DNA methylation profiling identifies epigenetic differences between ...

85 downloads 0 Views 2MB Size Report
May 7, 2010 - Personalized medicine has been defined as “the application of genomic and molecular data to better target the delivery of health care, facilitate ...
BRIEF REPORT

Epigenetics 6:1, 20-28; January 2011; © 2011 Landes Bioscience

DNA methylation profiling identifies epigenetic differences between diabetes patients with ESRD and diabetes patients without nephropathy Carmen Sapienza,1,2,* Jean Lee,3 Jasmine Powell,1 Oluwatoyin Erinle,1 Faahud Yafai,3 James Reichert,3 Elias S. Siraj3 and Michael Madaio4 Fels Institute for Cancer Research and Molecular Biology; Departments of 2Pathology and Laboratory Medicine; 3Medicine; Temple University Medical School; Philadelphia, PA USA; 4Department of Medicine; Medical College of Georgia; Augusta, GA USA

1

Key words: DNA methylation, end-stage renal disease, diabetic nephropathy, dialysis

We identified potential epigenetic biomarkers for chronic kidney disease progression by comparing site-specific DNA methylation levels in more than 14,000 genes between African American and Hispanic diabetes patients with end stage renal disease (ESRD) and diabetes patients without nephropathy. We identified 187 genes that are differentially methylated between the two groups on at least two CpG sites in each gene in DNA extracted from saliva. Of the 187 genes whose mean methylation levels differed between the two groups, 39 genes or closely related gene family members, have been reported to be involved in kidney development or diabetic nephropathy, per se, or have been associated with dialysis-induced changes in gene expression in peripheral blood cells. The fact that such a substantial fraction (21%) of the 187 candidate genes have been implicated previously through genome association or transcription profiling studies suggests strongly that the DNA methylation differences we observe are associated with disease predisposition and/ or treatment. The fact that these nephropathy and/or dialysis-associated differences between patients were identified in DNA extracted from saliva offers proof-of-principle that inter-individual epigenetic differences may prove useful as predictive biomarkers of disease susceptibility.

©201 1L andesBi os c i enc e. Donotdi s t r i but e. Introduction

Personalized medicine has been defined as “the application of genomic and molecular data to better target the delivery of health care, facilitate the discovery and clinical testing of new products, and help determine a person’s predisposition to a particular disease or condition.” 1 While there have been some notable successes in using genetic data to tailor treatment of disease, particularly in the area of cancer genetics, the utility of genetic information to stratify individuals according to disease predisposition, especially predisposition to common disease, has been limited. The limited utility of genetic information for predicting an individual’s risk for type 2 diabetes, for example, stems from the fact that multiple loci are involved,2,3 the relative risk associated with carrying any particular disease-predisposing allele is small3 and the influence of environmental factors is large.2,3 In fact, it has been argued that for many common diseases, precise genotype information adds no greater predictive power than information on family history alone. If the goals of personalized medicine are to be advanced, scientists and physicians must find ways to add significant predictive value to genotype information. Molecular profiling technologies,

such as proteomic profiling, metabolomic profiling and epigenetic profiling, offer the most likely options for obtaining additional relevant biomarker information. One difficulty with the use of such technologies is that, unlike genotype, which does not vary between the tissues of an individual, an individual’s proteomic, metabolomic or epigenetic profiles may differ between different tissues. The identification of biomarkers that have predictive value in disease predisposition requires that the biomarkers give information about the likelihood of pathology in a tissue that may not be available for analysis by assaying a tissue that can be accessed in a minimally invasive way. Biomarkers that predict the risk of dementia or kidney failure, for example, will only be useful clinically if they can be assayed by non-invasive techniques or in tissues such as blood or saliva, rather than brain and kidney. Diabetic nephropathy is a common complication of type 1 and type 2 diabetes and familial aggregation in multiple populations suggests that susceptibility is, in part, genetic.4-6 Linkage studies and association studies have identified candidate chromosomal regions and candidate genes that explain a small fraction of the nephropathy trait variance, but genes with major effect have not been identified.4-6 In this study, we assessed whether quantitative differences in CpG site-specific methylation in DNA

*Correspondence to: Carmen Sapienza; Email: [email protected] Submitted: 05/07/10; Accepted: 08/17/10 Previously published online: www.landesbioscience.com/journals/epigenetics/article/13362 DOI: 10.4161/epi.6.1.13362 20

Epigenetics

Volume 6 Issue 1

BRIEF REPORT

BRIEF REPORT

Table 1. Characteristics of diabetes patients without nephropathy [>10 years of diabetes, estimated glomerular filtration rate >60 (by MDRDS formula) and urine microalbumin 10 years, Table 1; henceforth called “patients without nephropathy”) and 24 diabetes patients with ESRD who were being treated by hemodialysis (Table 2; henceforth called “ESRD patients on HD”) using DNA extracted from saliva of each patient. We used the Illumina HumanMethylation27 BeadChip array to assay the fraction of molecules in each sample that was methylated at each of 27,578 CpG sites—the “b-value,” which may range from 0–1 (www.illumina.com/documents/icommunity/article_2008_05_infinium_methylation.pdf). One African American ESRD on HD patient’s array results were of insufficient quality (as judged by the fraction of array CpGs detected, see Methods), and this sample and the sample of one African

