Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 May 1.
Published in final edited form as: Nat Genet. 2009 Oct 11;41(11):1191–1198. doi: 10.1038/ng.466

Multiple loci influence erythrocyte phenotypes in the CHARGE Consortium

Santhi K Ganesh 1,2,3,*, Neil A Zakai 4,*, Frank J A van Rooij 5,6,*, Nicole Soranzo 7,8,*, Albert V Smith 9,*, Michael A Nalls 10,*, Ming-Huei Chen 11,12, Anna Kottgen 13, Nicole L Glazer 14,15, Abbas Dehghan 5,6, Brigitte Kuhnel 16, Thor Aspelund 9,17, Qiong Yang 11,18, Toshiko Tanaka 19,20, Andrew Jaffe 13, Joshua C M Bis 15, Germaine C Verwoert 5,6,21, Alexander Teumer 22, Caroline S Fox 2,11, Jack M Guralnik 23, Georg B Ehret 3,24, Kenneth Rice 25, Janine F Felix 5,6, Augusto Rendon 26, Gudny Eiriksdottir 9, Daniel Levy 2,11, Kushang V Patel 23, Eric Boerwinkle 27, Jerome I Rotter 28, Albert Hofman 5,6, Jennifer G Sambrook 26, Dena Hernandez 29, Gang Zheng 30, Stefania Bandinelli 31, Andrew B Singleton 10, Josef Coresh 13, Thomas Lumley 25, André G Uitterlinden 5,6,21,32, Janine M vanGils 33, Lenore J Launer 23, L Adrienne Cupples 11,18, Ben A Oostra 5,6,34, Jaap-Jan Zwaginga 35,36, Willem H Ouwehand 26,7, Swee-Lay Thein 37, Christa Meisinger 16, Panos Deloukas 7, Matthias Nauck 38, Tim Spector 8, Christian Gieger 16, Vilmundur Gudnason 9,17, Cornelia M van Duijn 5,6, Bruce M Psaty 14,15,39,40,41, Luigi Ferrucci 19, Aravinda Chakravarti 3, Andreas Greinacher 42,^, Christopher J O’Donnell 2,11,43,^, Jacqueline C M Witteman 5,6,^, Susan Furth 44,^, Mary Cushman 4,^, Tamara B Harris 23,^, Jing-Ping Lin 30,^
PMCID: PMC2778265  NIHMSID: NIHMS145796  PMID: 19862010

Abstract

Erythrocyte measures are heritable and have important health implications, yet their genetic determinants are largely unknown. We performed genome-wide association analyses in 24,167 European-ancestry individuals for six erythrocyte traits and identified associations at 23 loci (P values 5×10-8 to 1×10-57). Replication testing in an independent set of 9,456 European-ancestry individuals showed strong evidence of association in all 23 loci in meta-analysis of the discovery and replication cohorts. Our findings include previously identified loci (HBS1L/MYB, HFE, TMPRSS6, TFR2, SPTA1) and novel associations (EPO, TFRC, SH2B3, and 15 other loci). This study has identified novel determinants of erythrocyte traits, offering insight into common variants underlying variation in erythrocyte measures.

Introduction

Red blood cell disorders such as anemia and erythrocytosis are broadly associated with multiple comorbid conditions including hypertension and other cardiovascular diseases, yet the genetic determinants of erythrocyte traits in the general population are poorly defined. Erythrocytes, which comprise approximately 40% - 50% of blood volume, are a key component for the transport of oxygen and carbon dioxide for cellular respiration. In clinical practice, measures of erythrocyte quantity, size and composition are routinely tested to diagnose and monitor hematologic diseases as well as the overall health of patients. Variation in erythrocyte measures even within normal ranges are related to other non-hematologic diseases and mortality1-3.

Erythrocyte production and quality are under various environmental and genetic influences. While environmental exposures, dietary intake of vitamins and iron, and the anemia of chronic disease contribute substantially to abnormalities of erythrocyte measures, the heritability of erythrocyte traits ranges from 40% - 90%4-6. Disorders of hemoglobin production and hemoglobinopathies are some of the most common genetic diseases in the world, owing to natural selection. Some known low-frequency Mendelian variants also lead to inter-individual variability in erythrocyte traits in the general population7, 8. Candidate gene studies have identified a few non-hemoglobin loci, including EPOR and HBS1L, related to variation in erythrocyte traits8-10. Early genome-wide association and linkage studies of erythrocyte measures, which have identified a few associations, such as at chromosome 6q23, lacked statistical power for association and genetic resolution for testing competing hypotheses6, 11-13.

To investigate genetic determinants of erythrocyte traits in the general population, we carried out genome-wide association studies and meta-analysis within multiple community-based cohorts comprising the CHARGE consortium14, followed by replication in independent samples. We identified 23 genetic loci associated with these erythrocyte traits. We further extend these findings to investigate possible links between these traits and vascular diseases, reporting associations of a few of the 23 loci identified with blood pressure and hypertension.

Results

CHARGE Consortium study samples

The total sample size for the individual cohort genome-wide association analysis and the CHARGE meta-analysis was 24,167 (the Age, Gene/Environment Susceptibility Reykjavik Study (AGES), N=3,205; the Atherosclerosis Risk in Communities Study (ARIC), N=7,803; the Cardiovascular Health Study (CHS), N=3,256; the Framingham Heart Study (FHS), N=3,359; and the Rotterdam Study (RS), N=5,523). We also included the Invecchiare in Chianti Study (InCHIANTI, N=1,021), an Italian cohort study, in these analyses. Characteristics of the study participants, including age, sex and trait summaries, are presented in Table 1.

Table 1. CHARGE cohort description.

AGES ARIC CHS FHS RS InCHIANTI
Number of individuals eligible for GWAS 3219 8127 3275 3381 5523 1206
Percent women 58 53 61 54 60 56
Mean age (yrs) 51 (6) 54 (6) 72 (5) 38 (9) 68 (8) 68 (16)
Hb (g/dL) 13.40
(1.45)
14.80
(1.02)
14.11
(1.23)
14.46
(1.37)
14.12
(1.28)
13.77
(1.35)
Hct (% ) 40.35
(3.53)
43.60
(2.86)
42.14
(3.54)
43.00
(3.9)
41.36
( 3.35)
40.63
(3.49)
MCH (picogram) 30.91
(1.69)
NA NA 30.64
(1.85)
30.16
(1.81)
3.05
(1.99)
MCHC (%) 33.57
(0.70)
NA NA 33.67
(0.91)
34.17
(1.15)
33.84
(1.05)
MCV (femtoliter) 92.08
(4.49)
90.7
(4.22)
NA 90.7
(5.0)
88.29
(4.30)
90.22
(4.84)
RBC (1M cell/cmm) 4.39
(0.41)
NA NA 4.73
(0.46)
4.69 (0.44) 4.51
(0.43)
Years of baseline examinations 1968-1991 1987-1989 1989-90 1971-1975 1990-1993 1998
Years of DNA collection 2002-2006 1987-1998 1989-1990 1996-1999 1990-1993 1998-2001

Sample sizes and summary statistics of covariates and erythrocyte traits measured in each cohort in CHARGE. Erythrocyte traits are abbreviated as: hemoglobin concentration (Hgb), hematocrit (Hct), mean corpuscular hemoglobin (MCH), mean corpuscular hemoglobin concentration (MCHC), mean corpuscular volume (MCV), erythrocyte count (RBC). The sample sizes presented are the range of numbers of individuals with both genotype and erythrocyte measures, after the exclusion of individuals with values beyond three standard deviations of the population mean for each erythrocyte trait.

Values for age and each erythrocyte trait are presented as mean(sd).

