensives:the HyperGEN study. Am J Hum Genet 2001;68:136–144.

244. DeWan AT, Arnett DK, Miller MB, Peacock JM, Atwood LD,Province MA, et al. Refined mapping of suggestive linkage torenal function in African Americans: the HyperGEN study. AmJ Hum Genet 2002;71:204–205.

245. Doring A, Gieger C, Mehta D, Gohlke H, Prokisch H, Coassin S,et al. SLC2A9 influences uric acid concentrations with pro-nounced sex-specific effects. Nat Genet 2008;40:430–436.

246. Vitart V, Rudan I, Hayward C, Gray NK, Floyd J, Palmer CN,et al. SLC2A9 is a newly identified urate transporter influencingserum urate concentration, urate excretion and gout. Nat Genet2008;40:437–442.

247. Wallace C, Newhouse SJ, Braund P, Zhang F, Tobin M,Falchi M, et al. Genome-wide association study identifies genesfor biomarkers of cardiovascular disease: serum urate anddyslipidemia. Am J Hum Genet 2008;82:139–149.

248. Li S, Sanna S, Maschio A, Busonero F, Usala G, Mulas A, et al.The GLUT9 gene is associated with serum uric acid levels inSardinia and Chianti cohorts. PLoS Genet 2007;3:e194.

249. McArdle PF, Parsa A, Chang YP, Weir MR, O’Connell JR,Mitchell BD, et al. Association of a common nonsynonymousvariant in GLUT9 with serum uric acid levels in old order Amish.Arthritis Rheum 2008;58:2874–2881.

250. Bates JS, Lessard CJ, Leon JM, Nguyen T, Battiest LJ,Rodgers J, et al. Meta-analysis and imputation identifies a109 kb risk haplotype spanning TNFAIP3 associated withlupus nephritis and hematologic manifestations. Genes Immunol2009;10:470–477.

251. Yuan H, Pan HF, Li LH, Feng JB, Li WX, Li XP, et al.Meta analysis on the association between Fcγ RIIa–R/H131polymorphisms and systemic lupus erythematosus. Mol Biol Rep2009;36:1053–1058.

252. Karassa FB, Trikalinos TA, Ioannidis JP. Role of the Fcγreceptor IIa polymorphism in susceptibility to systemic lupuserythematosus and lupus nephritis: a meta-analysis. ArthritisRheum 2002;46:1563–1571.

253. Karassa FB, Bijl M, Davies KA, Kallenberg CG, Khamashta MA,Manger K, et al. Role of the Fcγ receptor IIA polymorphism inthe antiphospholipid syndrome: an international meta-analysis.Arthritis Rheum 2003;48:1930–1938.

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 18: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

Genetics of renal disease 215

254. Thakkinstian A, Dmitrienko S, Gerbase-Delima M, McDanielDO, Inigo P, Chow KM, et al. Association between cytokinegene polymorphisms and outcomes in renal transplantation: ameta-analysis of individual patient data. Nephrol Dial Transpl2008;23:3017–3023.

255. Pereira TV, Nunes AC, Rudnicki M, Magistroni R, Alber-tazzi A, Pereira AC, et al. Influence of ACE I/D gene polymor-phism in the progression of renal failure in autosomal dominantpolycystic kidney disease: a meta-analysis. Nephrol Dial Transpl2006;21:3155–3163.

256. Yong D, QingQing W, Hua L, Yang LX, QingLing Z, Ying H,et al. Association of uteroglobin G38A polymorphism withIgA nephropathy: a meta-analysis. Am J Kidney Dis 2006;48:1–7.

257. Wiwanitkit V. Angiotensin-converting enzyme gene polymor-phism is correlated to the progression of disease in patientswith IgA nephropathy: a meta-analysis. Ren Fail 2006;28:697–699.

258. Yong D, Qing WQ, Hua L, Kan JJ, Xi CJ, Jin QQ, et al. Associ-ation of angiotensin I-converting enzyme gene insertion/deletionpolymorphism and IgA nephropathy: a meta-analysis. Am JNephrol 2006;26:511–518.

259. Schena FP, D’Altri C, Cerullo G, Manno C, and Gesualdo L.ACE gene polymorphism and IgA nephropathy: an ethni-cally homogeneous study and a meta-analysis. Kidney Int2001;60:732–740.