American patient without nephropathy (a case-matched female with the lowest fraction of CpGs detected among the patients without nephropathy) were excluded from the analysis, leaving 23 individuals (21 African Americans and two Hispanics) in each group. Patients with diabetic nephropathy do not cluster separately by epigenotype but ~10% of CpG sites are differentially methylated. Unsupervised hierarchical cluster analysis using b-values for all CpGs did not cluster ESRD patients on HD separately from diabetic patients without nephropathy (Sup. Fig. 1). However, the clustering of most females separately from most males (Sup. Fig. 1) argues for robust detection of interindividual methylation differences between males and females, most likely at genes on the X chromosome (see Methods). When the 1,092 CpGs present on the X and Y chromosomes are removed from the analysis (Fig. 1), the ESRD on HD patients cluster somewhat better, in that eight of the 11 patients on the deepest branches (i.e., greatest differences from the rest of the group) are from the ESRD on HD group. However, as there is significant intermingling of patient groups within branches, we compared mean beta-values at each CpG between the 23 ESRD patients on HD and the 23 patients without nephropathy. A significant difference (a “DiffScore” of +20; see Methods)

www.landesbioscience.com Epigenetics

21

between mean beta-values in each group was detected for 2,870 CpGs (Sup. Table 1). Remarkably, 2,693 of these CpGs exhibited lower mean methylation levels in the ESRD on HD patients while only 177 CpGs were methylated at higher mean values in the dialysis patients. Epigenetic differences identify candidate genes related to candidate genes identified on the basis of genetic differences. Many of the CpG sites exhibiting significant methylation differences between the groups are found in genes that have been associated with diabetic nephropathy in more than one study (i.e., TIMP3, BCL2; Sup. Table 1). Because we have assayed methylation differences in only a small number of patients, we applied the additional more robust selection criterion that candidate genes must have significant differences in methylation at two or more CpG sites within the same gene in order to reduce the number of false-positive candidates. When this selection was applied, 389 CpGs in 187 genes [n.b.: using the “2 significant CpGs in the same gene” selection criterion and a significant “DiffScore” of +20 (see Methods for relationship to p value), we expect only five to six false positives in a scan of this size] were methylated significantly differently between the two groups (Table 3). Of the 187 genes in Table 3, 156 (83%) are assayed at only two CpGs on the array and both differ significantly between groups. Twenty-three of the remaining 32 genes have more than three CpGs assayed on the array but only two are significantly different. The remaining nine genes all have more than three CpGs that differ significantly. Twenty-one of the genes in Table 3 or related gene family members (Table 4), are among the 115 genes tested for linkage or association with diabetic nephropathy by Ewens et al.6 Although our analysis is not based on known differences in genotype between the two groups, it is interesting that a significant fraction of genes or like-gene family members, appear to exhibit epigenetic differences between the two groups (Table 4). This observation suggests that disease progression may be influenced by genetic and/or epigenetic differences in genes that operate in closely related or parallel pathways. Among the most interesting candidate genes (Table 4) are myosin light chain 9 (MYL9), matrix metalloprotease 10 (MMP10), tissue inhibitor of metalloprotease 4 (TIMP4) and methylenetetrahydrofolate reductase (MTHFR). Genetic variants in myosin heavy chain 9 (MYH9) have been reported to explain a significant fraction of focal glomerulosclerosis in African Americans,7 who also make up the majority of the patient population examined in this study. It is interesting that genetic variation in MYH9 is associated with nephropathy caused by hypertension8 but does not appear to be associated with diabetic nephropathy.9 Our identification of an association between epigenetic variation in a regulatory MYL9 and diabetic nephropathy is highly suggestive that similar pathways may operate in both diabetic and hypertensive nephropathy but that the defects involve different parts of the pathway. MMP10 is part of a cluster of nine matrix metalloprotease genes spread over approximately 500 kb on chromosome 11q (www.ensembl.org/Homo_sapiens/Location/View?g=ENSG

Table 2. Characteristics of ESRD patients on HD Sex

Age

Ethnicity

Type of diabetes

female

55

African American

2

female

57

African American

2

female

59

African American

2

female

63

African American

2

female

67

African American

2

female

71

African American

2

female

72

African American

2

female

72

African American

2

female

79

African American

2

female

80

African American

2

female

87

African American

2

female

38

African American

2

female

61

African American

2

male

41

African American

2

male

50

African American

2

male

54

African American

2

male

58

Hispanic

2

male

61

African American

2

male

61

Hispanic

2

male

63

African American

2

male

64

African American

2

male

66

African American

2

male

70

African American

2

male

74

African American

2

©201 1L andesBi os c i enc e. Donotdi s t r i but e.