Meta-analysis of genome-wide association studies for six erythrocyte traits in the CHARGE Consortium

We studied six erythrocyte traits: hemoglobin concentration (Hgb); hematocrit (Hct); mean corpuscular volume (MCV); mean corpuscular hemoglobin (MCH); mean corpuscular hemoglobin concentration (MCHC); and red blood cell count (RBC), as defined in Supplementary Table 1. When cohort results were combined, 831 SNP associations at 23 independent loci (r2 < 0.3 between loci) across the six traits reached the genome-wide (GW) significance threshold of P < 5×10-8. The -log10(P value) genome-wide association plots for the meta-analysis of each of the 6 traits are shown in Figure 1. Corresponding QQ-plots are shown in Supplementary Figure 1a and the genomic control lambda (λGC) values in Supplementary Table 2. The genomic control inflation factor post-meta-analysis, which was not corrected at the meta-analysis level, showed no systematic inflation (Hgb λGC = 1.066; Hct λGC = 1.045; MCH λGC = 1.014; MCHC λGC = 0.995; MCV λGC = 1.029; and RBC λGC = 1.029; Supplementary Table 2). The meta-analysis results for all traits are summarized in Table 2, which is organized by the 23 independent loci and includes gene annotation information for each locus. The table also lists for each trait the number of SNPs exceeding the GW significance level. Altogether, there were 45 trait-locus combinations with at least one GW significant SNP. The complete set of SNP associations identified by the CHARGE meta-analysis is provided in Supplementary Table 3. Replication and further analysis focused on the 45 SNPs that gave the smallest P values for each of the 45 trait-locus findings in CHARGE.

Figure 1.

Figure 1

Overview of CHARGE meta-analysis results for six erythrocyte traits: hemoglobin concentration (Hgb), hematocrit (Hct), mean corpuscular hemoglobin (MCH), mean corpuscular hemoglobin concentration (MCHC), mean corpuscular volume (MCV), erythrocyte count (RBC). -log10 (P value) is plotted on the y-axis against genomic position of each SNP. Genomic loci with significant association (P < 5 × 10-8) are plotted in red, and loci with suggestive evidence are in blue (P < 4 × 10-7).

Table 2. CHARGE discoverymeta-analysis results, ordered by genomic locus.

# SNPs per trait Gene Annotation

Locus# Chr Hgb Hct MCH MCHC MCV RBC In RefGene RefGeneswithin60kb Closest
RefGene
1 1q23.1 0 0 0 8 0 0 SPTA1 OR10Z1; SPTA1; OR10X1; OR6Y1
2 2p21 3 6 0 0 0 0 PRKCE PRKCE
3 2p16.1 0 0 0 0 27 0 BCL11A BCL11A
4 3q29 0 0 4 0 11 0 TFRC TFRC
5 4q12 0 0 0 0 8 0 none none KIT
6 6p22.2 49 3 72 0 133 0 HFE; LRRC16; SCGN; SLC17A1;
SLC17A3; SLC17A2; TRIM38;
HIST1H4B; ZNF322A
HFE; SCGN; LRRC16; SLC 17A1;
SLC17A2; SLC17A3; SLC17A4;
ZNF322A; ABT1; TRIM38
7 6p21.1 0 0 51 0 65 0 PRICKLE4;FRS3;TFEB;
MED20;USP49;CCND3;BYSL
PRICKLE4; FRS3; PGC; TFEB;
MED20; USP49; CCND3; BYSL;
TAF8; PGC; FRS3
8 6q21 0 0 0 0 5 0 none CD164
9 6q23.3 0 14 43 13 83 24 HBS1L; MYB ALDH8A1; HBS1L; MYB
10 6q24.1 0 0 9 0 13 0 none none CITED2
11 7p12.2 0 0 0 0 5 0 IKZF1 IKZF1
12 7q22.1 0 2 0 0 2 2 TFR2; ZAN GNB2; PCOLCE; FBXO24; TFR2;
ACTL6B; HRBL; MOSPD3; LRCH4;
ZAN; EPO; POP7; PERQ1; EPHB4
13 7q36.1 1 3 0 0 0 0 PRKAG2 PRKAG2
14 9p24.1 0 0 9 0 19 0 RCL1 RCL1; AK3
15 10q11.21 0 0 0 0 4 0 MARCH8 MARCH8; ALOX5
16 10q21.3 1 3 0 0 0 0 HK1 HK1
17 12q24.12 10 9 0 0 0 0 SH2B3; ATXN2; c12orf30; PTPN11 SH2B3; ATXN2; BRAP; ACAD10;
ERP29; TMEM116; C12orf30;
TRAFD1; C12orf30; RPL6; PTPN11
18 14q23.3 0 0 0 0 9 0 MAX;FNTB MAX; RAB15; FNTB
19 16p13.3 0 0 1 0 1 0 ITFG3 LUC7L; PDIA2; AXIN1; ITFG3;
RGS11; ARHGDIG
20 19p13.13 0 0 6 0 25 0 MAN2B1; RTBDN; MAST1; DNASE2;
GCDH; FARSA
MAN2B1; MORG1; ZNF490; TNPO2;
DHPS; FBXW9; ZNF791; C19orf56;
JUNB; HOOK2; PRDX2; RNASEH2A;
RTBDN; GADD45GIP1; KLF1;
FARSA; RAD23A; CALR; MAST1;
GCDH; DNASE2; DAND5; NFIX
21 20q13.2 3 0 0 0 0 0 none none TSHZ2
22 22q12.3 13 2 9 0 36 0 C22orf33; TST; MPST ;TMPRSS6 IL2RB; C22orf33; KCTD17; TST;
TMPRSS6; MPST
23 22q13.33 0 0 0 0 12 0 TMEM112B; NCAPH2; SCO2; ECGF1 TMEM112B; ADM2; MIOX; ECGF1;
KLHDC7B; LOC440836; SAPS2;
SCO2; NCAPH2; SBF1; MAPK8IP2;
MIOX; CHKB; CPT1B

CHARGE meta-analysis results, showing the chromosomal position of each locus identified the number of SNPs identified within each and for each erythrocyte traitwith meta-analysis P value < 5×10-8. Annotation for SNPs within genes (InRefGene), within +/- 60kb of annotated RefGenes (RefGenewithin60kb), or in cases where no annotated gene was identified within 60kb, the nearest gene is reported (ClosestRefGene).

Independent replication

Replication of the 45 SNPs was conducted using a meta-analysis of association data in 9,456 independent European-ancestry individuals from five population-based cohorts in the HaemGen Consortium (Supplementary Note). A joint analysis of the HaemGen and CHARGE data showed a decrease in P values for all but two SNPs selected for replication. For one of the two SNPs (rs1800562) that did not show an improvement in P value when associated with Hct, the association to the Hgb trait was significant after Bonferroni correction, and for the second SNP (rs4466998), the association to MCV in the joint analysis of CHARGE and HaemGen data remained genome wide significant (P = 4.91 × 10-8). Significant independent replication for at least one trait was observed at 13 of 23 loci, using a Bonferroni-corrected significance threshold of P < 0.0011, or 0.05/45. Taking the joint meta-analysis results in sum, these data provide supportive evidence that the 23 loci from the discovery meta-analysis are true positives. Table 3 provides the full replication results, including beta coefficients, standard errors, and P values for the primary CHARGE findings, the HaemGen replication, and a combined meta-analysis of the two consortia for the 45 CHARGE trait-locus SNPs.

Table 3. CHARGE meta-analysis results, ordered by locus and trait, and HaemGen replication analysis.

