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:

HAX1 deficiency causes autosomal recessive severe congenital neutropenia (Kostmann disease)

Abstract

Autosomal recessive severe congenital neutropenia (SCN)1 constitutes a primary immunodeficiency syndrome associated with increased apoptosis in myeloid cells2,3, yet the underlying genetic defect remains unknown. Using a positional cloning approach and candidate gene evaluation, we identified a recurrent homozygous germline mutation in HAX1 in three pedigrees. After further molecular screening of individuals with SCN, we identified 19 additional affected individuals with homozygous HAX1 mutations, including three belonging to the original pedigree described by Kostmann1. HAX1 encodes the mitochondrial protein HAX1, which has been assigned functions in signal transduction4 and cytoskeletal control5,6. Here, we show that HAX1 is critical for maintaining the inner mitochondrial membrane potential and protecting against apoptosis in myeloid cells. Our findings suggest that HAX1 is a major regulator of myeloid homeostasis and underline the significance of genetic control of apoptosis in neutrophil development.

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: Haplotypes on chromosome 1q.
Figure 2: Bone marrow phenotype, HAX1 genotype and HAX1 expression.
Figure 3: Apoptosis and mitochondrial membrane potential in HAX1-deficient granulocytes.
Figure 4: Reconstitution of ΔΨm in myeloid progenitor cells and fibroblasts after retroviral HAX1 gene transfer.

Similar content being viewed by others

Accession codes

Accessions

GenBank/EMBL/DDBJ

References

  1. Kostmann, R. Infantile genetic agranulocytosis (Agranulocystosis infantilis hereditaria): a new recessive lethal disease in man. Acta Paediatr. 45 (Suppl.), 1–78 (1956).

    CAS  Google Scholar 

  2. Carlsson, G. et al. Kostmann syndrome: severe congenital neutropenia associated with defective expression of Bcl-2, constitutive mitochondrial release of cytochrome c, and excessive apoptosis of myeloid progenitor cells. Blood 103, 3355–3361 (2004).

    Article  CAS  PubMed  Google Scholar 

  3. Cario, G. et al. Heterogeneous expression pattern of pro- and anti-apoptotic factors in myeloid progenitor cells of patients with severe congenital neutropenia treated with granulocyte colony-stimulating factor. Br. J. Haematol. 129, 275–278 (2005).

    Article  PubMed  Google Scholar 

  4. Suzuki, Y. et al. HAX-1, a novel intracellular protein, localized on mitochondria, directly associates with HS1, a substrate of Src family tyrosine kinases. J. Immunol. 158, 2736–2744 (1997).

    CAS  PubMed  Google Scholar 

  5. Gallagher, A.R., Cedzich, A., Gretz, N., Somlo, S. & Witzgall, R. The polycystic kidney disease protein PKD2 interacts with Hax-1, a protein associated with the actin cytoskeleton. Proc. Natl. Acad. Sci. USA 97, 4017–4022 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Radhika, V., Onesime, D., Ha, J.H. & Dhanasekaran, N. Gα13 stimulates cell migration through cortactin-interacting protein Hax-1. J. Biol. Chem. 279, 49406–49413 (2004).

    Article  CAS  PubMed  Google Scholar 

  7. Welte, K., Zeidler, C. & Dale, D. Severe congenital neutropenia. Semin. Hematol. 43, 189–195 (2006).

    Article  CAS  PubMed  Google Scholar 

  8. Dale, D.C. et al. Mutations in the gene encoding neutrophil elastase in congenital and cyclic neutropenia. Blood 96, 2317–2322 (2000).

    CAS  PubMed  Google Scholar 

  9. Sharp, T.V. et al. K15 protein of Kaposi's sarcoma-associated herpesvirus is latently expressed and binds to HAX-1, a protein with antiapoptotic function. J. Virol. 76, 802–816 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cilenti, L. et al. Regulation of HAX-1 anti-apoptotic protein by Omi/HtrA2 protease during cell death. J. Biol. Chem. 279, 50295–50301 (2004).

    Article  CAS  PubMed  Google Scholar 

  11. Elsner, J., Roesler, J., Emmendörffer, A., Lohmann-Matthes, M.L. & Welte, K. Abnormal regulation in the signal transduction in neutrophils from patients with severe congenital neutropenia: relation of impaired mobilization of cytosolic free calcium to altered chemotaxis, superoxide anion generation and F-actin content. Exp. Hematol. 21, 38–46 (1993).

    CAS  PubMed  Google Scholar 

  12. Carlsson, G. & Fasth, A. Infantile genetic agranulocytosis, morbus Kostmann: Presentation of six cases from the original “Kostmann family” and a review. Acta Paediatr. 90, 757–764 (2001).

    Article  CAS  PubMed  Google Scholar 

  13. Horwitz, M., Benson, K.F., Person, R.E., Aprikyan, A.G. & Dale, D.C. Mutations in ELA2, encoding neutrophil elastase, define a 21-day biological clock in cyclic haematopoiesis. Nat. Genet. 23, 433–436 (1999).

    Article  CAS  PubMed  Google Scholar 

  14. Gilman, P.A., Jackson, D.P. & Guild, H.G. Congenital agranulocytosis: prolonged survival and terminal acute leukemia. Blood 36, 576–585 (1970).

    CAS  PubMed  Google Scholar 

  15. Rosenberg, P.S. et al. The incidence of leukemia and mortality from sepsis in patients with severe congenital neutropenia receiving long-term G-CSF therapy. Blood 107, 4628–4635 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Dong, F. et al. Mutations in the gene for the granulocyte colony-stimulating-factor receptor in patients with acute myeloid leukemia preceded by severe congenital neutropenia. N. Engl. J. Med. 333, 487–493 (1995).

    Article  CAS  PubMed  Google Scholar 

  17. Green, D.R. & Kroemer, G. The pathophysiology of mitochondrial death. Science 305, 626–629 (2004).

    Article  CAS  PubMed  Google Scholar 

  18. Newmeyer, D.D. & Ferguson-Miller, S. Mitochondria: releasing power for life and unleashing the machineries of death. Cell 112, 481–490 (2003).

    Article  CAS  PubMed  Google Scholar 

  19. Maianski, N.A. et al. Functional characterization of mitochondria in neutrophils: a role restricted to apoptosis. Cell Death Differ. 11, 143–153 (2004).

    Article  CAS  PubMed  Google Scholar 

  20. Kroemer, G. & Reed, J.C. Mitochondrial control of cell death. Nat. Med. 6, 513–519 (2000).

    Article  CAS  PubMed  Google Scholar 

  21. Gross, A., McDonnell, J.M. & Korsmeyer, S.J. BCL-2 family members and the mitochondria in apoptosis. Genes Dev. 13, 1899–1911 (1999).

    Article  CAS  PubMed  Google Scholar 

  22. Opferman, J.T. & Korsmeyer, S.J. Apoptosis in the development and maintenance of the immune system. Nat. Immunol. 4, 410–415 (2003).

    Article  CAS  PubMed  Google Scholar 

  23. Hamasaki, A. et al. Accelerated neutrophil apoptosis in mice lacking A1-a, a subtype of the bcl-2-related A1 gene. J. Exp. Med. 188, 1985–1992 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Maianski, N.A., Mul, F.P.J., van Buul, J.D., Roos, D. & Kuijpers, T.W. Granulocyte colony-stimulating factor inhibits the mitochondria-dependent activation of caspase-3 in neutrophils. Blood 99, 672–679 (2002).

    Article  CAS  PubMed  Google Scholar 

  25. Dufva, M., Olsson, M. & Rymo, L. Epstein-Barr virus nuclear antigen 5 interacts with HAX-1, a possible component of the B-cell receptor signalling pathway. J. Gen. Virol. 82, 1581–1587 (2001).

    Article  CAS  PubMed  Google Scholar 

  26. Kawaguchi, Y. et al. Interaction of Epstein-Barr virus nuclear antigen leader protein (EBNA-LP) with HS1-associated protein X-1: implication of cytoplasmic function of EBNA-LP. J. Virol. 74, 10104–10111 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Yedavalli, V.S. et al. Human immunodeficiency virus type 1 Vpr interacts with antiapoptotic mitochondrial protein HAX-1. J. Virol. 79, 13735–13746 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Cottingham, R.W., Jr., Idury, R.M. & Schäffer, A.A. Faster sequential genetic linkage computations. Am. J. Hum. Genet. 53, 252–263 (1993).