22

00000166670). Another member of this group, MMP20, has been associated with human kidney aging and age-related decline in glomerular filtration rate.10 Regulation of matrix metalloproteases is likely to be an important factor in response to renal injury (reviewed in ref. 11) and ESRD patients on HD have significant kidney fibrosis. In this regard, the identification of epigenetic variation in both a matrix metalloprotease gene and a matrix metalloprotease inhibitor gene (TIMP4) may be significant. We also believe it is significant that MTHFR is identified as a differentially methylated gene in ESRD patients on HD. MTHFR is a central node in the one-carbon metabolism pathway,12,13 responsible for generating methyl groups for all cellular processes, including DNA methylation. Stenvinkel et al.14 have found that overall levels of DNA methylation are increased in patients with chronic kidney disease and inflammation (a secondary consequence of dialysis) and five of the six CpGs assayed in MTHFR are significantly less methylated in dialysis patients (Sup. Table 1), suggesting upregulation of this gene. The degree to which the ESRD patients on HD and the patients without nephropathy differ in methylation at two sites within each gene is illustrated for four representative genes in Figure 2; ESRD patients on HD have lower mean methylation in three of the genes (MMP10, MTFR and TIMP4; Figs. 2A–C, respectively) and higher mean methylation in one of the genes (CLIC6; Fig. 2D).

Epigenetics

Volume 6 Issue 1

©201 1L andesBi os c i enc e. Donotdi s t r i but e.

Figure 1. Cluster diagram using Pearson correlation and the 26,486 CpG probes that are not located on the X and Y chromosomes. All patients are African American except M ESRD on HD 19, M ESRD on HD 16, M PWON 12 and M PWON 3, who are Hispanic. All patients have type 2 diabetes except F PWON 19, F PWON 15 and M PWON 13, who have Type 1 diabetes. M ESRD on HD, male end stage renal disease on dialysis patient; F ESRD on HD, female end stage renal disease on dialysis patient; M PWON, male patient without nephropathy; F PWON, female patient without nephropathy.

Differentially methylated candidate genes are involved in pathways important in diabetic nephropathy and dialysis treatment. We also used the 187 ESRD/HD-associated candidate genes as the input core data for Ingenuity Pathway Analysis (www.ingenuity.com/) to determine whether specific metabolic pathways might be associated with the methylation differences in dialysis patients. Overall, this analysis suggests that dialysis patients have alterations in pathways involved in inflammation, oxidative stress, ubiquitination, fibrosis and drug metabolism (Table 5). These gene-specific methylation differences also

appear to occur non-randomly in genes involved in cancer (and we note that dialysis patients have a higher incidence of renal cell carcinoma; reviewed in ref. 15), as well as genes associated with cardiotoxicity, hepatotoxicity and nephrotoxicity (Table 5). Wilflingseder et al.16 have performed transcriptional profiling on peripheral blood from dialysis patients and found that more than 200 genes were upregulated in dialysis patients. Of the 187 candidate genes identified as differentially methylated in ESRD on HD patients (Table 3), 18 genes or closely related family members (Table 6) are among the genes identified as upregulated

www.landesbioscience.com Epigenetics

23

Table 3. Genes in which two or more CpGs are differentially methylated between ESRD patients on HD and diabetes patients without nephropathy ACSL6