CHARGE HaemGen CHARGE + HaemGen

Locus# Trait SNPID Chr PhysPos min_all MAF Gene % Var Beta s.e. P Beta s.e. P Beta s.e. P
2 Hct rs10168349 2 46208555 C 0.341667 PRKCE 0.16% 0.201 0.0283 1.176E-12 0.1524 0.0471 0.001212 0.1875 0.0238 3.748E-15
6 Hct rs1800562 6 26201120 A 0.041667 HFE 0.09% 0.3747 0.0608 7.204E-10 0.1167 0.1004 0.245 0.3073 0.0513 2.035E-09
9 Hct rs9483788 6 135477194 C 0.177966 HBS1L/MYB 0.13% 0.2172 0.0328 3.551E-11 0.2147 0.0527 4.55E-05 0.2166 0.0274 2.811E-15
12 Hct rs7385804 7 100073906 C 0.377358 TFR 2 0.17% -0.1592 0.0286 2.745E-08 -0.1269 0.0478 0.00799 -0.151 0.0242 4.45E-10
13 Hct rs10224002 7 151045974 G 0.258333 PRKAG2 0.22% 0.1691 0.0299 1.492E-08 0.2727 0.0493 3.08E-08 0.1963 0.0252 6.045E-15
16 Hct rs16926246 10 70763398 T 0.110169 HK1 0.13% 0.337 0.0513 4.986E-11 0.3094 0.096 0.00127 0.3315 0.0445 9.636E-14
17 Hct rs11065987 12 110556807 G 0.341667 SH 2B3/ATXN 2 0.15% -0.1809 0.0288 3.343E-10 -0.1438 0.0465 0.001983 -0.171 0.0241 1.363E-12
22 Hct rs2413450 22 35800170 T 0.381818 TMPRSS6 0.10% -0.162 0.0279 6.333E-09 -0.2082 0.0467 8.13E-06 -0.1736 0.0236 1.846E-13
2 Hgb rs10495928 2 46206670 G 0.341667 PRKCE 0.10% 0.011 0.011 5.926E-10 0.0537 0.0152 0.000414 0.0631 0.0088 7.052E-13
6 Hgb rs1800562 6 26201120 A 0.041667 HFE 0.17% 0.0224 0.0224 3.592E-16 0.1167 0.0322 0.000295 0.1617 0.0182 5.737E-19
13 Hgb rs10224002 7 151045974 G 0.258333 PRKAG2 0.14% 0.011 0.011 4.605E-08 0.0964 0.0164 4.19E-09 0.0714 0.0091 3.025E-15
16 Hgb rs16926246 10 70763398 T 0.110169 HK1 0.21% 0.0192 0.0192 1.036E-09 0.0863 0.0329 0.008721 0.1099 0.0164 2.116E-11
17 Hgb rs11065987 12 111076069 A 0.333333 TRAFD1 0.16% 0.0107 0.0107 1.345E-10 0.0427 0.0151 0.004591 0.0585 0.0086 1.159E-11
21 Hgb rs6013509 20 50751758 A 0.183673 TSHZ2 0.15% -0.0699 0.0116 1.963E-09 -0.0479 0.0206 0.01999 -0.0646 0.01 1.054E-10
22 Hgb rs855791 22 35792882 A 0.391667 TMPRSS6 0.20% -0.0962 0.011 2.044E-18 -0.0845 0.0157 7.97E-08 -0.0923 0.0089 3.25E-25
4 MCH rs11915082 3 197293536 A 0.425 TFRC 0.24% 0.0041 0.0007 4.888E-09 0.0035 0.0009 5.66E-05 0.0038 0.0005 7.729E-13
6 MCH rs1408272 6 25950930 G 0.033898 SLC17A3 0.41% -0.0134 0.0015 1.369E-18 -0.0183 0.0019 2.37E-22 -0.0153 0.0012 3.868E-39
7 MCH rs9349205 6 42033137 A 0.196429 CCND3/BYSL 0.29% -0.0066 0.0008 1.785E-16 -0.0037 0.0009 2.11E-05 -0.0053 0.0006 8.198E-20
9 MCH rs7776054 6 135460609 G 0.220339 HBS1L/MYB 1.02% -0.0092 0.0008 1.976E-33 -0.0107 0.0009 4.33E-36 -0.0099 0.0006 7.356E-69
10 MCH rs628751 6 139880112 C 0.491667 CITED2 0.34% -0.0049 0.0007 3.84E-13 -0.0034 0.0008 7.24E-06 -0.0043 0.0005 1.262E-17
14 MCH rs10758658 9 4846877 A 0.186441 RCL1 0.18% -0.0048 0.0008 1.634E-08 -0.0048 0.0009 5.14E-07 -0.0048 0.0006 2.166E-14
19 MCH rs1122794 16 249156 A 0.224138 ITFG 3 0.28% 0.0054 0.001 2.992E-08 0.0036 0.0012 0.00299 0.0047 0.0007 2.675E-10
20 MCH rs11085824 19 12862547 G 0.366667 GCDH 0.20% -0.0041 0.0007 8.105E-09 -0.003 0.0009 0.000532 -0.0037 0.0005 1.415E-11
22 MCH rs2413450 22 35800170 T 0.381818 TMPRSS6 0.41% -0.006 0.0007 8.818E-17 -0.0068 0.0008 5.34E-18 -0.0064 0.0005 8.77E-34
1 MCHC rs857721 1 156879172 A 0.316667 SPTA1 0.33% -0.0022 0.0004 3.414E-09 -0.0013 0.0004 0.00266 -0.0018 0.0003 1.033E-10
9 MCHC rs9373124 6 135464902 C 0.220339 HBS1L/MYB 0.30% -0.0023 0.0004 6.486E-10 -0.0018 0.0004 2.6E-05 -0.0021 0.0003 7.003E-14
3 MCV rs2540917 2 60462263 C 0.433333 BCL11A 0.24% -0.0031 0.0005 2.127E-11 -0.0022 0.0006 0.000192 -0.0028 0.0004 1.125E-14
4 MCV rs9859260 3 197284944 C 0.35 TFRC 0.23% 0.003 0.0005 3.246E-10 0.003 0.0008 0.000166 0.003 0.0004 8.499E-14
5 MCV rs172629 4 55102519 G 0.116667 KIT 0.27% -0.0043 0.0007 1.36E-09 -0.0051 0.001 2.22E-07 -0.0046 0.0006 9.816E-16
6 MCV rs1800562 6 26201120 A 0.041667 HFE 0.58% 0.0115 0.0011 1.425E-27 0.0137 0.0015 5.02E-20 0.0122 0.0009 1.012E-46
7 MCV rs9349205 6 42033137 A 0.196429 CCND3/BYSL 0.58% -0.0055 0.0005 1.756E-24 -0.0043 0.0008 8.48E-08 -0.0051 0.0004 1.121E-31
8 MCV rs9374080 6 109723113 C 0.375 CD164 0.22% -0.0026 0.0005 3.695E-08 -0.0017 0.0006 0.003738 -0.0023 0.0004 4.198E-10
9 MCV rs4895441 6 135468266 G 0.225 HBS1L/MYB 1.12% -0.008 0.0005 1.004E-57 -0.0083 0.0008 3.03E-27 -0.0081 0.0004 7.241E-86
10 MCV rs643381 6 139881116 A 0.489362 CITED2 0.50% -0.0039 0.0005 2.663E-18 -0.0037 0.0007 1.63E-07 -0.0039 0.0004 4.665E-25
11 MCV rs12718597 7 50395922 A 0.275 IKZF1 0.26% 0.0032 0.0005 8.138E-12 0.0018 0.0007 0.0145 0.0028 0.0004 4.689E-13
12 MCV rs7786877 7 100051951 G 0.208333 TFR 2 0.13% 0.0032 0.0005 5.452E-09 0.0024 0.0008 0.002081 0.003 0.0004 2.543E-11
14 MCV rs10758658 9 4846877 A 0.186441 RCL1 0.29% -0.0041 0.0006 4.354E-13 -0.0045 0.0008 4.65E-08 -0.0043 0.0005 3.184E-20
15 MCV rs11239550 10 45344735 G 0.228814 MARCH8 0.15% -0.0028 0.0005 1.873E-08 -0.0022 0.0007 0.003496 -0.0026 0.0004 1.346E-10
18 MCV rs4466998 14 64545293 A 0.478723 FNTB 0.17% 0.0027 0.0005 8.925E-09 0.0008 0.0006 0.2061 0.002 0.0004 4.907E-08
19 MCV rs7189020 16 244804 T 0.431034 ITFG3 0.19% -0.0031 0.0005 1.081E-09 -0.0024 0.0007 0.000871 -0.0029 0.0004 1.819E-12
20 MCV rs7255045 19 12793269 A 0.25 RTBDN 0.27% -0.0037 0.0006 1.233E-11 -0.0018 0.0008 0.0266 -0.0032 0.0004 2.173E-12
22 MCV rs2413450 22 35800170 T 0.381818 TMPRSS6 0.65% -0.0054 0.0005 1.078E-30 -0.0046 0.0007 7.62E-11 -0.0052 0.0004 2.772E-41
23 MCV rs131794 22 49318618 A 0.166667 ECGF1 0.22% -0.0044 0.0006 2.189E-13 -0.0029 0.0009 0.001795 -0.004 0.0005 1.033E-15
9 RBC rs9483788 6 135461324 G 0.225 HBS1L/MYB 0.65% 0.0141 0.0016 3.115E-19 0.0155 0.0013 2.19E-30 0.0141 0.001 1.148E-47
12 RBC rs2075671 7 100183042 A 0.225 EPO 0.20% 0.0086 0.0016 3.058E-08 0.0047 0.0016 0.003383 0.0068 0.0011 1.123E-09

Replication test results for the lead SNP per locus and per erythrocyte trait (45 SNPs). Results are organized by trait, the by locus, as indicated in Table 1. Results from a combined CHARGE and HaemGen Consortium meta-analysis are presented. Minor allele frequency (MAF) is presented based on HapMap CEU. % Var indicates the percent of variance explained by the lead SNP in the corresponding trait-locus. P values in bold font meet a Bonferroni-corrected significance threshold for replication of P < 0.0011 (0.05/45). Units were Hgb g/dl, Hct %, MCH picogram, MCHC g/dL, MCV femtoliter, RBC 1 M cells/ccm.

