Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Dec 1.
Published in final edited form as: Curr Opin Allergy Clin Immunol. 2013 Dec;13(6):589–595. doi: 10.1097/ACI.0000000000000011

Inborn errors of the development of human Natural Killer cells

Emmanuelle Jouanguy 1,2,3,*, Laure Gineau 1,2, Julien Cottineau 1,2, Vivien Beziat 4, Eric Vivier 5,6,7,8, Jean-Laurent Casanova 1,2,3,9
PMCID: PMC4077347  NIHMSID: NIHMS588885  PMID: 24135998

Abstract

Purpose of review

Inborn errors of human natural killer (NK) cells may affect the development of these cells, their function, or both. There are two broad categories of genetic defects of NK-cell development, depending on whether the deficiency is apparently specific to NK cells or clearly affects multiple hematopoietic lineages. We review here recent progress in the genetic dissection of NK deficiencies (NKDs).

Recent findings

Patients with severe combined immunodeficiencies (SCID) bearing mutations of ADA, AK2, IL2RG and JAK3 genes present NKDs and are prone to a broad range of infections. Patients with GATA2 deficiency are susceptible to both mycobacterial and viral infections and display NKD and a lack of monocytes. Patients with MCM4 deficiency display an apparently selective NKD associated with viral infections, but they also display various non hematopoietic phenotypes, including adrenal insufficiency and growth retardation.

Summary

These studies have initiated genetic dissection of the development of human NK cells. Further studies are warranted, including the search for genetic etiologies of NKD in particular. This research may lead to the discovery of molecules specifically controlling the development of NK cells and to improvements in our understanding of the hitherto elusive function of these cells in humans.

Keywords: Natural Killer cells, Immunodeficiencies, Genetic diseases

Introduction

Natural Killer (NK) cells are innate lymphoid cells (ILCs) constituting the third most abundant lymphocyte population in the peripheral blood [1]. They are thought to play an important role in the course of pregnancy, antiviral immunity and antitumoral immunity. Unlike B and T lymphocytes, they do not express somatically rearranged antigen receptors. Instead, they bear a diverse repertoire of receptors with activating or inhibitory properties [24]. Imbalances between these signals trigger NK cell activation, resulting in NK cell cytotoxicity and/or the production of cytokines [5, 6]. NK cells have been extensively characterized over almost four decades, but the genetic control of human NK cell development and the function of these cells in host defense remain to be elucidated.

Major advances have been made in the dissection of NK cell development and maturation in mice (7•]. The developmental sequence from NK cell precursors to mature NK cells has been divided into seven stages, each corresponding to the expression of a different pattern of surface markers and requiring different transcription factors [8•, 9]. NK cell development begins in the bone marrow and is completed in secondary lymphoid tissues (SLT). Indeed, NK cell progenitors may leave the bone marrow at a very early stage to seed SLT within which they differentiate [10]. Some of the maturation steps require interaction with other cells, such as neutrophils and monocytes in particular [11, 12••]. The requirement of NK cells for antiviral immunity is well established in mice [1315]. It has been recently reported that mouse NK cells also display features of adaptive immunity, particularly during viral infections [1618]. Through their cytokine production and cytotoxic functions, NK cells can act at various steps of the immune response exerting their effects on antigen loads, dendritic cells and T cells [19•21].

In humans, a model of NK cell differentiation has been proposed based on the expression of various markers, such as CD34, CD56, CD57, CD62L, CD94, CD117 and KIRs [2225]. However, the development and function of these cells remain elusive, due to the rarity of pure NK cell deficiencies (NKDs) [2628]. Inherited NKDs, in which NK cells counts are below 100 cells/mm3, are generally associated with a lack of other lymphoid subsets, as in patients with severe combined immunodeficiency (SCID), which is defined as a lack of autologous T-cells. There are also combined NKDs associated with normal T-cell counts but low levels of myeloid cells, as in patients with the MonoMac syndrome [29, 30]. Finally, selective NKDs have been reported in individuals with no other hematopoietic phenotype and, in at least one case, the genetic basis of the deficit has been determined [3135]. We will review here the known forms of inherited NKDs (selective or combined). We will not review the inborn errors of NK cell function in which NK cells are present, such as familial hemophagocytic lymphohistiocytosis (e.g. perforin deficiency), which have been reviewed elsewhere [27].

Deficits of early hematopoiesis: AK2 and ADA deficiency

Patients with adenylate kinase 2 (AK2) deficiency (also known as reticular dysgenesis), or with adenosine deaminase (ADA) deficiency display severe immunodeficiency, with T B NK SCID associated with an absence of granulocytes in AK2-deficient patients and hypogranular neutrophils in ADA-deficient patients [36]. Both morbid genes are involved in metabolism: AK2 plays a key role in the adenosine diphosphate (ADP) generation and ADA is a key enzyme of the pathway for adenosine and deoxyadenosine deamination [37, 38]. The pathogenesis of the SCID phenotype in patients with AK2 and ADA deficiencies remains unclear. Moreover, NK cell lymphopenia has not been investigated in patients or the corresponding knockout mice. AK2-deficient patients have no cells developing beyond the myelocyte stage and scattered maturing lymphocytes in bone marrow (BM) and a dysplastic thymus [39, 40]. They display spontaneously high fibroblast apoptosis rates, probably due to impaired ADP flux in the mitochondrial matrix [41]. These findings suggest that NK cell differentiation in BM is sensitive to the mitochondrial control of energy balance, apoptosis, or both. In ADA-deficient patients, the accumulation of purine metabolites is toxic to thymocytes and splenic B cells leading to their apoptosis [4244]. The BM has a normal number of NK cell progenitors, suggesting that the lack of NK cells results from high rates of apoptosis during the maturation in SLT [45, 46]. All patients display a broad spectrum of life-threatening infections, including viral, bacterial and fungal infections, with other clinical signs, probably due to the metabolic dysregulation (Table 1). The broad nature of the lymphoid and myeloid defect in these patients and their non hematopoietic phenotypes make it impossible to determine whether and which infections result from the lack of NK cells.