APC

DLX3

JUN

ADAMTS10

CDKN1C

DNAJC18

AKR1B1

CFL1

DNM2

SLC2A4

KIAA0922

PCSK4

SLC4A3

KIAA1199

PDLIM3

SOAT2 SOCS1

ALDH3B2

CHPF

DTL

KIAA1838

PFKFB4

ANKRD15

CHRNB3

EDA2R

KIF1B

PFTK1

SOLH

ANXA13

CLIC3

EIF4B

KLK10

PHF2

SOSTDC1

ARHGAP22

CLIC6

EMG1

LAPTM4A

PHLDA2

SPINLW1

ARL4

CNAP1

EMX1

LGALS3BP

PHYH

SPINT2

ASPA

CRLF1

EPHA3

LMO4

PIP5KL1

SPRR2A

ATP10A

CSNK1E

FAM105A

LOC284837

PKNOX1

SPRY4

ATP5I

CTBP1

FBXO32

LRRC3

PLCL1

SRF

BAIAP2

CTSZ

FBXL16

LYST

POLE

STK6

BIK

CYBASC3

FEN1

MAF1

POLR2F

STMN1

BOC

CYLN2

FGFRL1

MAP3K3

POLR3D

SURF5

BPHL

CYP2S1

FHIT

MBNL3

POMGNT1

TCF15

BRD3

CYP8B1

FLJ12118

MCAM

PSME2

TFAP2E

BTNL2

DAAM1

FLJ21103

MEST

PTGER3

TIMP4 TMEM79

C1orf142

DAB2IP

FLJ32569

MKNK2

RAP1B

C1orf149

CMTM2

FZD2

MMP10

RARRES2

TNXB

C20orf100

COX8C

GAS1

MSX1

RASSF1

TSPAN14

GNAS

GLOXD1

MTHFR

RASSF5

TUBA6

HIST2H4

GMFB

MYL9

RBM14

TUSC4

UBE2J1

C20orf29 C5 C8orf34 C9orf127 C9orf26 CACNB3

©201 1L andesBi os c i enc e. Donotdi s t r i but e. HIST3H3

GRB10

MYST4

RBM35B

LILRB5

GSDML

NARG1

RGS19

UGDH

RB1

HM13

NDUFB7

RNF166

USMG5

ZIM2

IGF2AS

NIP

RPL7A

VKORC1

CBX7

ZPBP2

INHBA

NSD1

RPRML

WDR37

CCND1

DCAKD

ISYNA1

NTN4

RRAS2

WSB2

CCND2

DDX5

ITGB4

OVOL1

SFRS9

ZDHHC3

CDC16

DHRS4

JAG1

PCAF

SH2D3A

ZFP36L2

CDKL3

DHX9

JUB

SHB

ZNF688

DIRAS2

SLC1A1

more than 2-fold in dialysis patients.16 Seven of the genes identified by methylation profiling (in bold in Table 6) appear associated with both diabetic nephropathy (Table 4) and dialysis (Table 6). Discussion At first sight, our results appear to be in contrast to those of Stenvinkel et al.14 who found that overall levels of DNA methylation were higher in patients with chronic kidney disease and inflammation, whether they were being treated by dialysis or not. However, these investigators measured relative levels of methylation at HpaII sites (CCGG), genome-wide, by the “LUMA” assay.17 Although both CpG sites and HpaII sites are enriched in gene promoters, the majority of HpaII sites in the genome are present in repeated sequences, such as the Alu family of SINE elements. Any inflammation-based global or genome-wide

24

PCGF1

increases in methylation may be more likely to affect repeated sequences than the largely gene promoter-based CpGs assayed on the Illumina BeadChip used in our study. We also note that Wilflingseder et al.16 found that dialysis was associated, overwhelmingly, with upregulation of gene expression and we found that genes that were differentially methylated in ESRD patients on HD were significantly less methylated in more than 90% of cases. Gene promoter methylation is, in general, inversely correlated with transcript level18 so the overall results of our study and Wilflingseder et al.16 are in agreement. Unfortunately, we were unable to determine whether the between-group methylation differences we observed were associated with differences in transcript level between the two groups because appropriate samples were unavailable. However, we have shown previously19 that 25–40% of methylation differences between groups are associated with moderate cis effects on mean transcript level. Even if only a fraction of the methylation

Epigenetics

Volume 6 Issue 1

Table 4. Genes differentially methylated in ESRD on HD patients that have been implicated in diabetic nephropathy or kidney development/ disease3,9-11 Genes implicated in diabetic nephropathy or kidney disease AKR1B1

Genes associated with ESRD or HD by methylation profiling

Genes expressed in kidney, identified by methylation profiling

AKR1B1 ANKRD15

CTSD, CTSL

CTSZ

CYBA

CYBASC3, CYP2S1, CYP8B1, COX8C

ANKRD15

FZD2

FZD2

ITGA1, ITGA3

ITGB4

ITGB4

IGF1

IGF2AS

LGALS3

LGALS3BP

MMP1, MMP2, MMP3, MMP9

MMP10

MTHFR

MTHFR

MYH9

MYL9

SLC2A2, SLC12A3 TIMP2, TIMP3 UBA52

NTN4

NTN4

PTGER3

PTGER3

SLC1A1, SLC2A4, SLC4A3

33% (data not shown). Although only two tissues are compared in our study, the two tissues diverge early in development (extraembryonic vs. embryonic) and might be expected to differ greatly from one another. Although our estimate would be expected to rise by comparing additional tissues, both our estimate and that of Rakyan et al.18 suggest that the majority of CpG sites should be methylated similarly between most tissues in most individuals. With respect to their utility as biomarkers, the most important variable is not the level of variability between tissues within an individual but the level of variability in the same tissue between individuals of different disease state or disease predisposition. We note that when Benjamini-Hochberg correction for multiple testing is applied to our data set, the number of CpGs identified as significantly different falls dramatically. For instance, the number of CpGs that have a DiffScore of greater/less than 20 falls to 30, from 2,870 detected without correction. However, we were well aware that nearly 50% of the original 2,870 CpGs were likely false-positives (1,379 expected at p < 0.05), and chose to apply the additional criterion of identifying candidate genes by selecting only those with two or more CpGs significantly different. This “common sense” biological criterion resulted in the elimination of 87% of the original CpGs, leaving only 389 of the 2,870. As a measure of the relative difference between a strict statistical selection and the selection used to identify the genes in Table 3, if we take all Benjamini-Hochberg corrected CpGs with DiffScores of greater/less than 15, there are 212 CpGs that identify 30 of the same genes identified by the “2 CpGs in the same gene” procedure. We note, also, that our sample comparison groups contain mostly African American patients, with only a small number of Hispanic patients. If the Hispanic patients are removed from the analysis, under the possibility that they introduce additional variance into the data set because of genetic heterogeneity, a larger number of candidate genes in which two CpGs are significantly different is identified. However, 146 of the original 187 candidates genes (76%) and 2,359 of the 2,870 original significantly different CpGs (82%) are the same when the Hispanic patients are excluded from the analysis, arguing that our conclusions on the original data set are conservative. We hope to address these and other concerns in a larger study in which patient groups may be subdivided by appropriate clinical considerations (comparison of patients with diabetic nephropathy who are pre-dialysis with diabetic nephropathy dialysis patients, for example) and in which environmental factors are taken into consideration.