For each lead SNP in the 23 independent loci, percent variance explained for each of the lead SNPs in the corresponding trait is provided in Table 3, averaging the percent variance explained for each SNP across the CHARGE cohorts. The combination of lead SNPs from each of the trait loci showed that average percent variance explained by the combination of lead SNPs, beyond the variance explained by age and gender, was 1.14% of Hgb variation (rs10495928, rs1800562, rs10224002, rs16926246, rs11065987, rs6013509, rs855791); 1.16% of Hct variation (rs10168349, rs1800562, rs7385804, rs10224002, rs16926246, rs11065987, rs9483788, rs2413450); 4.53% of MCH variation (rs11915082, rs1408272, rs9349205, rs7776054, rs628751, rs10758658, rs1122794, rs11085824, rs2413450); 0.63% of MCHC variation (rs857721, rs9373124); 5.98% of MCV variation (rs2540917, rs9859260, rs172629, rs1800562, rs9349205, rs9374080, rs4895441, rs643381, rs12718597, rs7786877, rs10758658, rs11239550, rs4466998, rs7189020, rs7255045, rs2413450, rs131794); and 0.85% of RBC variation (rs9483788, rs2075671).

Annotation of associated loci

For 20 of the identified loci, top associated SNPs were identified within a +/- 60 Kb window of a RefSeq gene (Table 2). For three loci, chromosomes 4q12, 6q24.1, 20q13.2, no genes were identified within this window, with the nearest genes approximately 116 Kb, 89 Mb, and 50 Mb away, respectively. Of the 23 loci, previously reported mutations or genetic associations for erythrocyte traits, markers of iron status or fetal hemoglobin levels, have been noted at six loci containing the genes HFE, TFR2, TMPRSS6, SPTA1, HBS1L-MYB, and BCL11A. Most of the remaining loci have not previously been reported to be associated with erythrocyte traits, though several genes are known to have important roles in erythrocyte biology or erythropoiesis. Genes identified near the associated loci, and their associated erythrocyte traits, are presented in Table 2 and Figure 2. Gene annotations, including gene information, known genetic mutations causing hematologic and non-hematologic diseases, and previously defined roles in hematologic and cardiovascular systems are listed in Supplementary Table 4. We confirmed the association of the known C282Y (rs1800562) and H63D (rs1799945) mutations in the HFE gene, mutations that are already known to underlie hereditary hemochromatosis, with Hgb, Hct, MCH and MCV.

Figure 2.

Figure 2

Results of the CHARGE meta-analysis are organized into a Venn diagram, demonstrating overlap of loci meeting a genome-wide significance threshold of P < 5×10-8.

Gene expression in blood and endothelial cell lines

RNA expression levels for genes within a 1 Mb interval of each of the 23 loci we identified are presented in Supplementary Figure 2, for erythroid bodies (EBs), human umbilical vein endothelial cells (HUVECs), and seven other blood cell lines. For the top associated locus, chromosome 6q23.3, four genes were identified (ALDH8A1, HBS1L, MYB, AHI1) within the 1 Mb interval. A heatmap showing gene expression levels for each of these four genes is shown in Figure 3, demonstrating approximately 2-fold expression of MYB in EBs compared to other cell lines. Gene expression was detected for genes in multiple loci (Supplementary Figure 2). Since the identification of gene expression in biologically relevant tissues provides a rationale for prioritization of candidate genes for further genetic or functional investigations, we noted broad categories of expression patterns in EBs and HUVECs. The most highly expressed genes in EBs were in chromosomes 3q29 (TFRC), 6p22.2 (HIST1H4C, which is near HFE), 6p21.1 (CCND3), 10q21.3 (HK1), 12q24.12 (RPL6P27), 16p13.3 (HBZ, HBA1), and 22q12.3 (TST, RAC2). The most highly expressed genes in HUVECs were in chromosomes 6p22.2 (HIST14HC), 7q22.1 (SERPINE1), and 22q13.3 (MFNG).

Figure 3.

Figure 3

Gene expression in blood and endothelial cells for genes in the chromosome 6q23.3 region. (a) SNPs in the locus are plotted against recombination rates as observed in HapMap CEU, using a window of +/-500kb around the lead SNP identified in this locus, which is plotted in blue. SNPs identified by the CHARGE meta-analyses are colored according to correlation with the lead SNP (r2 ≥ 0.8 red; 0.5 ≤ r2 < 0.8 orange; 0.2 ≤ r2 < 0.5 yellow; r2 < 0.2 white; no r2 value provided). The P value for the lead SNP in this region is provided. (b) A heatmap of gene expression levels in nine blood and endothelial cell lines is shown, including all genes, as annotated by ENSEMBL 54, within the +/- 500kb window of the locus (MK = megakaryocyte; EB = erythroid bodies; HUVEC = human umbilical vein endothelial cells; CD14 = monocytes; CD66b = granulocytes; CD19 = B lymphocytes; CD56 = NK cells; CD8 = Tc lymphocytes; CD4 = Th lymphocytes).

Blood pressure analysis of SNPs associated with erythrocyte traits

The results of association testing for the 45 lead SNPs from the CHARGE analysis of erythrocyte traits within the 23 loci are summarized in Supplementary Table 5a. We identified associations at a Bonferroni-corrected significance threshold P < 0.00135 (0.05/37, since 37 of the 45 SNPs are unique) in chromosomes 12q24.1 (SH2B3) and 7q36.1 (PRKAG2). The previously reported association of the chromosome 12q24.1 (SH2B3) locus with systolic blood pressure (SBP) and diastolic blood pressure (DBP) was the most significant association (rs1106598, SBP P = 1.2×10-6, HTN P = 0.0035; rs1763023, DBP P = 4.2×10-8). In the reported BP and HTN analysis, signals at this locus spanned 700kb from rs3184504 to rs1106618815, and association signals in Hgb and Hct spanned 987kb, from rs3184504 in SH2B3 to rs11066301 in PTPN11 and contained multiple genes15. Inspection of RNA expression data (Supplementary Figure 2) showed that in this region, SH2B3 and ATXN2 show high levels of gene expression in erythroid bodies and endothelial cells. Nominal associations (P < 0.05) were identified in the chromosomes 6p22.2 (HFE), 6q24.1, 7q22.1 (TFR2), and 20q12.3 (Supplementary Table 5a), and results of the evaluation of SNPs associated with BP and hypertension are presented in Supplementary Table 5b.

Discussion

In this meta-analysis of genome-wide association data from 24,167 European-ancestry individuals from six cohort studies in the CHARGE Consortium, we identified 23 loci associated with at least one of the six erythrocyte traits Hgb, Hct, MCH, MCHC, MCV, and RBC. We sought evidence for replication in an independent analysis of data from 9,456 European-ancestry individuals in the HaemGen Consortium. In the joint meta-analysis, merging CHARGE and HaemGen data, all 23 loci had P values less than P < 5×10-8, implying strong associations that merit further study. Among the 23 loci, six were previously known QTLs, and 17 are novel loci, some of which contain genes known to be involved with iron homeostasis, erythropoiesis, globin synthesis and erythrocyte membrane function. Finally, an investigation of possible links between these erythrocyte traits and blood pressure and hypertension confirmed overlap at the previously known SH2B3 locus, and identified additional suggestive associations, none of which met a genome-wide significance threshold.