260. Xu M, Chen X, Yan L, Cheng H, Chen W. Association between(AC)n dinucleotide repeat polymorphism at the 5′-end of thealdose reductase gene and diabetic nephropathy: a meta-analysis.J Mol Endocrinol 2008;40:243–251.

261. Neamat-Allah M, Feeney SA, Savage DA, Maxwell AP, Han-son RL, Knowler WC, et al. Analysis of the association betweendiabetic nephropathy and polymorphisms in the aldose reduc-tase gene in Type 1 and Type 2 diabetes mellitus. Diabet Med2001;18:906–914.

262. Zintzaras E, Uhlig K, Koukoulis GN, Papathanasiou AA, Ste-fanidis I. Methylenetetrahydrofolate reductase gene polymor-phism as a risk factor for diabetic nephropathy: a meta-analysis.J Hum Genet 2007;52:881–890.

263. Zintzaras E, Stefanidis I. Association between the GLUT1 genepolymorphism and the risk of diabetic nephropathy: a meta-analysis. J Hum Genet 2005;50:84–91.

264. Ng DP, Tai BC, Lim XL. Is the presence of retinopathy ofpractical value in defining cases of diabetic nephropathy ingenetic association studies? The experience with the ACEinsertion/deletion polymorphism in 53 studies comprising 17 791subjects. Diabetes 2008;57:2541–2546.

265. Ng DP, Tai BC, Koh D, Tan KW, Chia KS. Angiotensin-Iconverting enzyme insertion/deletion polymorphism and itsassociation with diabetic nephropathy: a meta-analysis of studiesreported between 1994 and 2004 and comprising 14 727 subjects.Diabetologia 2005;48:1008–1016.

266. Kennon B, Petrie JR, Small M, Connell JM. Angiotensin-converting enzyme gene and diabetes mellitus. Diabet Med1999;16:448–458.

267. Tarnow L, Parving HH, Jacobsen P, Rossing P, LecerfL, Poirier O, et al. [The significance of deletion poly-morphism in the ACE gene for progression of diabeticnephropathies treated with ACE inhibitors]. Ugeskr Laeger1998;160:4886–4889.

268. Fujisawa T, Ikegami H, Kawaguchi Y, Hamada Y, Ueda H,Shintani M, et al. Meta-analysis of association of inser-tion/deletion polymorphism of angiotensin I-converting enzymegene with diabetic nephropathy and retinopathy. Diabetologia1998;41:47–53.

269. Staessen JA, Wang JG, Ginocchio G, Petrov V, Saavedra AP,Soubrier F, et al. The deletion/insertion polymorphism of theangiotensin converting enzyme gene and cardiovascular–renalrisk. J Hypertens 1997;15:1579–1592.

270. Parving HH, Tarnow L, Rossing P. Genetics of diabeticnephropathy. J Am Soc Nephrol 1996;7:2509–2517.

271. * Prakash S, Chung KW, Sinha S, Barmada M, Ellis D,Ferrell RE, et al. Autosomal dominant progressive nephropathywith deafness: linkage to a new locus on chromosome 11q24.J Am Soc Nephrol 2003;14:1794–1803.

272. * Arar NH, Voruganti VS, Nath SD, Thameem F, Bauer R,Cole SA, et al. A genome-wide search for linkage to chronickidney disease in a community-based sample: the SAFHS.Nephrol Dial Transpl 2008;23:3184–3191.

273. * Sale MM, Freedman BI, Langefeld CD, Williams AH, HicksPJ, Colicigno CJ, et al. A genome-wide scan for type 2 diabetesin African-American families reveals evidence for a locus onchromosome 6q. Diabetes 2004;53:830–837.

274. * Freedman BI, Bowden DW, Rich SS, Valis CJ, Sale MM,Hicks PJ, et al. A genome scan for all-cause end-stagerenal disease in African Americans. Nephrol Dial Transpl2005;20:712–718.

275. * Neary JJ, Conlon PJ, Croke D, Dorman A, Keogan M,Zhang FY, et al. Linkage of a gene causing familial membra-noproliferative glomerulonephritis type III to chromosome 1.J Am Soc Nephrol 2002;13:2052–2057.