©201 1L andesBi os c i enc e. Donotdi s t r i but e. TIMP4

UBE2J1

differences that we detect are associated with differences in transcript level, the metabolic networks likely to be affected (Table 5) appear to be those that are involved in diabetic nephropathy and/ or causes or effects of dialysis. At this point, it is not possible to distinguish whether any particular difference we observe between patient-groups is caused by the ESRD state, per se, treatments associated with ESRD, or are predisposing factors that lead to ESRD. However, the fact that we observe methylation differences in genes that are involved in kidney disease progression, and that we observe these differences in non-renal tissues, suggests that either the patients who develop ESRD have constitutional epigenetic differences (similar to the constitutional “loss of imprinting” observed at IGF2/H19 in colon cancer patients)20 that may, themselves, have a genetic basis or that the differences are the result of environmental factors that alter epigenetic marks in a similar way in all tissues exposed. In this regard we note that although there is a common perception that CpG sites are differentially methylated between tissues, the fraction of CpG sites that are actually methylated differently appears modest. Rakyan et al.18 have estimated that only 18% of the ~25,000 genes analyzed in 16 different tissues show “tissue-specific” patterns of methylation, with between tissue differences of more than 33%.18 We have corroborated this result by comparing cord blood and placenta samples from 71 children on either an Illumina GoldenGate array (15) or the Illumina Infinium array we have used in this study. Of the 58,228 CpGs queried between the two tissues, only 11% varied by more than

Methods Saliva sample collection and DNA extraction. Diabetes patients with ESRD were recruited from the DCI and Fresenius Dialysis Clinics in Philadelphia and diabetes patients without nephropathy were recruited through the Department of Endocrinology at Temple University Medical School. Those who gave informed consent (Temple University IRB approved protocol 11061) to participate in the study were provided with an “Oragene DNA” saliva collection kit (OG-100) (www.dnagenotek.com/index.

www.landesbioscience.com Epigenetics

25

©201 1L andesBi os c i enc e. Donotdi s t r i but e. Figure 2. Each plot shows methylation levels at two CpGs in ESRD/HD-associated candidate genes MMP10 (A), MTHFR (B), TIMP4 (C) and CLIC6 (D) in all 46 patients. For MTHFR, the graph shows the two CpGs with the largest difference between patient groups.

html) into which the patients expectorated the required amount (~2 ml) of saliva. All interactions with patients conformed to the principles of the Declaration of Helsinki. DNA was prepared from saliva according to the manufacturer’s protocol. Sample preparation and array processing. 500 ng of each DNA sample was treated with sodium bisulfite using the EZ DNA Methylation-Gold Kit D5005 (Zymo Research, Orange, CA). Completeness of conversion was monitored by bisulfite pyrosequencing of 12 CpG sites in the imprinted SNRPN gene, using a commercially available assay (EpigenDx, Worcester, MA). “HumanMethylation27 BeadChip” arrays were purchased from Illumina, Inc., (San Diego, CA) and samples were processed at the University of Pennsylvania Microarray Facility. Data analysis. Data analysis was performed using the Illumina Genome Studio 2008.1 software. Greater than 23,000 of the 27,578 CpGs on the array were detected in all samples but one. This ESRD on HD sample and a diabetes patient without nephropathy, matched for sex and having the lowest number of CpGs detected in this group (24,849), were excluded from the analysis. Unsupervised hierarchical clustering using all CpGs clustered samples largely on the basis of sex (Sup. Fig. 1),