Table 1.

Combined and isolated quantitative NK cells deficiencies

Mode of
transmission
Disease
Mutated Gene NK cell
number/phenotype
Other immunological
abnormalities
Other clinical
phenotypes
Infectious susceptibility Cancer phenotype References
Autosomal recessive

Metabolic
T-B-SCID
ADA Markedly decreased No T and B cells
Hypogranular neutrophils
Autoimmunity
Hepatic and renal diseases
Neurological abnormalities
Skeletal alterations
Multiple life-threatening infections 36
Autosomale recessive
Reticular dysgenesis
AK2 Absent No leukocytes Deafness Multiple life-threatening infections 3941
X-linked
T-B+ SCID
IL2RG Markedly decreased No T cells Multiple life-threatening infections 47, 48, 49
Autosomal recessive
T-B+ SCID
JAK3 Markedly decreased No T cells Multiple life-threatening infections 48, 49, 50
Autosomal recessive
Growth hormone insensitivity syndrome
STAT5b Decreased Low T cell numbers
Treg dysfunction
Hyper-gammaglobulinemia
Growth retardation
Autoimmunity
Allergy
Chronic pulmonary diseases
Herpes infections
Eczema
5155
Autosomal dominant
Mono-Mac syndrome
GATA-2 Markedly decreased
No CD56bright
Fewer CD56dim
Few or no monocytes, dendritic cells, B lymphocytes Autoimmunity
Primary lymphedema
Pulmonary proteinosis
Papillomavirus infections
Herpes infections
Non-tuberculous mycobacteria
Fungal infections
Myelodysplastic syndrome
Leukemia
EBV-associated smooth muscle tumors
29, 30, 5662
Autosomal recessive MCM4 Markedly decreased
Normal CD56bright
Fewer CD56dim
Growth retardation
Adrenal insufficiency
Herpes infections EBV lymphoma 31, 63, 64

Deficits of early T and NK cell development: IL-2Rγ and JAK3 deficiency

Janus kinase 3 (JAK3) is the signaling adaptor associated with the common γ chain (γc), a component of the multichain receptors for IL-2, -4, -7, -9, -15 and -21 [65]. Patients with JAK3 or IL-2Rγc deficiency generally have no T and NK cells, but normal or particularly large numbers of nonfunctional B cells (T B+ NK SCID phenotype) [47, 50, 66]. Six cytokines are known to use the common γ chain. Two of these cytokines, IL-2 and IL-7 are essential for T-cell development and function, as patients with IL-2Rα (CD25) have a T cell immunodeficiency associated with autoimmunity and those with IL-7Rα deficiency have T B+ NK+ SCID [67, 68]. By contrast, patients with IL-21R deficiency have normal numbers of T, B and NK lymphocytes, but display impaired T-cell proliferation, memory B-cell generation and NK cell antibody-dependent cellular cytotoxicity [69]. For the remaining cytokines (IL-4, -9 and -15), a defective IL-15 response is the most plausible mechanism underlying impaired NK cell development in IL2Rγ- and JAK3-deficient patients. Indeed, IL-15, alone or with stem cell factor, drives the generation of CD56+ NK cells from BM CD34+ progenitors in vitro [70, 71]. Moreover, some patients with specific IL2RG mutations not affecting IL-15 signaling have functional NK cells [72, 73]. Overall, the NKD probably results from IL-15 signaling failure, leading to a blockade of BM and/or SLT development. Clinically, all patients present a broad spectrum of life-threatening infections, including viral, bacterial and fungal infections (Table 1)[48, 49]. It is difficult to determine whether and which infections result from the NKD, even by comparing these patients with patients with NK+ SCID, in part because they have a BM transplantation very early in their life.

Impaired IL-2, IL-15 signaling: STAT5b

Signal transducer and activator of transcription (STAT)-5 is involved in the growth hormone (GH), IL-2 and IL-15 receptor signaling cascades. Following cytokine stimulation, STAT5 is phosphorylated by JAK3, inducing target gene transcription [74]. Patients with STAT5b deficiency present T and NK cell lymphopenia [5155]. Unfortunately, their circulating and BM NK cells have not yet been thoroughly characterized. NK cell cytotoxicity is weak in basal conditions in these patients, partly due to the small number of cells, but IL-2 stimulation increases cytotoxicity and results in a normal perforin induction, suggesting that the IL-2-dependent STAT5b activation is not essential for mature NK cell activity [52, 53]. An in vitro study in the YT human NK cell line showed that the molecular disruption of both STAT5a and STAT5b leads to higher levels of cell death and DNA degradation, but normal progression through the cell cycle [75]. These results suggest that the NK cell lymphopenia in STAT5b deficiency results from impaired IL-15-dependent survival signaling or the inhibition of apoptotic signaling. The clinical phenotype of STAT5b-deficient patients is summarized in Table 1. They present a GH insensitivity syndrome, with facial dysmorphia and a particular susceptibility to respiratory tract infections. They also develop eczema and viral infections. The contribution of the NKD to these infectious and immunological phenotypes is unclear.

Deficits of myeloid, B-, and NK-cell development: GATA2 deficiency

Autosomal dominant GATA2 deficiency was first described in 2011 [56, 57]. It causes the monocytopenia and mycobacterial infections (MonoMAC) syndrome [29, 30], which typically combines deficiencies of dendritic cells, monocytes, B and NK lymphocytes [29, 30,5660]. A GATA2 mutation was identified in the first case report for a patient with an apparently selective NKD [61], who has since developed many other cytopenias [62••]. In humans, NK cells can be identified by the surface expression of CD56 or neural cell adhesion molecule (N-CAM). In the linear differentiation model for human NK cells, CD56bright and CD56dim NK cells correspond to sequential steps in NK cell differentiation. CD56bright NK cells are immature and only weakly cytolytic but have a high cytokine production capacity, whereas CD56dim NK cells exert both effector functions [24, 76]. The NK cell phenotype in GATA2-deficient patients is characterized by the lack of the CD56bright population and a strong decrease in the size of the CD56dim [62••], suggesting a survival defect of the CD56bright population. Moreover, despite the normal expression of maturation markers, the CD56dim population is also functionally impaired [62••]. Analyses of BM from patients frequently show hypocellularity, myeloid dysplasia, and an absence of multilymphoid progenitors and CD38+-CD10+ B/NK cell precursor [29,59]. The NKD in patients with GATA2 deficiency probably results from a series of failures in (i) the maintenance of primitive NK progenitors, (ii) the survival and homeostasis of CD56bright cells in SLT and blood and/or (iii) the maintenance of circulating CD56dim cells. Further studies are required to determine whether GATA2 is involved in IL-15-dependent survival signaling for NK cell development. Clinically, these patients present viral infections, leukemia and cytogenetic abnormalities (Table 1). The contribution of the NKD to these manifestations is unclear.