    PubMed  PubMed Central  Google Scholar 

  29. Schäffer, A.A., Gupta, S.K., Shriram, K. & Cottingham, R.W. Jr. Avoiding recomputation in linkage analysis. Hum. Hered. 44, 225–237 (1994).

    Article  PubMed  Google Scholar 

  30. Klein, C., Bueler, H.R. & Mulligan, R.C. Comparative analysis of genetically modified dendritic cells and tumor cells as therapeutic cancer vaccines. J. Exp. Med. 191, 1699–1708 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are indebted to the participants and their families and to M. Ballmaier and C. Reimers (Central Medical School Hannover, Flow Cytometry Laboratory) for their assistance. We thank all colleagues referring and registering patients at the International SCN Registry. We wish to acknowledge the genetic studies performed by M. Entesarian, K. Ericson and M. Nordenskjöld. Plasmid K83.pHCMV-GALVenv was a gift from C. Baum (Hannover Medical School). This study was supported by a grant from the Deutsche Forschungsgemeinschaft (DFG-KliFo 110), by the German José Carreras Leukemia Foundation, by the Bundesministerium für Bildung und Forschung (Congenital Bone Marrow Failure Syndromes) and in part by the Intramural Research program of the US National Institutes of Health, National Library of Medicine.

Author information

Authors and Affiliations

Authors

Contributions

C.K. designed and directed the study; obtained clinical samples; taught and supervised G.A., I.S., K.B., C.R. and G.B.; provided laboratory resources and wrote the manuscript with help from B.G. and A.A.S. The manuscript was then reviewed and approved by all authors. M. Grudzien did the genotyping for linkage analysis and sequenced candidate genes. G.A. performed all gene transfer studies and functional assays on myeloid cells and fibroblasts. M. Germeshausen sequenced HAX1, ELA2 and CSFR3 and comprehensively analyzed genetic data. I.S. discovered the first HAX1 mutation and performed sequencing and protein blotting. A.A.S. chose markers to genotype in the linkage region and performed linkage analysis computations. K.B. performed functional immunological assays. C.R. performed functional neutrophil studies and taught G.A. C.Z. cared for patients and collected and curated data in the SCN patient registry. B.S. collected and curated data in the SCN patient registry. N.R. treated patients in Iran and ascertained their samples for this study. G.B. performed functional neutrophil studies and sequenced candidate genes. G.C. and J.-I.H. initiated the Swedish Kostmann family project; G.C. treated the patients, and J.-I.H. and J.P. supervised the project with the support of B.F. N.D. was responsible for Swedish Kostmann gene studies. M.M. sequenced genomic samples from the Kostmann family. B.G. provided laboratory resources, organized patient samples, supervised M. Grudzien and assisted A.A.S. K.W. provided laboratory resources and resources for SCN registry and helped to initiate and carry out the study. M. Grudzien and G.A. contributed equally to this work and are considered acquo loco.

Corresponding author

Correspondence to Christoph Klein.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Fig. 1

Germline HAX1 mutation in members of the original Kostmann pedigree. (PDF 433 kb)

Supplementary Fig. 2

Increased apoptosis and enzymatic caspase 3/7 activity in HAX1-deficient granulocytes. (PDF 965 kb)

Supplementary Fig. 3

Increased loss of mitochondrial membrane potential ΔΨm in HAX1-deficient fibroblasts. (PDF 743 kb)

Supplementary Table 1

Immunological analysis of HAX1-deficient patients. (PDF 65 kb)

Supplementary Table 2

HAX2 genomic PCR primers and conditions. (PDF 52 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Klein, C., Grudzien, M., Appaswamy, G. et al. HAX1 deficiency causes autosomal recessive severe congenital neutropenia (Kostmann disease). Nat Genet 39, 86–92 (2007). https://doi.org/10.1038/ng1940

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng1940

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