26

suggesting methylation differences on the inactive X chromosome in females was the largest influence. In fact, 728 of the 1,085 X-linked CpGs exhibited significant differences in mean beta-value between males and females, again arguing for high quality of sample preparation and array processing. The 1,085 CpGs on the X chromosome and 7 CpGs on the Y chromosome were then excluded from the cluster analysis to eliminate the effect of sex chromosome differences in methylation (Fig. 1). Patient group mean b-value methylation at each site was compared and “DiffScores” computed. We note that all analyses were performed using the background normalized b-values to correct for background differences between arrays; however, additional normalization procedures were not employed because beta values are fractional and based on ratios of signals between different probes on the same array and are constrained between 0 and 1. In addition, normalization procedures have the effect of discounting small but significant differences between groups on the basis that they are unlikely to be of biological significance. However, we have found that some small absolute differences in methylation are correlated with between group differences in transcript levels19 and we did not wish to bias our selection of genes as

Epigenetics

Volume 6 Issue 1

Table 5. Top gene functions of the 187 ESRD/HD-associated gene candidates in ingenuity pathway analysis IPA functional category

Genes that are differentially methylated between diabetes patients without nephropathy and ESRD patients on HD

Renal fibrosis, renal proliferation, renal dysplasia

STMN1, CDKN1C

Cardiac hypertrophy, cardiac hemorrhaging

FBXO32, IL33, JUN, TCF14, CTBP1, INHBA

Liver proliferation, liver fibrosis, liver necrosis/cell death, liver hematopoiesis, liver dysplasia

JUN, CCND1, RB1, C5, SOCS1

Cell cycle, developmental disorder, genetic disorder

APC, AURKA, CCND2, CDC16, CDKN1C, Cyclin A, Cyclin D, Cyclin E, DAB2IP, ERK, FEN1, GC-GCR dimer, GRB10, Growth hormone, HIST3H3 (includes EG:8290), Histone h3, Histone h4, INHBA, JAG1, JUB, KAT2B, MEF2, Mhc ii, MKNK2, NSD1, NTN4, PFTK1, POLE, Rar, Rb, RB1, RPL7A, SOCS1, SPINT2, SWI-SNF

Amino acid metabolism, small molecule biochemistry, cancer

Actin, Akt, ALP, ARHGAP22, BAIAP2, CFL1, CYP8B1, DHX9, DTL, Fgf, FSH, GNAS, hCG, ITGB4, JUN, MCAM, Mmp, MMP10, MTHFR, Pdgf, PDGF BB, PDLIM3, Pka, PLC, PP2A, SLC1A1, SPRR2A, SPRY4, STMN1, Tgf beta, TIMP4, TUBA1C, Tubulin, UGDH, Vegf

Connective tissue development and function, tissue development, cancer

ADCY, ALDH3B2 (includes EG:222), Ap1, APC, Ck2, CSNK1E, Ctbp, CTBP1, DDX5, EIF4B, FBXO32, FZD2, GAS1, Gsk3, HIST2H4A, IgG, IL1, IL12 (complex), Interferon a, Jnk, LMO4, MAP3K3, MED22, MIR1, P38 MAPK, PHLDA2, Pias, Pkc(s), POLR2F, POLR3D, Proteasome, PTGER3, RGS19, RNA polymerase II, SFRS9

Cancer, respiratory disease, tumor morphology

ANXA13, BRD3, C5, Cbp/p300, CCND1, CLIP2, Creb, EDA2R, ERK1/2, FHIT, GMFB, Hat, IL33, Integrin, Laminin, MAP2K1/2, Mek, MYST4, NFκB (complex), Nuclear factor 1, PI3K, PLCL1, Rac, Raf, Rap, Rap1, RAP1B, Ras, Ras homolog, RASSF1, RASSF5, RRAS2, SRF, TCR

Organismal injury and abnormalities, cellular development, nervous system development and function

ADAMTS10 (includes EG:81794), AKR1B1, AKR1B10, ATP6V1B2, BOC, BTNL2, CDON, CLC, COX15, COX8C, CRLF1, CTSZ (includes EG:1522), CYP2S1, Cytochrome c oxidase, DDAH2, EPO, FGF2, FGFRL1, GDF3, IFNG, MC1R, MEST, NDUFB7, NDUFV1, NR3C1, PSMB9, PSME2, RARRES2, retinoic acid, SHB, STAT3, TNF, TNFAIP2, WDR37

Cellular growth and proliferation, hematological system development and function, dental disease

ALDH3A2, ARL4A, ATN1, BMP4, BTNL2, C9ORF127, CASP1, CASP14, CORO1C, CRIP2, DLX2, DLX3, DLX5, FURIN, HRH2, IL4, KIAA1199, KLK10, LEFTY1, LGALS3BP, LTA4H, LYST, MSX1, NODAL, OVOL1, PAX9, PCSK4, PCSK7, SOLH, SOSTDC1, SP1, TGFβ1, Timp, TPSB2, UBE2J1

Cellular compromise, neurological disease, organismal injury and abnormalities

ACOT2, ATP5I, ATP6AP1, ATP6V1D, CHRNB3, DCAKD, DIRAS2, DNM, DNM2, dopamine, FAM105A, G-protein beta, HM13, HTT, ISYNA1, JPH2, KANK1, LEPRE1, Mapk, MIR124, MOGS, MYO1E, NARG1, NME2, PKNOX1, PRKAR1A, REST, SFXN3, SLC2A4, SUCLG2, SYNGR2, TSPAN14, TUSC4, Ubiquitin, XBP1

