Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Rare and low-frequency coding variants in CXCR2 and other genes are associated with hematological traits

Abstract

Hematological traits are important clinical parameters. To test the effects of rare and low-frequency coding variants on hematological traits, we analyzed hemoglobin concentration, hematocrit levels, white blood cell (WBC) counts and platelet counts in 31,340 individuals genotyped on an exome array. We identified several missense variants in CXCR2 associated with reduced WBC count (gene-based P = 2.6 × 10−13). In a separate family-based resequencing study, we identified a CXCR2 frameshift mutation in a pedigree with congenital neutropenia that abolished ligand-induced CXCR2 signal transduction and chemotaxis. We also identified missense or splice-site variants in key hematopoiesis regulators (EPO, TFR2, HBB, TUBB1 and SH2B3) associated with blood cell traits. Finally, we were able to detect associations between a rare somatic JAK2 mutation (encoding p.Val617Phe) and platelet count (P = 3.9 × 10−22) as well as hemoglobin concentration (P = 0.002), hematocrit levels (P = 9.5 × 10−7) and WBC count (P = 3.1 × 10−5). In conclusion, exome arrays complement genome-wide association studies in identifying new variants that contribute to complex human traits.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Association results in the meta-analysis of the MHI and WHI cohorts (n = 24,814) for hematocrit levels at the TFR2-EPO locus on chromosome 7 and for platelet count at the SH2B3 locus on chromosome 12.
Figure 2: Rare and low-frequency missense variants in CXCR2 are associated with lower WBC count.
Figure 3: Functional characterization of the CXCR2fs mutant receptor.

Similar content being viewed by others

References

  1. Nalls, M.A. et al. Multiple loci are associated with white blood cell phenotypes. PLoS Genet. 7, e1002113 (2011).

    Article  CAS  Google Scholar 

  2. Gieger, C. et al. New gene functions in megakaryopoiesis and platelet formation. Nature 480, 201–208 (2011).

    Article  CAS  Google Scholar 

  3. van der Harst, P. et al. Seventy-five genetic loci influencing the human red blood cell. Nature 492, 369–375 (2012).

    Article  CAS  Google Scholar 

  4. Zhou, Z. et al. USF and NF-E2 cooperate to regulate the recruitment and activity of RNA polymerase II in the β-globin gene locus. J. Biol. Chem. 285, 15894–15905 (2010).

    Article  CAS  Google Scholar 

  5. de la Chapelle, A., Sistonen, P., Lehvaslaiho, H., Ikkala, E. & Juvonen, E. Familial erythrocytosis genetically linked to erythropoietin receptor gene. Lancet 341, 82–84 (1993).

    Article  CAS  Google Scholar 

  6. Biasiotto, G. et al. New TFR2 mutations in young Italian patients with hemochromatosis. Haematologica 93, 309–310 (2008).

    Article  CAS  Google Scholar 

  7. Pelucchi, S. et al. Expression of hepcidin and other iron-related genes in type 3 hemochromatosis due to a novel mutation in transferrin receptor-2. Haematologica 94, 276–279 (2009).

    Article  CAS  Google Scholar 

  8. Qayyum, R. et al. A meta-analysis and genome-wide association study of platelet count and mean platelet volume in African Americans. PLoS Genet. 8, e1002491 (2012).

    Article  CAS  Google Scholar 

  9. Forand, A. et al. EKLF-driven PIT1 expression is critical for mouse erythroid maturation in vivo and in vitro. Blood 121, 666–678 (2013).

    Article  CAS  Google Scholar 

  10. Baxter, E.J. et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 365, 1054–1061 (2005).

    Article  CAS  Google Scholar 

  11. James, C. et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 434, 1144–1148 (2005).

    Article  CAS  Google Scholar 

  12. Kralovics, R. et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N. Engl. J. Med. 352, 1779–1790 (2005).

    Article  CAS  Google Scholar 

  13. Levine, R.L. et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell 7, 387–397 (2005).

    Article  CAS  Google Scholar 

  14. Bohinjec, J. Myelokathexis: chronic neutropenia with hyperplastic bone marrow and hypersegmented neutrophils in two siblings. Blut 42, 191–196 (1981).

    Article  CAS  Google Scholar 

  15. Xu, X. et al. JAK2V617F: prevalence in a large Chinese hospital population. Blood 109, 339–342 (2007).

    Article  CAS  Google Scholar 

  16. Sidon, P., El Housni, H., Dessars, B. & Heimann, P. The JAK2V617F mutation is detectable at very low level in peripheral blood of healthy donors. Leukemia 20, 1622 (2006).