276. * Mathis BJ, Kim SH, Calabrese K, Haas M, Seidman JG,Seidman CE, et al. A locus for inherited focal segmentalglomerulosclerosis maps to chromosome 19q13. Kidney Int1998;53:282–286.

277. * Gharavi AG, Yan Y, Scolari F, Schena FP, Frasca GM,Ghiggeri GM, et al. IgA nephropathy, the most commoncause of glomerulonephritis, is linked to 6q22–23. Nat Genet2000;26:354–357.

278. * Bisceglia L, Cerullo G, Forabosco P, Torres DD, Scolari F, DiPerna M, et al. Genetic heterogeneity in Italian families with IgAnephropathy: suggestive linkage for two novel IgA nephropathyloci. Am J Hum Genet 2006;79:1130–1134.

279. * Paterson AD, Liu XQ, Wang K, Magistroni R, SongX, Kappel J, et al. Genome-wide linkage scan of alarge family with IgA nephropathy localizes a novelsusceptibility locus to chromosome 2q36. J Am Soc Nephrol2007;18:2408–2415.

280. * Freedman BI, Bowden DW, Rich SS, Xu J, Wagenknecht LE,Ziegler J, et al. Genome-wide linkage scans for renal functionand albuminuria in Type 2 diabetes mellitus: the Diabetes HeartStudy. Diabet Med 2008;25:268–276.

281. * Mottl AK, Vupputuri S, Cole SA, Almasy L, Goring HH,Diego VP, et al. Linkage analysis of glomerular filtration ratein American Indians. Kidney Int 2008;74:1185–1191.

282. * Leon JM, Freedman BI, Miller MB, North KE, Hunt SC,Eckfeldt JH, et al. Genome scan of glomerular filtration rateand albuminuria: the HyperGEN study. Nephrol Dial Transpl2007;22:763–771.

283. * Puppala S, Arya R, Thameem F, Arar NH, Bhandari K,Lehman DM, et al. Genotype by diabetes interaction effectson the detection of linkage of glomerular filtration rate to aregion on chromosome 2q in Mexican Americans. Diabetes2007;56:2818–2828.

284. * Turner ST, Kardia SL, Mosley TH, Rule AD, Boerwinkle E,de Andrade M. Influence of genomic loci on measures of chronickidney disease in hypertensive sibships. J Am Soc Nephrol2006;17:2048–2055.

285. * Fox CS, Yang Q, Cupples LA, Guo CY, Larson MG,Leip EP, et al. Genomewide linkage analysis to serumcreatinine, GFR, and creatinine clearance in a community-basedpopulation: the Framingham Heart Study. J Am Soc Nephrol2004;15:2457–2461.

286. * Hunt SC, Coon H, Hasstedt SJ, Cawthon RM, Camp NJ,Wu LL, et al. Linkage of serum creatinine and glomerularfiltration rate to chromosome 2 in Utah pedigrees. Am J Hypertens2004;17:511–515.

287. * DeWan AT, Arnett DK, Atwood LD, Province MA, Lewis CE,Hunt SC, et al. A genome scan for renal function amonghypertensives: the HyperGEN study. Am J Hum Genet2001;68:136–144.

288. * Krolewski AS, Poznik GD, Placha G, Canani L, Dunn J,Walker W, et al. A genome-wide linkage scan for genes

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Page 19: Unravelling the genetic basis of renal diseases; from single gene to multifactorial disorders

216 AJ McKnight et al

controlling variation in urinary albumin excretion in type IIdiabetes. Kidney Int 2006;69:129–136.

289. * Arar N, Nath S, Thameem F, Bauer R, Voruganti S, ComuzzieA, et al. Genome-wide scans for microalbuminuria in Mexican

Americans: the San Antonio Family Heart Study. Genet Med2007;9:80–87.

∗ References cited in supporting information online only.

SUPPORTING INFORMATION ON THE INTERNET

The following supporting information may be found in the online version of this article:

Table S1. Chromosomal regions and associated genes for common forms of renal disease identified fromgenome-wide studies.

J Pathol 2010; 220: 198–216 DOI: 10.1002/pathCopyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.