Cancer, cellular compromise, cellular movement

ATP10A, BAT1, CDKL3, CNN1, COPB1, CRY1, CTSZ (includes EG:1522), DHRS4, EMG1, F9, FAM120B, GRIP1, HNF4A, KHDRBS1, KIF1B, LAPTM4A, NSF, PFKFB4, PSMB7, PSMC4, PTPRN, SFRS2, SLC25A5, SMARCA5, TMEM79, TRAF2, TRAIP, TUBB4, UBE2N, UBQLN1, UBQLN4, USMG5, VKORC1, ZDHHC3

Cellular development, cellular growth and proliferation, reproductive system development and function

Alpha actin, Bcl9-Cbp/p300-Ctnnb1-Lef/Tcf, CAPZA2, CCNE2, CHPF, COL5A2, CPSF6, DAAM1, EGFR, ELF3, EP300, ERBB2, HABP2, HNF1A, HSPB8, LAMC2, LTA4H, MACF1, MXI1, MYL9 (includes EG:10398), PHF2, PHYH, PIK3C2A, PIN1, PTGER1, RBM14, RIN2, SH2D3A, SLPI, SOAT2, SOLH, TFF3, TNXB, WSB2, ZFP36L2

Lipid metabolism, molecular transport, small molecule biochemistry

ACSL3, ACSL6, ACTL6A, APP, ASXL1, ASXL2, ASXL3, BCOR, BPHL, C1ORF149, C20ORF20, C8ORF34, CBX2, CBX7, CBX8, CDKN2A, CLIC6, DAG1, DCTN4, EFNA3, EMX1, EPHA3, ESRP2, FBXL16, L3MBTL2, MIR220A, MIRLET7E (includes EG:406887), MIRLET7G (includes EG:406890), oleic acid, PCGF1, PCGF6, PHF19, POMGNT1, RING1, RNF2

Cell cycle, cell signaling, molecular transport

AGT, AGTR2, ANXA3, AR, BCL2L10, b-estradiol, BIK, CACNA1C, CACNA2D1, CACNB3, CACNB4, CALD1, Caspase, CCND3, CLIC3, COX5A, CYTH3, F Actin, GHRHR, HBXIP, KIAA0922, LEFTY2, LIMA1, MAF1, MAPK15, phosphatidylinositol-3,4,5-triphosphate, PIP5KL1, RASGRP1, SLC4A3, SPINLW1, STK3, TCF15, TERT, TMOD3, TOX2

©201 1L andesBi os c i enc e. Donotdi s t r i but e.

candidates on this basis. DiffScores are related to p-values by the relationship: DiffScore = 10sgn(b-valueESRD - b-valuediabetes no nephropathy)log10p All CpGs with DiffScores greater than 20 (significantly more methylated in ESRD patients on HD) or less than -20 (significantly less methylated in ESRD patients on HD) were considered as potential candidates for genes associated with ESRD and/or HD. This selection resulted in 2,870 CpGs in more than 2,000 genes. However, the majority of these genes were significantly different at only a single CpG site (in fact, 2549 of the genes

on the array are represented by only a single CpG and 191 of these were among the more than 2,000 selected on the basis of DiffScores). We have used the criterion requiring two or more CpGs significantly different in a previous study19 and found that 25–40% of candidates selected adding this criterion show significant differences in transcript level. Applying this additional criterion, 389 CpGs in 187 genes were selected as candidates on the basis of strong epigenetic differences between groups. Calculation of fold-enrichment of genes involved in diabetic nephropathy or HD. 2,549 of the 14,495 genes on the

www.landesbioscience.com Epigenetics

27

HumanMethylation27 array are represented by only a single CpG and so cannot fulfill the “two or more CpGs” criterion. The 187 genes that do fulfill this criterion are, therefore, selected from a possible pool of 11,946 genes and represent 0.016 of the total. By contrast, the 21 genes known or suspected to be involved in diabetic nephropathy (Table 2) represent 0.11 of the 187 genes or a 6.9-fold enrichment.

Table 6. Comparison of differentially methylated genes found in ­dialysis vs. upregulated in Wilflingseder et al. 2008

Acknowledgements

Helen Elivera R.N., assisted with collection of the specimens and consenting of patients and Drs. Sridhar Reddy and Colleen Veloski were extremely helpful in selecting patients and reviewing chart data. We gratefully acknowledge the assistance of Lior Benjamin in preparing samples. Financial Support

This research was supported by a grant from the Paul Teschan Fund, Dialysis Clinic, Inc., Nashville, TN. Note

Supplementary materials can be found at: www.landesbioscience.com/supplement/Sapienza-EPI6-1-Sup. pdf www.landesbioscience.com/supplement/Sapienza-EPI6-1-Sup. xls

2.