The six erythrocyte traits studied included some that are highly correlated, and as expected, we observed a high degree of concordance in the results across the six traits. Among the many genome-wide significant associations identified, the patterns of association are likely to reflect the correlations among these related traits. Interestingly, MCHC, a ratio of Hgb and Hct, two directly measured traits, is uniquely associated with chromosome 1q23.1 (SPTA1), a gene with several rare mutations known to cause deformation of erythrocytes16, 17. Reviewing the results in total, we observed there were generally three patterns of significant associations among the six traits (Figure 2). Results were generally similar for: (1) Hgb and Hct, which are mainly quantitative measures of hemoglobin in the blood; (2) MCH and MCV, representing erythrocyte size and quantity of hemoglobin per erythrocyte; and (3) MCHC, the ratio of Hgb to Hct, which appears somewhat distinct from the other traits. Across the six traits studied, the strongest signal was found in the HBS1L/MYB locus on chromosome 6q23, which was observed for five of the six individual traits (Hct, MCH, MCHC, MCV, RBC) at the genome-wide significance level. This locus also provided a modest but non-significant result for Hgb (rs4895441, P = 4.8×10-4). In addition, the Hgb/Hct and MCH/MCV patterns overlapped for associations in the chromosome 6p22.2 (HFE), 22q12.3 (TMPRSS6) and 7q22.1 (TFR2/EPO) loci. The RBC results represent a subset of the overlap between the Hgb/Hct and MCH/MCV patterns, with associations observed in the 7q22.1 (TFR2/EPO) and 6q23 (HBS1L/MYB) loci. Across the erythrocyte traits, overlap occurs where known patterns of traits characterize various clinically observed hematologic diseases, providing a possible context in which to interpret the overlap of associations.

We annotated and categorized the findings of our analyses by association with known genetic disorders, biologic function, or altered function of the hematopoietic system, to assist with interpretation of the findings (Supplementary Table 4). We here consider these multiple findings in light of their potential role in several processes critical to erythrocyte biology, including iron homeostasis, erythrocyte membrane function, erythropoiesis and globin synthesis.

Iron homeostasis

We identified genome-wide significant association of SNPs within the HFE gene with Hgb, Hct, MCH, and MCV. C282Y mutation in HFE is the principal cause of hereditary hemochromatosis, a common autosomal recessive iron overload disease in individuals of northern European descent18. This mutation was associated with increased MCV and Hgb concentrations in a study of individuals drawn from a hemochromatosis and iron overload screening study7, and this variant was the lead association result for both Hgb and Hct in our study. Heterozygotes for either allele do not manifest clinical iron overload but may display an increased iron uptake and “resistance” to anemia, and the C282Y mutation may increase risk of coronary heart disease by increasing iron stores and lipid oxidation19, 20. The HFE gene induces expression of the iron regulatory hormone hepcidin. Hepcidin has recently emerged as the likely link between the inflammatory response and the handling of iron for erythropoiesis by both downregulating the absorption of iron in the intestine and by inhibiting the release of iron from macrophages21-24. SNPs within the TMPRSS6 gene were associated with Hgb, Hct, MCH and MCV. TMPRSS6 was identified by linkage and association studies in five families and two sporadic cases with iron-refractory iron deficiency anemia, a rare Mendelian disease25. TMPRSS6 encodes a type II transmembrane serine protease produced by the liver that regulates the expression of hepcidin25. The transferrin receptor (encoded by TFRC(TFR1)) and transferrin receptor 2 (TFR2) are highly homologous type II trans-membrane proteins in the transferrin protein family. SNPs within TFRC were associated with MCH and MCV, and SNPs within TFR2 were associated with Hct and MCV. Reduced TFRC expression is associated with anemia26. Existing evidence indicates that TFR2 is also a modulator of hepcidin expression, and mutations in TFR2 cause hemochromatosis type 327.

Erythropoiesis and globin synthesis

Two loci, chromosome 2p16.1 (BCL11A) associated with MCV, and chromosome 6q23.3 (HBS1L-MYB) associated with all traits except for Hgb, are related to variation in fetal hemoglobin and hemoglobin beta levels28, 29. BCL11A is an oncogene related to B-cell malignancies30 and regulates fetal hemoglobin expression31. BCL11A is expressed in erythroid precursors, and we observed BCL11A expression in EBs (Supplementary Figure 2) making it a biologically plausible candidate gene for erythrocyte trait variation32. A healthy population study showed polymorphisms in HBS1L and MYB influences erythrocyte, platelet, and monocyte counts10. Although the role of HBS1L is unknown, MYB has been associated with proliferation, survival, and differentiation of hematopoietic progenitor cells33, 34. MYB is also associated with eosinophil counts in blood and atopic asthma35. There are strong associations between SNPs within this locus and multiple erythrocyte traits (lead SNP rs4895441, Hgb P = 4.8×10-4; Hct P = 9.7×10-10; MCH P = 7.8×10-32; MCHC P = 4.5×10-9; MCV P = 1.0×10-57; and RBC P = 2.2×10-15). These strong genetic effects may explain why several prior linkage analyses of erythrocyte traits have identified this chromosomal region6, 11, 13. SNPs within SH2B3 are associated with Hct and Hgb. Interestingly, the SNPs within this gene are associated with blood pressure, myocardial infarction, type 1 diabetes, and celiac disease15, 35-39. SH2B3 is expressed in hematopoietic precursor cells and increases hematopoietic progenitors of erythroid, megakaryocytic, and myeloid lineages35, 40. SH2B3 is also expressed in vascular endothelium, where it promotes inflammation and may thereby contribute to vascular disease. Expression in different cell lineages and tissues may underlie the diverse pleiotropic consequences of SH2B3 on hematopoietic traits, autoimmune diseases, and vascular diseases. In the same locus, the PTPN11 gene product interacts with the transcription factor SHP2, which has an essential role in blood development that has been demonstrated in a murine Shp2-/- model41, and PTPN11 mutations cause Noonan’s and LEOPARD syndromes and juvenile myelomonocytic leukemia42-44. Lastly, we identified associations for Hct, MCV and RBC near the EPO gene. Erythropoietin, a glycoprotein hormone that controls erythropoiesis, is the first human recombinant hematopoietic protein approved for human use and is now used widely for the treatment of anemia. EPO variants have previously been described in association with diabetic retinal and renal vascular complications45 but not with erythrocyte traits.

Erythrocyte membrane

SNPs within the SPTA1 gene were associated with MCHC. SPTA1 encodes erythroid spectrin, a protein in the erythrocyte membrane, and is essential in determining the shape and deformability of erythrocytes. Spectrin mutations have been previously associated with hemolytic anemia, elliptocytosis, spherocytosis, and propoikilocytosis, but not with variations in MCHC outside of disease states16, 17.

Gene expression

The gene expression data may be viewed as additional annotation of the 23 loci we identified, confirming which genes are expressed in cell types of interest. These data may be used to generate further specific hypotheses that can then be tested at a functional and molecular level.

Multiple lines of evidence formed the basis for our rationale to study the relationship of the SNPs identified through the study of erythrocyte traits to blood pressure (BP) and hypertension. Prior studies have shown that Hgb and Hct levels are associated with increased risk for hypertension and a variety of other vascular diseases and mortality1-3, 46-49. From a rheologic perspective, blood viscosity depends largely on Hgb or Hct levels and is a determinant of blood pressure50-53. There is an inverse relationship between viscosity and vascular blood flow54, and elevated Hct thereby hampers organ perfusion. Given these findings, we are intrigued by the overlap between the association results for Hgb and Hct from our study and the recently reported associations for BP and hypertension15, 36. We observed overlap of associations in the chromosome 12q24.12 region across a 987 kb linkage disequilibrium block, containing SH2B3, ATXN2, BRAP, C12orf03, TRAFD1, ACAD10, TMEM116 and PTPN11. We also identified associations within the 7q36.1 region containing PRKAG2, which does not have specifically known hematologic or vascular roles, but mutations in this gene cause cardiomyopathy and cardiac conduction system disorders55, 56. Neither causality nor independence of these associations is necessarily supported by these findings. However, these associations suggest that common genetic bases may underlie some of the correlation seen between erythrocyte, BP and hypertension traits. Further confirmation in large independent cohorts may provide stronger evidence for the strength and consistency of the associations with hypertension.

Limitations of this study include restriction of the discovery and replication analyses to individual of European ancestry. Several spectrin and globin mutations have been identified in African American kindreds and the prevalence of hemoglobinopathies of various types is generally higher among individuals of African ancestry, highlighting the need for further investigation of these findings in individuals of non-European-ancestry57. As with any meta-analysis of genome-wide association results across different cohorts, population structure and other sources of heterogeneity may have caused false positives or false negatives. To assess population structure, we examined the per-cohort λGC, demonstrating that these values were consistently below 1.08, and we applied genomic control to the cohort-level test statistics. The final meta-analyses also showed no systematic inflation of the distribution of the final association statistics. In the replication analysis, for those loci that did not meet a conservative replication test, power may have been limited, and many are likely to improve with additional study. Finally, the interpretation of multiple analyses of correlated traits requires caution, particularly in attributing causality or independence of effects. We take the findings from our analyses of the six erythrocyte traits to indicate a set of loci that are of interest with regards to erythrocyte production, homeostasis and function. Specific differences in association patterns may highlight different pathways, and to understand this more deeply, further studies are needed.