Isolated NK deficiency: MCM4 deficiency

Mini chromosomal maintenance (MCM)-4 is a member of the highly conserved hexameric MCM complex involved in DNA replication [77]. Patients with autosomal recessive MCM4 deficiency have normal numbers of T and B cells but very few circulating NK cells [31, 63••, 64]. The biochemical defect is partial as the homozygous mutation is hypomorphic. In these patients, the NK CD56bright population is present at normal levels but NK CD56dim cells are almost completely absent. The NK CD56bright subset displays a strong proliferation defect following IL-2 or IL-15 stimulation and an excess of spontaneous apoptosis [63••]. By contrast, the few NK CD56dim cells proliferate normally upon stimulation with IL-2 and IL-15, but present excess spontaneous apoptosis, that is not prevented by IL-2 or IL-15 activation [63••]. These findings confirm that the NK CD56dim subset originates from the NK CD56bright population and indicate that this last step of differentiation requires the hyperproliferation of NK CD56bright cells. Partial MCM4 deficiency affects DNA replication, by disrupting control of the prevention of re-replication, and leads to genomic instability, characterized by an accumulation of chromosomal aberrations, potentially accounting for the loss of the CD56dim NK cell subset. Clinically, patients present growth retardation, adrenal insufficiency due to abnormal adrenal morphology [64] and susceptibility to viral infections, probably resulting at least partly from the NKD, although better documentation is required to confirm this (Table 1). It is interesting that partial MCM4 deficiency results in such a specific hematopoietic (NK CD56dim cell deficiency) and endocrine (adrenal insufficiency) phenotype, reflecting different requirements for MCM4 in different tissues.

Other isolated NK deficiencies

Finally, several case reports for patients with unexplained NKD have been published. One such patient was an Israeli consanguineous patient who developed a severe varicella [33]. She presented an isolated NKD with normal numbers of T and B cells and normal T-cell proliferation. Functional studies showed a normal IL-15 response in T cells, implying that the genetic defect affects an IL-15-independent pathway in NK homeostasis [33]. A similar clinical case, with severe varicella and a total absence of NK cells, was reported in a non consanguineous family [34]. Another report concerned a French, multiplex, non-consanguineous family in which one patient died from severe cytomegalovirus infection [78]. Like STAT5b- and MCM4-deficient patients, the two patients from this family had an intra- and extrauterine growth retardation, without autoimmunity or adrenal insufficiency [78]. They also had neutropenia, but to a lesser extent than GATA2-deficient patients. Studies of the patients’ T cells in vitro showed impaired IL-2- and IL-15-dependent survival [32], but no genetic etiology has yet been identified. Another recently reported case of NKD concerned a patient with normal number of T and B lymphocytes and normal T-cell proliferation [35]. This patient has. bilateral adrenal EBV-associated smooth muscle tumors, as described in GATA2-deficient patients. Both CD56bright and the CD56dim NK cells are detectable, but with particularly high levels of CD117 (human homolog of c-kit) expression [35], suggesting incomplete development, as the CD56bright and the CD56dim NK cells are CD117+ and CD117, respectively [79]. However, some cases have been identified during the first infectious episode. They have to be considered carefully as some herpes viruses may lead to a decrease of NK cell count [80].

Conclusion

NK cells were first described, 40 years ago, as cytotoxic lymphocytes of the innate immune system [8183]. Their function has recently been re-evaluated, as they also display adaptive behaviors [18]. The development of NK cells and the function of these cells, in antiviral immunity in particular, have been studied in detail [8, 19•]. However, we still know little about the development and function of NK cells in humans, due partly to the rarity of inherited forms of NKDs. Recent discoveries in this field have confirmed previous findings but have also provided new insights. First, the IL-15/JAK3-and GATA2-dependent signaling pathways are essential for NK cell development, through mechanisms that are only partially understood. Second, the final step in NK maturation involves a hyperproliferation of the CD56bright NK cells that is highly dependent on MCM4. However, further investigations are required in patients with NKD: (i) in-depth phenotyping, including evaluations of the expression of different markers, such as CD34, CD117 and CD94, in the CD56bright population in blood and SLT [24], (ii) studies of proliferation and apoptosis in response to IL-15 activation, (iii) in vitro differentiation assays of CD34+ precursors, from patients or from controls transfected with siRNA, upon activations with various molecules, including IL-15. These investigations should shed light on the precise mechanism underlying human NK cell development. Needless to say, the search for new genetic etiologies of NKD is also important. Identification of the genetic defects underlying NKD will provide new insight into the development and function of NK cells in humans. Clinically, the only phenotype common to all patients with NKD, whether isolated or combined, is predisposition to viral infections, especially herpes virus infections (Table 1), contributing to the concept of primary immunodeficiencies associated with a restrictive susceptibility to infection [8487]. GATA2-deficient patients display susceptibility to papillomaviruses, but it is unclear to what extent this is due to the NKD. Some patients with MCM4 and GATA2 deficiencies have also developed cancers suggesting a possible role of NK cells in antitumoral immunity. However, these patients also present genomic instability, which may be responsible for these cancers. Long-term clinical and immunological monitoring is required to evaluate the patient outcome, changes in the immune system and correlations between the NKD and clinical signs.

Key points.