3.

4.

5.

6.

7.

8.

28

Wilflingseder et al.

WSB2

WSB2

JUN

JUN

KIF1B

KIF11

ITGB4

ITGB1

LGALS3BP

LGALS9

RGS19

RGS1, RGS6, RGS18

ARHGAP22

RHGAP30

MAP3K3

MAP3K8, MAPK14

MYL9

MYLK

RASSF1, RASSF5

RASSF4

SOCS1

SOCS3

STK6

STK17B

AKR1B1

AKR1C3

SLC1A1, SLC2A4, SLC4A3

SLC27A3

LILRB5

LILRB3

Genes in bold may be associated with diabetic nephropathy, per se, as well as dialysis.

©201 1L andesBi os c i enc e. Donotdi s t r i but e.

References 1.

Our report

USA Senate bill S. 976: “Genomics and Personalized Medicine Act of 2007.” www.govtrack.us/congress/bill. xpd?bill=s110-976 Sladek R, Rocheleau G, Rung J, Dina C, Shen L, Serre D, et al. A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature 2007; 445:828-30. Scott LJ, Mohlke KL, Bonnycastle LL, Willer CJ, Li Y, Duren WL, et al. A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science 2007; 316:1341-5. Freedman BI, Bostrom M, Daeihagh P, Bowden DW. Genetic factors in diabetic nephropathy. Clin J Am Soc Nephrol 2007; 2:1306-16. Imperatore G, Knowler WC, Nelson RG, Hanson RL. Genetics of diabetic nephropathy in the Pima Indians. Curr Diab Rep 2001; 1:275-81. Ewens KG, George RA, Sharma K, Ziyadeh FN, Spielman RS. Assessment of 115 candidate genes for diabetic nephropathy by transmission/disequilibrium test. Diabetes 2005; 54:3305-18. Kopp JB, Smith MW, Nelson GW, Johnson RC, Freedman BI, Bowden DW. MYH9 is a major-effect risk gene for focal segmental glomerulosclerosis. Nat Genet 2008; 40:1175-84. Freedman BI, Hicks PJ, Bostrom MA, Cunningham ME, Liu Y, Divers J, et al. Polymorphisms in the non-muscle myosin heavy chain 9 gene (MYH9) are strongly associated with end-stage renal disease historically attributed to hypertension in African Americans. Kidney Int 2009; 75:736-45.

9. Kao WH, Klag MJ, Meoni LA, Reich D, BerthierSchaad Y, Li M, et al. MYH9 is associated with nondiabetic end-stage renal disease in African Americans. Nat Genet 2008; 40:1185-92. 10. Wheeler HE, Metter EJ, Tanaka T, Absher D, Higgins J, Zahn JM, et al. Sequential use of transcriptional profiling, expression quantitative trait mapping and gene association implicates MMP20 in human kidney aging. PLoS Genet 2009; 5:1000685. 11. Liu Y. Advanced oxidation protein products: a causative link between oxidative stress and podocyte depletion. Kidney Int 2006; 69:213-7. 12. Friso S, Choi SW. Gene-nutrient interactions in onecarbon metabolism. Curr Drug Metab 2005; 6:37-46. 13. Schwahn B, Rozen R. Polymorphisms in the methylenetetrahydrofolate reductase gene: clinical consequences. Am J Pharmacogenomics 2001; 1:189-201. 14. Stenvinkel P, Karimi M, Johansson S, Axelsson J, Suliman M, Lindholm B, et al. Impact of inflammation on epigenetic DNA methylation—a novel risk factor for cardiovascular disease? J Intern Med 2007; 261:488-99. 15. Bonsib SM. Renal cystic diseases and renal neoplasms: a mini-review. Clin J Am Soc Nephrol 2009; 4:19982007.

Epigenetics

16. Wilflingseder J, Perco P, Kainz A, Korbély R, Mayer B, Oberbauer R. Biocompatibility of haemodialysis membranes determined by gene expression of human leucocytes: a crossover study. Eur J Clin Invest 2008; 38:918-24. 17. Karimi M, Johansson S, Ekström TJ. Using LUMA: a Luminometric-based assay for global DNAmethylation. Epigenetics 2006; 1:45-8. 18. Rakyan VK, Down TA, Thorne NP, Flicek P, Kulesha E, Gräf S, et al. An integrated resource for genomewide identification and analysis of human tissue-specific differentially methylated regions (tDMRs). Genome Res 2008; 18:1518-29. 19. Katari S, Turan N, Bibikova M, Erinle O, Chalian R, Foster M, et al. DNA methylation and gene expression differences in children conceived in vitro or in vivo. Hum Mol Genet 2009; 18:3769-78. 20. Cui H, Cruz-Correa M, Giardiello FM. Loss of IGF2 imprinting: a potential marker of colorectal cancer risk. Science 2003; 299:1753-5.

Volume 6 Issue 1