In summary, we have identified and validated common variants at several known and novel loci that influence the levels of six clinically relevant red blood cell measures in population-based cohorts. These QTLs have implications for understanding a variety of hematologic diseases as well as correlates of erythrocyte traits, such as BP and hypertension. Further studies are warranted to define these variants in the extremes of the distributions of these traits and ethnically diverse populations and to understand the functional impact of variants at the implicated candidate genes.

Supplementary Material

1
2

Acknowledgements

Complete study acknowledgments are listed in the Supplementary Note. The authors thank the studies’ participants, staff and the funding agencies for their support.

Appendix

Methods

CHARGE Consortium organization and study samples

We performed a cross-sectional analysis of genotype and phenotypic data on erythrocyte traits in the CHARGE consortium14, which includes five cohort studies that have genotyped high density SNP markers and have phenotypic data on erythrocyte traits (AGES N=3,205; ARIC N=7,803; CHS N=3,256; FHS N=3,359; RS N=5,523) and InCHIANTI (N=1,021). Each participating study was approved by its corresponding Institutional Review Board, and all study subjects provided informed consent for participation in the study and genetic research. Participants were excluded if they were of non-European ancestry, as determined by self-report, and also by principal component analysis in ARIC and RS. Detailed methods for each of the participating cohorts are provided in the Supplementary Note.

Genotyping and imputation

Briefly, each study genotyped samples using high-density SNP marker platforms (Affymetrix SNP6.0 - ARIC; Affymetrix 500K - FHS; Illumina 370K - AGES, CHS; Illumina 550K-InCHIANTI, RS). Genotypes were then imputed to a set of approximately 2.5M HapMap SNPs using Phase II CEU HapMap individuals for reference using either MACH (http://www.sph.umich.edu/csg/abecasis /MACH) (ARIC, AGES, FHS, InCHIANTI, RS) or BimBam58 (CHS) software.

Erythrocyte phenotypes

Erythrocyte parameters studied were 1) hemoglobin concentration (Hgb), the concentration of hemoglobin within whole blood; 2) hematocrit (Hct), the percent of whole blood comprised of cellular erythrocyte elements 3) red blood cell count (RBC), the number of red blood cells per volume of blood. 4) mean corpuscular volume (MCV), the average erythrocyte volume; 5) mean corpuscular hemoglobin (MCH), the average quantity of Hgb per erythrocyte; and 6) mean corpuscular hemoglobin concentration (MCHC), the ratio of Hgb to Hct. The definition and units of each trait are provided in Supplementary Table 1. Blood was drawn from each participant using standard phlebotomy methods, and erythrocyte measures were obtained using standard clinical assays in certified laboratories.

Phenotype modeling and statistical analysis

All traits studied were continuous. Based on prior convention and visual inspection of the data, MCH, MCHC and MCV were natural log transformed, RBC was square root transformed, and Hgb and Hct were not transformed prior to analyses. In order to focus on determinants of variation of these traits in the general population, rather than on specific hematologic diseases which are over-represented at the tails of the distribution for each of the traits, we restricted analysis to those individuals within three standard deviations of the sample mean within each cohort. For each SNP meeting QC criteria, linear regression was used to assess association with trait, separately for all six traits. An additive genetic model was used throughout. These regressions were adjusted for age, gender, and site in the multi-center cohorts. In FHS, linear mixed effects models were used to account for relatedness, and these models included adjustment for principal components, computed using Eigenstrat 2.059. The genome-wide level of significance threshold was set at P < 5×10-8.

Each cohort’s results were forwarded to a central repository, including regression coefficients, standard errors, sample size, imputation quality, minor allele designation and minor allele frequency. Prior to meta-analysis, we performed genomic control on each cohort-specific distribution of the association test statistics for each trait60. We also filtered out SNPs with allelic frequency less than 1% or poor imputation quality (the ratio of observed variance of imputed allele counts to the expected variance of imputed allele counts > 1.1 from the imputation software output). Separately for each SNP and trait, within-cohort association results were combined in an inverse variance weighted meta-analysis, as implemented in METAL (http://www.sph.umich.edu/csg/abecasis/Metal/index.html). After meta-analysis, genomic control inflation factor (λGC) was again calculated to assess stratification between the cohorts and resulting inflation of the test statistics. Genomic control was not applied to the final meta-analysis results. The SNAP program (http://www.broadinstitute.org/ mpg/snap) was used to estimate linkage disequilibrium between the associated loci. Percent variance explained within each cohort was calculated from the r2 estimate derived from a linear regression model for individual lead SNPs at each trait locus and the combination of all leads SNPs per trait, accounting for age and gender as well.

Replication samples and analysis

The replication set included samples from the five population-based cohorts of individuals of European ancestry that comprise the HaemGen Consortium (Study of Health in Pomerania (SHIP) N=3,200; UK Blood Services Common Control (UKBS1-CC1) N=1,290; Twins UK adult twin registry N=1,510; KORAF3 500K study population N =1,643; and KORA-F4 N=1,814). Further information on these cohorts is provided in the Supplementary Note. Trait definitions were identical to our initial study. Analysis for each of the six traits undergoing replication was implemented in the same way as for the CHARGE analyses. For replication, we used a Bonferroni correction for the number of SNPs tested by the HaemGen Consortium. Given the smaller sample size available for the HaemGen replication analysis relative to the CHARGE discovery analysis, we performed a combined meta-analysis on the top trait-locus SNPs identified by the CHARGE meta-analysis, to assess impact on the association signals.

Gene expression in blood cell subtypes and endothelial cells

To assist with prioritization of the candidate genes identified in this study, we evaluated RNA expression in eight blood cell lines (stem-cell derived erythroblasts (EB) and megakaryocytes (MK), CD14+ monocytes, CD56+ natural killer (NK) cells, CD4+ T helper (Th) lymphocytes, CD8+ cytotoxic T (Tc) lymphocytes, and CD66b+ granulocytes), using an established catalogue of gene expression in these lineages61; we also examined gene expression in cultured human umbilical vein endothelial cells (HUVECs)62. For each of the 23 identified loci, we examined the transcript levels in the nine cell types of all genes within a +/-500kb window centered on the lead association SNP.

Briefly, EB and MK samples were obtained by culturing of CD34-positive haematopoietic stem cells purified from cord blood cultured with EPO-IL3-SCF for 10 days and with TPO, respectively61. Cultured cells were sorted by fluorescence-activated cell sorting using either a monoclonal antibody against CD235a (glycophorin A) or against CD41 (άIIb integrin). The other six blood cell types were purified from the peripheral blood of seven healthy subjects using the corresponding CD markers. Blood cell types were hybridized onto Illumina V2 Ref6 gene expression beadarrays, and the detailed methods for isolation of other hematologic cell types, RNA extraction and microarray analysis are described elsewhere61. Cultured human umbilical vein endothelial cells (HUVECs) and expression profiles were ascertained as has been described elsewhere62 and using the same microarray platform61.

Raw data was VST transformed using the R/Bioconductor package “lumi,”63 followed by quantile normalization. The mode of the transformed signal intensity is 7.94 and can be taken as the background intensity. The maximum probe intensity was 15.85 corresponding to a signal intensity of 59,000 in a linear scale. For each cell type and probe, a Grubbs test for outlier identification was used and samples with P values less than 0.01 were removed (implemented in the R package “outliers”). This was not performed for the MK, EB or HUVEC cell types due to their smaller sample size (4, 4, and 3 respectively). Probe mappings were obtained from re-annotation efforts available at http://www.compbio.group.cam.ac.uk/Resources/Annotation/.

Annotation of genetic loci

To determine genes within or neighboring each locus, we examined RefSeq gene annotations, build 36. To annotate the loci and genes, we reviewed the literature, OMIM64 and the Genetic Association Database65 (Supplementary Table 4).