    Article  CAS  Google Scholar 

  17. Vannucchi, A.M., Pieri, L. & Guglielmelli, P. JAK2 allele burden in the myeloproliferative neoplasms: effects on phenotype, prognosis and change with treatment. Ther. Adv. Hematol. 2, 21–32 (2011).

    Article  CAS  Google Scholar 

  18. Oh, S.T. When the brakes are lost: LNK dysfunction in mice, men, and myeloproliferative neoplasms. Ther. Adv. Hematol. 2, 11–19 (2011).

    Article  CAS  Google Scholar 

  19. Bersenev, A., Wu, C., Balcerek, J. & Tong, W. Lnk controls mouse hematopoietic stem cell self-renewal and quiescence through direct interactions with JAK2. J. Clin. Invest. 118, 2832–2844 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Beck, L. et al. The phosphate transporter PiT1 (Slc20a1) revealed as a new essential gene for mouse liver development. PLoS ONE 5, e9148 (2010).

    Article  Google Scholar 

  21. Gerstein, M.B. et al. Architecture of the human regulatory network derived from ENCODE data. Nature 489, 91–100 (2012).

    Article  CAS  Google Scholar 

  22. McMullin, M.F., Wu, C., Percy, M.J. & Tong, W. A nonsynonymous LNK polymorphism associated with idiopathic erythrocytosis. Am. J. Hematol. 86, 962–964 (2011).

    Article  CAS  Google Scholar 

  23. Macari, E.R. & Lowrey, C.H. Induction of human fetal hemoglobin via the NRF2 antioxidant response signaling pathway. Blood 117, 5987–5997 (2011).

    Article  CAS  Google Scholar 

  24. Zeller, T. et al. Genetics and beyond—the transcriptome of human monocytes and disease susceptibility. PLoS ONE 5, e10693 (2010).

    Article  Google Scholar 

  25. Stranger, B.E. et al. Population genomics of human gene expression. Nat. Genet. 39, 1217–1224 (2007).

    Article  CAS  Google Scholar 

  26. Barrett, J.C. et al. Genome-wide association study and meta-analysis find that over 40 loci affect risk of type 1 diabetes. Nat. Genet. 41, 703–707 (2009).

    Article  CAS  Google Scholar 

  27. Sawcer, S. et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature 476, 214–219 (2011).

    Article  CAS  Google Scholar 

  28. Jostins, L. et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 491, 119–124 (2012).

    Article  CAS  Google Scholar 

  29. Stasi, R. Immune thrombocytopenia: pathophysiologic and clinical update. Semin. Thromb. Hemost. 38, 454–462 (2012).

    Article  CAS  Google Scholar 

  30. Johnston, C.M. et al. Large-scale population study of human cell lines indicates that dosage compensation is virtually complete. PLoS Genet. 4, e9 (2008).

    Article  Google Scholar 

  31. Cernelc, P. et al. Effects of molgramostim, filgrastim and lenograstim in the treatment of myelokathexis. Pflugers Arch. 440, R81–R82 (2000).

    Article  CAS  Google Scholar 

  32. Park, S.H. et al. Structure of the chemokine receptor CXCR1 in phospholipid bilayers. Nature 491, 779–783 (2012).

    Article  CAS  Google Scholar 

  33. Salchow, K. et al. A common intracellular allosteric binding site for antagonists of the CXCR2 receptor. Br. J. Pharmacol. 159, 1429–1439 (2010).

    Article  CAS  Google Scholar 

  34. Marioni, J.C. et al. Breaking the waves: improved detection of copy number variation from microarray-based comparative genomic hybridization. Genome Biol. 8, R228 (2007).

    Article  Google Scholar 

  35. Sabeti, P.C. et al. Genome-wide detection and characterization of positive selection in human populations. Nature 449, 913–918 (2007).

    Article  CAS  Google Scholar 

  36. Bird, C.P. et al. Fast-evolving noncoding sequences in the human genome. Genome Biol. 8, R118 (2007).

    Article  Google Scholar 

  37. ENCODE Project Consortium. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature 447, 799–816 (2007).

  38. Hernandez, P.A. et al. Mutations in the chemokine receptor gene CXCR4 are associated with WHIM syndrome, a combined immunodeficiency disease. Nat. Genet. 34, 70–74 (2003).

    Article  CAS  Google Scholar 

  39. Beaudoin, M. et al. Pooled DNA resequencing of 68 myocardial infarction candidate genes in French Canadians. Circ. Cardiovasc. Genet. 5, 547–554 (2012).