  • This article provides an overview of the genetic etiologies of isolated or combined NK deficiency in humans.

  • Recent studies have revealed an unexpected role for GATA2 and MCM4 in human NK cell development.

  • The genetic dissection of various forms of inherited NK cell deficiency should make it possible to decipher the factors controlling human NK cell development.

  • These genetic studies should also help to define the hitherto elusive function of human NK cells in host defense.

Acknowledgments

We thank the members of the laboratory for helpful discussions, especially Capucine Picard and Anne Puel for their critical comments, and Yelena Nemirovskaya, Eric Anderson, Martine Courat, Michele N’Guyen for secretarial assistance.

This work was supported by grants from INSERM, University Paris Descartes, the Rockefeller University, the St. Giles Foundation, and grant number UL1TR000043 from the National Center for Advancing Translational Sciences, National Institutes of Health Clinical and Translational Science Award (CTSA) program at the Rockefeller University. The lab of E.V. is supported by the European Research Council (THINK Advanced Grant), by the Institut Universitaire de France and by institutional grants from INSERM, CNRS and Aix-Marseille University to CIML. L.G. was supported by a La Ligne Nationale Contre le Cancer (LNCC) grant and INSERM. V.B. is supported by a Wenner Gren foundation grant.

Footnotes

Conflicts of interest

E.V. is cofounder and shareholder of Innate Pharma. The other authors declare no conflict of interest.

References

Papers of particular interest, published within the annual period of review, have been highlighted as:

• of special interest

•• of outstanding interest

  • 1.Spits H, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, et al. Innate lymphoid cells--a proposal for uniform nomenclature. Nat Rev Immunol. 2013 Feb;13(2):145–9. doi: 10.1038/nri3365. [DOI] [PubMed] [Google Scholar]
  • 2.Yokoyama WM. Natural killer cell receptors. Curr Opin Immunol. 1998 Jun;10(3):298–305. doi: 10.1016/s0952-7915(98)80168-4. [DOI] [PubMed] [Google Scholar]
  • 3.Lanier LL. Natural killer cell receptor signaling. Curr Opin Immunol. 2003 Jun;15(3):308–14. doi: 10.1016/s0952-7915(03)00039-6. [DOI] [PubMed] [Google Scholar]
  • 4.Moffett-King A. Natural killer cells and pregnancy. Nat Rev Immunol. 2002 Sep;2(9):656–63. doi: 10.1038/nri886. [DOI] [PubMed] [Google Scholar]
  • 5.Long EO, Sik Kim H, Liu D, Peterson ME, Rajagopalan S. Controlling natural killer cell responses: integration of signals for activation and inhibition. Annu Rev Immunol. 2013;31:227–58. doi: 10.1146/annurev-immunol-020711-075005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kim S, Poursine-Laurent J, Truscott SM, Lybarger L, Song YJ, Yang L, et al. Licensing of natural killer cells by host major histocompatibility complex class I molecules. Nature. 2005 Aug 4;436(7051):709–13. doi: 10.1038/nature03847. [DOI] [PubMed] [Google Scholar]
  • 7•.Narni-Mancinelli E, Ugolini S, Vivier E. Tuning the threshold of natural killer cell responses. Curr Opin Immunol. 2013 Feb;25(1):53–8. doi: 10.1016/j.coi.2012.11.005. A clear and comprehensive review on the regulatory role of the NK cells on the immune response in mice. [DOI] [PubMed] [Google Scholar]
  • 8•.Hesslein DG, Lanier LL. Transcriptional control of natural killer cell development and function. Adv Immunol. 2011;109:45–85. doi: 10.1016/B978-0-12-387664-5.00002-9. A clear and comprehensive review on the transcription factors involved in the NK cell development and function. [DOI] [PubMed] [Google Scholar]
  • 9.Narni-Mancinelli E, Chaix J, Fenis A, Kerdiles YM, Yessaad N, Reynders A, et al. Fate mapping analysis of lymphoid cells expressing the NKp46 cell surface receptor. Proceedings of the National Academy of Sciences of the United States of America. 2011 Nov 8;108(45):18324–9. doi: 10.1073/pnas.1112064108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Di Santo JP. Natural killer cell developmental pathways: a question of balance. Annu Rev Immunol. 2006;24:257–86. doi: 10.1146/annurev.immunol.24.021605.090700. [DOI] [PubMed] [Google Scholar]
  • 11.Soderquest K, Powell N, Luci C, van Rooijen N, Hidalgo A, Geissmann F, et al. Monocytes control natural killer cell differentiation to effector phenotypes. Blood. 2011 Apr 28;117(17):4511–8. doi: 10.1182/blood-2010-10-312264. [DOI] [PubMed] [Google Scholar]
  • 12••.Jaeger BN, Donadieu J, Cognet C, Bernat C, Ordonez-Rueda D, Barlogis V, et al. Neutrophil depletion impairs natural killer cell maturation, function, and homeostasis. J Exp Med. 2012 Mar 12;209(3):565–80. doi: 10.1084/jem.20111908. The authors show that neutrophils are required for the NK cell maturation and education in humans and mice. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Arase H, Mocarski ES, Campbell AE, Hill AB, Lanier LL. Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors. Science. 2002 May 17;296(5571):1323–6. doi: 10.1126/science.1070884. [DOI] [PubMed] [Google Scholar]
  • 14.Brown MG, Dokun AO, Heusel JW, Smith HR, Beckman DL, Blattenberger EA, et al. Vital involvement of a natural killer cell activation receptor in resistance to viral infection. Science. 2001 May 4;292(5518):934–7. doi: 10.1126/science.1060042. [DOI] [PubMed] [Google Scholar]
  • 15.Lee SH, Girard S, Macina D, Busa M, Zafer A, Belouchi A, et al. Susceptibility to mouse cytomegalovirus is associated with deletion of an activating natural killer cell receptor of the C-type lectin superfamily. Nat Genet. 2001 May;28(1):42–5. doi: 10.1038/ng0501-42. [DOI] [PubMed] [Google Scholar]
  • 16.Min-Oo G, Kamimura Y, Hendricks DW, Nabekura T, Lanier LL. Natural killer cells: walking three paths down memory lane. Trends Immunol. 2013 Jun;34(6):251–8. doi: 10.1016/j.it.2013.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Moretta A, Marcenaro E, Parolini S, Ferlazzo G, Moretta L. NK cells at the interface between innate and adaptive immunity. Cell Death Differ. 2008 Feb;15(2):226–33. doi: 10.1038/sj.cdd.4402170. [DOI] [PubMed] [Google Scholar]
  • 18.Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, et al. Innate or adaptive immunity? The example of natural killer cells. Science. 2011 Jan 7;331(6013):44–9. doi: 10.1126/science.1198687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19•.Jost S, Altfeld M. Control of human viral infections by natural killer cells. Annu Rev Immunol. 2013;31:163–94. doi: 10.1146/annurev-immunol-032712-100001. A clear review on the function of NK cells in the antiviral immunity in mice. [DOI] [PubMed] [Google Scholar]
  • 20.Narni-Mancinelli E, Jaeger BN, Bernat C, Fenis A, Kung S, De Gassart A, et al. Tuning of natural killer cell reactivity by NKp46 and Helios calibrates T cell responses. Science. 2012 Jan 20;335(6066):344–8. doi: 10.1126/science.1215621. [DOI] [PubMed] [Google Scholar]
  • 21.Walzer T, Dalod M, Robbins SH, Zitvogel L, Vivier E. Natural-killer cells and dendritic cells: “l’union fait la force”. Blood. 2005 Oct 1;106(7):2252–8. doi: 10.1182/blood-2005-03-1154. [DOI] [PubMed] [Google Scholar]
  • 22.Beziat V, Descours B, Parizot C, Debre P, Vieillard V. NK cell terminal differentiation: correlated stepwise decrease of NKG2A and acquisition of KIRs. PLoS One. 2010;5(8):e11966. doi: 10.1371/journal.pone.0011966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bjorkstrom NK, Riese P, Heuts F, Andersson S, Fauriat C, Ivarsson MA, et al. Expression patterns of NKG2A, KIR, and CD57 define a process of CD56dim NK-cell differentiation uncoupled from NK-cell education. Blood. 2010 Nov 11;116(19):3853–64. doi: 10.1182/blood-2010-04-281675. [DOI] [PubMed] [Google Scholar]