Blood pressure and hypertension analyses

Following the observation that epidemiologic studies have identified a strong yet unexplained link between Hgb and Hct and blood pressure, we examined the associations of these SNPs to those previously reported meta-analysis within the CHARGE consortium for systolic blood pressure (SBP), diastolic blood pressure (DBP) and hypertension1, 2, 15. The CHARGE cohorts that contributed samples to the BP and hypertension analyses were AGES, ARIC, CHS, FHS, and RS, with a total sample size of N=29,136. SBP and DBP measured at the first visit attended were continuous traits, and hypertension was analyzed as a dichotomous trait15. We tested the association of the lead SNP per locus for each trait (45 SNPs) for association with SBP, DBP and hypertension15, using a Bonferroni corrected threshold for the number of SNPs tested. We additionally reversed the analysis, examining the test statistics within each of the six erythrocyte traits for those SNPs reported to be associated with SBP, DBP or hypertension15.

Footnotes

Competing Interest Statement: Aravinda Chakravarti is a paid consultant with Affymetrix in accordance with the policies of Johns Hopkins.

References

  • 1.Havlik RJ, et al. Evidence for additional blood pressure correlates in adults 20-56 years old. Circulation. 1980;61:710–5. doi: 10.1161/01.cir.61.4.710. [DOI] [PubMed] [Google Scholar]
  • 2.Tell GS, et al. The Cardiovascular Health Study. Cardiovascular Health Study (CHS) Collaborative Research Group Correlates of blood pressure in community-dwelling older adults. Hypertension. 1994;23:59–67. doi: 10.1161/01.hyp.23.1.59. [DOI] [PubMed] [Google Scholar]
  • 3.Zakai NA, et al. A prospective study of anemia status, hemoglobin concentration, and mortality in an elderly cohort: the Cardiovascular Health Study. Arch Intern Med. 2005;165:2214–20. doi: 10.1001/archinte.165.19.2214. [DOI] [PubMed] [Google Scholar]
  • 4.Whitfield JB, Martin NG. Genetic and environmental influences on the size and number of cells in the blood. Genet Epidemiol. 1985;2:133–44. doi: 10.1002/gepi.1370020204. [DOI] [PubMed] [Google Scholar]
  • 5.Evans DM, Frazer IH, Martin NG. Genetic and environmental causes of variation in basal levels of blood cells. Twin Res. 1999;2:250–7. doi: 10.1375/136905299320565735. [DOI] [PubMed] [Google Scholar]
  • 6.Lin JP, et al. Evidence for linkage of red blood cell size and count: genome-wide scans in the Framingham Heart Study. Am J Hematol. 2007;82:605–10. doi: 10.1002/ajh.20868. [DOI] [PubMed] [Google Scholar]
  • 7.McLaren CE, et al. Determinants and characteristics of mean corpuscular volume and hemoglobin concentration in white HFE C282Y homozygotes in the hemochromatosis and iron overload screening study. Am J Hematol. 2007;82:898–905. doi: 10.1002/ajh.20937. [DOI] [PubMed] [Google Scholar]
  • 8.Zeng SM, Yankowitz J, Widness JA, Strauss RG. Etiology of differences in hematocrit between males and females: sequence-based polymorphisms in erythropoietin and its receptor. J Gend Specif Med. 2001;4:35–40. [PubMed] [Google Scholar]
  • 9.Zeng SM, Yankowitz J, Widness JA, Strauss RG. Sequence-based polymorphisms in members of the apoptosis Bcl-2 gene family and their association with hematocrit level. J Gend Specif Med. 2003;6:36–42. [PubMed] [Google Scholar]
  • 10.Menzel S, et al. The HBS1L-MYB intergenic region on chromosome 6q23.3 influences erythrocyte, platelet, and monocyte counts in humans. Blood. 2007;110:3624–6. doi: 10.1182/blood-2007-05-093419. [DOI] [PubMed] [Google Scholar]
  • 11.Iliadou A, et al. Genomewide scans of red cell indices suggest linkage on chromosome 6q23. J Med Genet. 2007;44:24–30. doi: 10.1136/jmg.2006.043521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Yang Q, Kathiresan S, Lin JP, Tofler GH, O’Donnell CJ. Genome-wide association and linkage analyses of hemostatic factors and hematological phenotypes in the Framingham Heart Study. BMC Med Genet. 2007;8(Suppl 1):S12. doi: 10.1186/1471-2350-8-S1-S12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lin JP, O’Donnell CJ, Levy D, Cupples LA. Evidence for a gene influencing haematocrit on chromosome 6q23-24: genomewide scan in the Framingham Heart Study. J Med Genet. 2005;42:75–9. doi: 10.1136/jmg.2004.021097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Psaty BM, O’Donnell CJ, Gudnason V. Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium: Design of prospective meta-analyses of genome-wide association studies from five cohrots. Circ Cardiovascular Genetics. 2008 doi: 10.1161/CIRCGENETICS.108.829747. al., e. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Levy D, et al. Genome-wide association study of blood pressure and hypertension. Nat Genet. 2009 doi: 10.1038/ng.384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Agre P, Casella JF, Zinkham WH, McMillan C, Bennett V. Partial deficiency of erythrocyte spectrin in hereditary spherocytosis. Nature. 1985;314:380–3. doi: 10.1038/314380a0. [DOI] [PubMed] [Google Scholar]
  • 17.Marchesi SL, et al. Mutant forms of spectrin alpha-subunits in hereditary elliptocytosis. J Clin Invest. 1987;80:191–8. doi: 10.1172/JCI113047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Feder JN, et al. A novel MHC class I-like gene is mutated in patients with hereditary haemochromatosis. Nat Genet. 1996;13:399–408. doi: 10.1038/ng0896-399. [DOI] [PubMed] [Google Scholar]
  • 19.Roest M, et al. Heterozygosity for a hereditary hemochromatosis gene is associated with cardiovascular death in women. Circulation. 1999;100:1268–73. doi: 10.1161/01.cir.100.12.1268. [DOI] [PubMed] [Google Scholar]
  • 20.Tuomainen TP, et al. Increased risk of acute myocardial infarction in carriers of the hemochromatosis gene Cys282Tyr mutation: a prospective cohort study in men in eastern Finland. Circulation. 1999;100:1274–9. doi: 10.1161/01.cir.100.12.1274. [DOI] [PubMed] [Google Scholar]
  • 21.Vecchi C, et al. ER stress controls iron metabolism through induction of hepcidin. Science. 2009;325:877–80. doi: 10.1126/science.1176639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Swinkels DW, Wetzels JF. Hepcidin: a new tool in the management of anaemia in patients with chronic kidney disease? Nephrol Dial Transplant. 2008;23:2450–3. doi: 10.1093/ndt/gfn267. [DOI] [PubMed] [Google Scholar]
  • 23.Roy CN, et al. Hepcidin antimicrobial peptide transgenic mice exhibit features of the anemia of inflammation. Blood. 2007;109:4038–44. doi: 10.1182/blood-2006-10-051755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Feder JN, et al. The hemochromatosis gene product complexes with the transferrin receptor and lowers its affinity for ligand binding. Proc Natl Acad Sci U S A. 1998;95:1472–7. doi: 10.1073/pnas.95.4.1472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Finberg KE, et al. Mutations in TMPRSS6 cause iron-refractory iron deficiency anemia (IRIDA) Nat Genet. 2008;40:569–71. doi: 10.1038/ng.130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kerenyi MA, et al. Stat5 regulates cellular iron uptake of erythroid cells via IRP-2 and TfR-1. Blood. 2008;112:3878–88. doi: 10.1182/blood-2008-02-138339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Roetto A, et al. Hemochromatosis due to mutations in transferrin receptor 2. Blood Cells Mol Dis. 2002;29:465–70. doi: 10.1006/bcmd.2002.0585. [DOI] [PubMed] [Google Scholar]
  • 28.Uda M, et al. Genome-wide association study shows BCL11A associated with persistent fetal hemoglobin and amelioration of the phenotype of beta-thalassemia. Proc Natl Acad Sci U S A. 2008;105:1620–5. doi: 10.1073/pnas.0711566105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Menzel S, et al. A QTL influencing F cell production maps to a gene encoding a zinc-finger protein on chromosome 2p15. Nat Genet. 2007;39:1197–9. doi: 10.1038/ng2108. [DOI] [PubMed] [Google Scholar]
  • 30.Bea S, et al. Diffuse large B-cell lymphoma subgroups have distinct genetic profiles that influence tumor biology and improve gene-expression-based survival prediction. Blood. 2005;106:3183–90. doi: 10.1182/blood-2005-04-1399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Sankaran VG, et al. Human fetal hemoglobin expression is regulated by the developmental stage-specific repressor BCL11A. Science. 2008;322:1839–42. doi: 10.1126/science.1165409. [DOI] [PubMed] [Google Scholar]
  • 32.Higgs DR, Wood WG. Genetic complexity in sickle cell disease. Proc Natl Acad Sci U S A. 2008;105:11595–6. doi: 10.1073/pnas.0806633105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sandberg ML, et al. c-Myb and p300 regulate hematopoietic stem cell proliferation and differentiation. Dev Cell. 2005;8:153–66. doi: 10.1016/j.devcel.2004.12.015. [DOI] [PubMed] [Google Scholar]
  • 34.Emambokus N, et al. Progression through key stages of haemopoiesis is dependent on distinct threshold levels of c-Myb. Embo J. 2003;22:4478–88. doi: 10.1093/emboj/cdg434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gudbjartsson DF, et al. Sequence variants affecting eosinophil numbers associate with asthma and myocardial infarction. Nat Genet. 2009;41:342–7. doi: 10.1038/ng.323. [DOI] [PubMed] [Google Scholar]
  • 36.Newton-Cheh C, et al. Genome-wide association study identifies eight loci associated with blood pressure. Nat Genet. 2009 doi: 10.1038/ng.361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Todd JA, et al. Robust associations of four new chromosome regions from genome-wide analyses of type 1 diabetes. Nat Genet. 2007;39:857–64. doi: 10.1038/ng2068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hunt KA, et al. Newly identified genetic risk variants for celiac disease related to the immune response. Nat Genet. 2008;40:395–402. doi: 10.1038/ng.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Smyth DJ, et al. Shared and distinct genetic variants in type 1 diabetes and celiac disease. N Engl J Med. 2008;359:2767–77. doi: 10.1056/NEJMoa0807917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Velazquez L, et al. Cytokine signaling and hematopoietic homeostasis are disrupted in Lnk-deficient mice. J Exp Med. 2002;195:1599–611. doi: 10.1084/jem.20011883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Saxton TM, et al. Abnormal mesoderm patterning in mouse embryos mutant for the SH2 tyrosine phosphatase Shp-2. Embo J. 1997;16:2352–64. doi: 10.1093/emboj/16.9.2352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Jongmans M, et al. Genotypic and phenotypic characterization of Noonan syndrome: new data and review of the literature. Am J Med Genet A. 2005;134A:165–70. doi: 10.1002/ajmg.a.30598. [DOI] [PubMed] [Google Scholar]
  • 43.Kalidas K, et al. Genetic heterogeneity in LEOPARD syndrome: two families with no mutations in PTPN11. J Hum Genet. 2005;50:21–5. doi: 10.1007/s10038-004-0212-x. [DOI] [PubMed] [Google Scholar]
  • 44.Tartaglia M, et al. Somatic mutations in PTPN11 in juvenile myelomonocytic leukemia, myelodysplastic syndromes and acute myeloid leukemia. Nat Genet. 2003;34:148–50. doi: 10.1038/ng1156. [DOI] [PubMed] [Google Scholar]
  • 45.Tong Z, et al. Promoter polymorphism of the erythropoietin gene in severe diabetic eye and kidney complications. Proc Natl Acad Sci U S A. 2008;105:6998–7003. doi: 10.1073/pnas.0800454105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Wannamethee G, Shaper AG. Haematocrit: relationships with blood lipids, blood pressure and other cardiovascular risk factors. Thromb Haemost. 1994;72:58–64. [PubMed] [Google Scholar]
  • 47.Gagnon DR, Zhang TJ, Brand FN, Kannel WB. Hematocrit and the risk of cardiovascular disease--the Framingham study: a 34-year follow-up. Am Heart J. 1994;127:674–82. doi: 10.1016/0002-8703(94)90679-3. [DOI] [PubMed] [Google Scholar]
  • 48.Elwood PC, Waters WE, Benjamin IT, Sweetnam PM. Mortality and anaemia in women. Lancet. 1974;1:891–4. doi: 10.1016/s0140-6736(74)90346-8. [DOI] [PubMed] [Google Scholar]
  • 49.Sarnak MJ, et al. Anemia as a risk factor for cardiovascular disease in The Atherosclerosis Risk in Communities (ARIC) study. J Am Coll Cardiol. 2002;40:27–33. doi: 10.1016/s0735-1097(02)01938-1. [DOI] [PubMed] [Google Scholar]
  • 50.de Simone G, et al. Association of blood pressure with blood viscosity in american indians: the Strong Heart Study. Hypertension. 2005;45:625–30. doi: 10.1161/01.HYP.0000157526.07977.ec. [DOI] [PubMed] [Google Scholar]
  • 51.Letcher RL, Chien S, Pickering TG, Laragh JH. Elevated blood viscosity in patients with borderline essential hypertension. Hypertension. 1983;5:757–62. doi: 10.1161/01.hyp.5.5.757. [DOI] [PubMed] [Google Scholar]
  • 52.Sharp DS, et al. Mean red cell volume as a correlate of blood pressure. Circulation. 1996;93:1677–84. doi: 10.1161/01.cir.93.9.1677. [DOI] [PubMed] [Google Scholar]
  • 53.Wells RE, Jr., Merrill EW. Influence of flow properties of blood upon viscosity-hematocrit relationships. J Clin Invest. 1962;41:1591–8. doi: 10.1172/JCI104617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Ott EO, Lechner H, Aranibar A. High blood viscosity syndrome in cerebral infarction. Stroke. 1974;5:330–3. doi: 10.1161/01.str.5.3.330. [DOI] [PubMed] [Google Scholar]
  • 55.Gollob MH, et al. Identification of a gene responsible for familial Wolff-Parkinson-White syndrome. N Engl J Med. 2001;344:1823–31. doi: 10.1056/NEJM200106143442403. [DOI] [PubMed] [Google Scholar]
  • 56.Gollob MH, et al. Novel PRKAG2 mutation responsible for the genetic syndrome of ventricular preexcitation and conduction system disease with childhood onset and absence of cardiac hypertrophy. Circulation. 2001;104:3030–3. doi: 10.1161/hc5001.102111. [DOI] [PubMed] [Google Scholar]
  • 57.Beutler E, Waalen J. The definition of anemia: what is the lower limit of normal of the blood hemoglobin concentration? Blood. 2006;107:1747–50. doi: 10.1182/blood-2005-07-3046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Servin B, Stephens M. Imputation-based analysis of association studies: candidate regions and quantitative traits. PLoS Genet. 2007;3:e114. doi: 10.1371/journal.pgen.0030114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Price AL, et al. Principal components analysis corrects for stratification in genome-wide association studies. Nat Genet. 2006;38:904–9. doi: 10.1038/ng1847. [DOI] [PubMed] [Google Scholar]
  • 60.Devlin B, Roeder K. Genomic control for association studies. Biometrics. 1999;55:997–1004. doi: 10.1111/j.0006-341x.1999.00997.x. [DOI] [PubMed] [Google Scholar]
  • 61.Watkins NA, et al. A HaemAtlas: characterizing gene expression in differentiated human blood cells. Blood. 2009;113:e1–9. doi: 10.1182/blood-2008-06-162958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.da Costa Martins P, et al. P-selectin glycoprotein ligand-1 is expressed on endothelial cells and mediates monocyte adhesion to activated endothelium. Arterioscler Thromb Vasc Biol. 2007;27:1023–9. doi: 10.1161/ATVBAHA.107.140442. [DOI] [PubMed] [Google Scholar]
  • 63.Du P, Kibbe WA, Lin SM. lumi: a pipeline for processing Illumina microarray. Bioinformatics. 2008;24:1547–8. doi: 10.1093/bioinformatics/btn224. [DOI] [PubMed] [Google Scholar]
  • 64.Online Mendelian Inheritance in Man, OMIM (TM) McKusick-Nathans Institute of Genetic Medicine. Johns Hopkins University; National Center for Biotechnology Information, National Library of Medicine; Baltimore, MD: Bethesda, MD: date of download. [Google Scholar]
  • 65.Becker KG, Barnes KC, Bright TJ, Wang SA. The genetic association database. Nat Genet. 2004;36:431–2. doi: 10.1038/ng0504-431. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1
2

RESOURCES