    Article  CAS  Google Scholar 

  40. Women's Health Initiative Study Group. Design of the Women's Health Initiative clinical trial and observational study. Control. Clin. Trials 19, 61–109 (1998).

  41. Völzke, H. et al. Cohort profile: the study of health in Pomerania. Int. J. Epidemiol. 40, 294–307 (2011).

    Article  Google Scholar 

  42. Goldstein, J.I. et al. zCall: a rare variant caller for array-based genotyping: genetics and population analysis. Bioinformatics 28, 2543–2545 (2012).

    Article  CAS  Google Scholar 

  43. Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 81, 559–575 (2007).

    Article  CAS  Google Scholar 

  44. 1000 Genomes Project Consortium. An integrated map of genetic variation from 1,092 human genomes. Nature 491, 56–65 (2012).

  45. Price, A.L. et al. Principal components analysis corrects for stratification in genome-wide association studies. Nat. Genet. 38, 904–909 (2006).

    Article  CAS  Google Scholar 

  46. Yang, J., Lee, S.H., Goddard, M.E. & Visscher, P.M. GCTA: a tool for genome-wide complex trait analysis. Am. J. Hum. Genet. 88, 76–82 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. R Core Team. R: A Language and Environment for Statistical Computing (R Foundation for Statistical Computing, Vienna, 2013).

  48. Liu, D.J. et al. Meta-analysis of gene-level tests for rare variant association. Nat. Genet. 46, 200–204 (2014).

    Article  CAS  Google Scholar 

  49. Willer, C.J., Li, Y. & Abecasis, G.R. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics 26, 2190–2191 (2010).

    Article  CAS  Google Scholar 

  50. Kent, W.J. et al. The human genome browser at UCSC. Genome Res. 12, 996–1006 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Rozen, S. & Skaletsky, H. Primer3 on the WWW for general users and for biologist programmers. Methods Mol. Biol. 132, 365–386 (2000).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank all participants and staff of the MHI Biobank and acknowledge the technical support of the Beaulieu-Saucier MHI Pharmacogenomic Center. This work was supported by the Centre of Excellence in Personalized Medicine (CEPMed), Fonds de Recherche du Québec–Santé (FRQS), the Canada Research Chair program and the MHI Foundation. The WHI program is funded by the National Heart, Lung, and Blood Institute, the US National Institutes of Health and the US Department of Health and Human Services (HHSN268201100046C, HHSN268201100001C, HHSN268201100002C, HHSN268201100003C, HHSN268201100004C and HHSN271201100004C). Exome chip data and analysis were supported through the Exome Sequencing Project (NHLBI RC2 HL-102924, RC2 HL-102925 and RC2 HL-102926), the Genetics and Epidemiology of Colorectal Cancer Consortium (NCI CA137088), the Genomics and Randomized Trials Network (NHGRI U01-HG005152) and a National Cancer Institute training grant (R25CA094880). The authors thank the WHI investigators and staff for their dedication and the study participants for making the program possible. SHIP is part of the Community Medicine Research network of the University of Greifswald, Germany, which is funded by the Federal Ministry of Education and Research (grants 01ZZ9603, 01ZZ0103 and 01ZZ0403), the Ministry of Cultural Affairs, as well as the Social Ministry of the Federal State of Mecklenburg–West Pomerania, and the network Greifswald Approach to Individualized Medicine (GANI_MED) funded by the Federal Ministry of Education and Research (grant 03IS2061A). Generation of ExomeChip data has been supported by the Federal Ministry of Education and Research (grant 03Z1CN22) and the Federal State of Mecklenburg–West Pomerania. The University of Greifswald is a member of the Center of Knowledge Interchange program of Siemens AG and the Caché Campus program of InterSystems. G.A.D. acknowledges support from US National Institutes of Health award P01AI061093.

Author information

Authors and Affiliations

Authors

Contributions

P.L.A., G.A.D., A.P.R. and G.L. conceived and designed the experiments. P.L.A., A.T., U.S., A.O., K.S.L., G.A.D., A.P.R. and G.L. performed the experiments. P.L.A., A.T., U.S., A.O., K.S.L., G.A.D., A.P.R. and G.L. analyzed the data. All authors contributed reagents and materials. P.L.A., G.A.D., A.P.R. and G.L. wrote the manuscript with contributions from all authors.

Corresponding authors

Correspondence to George A Diaz, Alexander P Reiner or Guillaume Lettre.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Quantile-quantile plots of single-variant association results.

(a) Hematocrit. (b) Hemoglobin. (c) White blood cell count. (d) Platelet count.

Supplementary Figure 2 Quantile-quantile plots of gene-based burden T1 results.