  • 24.Freud AG, Caligiuri MA. Human natural killer cell development. Immunol Rev. 2006 Dec;214:56–72. doi: 10.1111/j.1600-065X.2006.00451.x. [DOI] [PubMed] [Google Scholar]
  • 25.Juelke K, Killig M, Luetke-Eversloh M, Parente E, Gruen J, Morandi B, et al. CD62L expression identifies a unique subset of polyfunctional CD56dim NK cells. Blood. 2010 Aug 26;116(8):1299–307. doi: 10.1182/blood-2009-11-253286. [DOI] [PubMed] [Google Scholar]
  • 26.Orange JS. Human natural killer cell deficiencies and susceptibility to infection. Microbes Infect. 2002 Dec;4(15):1545–58. doi: 10.1016/s1286-4579(02)00038-2. [DOI] [PubMed] [Google Scholar]
  • 27.Orange JS. Human natural killer cell deficiencies. Curr Opin Allergy Clin Immunol. 2006 Dec;6(6):399–409. doi: 10.1097/ACI.0b013e3280106b65. [DOI] [PubMed] [Google Scholar]
  • 28.Orange JS, Ballas ZK. Natural killer cells in human health and disease. Clin Immunol. 2006 Jan;118(1):1–10. doi: 10.1016/j.clim.2005.10.011. [DOI] [PubMed] [Google Scholar]
  • 29.Bigley V, Haniffa M, Doulatov S, Wang XN, Dickinson R, McGovern N, et al. The human syndrome of dendritic cell, monocyte, B and NK lymphoid deficiency. J Exp Med. 2011 Feb 14;208(2):227–34. doi: 10.1084/jem.20101459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Calvo KR, Vinh DC, Maric I, Wang W, Noel P, Stetler-Stevenson M, et al. Myelodysplasia in autosomal dominant and sporadic monocytopenia immunodeficiency syndrome: diagnostic features and clinical implications. Haematologica. 2011 Aug;96(8):1221–5. doi: 10.3324/haematol.2011.041152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Eidenschenk C, Dunne J, Jouanguy E, Fourlinnie C, Gineau L, Bacq D, et al. A novel primary immunodeficiency with specific natural-killer cell deficiency maps to the centromeric region of chromosome 8. Am J Hum Genet. 2006 Apr;78(4):721–7. doi: 10.1086/503269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Eidenschenk C, Jouanguy E, Alcais A, Mention JJ, Pasquier B, Fleckenstein IM, et al. Familial NK cell deficiency associated with impaired IL-2- and IL-15-dependent survival of lymphocytes. J Immunol. 2006 Dec 15;177(12):8835–43. doi: 10.4049/jimmunol.177.12.8835. [DOI] [PubMed] [Google Scholar]
  • 33.Etzioni A, Eidenschenk C, Katz R, Beck R, Casanova JL, Pollack S. Fatal varicella associated with selective natural killer cell deficiency. The Journal of pediatrics. 2005 Mar;146(3):423–5. doi: 10.1016/j.jpeds.2004.11.022. [DOI] [PubMed] [Google Scholar]
  • 34.Notarangelo LD, Mazzolari E. Natural killer cell deficiencies and severe varicella infection. J Pediatr. 2006 Apr;148(4):563–4. doi: 10.1016/j.jpeds.2005.06.028. author reply 4. [DOI] [PubMed] [Google Scholar]
  • 35.Shaw RK, Issekutz AC, Fraser R, Schmit P, Morash B, Monaco-Shawver L, et al. Bilateral adrenal EBV-associated smooth muscle tumors in a child with a natural killer cell deficiency. Blood. 2012 Apr 26;119(17):4009–12. doi: 10.1182/blood-2011-10-385377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Blackburn MR, Kellems RE. Adenosine deaminase deficiency: metabolic basis of immune deficiency and pulmonary inflammation. Adv Immunol. 2005;86:1–41. doi: 10.1016/S0065-2776(04)86001-2. [DOI] [PubMed] [Google Scholar]
  • 37.Lee HJ, Pyo JO, Oh Y, Kim HJ, Hong SH, Jeon YJ, et al. AK2 activates a novel apoptotic pathway through formation of a complex with FADD and caspase-10. Nat Cell Biol. 2007 Nov;9(11):1303–10. doi: 10.1038/ncb1650. [DOI] [PubMed] [Google Scholar]
  • 38.Hirschhorn R, Roegner V, Rubinstein A, Papageorgiou P. Plasma deoxyadenosine, adenosine, and erythrocyte deoxyATP are elevated at birth in an adenosine deaminase-deficient child. J Clin Invest. 1980 Mar;65(3):768–71. doi: 10.1172/JCI109725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lagresle-Peyrou C, Six EM, Picard C, Rieux-Laucat F, Michel V, Ditadi A, et al. Human adenylate kinase 2 deficiency causes a profound hematopoietic defect associated with sensorineural deafness. Nat Genet. 2009 Jan;41(1):106–11. doi: 10.1038/ng.278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Heltzer ML, Paessler M, Raffini L, Bunin N, Perez EE. Successful haploidentical bone marrow transplantation in a patient with reticular dysgenesis: three-year follow-up. J Allergy Clin Immunol. 2007 Oct;120(4):950–2. doi: 10.1016/j.jaci.2007.07.026. [DOI] [PubMed] [Google Scholar]
  • 41.Pannicke U, Honig M, Hess I, Friesen C, Holzmann K, Rump EM, et al. Reticular dysgenesis (aleukocytosis) is caused by mutations in the gene encoding mitochondrial adenylate kinase 2. Nat Genet. 2009 Jan;41(1):101–5. doi: 10.1038/ng.265. [DOI] [PubMed] [Google Scholar]
  • 42.Thompson LF, Van de Wiele CJ, Laurent AB, Hooker SW, Vaughn JG, Jiang H, et al. Metabolites from apoptotic thymocytes inhibit thymopoiesis in adenosine deaminase-deficient fetal thymic organ cultures. J Clin Invest. 2000 Nov;106(9):1149–57. doi: 10.1172/JCI9944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Apasov SG, Blackburn MR, Kellems RE, Smith PT, Sitkovsky MV. Adenosine deaminase deficiency increases thymic apoptosis and causes defective T cell receptor signaling. J Clin Invest. 2001 Jul;108(1):131–41. doi: 10.1172/JCI10360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Aldrich MB, Chen W, Blackburn MR, Martinez-Valdez H, Datta SK, Kellems RE. Impaired germinal center maturation in adenosine deaminase deficiency. J Immunol. 2003 Nov 15;171(10):5562–70. doi: 10.4049/jimmunol.171.10.5562. [DOI] [PubMed] [Google Scholar]
  • 45.Freud AG, Yokohama A, Becknell B, Lee MT, Mao HC, Ferketich AK, et al. Evidence for discrete stages of human natural killer cell differentiation in vivo. J Exp Med. 2006 Apr 17;203(4):1033–43. doi: 10.1084/jem.20052507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ficara F, Superchi DB, Hernandez RJ, Mocchetti C, Carballido-Perrig N, Andolfi G, et al. IL-3 or IL-7 increases ex vivo gene transfer efficiency in ADA-SCID BM CD34+ cells while maintaining in vivo lymphoid potential. Mol Ther. 2004 Dec;10(6):1096–108. doi: 10.1016/j.ymthe.2004.08.014. [DOI] [PubMed] [Google Scholar]
  • 47.Noguchi M, Yi H, Rosenblatt HM, Filipovich AH, Adelstein S, Modi WS, et al. Interleukin-2 receptor gamma chain mutation results in X-linked severe combined immunodeficiency in humans. Cell. 1993 Apr 9;73(1):147–57. doi: 10.1016/0092-8674(93)90167-o. [DOI] [PubMed] [Google Scholar]
  • 48.Buckley RH, Schiff RI, Schiff SE, Markert ML, Williams LW, Harville TO, et al. Human severe combined immunodeficiency: genetic, phenotypic, and functional diversity in one hundred eight infants. J Pediatr. 1997 Mar;130(3):378–87. doi: 10.1016/s0022-3476(97)70199-9. [DOI] [PubMed] [Google Scholar]
  • 49.Stephan JL, Vlekova V, Le Deist F, Blanche S, Donadieu J, De Saint-Basile G, et al. Severe combined immunodeficiency: a retrospective single-center study of clinical presentation and outcome in 117 patients. J Pediatr. 1993 Oct;123(4):564–72. doi: 10.1016/s0022-3476(05)80951-5. [DOI] [PubMed] [Google Scholar]
  • 50.Macchi P, Villa A, Giliani S, Sacco MG, Frattini A, Porta F, et al. Mutations of Jak-3 gene in patients with autosomal severe combined immune deficiency (SCID) Nature. 1995 Sep 7;377(6544):65–8. doi: 10.1038/377065a0. [DOI] [PubMed] [Google Scholar]
  • 51.Nadeau K, Hwa V, Rosenfeld RG. STAT5b deficiency: an unsuspected cause of growth failure, immunodeficiency, and severe pulmonary disease. J Pediatr. 2011 May;158(5):701–8. doi: 10.1016/j.jpeds.2010.12.042. [DOI] [PubMed] [Google Scholar]
  • 52.Bernasconi A, Marino R, Ribas A, Rossi J, Ciaccio M, Oleastro M, et al. Characterization of immunodeficiency in a patient with growth hormone insensitivity secondary to a novel STAT5b gene mutation. Pediatrics. 2006 Nov;118(5):e1584–92. doi: 10.1542/peds.2005-2882. [DOI] [PubMed] [Google Scholar]
  • 53.Scaglia PA, Martinez AS, Feigerlova E, Bezrodnik L, Gaillard MI, Di Giovanni D, et al. A novel missense mutation in the SH2 domain of the STAT5B gene results in a transcriptionally inactive STAT5b associated with severe IGF-I deficiency, immune dysfunction, and lack of pulmonary disease. J Clin Endocrinol Metab. 2012 May;97(5):E830–9. doi: 10.1210/jc.2011-2554. [DOI] [PubMed] [Google Scholar]
  • 54.Vidarsdottir S, Walenkamp MJ, Pereira AM, Karperien M, van Doorn J, van Duyvenvoorde HA, et al. Clinical and biochemical characteristics of a male patient with a novel homozygous STAT5b mutation. J Clin Endocrinol Metab. 2006 Sep;91(9):3482–5. doi: 10.1210/jc.2006-0368. [DOI] [PubMed] [Google Scholar]
  • 55.Kofoed EM, Hwa V, Little B, Woods KA, Buckway CK, Tsubaki J, et al. Growth hormone insensitivity associated with a STAT5b mutation. N Engl J Med. 2003 Sep 18;349(12):1139–47. doi: 10.1056/NEJMoa022926. [DOI] [PubMed] [Google Scholar]
  • 56.Dickinson RE, Griffin H, Bigley V, Reynard LN, Hussain R, Haniffa M, et al. Exome sequencing identifies GATA-2 mutation as the cause of dendritic cell, monocyte, B and NK lymphoid deficiency. Blood. 2011 Sep 8;118(10):2656–8. doi: 10.1182/blood-2011-06-360313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hsu AP, Sampaio EP, Khan J, Calvo KR, Lemieux JE, Patel SY, et al. Mutations in GATA2 are associated with the autosomal dominant and sporadic monocytopenia and mycobacterial infection (MonoMAC) syndrome. Blood. 2011 Sep 8;118(10):2653–5. doi: 10.1182/blood-2011-05-356352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Bigley V, Collin M. Dendritic cell, monocyte, B and NK lymphoid deficiency defines the lost lineages of a new GATA-2 dependent myelodysplastic syndrome. Haematologica. 2011 Aug;96(8):1081–3. doi: 10.3324/haematol.2011.048355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Camargo JF, Lobo SA, Hsu AP, Zerbe CS, Wormser GP, Holland SM. MonoMAC Syndrome in a Patient With a GATA2 Mutation: Case Report and Review of the Literature. Clin Infect Dis. 2013 Sep;57(5):697–9. doi: 10.1093/cid/cit368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kazenwadel J, Secker GA, Liu YJ, Rosenfeld JA, Wildin RS, Cuellar-Rodriguez J, et al. Loss-of-function germline GATA2 mutations in patients with MDS/AML or MonoMAC syndrome and primary lymphedema reveal a key role for GATA2 in the lymphatic vasculature. Blood. 2012 Feb 2;119(5):1283–91. doi: 10.1182/blood-2011-08-374363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Biron CA, Byron KS, Sullivan JL. Severe herpesvirus infections in an adolescent without natural killer cells. N Engl J Med. 1989 Jun 29;320(26):1731–5. doi: 10.1056/NEJM198906293202605. [DOI] [PubMed] [Google Scholar]
  • 62••.Mace EM, Hsu AP, Monaco-Shawver L, Makedonas G, Rosen JB, Dropulic L, et al. Mutations in GATA2 cause human NK cell deficiency with specific loss of the CD56(bright) subset. Blood. 2013 Apr 4;121(14):2669–77. doi: 10.1182/blood-2012-09-453969. The authors report that GATA2 deficiency is associated with the specific lack of NK CD56bright subset and an impaired function of the NK CD56dim population. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63••.Gineau L, Cognet C, Kara N, Lach FP, Dunne J, Veturi U, et al. Partial MCM4 deficiency in patients with growth retardation, adrenal insufficiency, and natural killer cell deficiency. J Clin Invest. 2012 Mar 1;122(3):821–32. doi: 10.1172/JCI61014. The authors describe the first genetic etiology associated with isolated NKD, partial autosomal recessive MCM4 deficiency. The mutation leads to the lack of prevention of re-replication and genomic instability. The NKD is characterized by a strong decrease of NK CD56dim subset but a normal count of NK CD56bright subset but with a decreased proliferation ability. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hughes CR, Guasti L, Meimaridou E, Chuang CH, Schimenti JC, King PJ, et al. MCM4 mutation causes adrenal failure, short stature, and natural killer cell deficiency in humans. J Clin Invest. 2012 Mar 1;122(3):814–20. doi: 10.1172/JCI60224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Russell SM, Johnston JA, Noguchi M, Kawamura M, Bacon CM, Friedmann M, et al. Interaction of IL-2R beta and gamma c chains with Jak1 and Jak3: implications for XSCID and XCID. Science. 1994 Nov 11;266(5187):1042–5. doi: 10.1126/science.7973658. [DOI] [PubMed] [Google Scholar]
  • 66.Russell SM, Tayebi N, Nakajima H, Riedy MC, Roberts JL, Aman MJ, et al. Mutation of Jak3 in a patient with SCID: essential role of Jak3 in lymphoid development. Science. 1995 Nov 3;270(5237):797–800. doi: 10.1126/science.270.5237.797. [DOI] [PubMed] [Google Scholar]
  • 67.Puel A, Ziegler SF, Buckley RH, Leonard WJ. Defective IL7R expression in T(−)B(+)NK(+) severe combined immunodeficiency. Nat Genet. 1998 Dec;20(4):394–7. doi: 10.1038/3877. [DOI] [PubMed] [Google Scholar]
  • 68.Sharfe N, Dadi HK, Shahar M, Roifman CM. Human immune disorder arising from mutation of the alpha chain of the interleukin-2 receptor. Proc Natl Acad Sci U S A. 1997 Apr 1;94(7):3168–71. doi: 10.1073/pnas.94.7.3168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Kotlarz D, Zietara N, Uzel G, Weidemann T, Braun CJ, Diestelhorst J, et al. Loss-of-function mutations in the IL-21 receptor gene cause a primary immunodeficiency syndrome. J Exp Med. 2013 Mar 11;210(3):433–43. doi: 10.1084/jem.20111229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Cavazzana-Calvo M, Hacein-Bey S, de Saint Basile G, De Coene C, Selz F, Le Deist F, et al. Role of interleukin-2 (IL-2), IL-7, and IL-15 in natural killer cell differentiation from cord blood hematopoietic progenitor cells and from gamma c transduced severe combined immunodeficiency X1 bone marrow cells. Blood. 1996 Nov 15;88(10):3901–9. [PubMed] [Google Scholar]
  • 71.Leclercq G, Debacker V, de Smedt M, Plum J. Differential effects of interleukin-15 and interleukin-2 on differentiation of bipotential T/natural killer progenitor cells. J Exp Med. 1996 Aug 1;184(2):325–36. doi: 10.1084/jem.184.2.325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Ginn SL, Smyth C, Wong M, Bennetts B, Rowe PB, Alexander IE. A novel splice-site mutation in the common gamma chain (gammac) gene IL2RG results in X-linked severe combined immunodeficiency with an atypical NK+ phenotype. Hum Mutat. 2004 May;23(5):522–3. doi: 10.1002/humu.9235. [DOI] [PubMed] [Google Scholar]
  • 73.Kumaki S, Ishii N, Minegishi M, Tsuchiya S, Cosman D, Sugamura K, et al. Functional role of interleukin-4 (IL-4) and IL-7 in the development of X-linked severe combined immunodeficiency. Blood. 1999 Jan 15;93(2):607–12. [PubMed] [Google Scholar]
  • 74.Lin JX, Leonard WJ. The role of Stat5a and Stat5b in signaling by IL-2 family cytokines. Oncogene. 2000 May 15;19(21):2566–76. doi: 10.1038/sj.onc.1203523. [DOI] [PubMed] [Google Scholar]
  • 75.Behbod F, Nagy ZS, Stepkowski SM, Karras J, Johnson CR, Jarvis WD, et al. Specific inhibition of Stat5a/b promotes apoptosis of IL-2-responsive primary and tumor-derived lymphoid cells. J Immunol. 2003 Oct 15;171(8):3919–27. doi: 10.4049/jimmunol.171.8.3919. [DOI] [PubMed] [Google Scholar]
  • 76.Romagnani C, Juelke K, Falco M, Morandi B, D’Agostino A, Costa R, et al. CD56brightCD16- killer Ig-like receptor- NK cells display longer telomeres and acquire features of CD56dim NK cells upon activation. J Immunol. 2007 Apr 15;178(8):4947–55. doi: 10.4049/jimmunol.178.8.4947. [DOI] [PubMed] [Google Scholar]
  • 77.Bell SP, Dutta A. DNA replication in eukaryotic cells. Annu Rev Biochem. 2002;71:333–74. doi: 10.1146/annurev.biochem.71.110601.135425. [DOI] [PubMed] [Google Scholar]
  • 78.Bernard F, Picard C, Cormier-Daire V, Eidenschenk C, Pinto G, Bustamante JC, et al. A novel developmental and immunodeficiency syndrome associated with intrauterine growth retardation and a lack of natural killer cells. Pediatrics. 2004 Jan;113(1 Pt 1):136–41. doi: 10.1542/peds.113.1.136. [DOI] [PubMed] [Google Scholar]
  • 79.Matos ME, Schnier GS, Beecher MS, Ashman LK, William DE, Caligiuri MA. Expression of a functional c-kit receptor on a subset of natural killer cells. J Exp Med. 1993 Sep 1;178(3):1079–84. doi: 10.1084/jem.178.3.1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Vossen MT, Biezeveld MH, de Jong MD, Gent MR, Baars PA, von Rosenstiel IA, et al. Absence of circulating natural killer and primed CD8+ cells in life-threatening varicella. J Infect Dis. 2005 Jan 15;191(2):198–206. doi: 10.1086/426866. [DOI] [PubMed] [Google Scholar]
  • 81.Kiessling R, Klein E, Wigzell H. “Natural” killer cells in the mouse. I. Cytotoxic cells with specificity for mouse Moloney leukemia cells. Specificity and distribution according to genotype. Eur J Immunol. 1975 Feb;5(2):112–7. doi: 10.1002/eji.1830050208. [DOI] [PubMed] [Google Scholar]
  • 82.Takasugi M, Mickey MR, Terasaki PI. Reactivity of lymphocytes from normal persons on cultured tumor cells. Cancer Res. 1973 Nov;33(11):2898–902. [PubMed] [Google Scholar]
  • 83.Jondal M, Pross H. Surface markers on human b and t lymphocytes. VI. Cytotoxicity against cell lines as a functional marker for lymphocyte subpopulations. Int J Cancer. 1975 Apr 15;15(4):596–605. doi: 10.1002/ijc.2910150409. [DOI] [PubMed] [Google Scholar]
  • 84.Alcais A, Quintana-Murci L, Thaler DS, Schurr E, Abel L, Casanova JL. Life-threatening infectious diseases of childhood: single-gene inborn errors of immunity? Ann N Y Acad Sci. 2010 Dec;1214:18–33. doi: 10.1111/j.1749-6632.2010.05834.x. [DOI] [PubMed] [Google Scholar]
  • 85.Casanova J, Abel L. The genetic theory of infectious diseases: brief history and selected illustrations. Ann Rev Genomics. 2013 doi: 10.1146/annurev-genom-091212-153448. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Casanova JL, Abel L. Primary immunodeficiencies: a field in its infancy. Science. 2007 Aug 3;317(5838):617–9. doi: 10.1126/science.1142963. [DOI] [PubMed] [Google Scholar]
  • 87.Quintana-Murci L, Alcais A, Abel L, Casanova JL. Immunology in natura: clinical, epidemiological and evolutionary genetics of infectious diseases. Nat Immunol. 2007 Nov;8(11):1165–71. doi: 10.1038/ni1535. [DOI] [PubMed] [Google Scholar]

RESOURCES