(a) Hematocrit. (b) Hemoglobin. (c) White blood cell count. (d) Platelet count. For each gene, we only considered missense, nonsense and splice-site variants with a minor allele frequency of <1%.

Supplementary Figure 3 Quantile-quantile plots of gene-based SKAT results.

(a) Hematocrit. (b) Hemoglobin. (c) White blood cell count. (d) Platelet count. For each gene, we only considered missense, nonsense and splice-site variants with a minor allele frequency of <5%.

Supplementary Figure 4 Impact of the mutation p.Asp70Asn in EPO.

(a) Crystal structure of erythropoietin (EPO, yellow) with two erythropoietin receptor subunits (EPOR, green and red)24. The white circle indicates the interface between EPO and EPOR magnified in (b) and (c). In (b) and (c), the white circles highlight residue 70 (43 after cleavage of the propeptide). Replacing Asp70 (b) by Asn70 (c) may destabilize the EPO loop that interacts with EPOR because it disrupts a hydrogen bond between Asp70 and Thr71.

Supplementary Figure 5 Intensity plots for JAK2 p.Val617Phe in the Montreal Heart Institute (MHI; left) and the Study of Health in Pomerania (SHIP; right) genotyped on the Illumina HumanExome BeadChip.

Density plots for the Women's Health Initiative were not available.

Supplementary Figure 6 Mutations in SH2B3 may interfere with JAK2 binding.

Crystal structure of the SH2-B SH2 domain (beige) with JAK2 phosphotyrosine peptide (pTyr813, yellow)25. JAK2 pTyr813 interacts with arginine residues in the SH2-B BC loop. The glutamate residues identified in our association study (Glu395 and Glu400, green) correspond to Glu558 and Glu563 in the SH2-B sequence. They are directly in the BC loop that interacts with JAK2. Glu563 interacts with Arg578.

Supplementary Figure 7 UCSC ENCODE tracks for the rs1465788 variant near ZFP36L1.

The rs1465788 variant appears to be in a K562 FAIRE nucleosome depletion peak. rs1465788 is also in high LD with rs2236263 (r2 = 0.91 in CEU), rs3742887 (r2 = 0.99 in CEU) and rs6573857 (r2 = 1 in CEU), which all lie in an active promoter for ZFP36L1.

Supplementary Figure 8 Crystal structure of CXCR1 in a phospholipid bilayer.

CXCR2, like CXCR1 and CXCR4, is a protein with seven transmembrane (TM) domains. In purple are the CXCR1 residues that correspond to the missense mutations identified in CXCR2 (two CXCR2 residues, Met6 and Ala37, do not have their CXCR1 equivalent on this structure). In parentheses are the corresponding CXCR2 residues. CXCR2 Arg289 and Arg294 are in TM domains. The remaining four CXCR2 residues (Arg144, Arg236, Arg248, His332) are in intracellular loops.

Supplementary Figure 9 Analysis of CXCR2 in a myelokathexis pedigree.

(a) A myelokathexis pedigree with two affected (filled) and two unaffected (open) daughters is shown at left. The CXCR2 sequence trace shows a single-base deletion corresponding to coding sequence position 968 in individual I-2 (unaffected) compared to an unrelated control (arrow). (b) An NcoI recognition site (CCATGG) that gives a 325-bp fragment when a wild-type amplicon is digested is destroyed by the c.968delA mutation so that the 375-bp undigested amplicon persists. Affected siblings II-2 and II-4 are homozygous for the deletion mutation, parents and sibling II-1 are heterozygous for the mutation, and sibling II-3 is homozygous for the wild-type allele. White blood cell (WBC) counts for each family member are shown below. (c) A structural model of CXCR2 with the conserved GPCR motifs (purple, intracellular loop 2; dark blue, DRY; green, NPXXY), the mutant frameshift peptide (orange) and the truncated wild-type tail (red) indicated. The frameshift is located in helix 8, which is predicted to be parallel to the plasma membrane. Amino acid sequences from the wild-type and mutant CXCR2 constructs are shown. The CXCR2fs construct contains a six-amino-acid frameshift peptide (LDSSRF). Amino acid colors correspond to those used in the structure model. The black bar over the primary sequence indicates the location of a highly conserved LLKIL dileucine motif that is critical for receptor-mediated chemotaxis27.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9, Supplementary Tables 1–7 and Supplementary Note (PDF 2538 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Auer, P., Teumer, A., Schick, U. et al. Rare and low-frequency coding variants in CXCR2 and other genes are associated with hematological traits. Nat Genet 46, 629–634 (2014). https://doi.org/10.1038/ng.2962

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2962

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing