Abstract
For many years, Severe Combined Immune Deficiency (SCID) diseases, characterized by virtual lack of circulating T cells and severe predisposition to infections since early in life, have been considered the prototypic forms of genetic defects of T cell development. More recently, advances in genome sequencing have allowed identification of a growing number of gene defects that cause severe, but incomplete, defects in T cell development and/or function. Along with recurrent and severe infections, and especially cutaneous viral infections, the clinical phenotype of these conditions is characterized by prominent immune dysregulation.
Keywords: Immunodeficiency, T cell development, thymus, autoimmunity, tolerance
Introduction
Severe combined immune deficiency (SCID) includes a heterogeneous group of genetic disorders characterized by severe impairment of T cell development, with virtual lack of circulating autologous T lymphocytes and absence of functional T cell responses1. Clinical manifestations of SCID include early-onset life-threatening infections and failure to thrive. In contrast, especially in recent years, several conditions have been described, in which T cell development and function are impaired, but not abrogated. These disorders have a broader spectrum of clinical manifestations that often includes autoimmunity, inflammation and lymphoproliferative disease, indicating disturbance of immune homeostasis. In this review we will focus on this heterogeneous group of disorders. We will focus our attention to diseases whose molecular basis has been defined in the last few years, for which novel clinical and immunological phenotypes have been reported, or novel insights into the pathophysiology of clinical manifestations of immune dysregulation associated with these conditions have been recently provided. Purine nucleoside phosphorylase deficiency2, Major Histocompatibility Complex (MHC) class I3 and class II4 deficiencies, and CD8α deficiency5 also belong to this group of partial defects of T cell development and function, but their clinical and immunological phenotype is well known and will not be discussed further here.
Combined immunodeficiencies due to hypomorphic mutations in SCID-associated genes
Although SCID is typically associated with lack of T cell development and function, hypomorphic mutations in SCID-causing genes may allow residual development of autologous T cells and reduced (but not absent) T cell function. The clinical phenotype associated with this condition is not limited to typical manifestations of SCID, but often includes immune dysregulation and lymphoproliferative disease1,6. Omenn syndrome (OS) represents the prototypic form of severe inflammatory/autoimmune condition associated with hypomorphic mutations in SCID-causing genes, with severe restriction of T cell development, associated with profound abnormalities of the mechanisms that govern immune tolerance7. In particular, there is reduced number of medullary thymic epithelial cells expressing Autoimmune Regulator (AIRE), a transcription factor that permits expression of tissue-restricted peptides that are presented to nascent thymocytes, enabling clonal deletion of self-reactive T cells8,9. Moreover, development and function of regulatory T (Treg) lymphocytes are severely compromised10. As a result of inefficient deletion and suppression of self-reactive T cells, oligoclonal expansion of distinct T cell clonotypes is observed in patients with OS7,9. From a mechanistic standpoint, the abnormalities of immune tolerance in OS reflect defective lymphostromal cross-talk in the thymus, and have been also observed in cases of OS due to genetic defects other than RAG mutations8, indicating that normal T cell development is essential to support maturation and function of thymic epithelial and dendritic cells. It is likely that similar abnormalities may contribute to clinical manifestations of immune dysregulation in other conditions with impaired T cell development and function described below.
More recently, hypomorphic RAG mutations have been identified in patients with milder phenotypes, such as delayed-onset disease and granulomatous or autoimmune manifestations10–13, dysgammaglobulinemia with hyper-IgM phenotype14, and idiopathic CD4 lymphopenia15. In these cases, the functional activity of mutated RAG proteins was significantly higher than in patients with SCID or OS.
Somatic mutations are another mechanism that may modify the disease phenotype in patieints with SCID-associated gene defects. Originally reported in adenosine deaminase deficiency16, the occurrence of somatic mutations that may restore, at least in part, expression and function of the mutated protein, have been subsequently demonstrated in other SCID disorders17. In some cases, this resulted in a shift of phenotype, from SCID to OS18,19; more rarely, restoration of T cell function has been observed20,21, providing a strong basis for the development of gene therapy.
Finally, environmental factors may also shape the phenotypic spectrum of SCID and associated disorders, especially in patients with hypomorphic mutations, as indicated by expansion of T cells expressing TCRγδ following CMV infection22,23, and by conversion of SCID into OS phenotype following viral infection24.
TCRα gene (TRAC) defects leading to selective lack of TCRαβ+ T lymphocytes
TCRαβ+ lymphocytes comprise approximately 80–85% of all circulating T cells. Morgan et al. have reported on two patients who presented with a history of recurrent respiratory infections, candidiasis diarrhea, failure to thrive, autoimmunity (hemolytic anemia, vitiligo), eczema, eosinophilia, lymphadenopathy and organomegaly25. One of the patients showed increased susceptibility to severe herpes virus (EBV, VZV, HHV6) infections. All T cells expressing CD3 at normal density also co-expressed TCRγδ; an unusual population of CD3lo cells expressed TCRαβ at very low level. In vitro proliferative responses to mitogens and antigens were decreased, but specific antibody production was preserved. Mutation analysis disclosed in both patients a homozygous splicing mutation in the TCRα constant (TRAC) gene, with loss of TCRα transmembrane and intracytoplasmic domains. Although several aspects of the disease remain unclear (including the developmental history of CD3lo cells and the nature of signals that may promote their expansion in vivo), this novel combined immunodeficiency illustrates the non-redundant role played by TCRαβ+ T cells in immune defense and homeostasis.
Lck deficiency
The Lymphocyte-specific protein tyrosine kinase (Lck) is constitutively associated with CD4 and CD8 proteins, and plays a key role in the initial steps of T cell receptor (TCR) signaling process, by mediating phosphorylation of Immunoreceptor Tyrosine Activation Motifs (ITAMs) in the intracytoplasmic domains of CD3 subunits and of the Zeta-Associated Protein of 70 kDa (ZAP70) 26.
Defective expression of Lck had been reported in three patients with variable clinical and immunological phenotypes27–29, however, LCK gene mutations could not be demonstrated. More recently, Hauck et al. have reported on a child with recurrent respiratory infections, protracted diarrhea, failure to thrive, nodular skin lesions, arthritis, retinal vasculitis, and autoimmune thrombocytopenia30. Genetic studies showed that the child carried a maternal uniparental isodisomy of chromosome 1, and was homozygous for a missense mutation (L341P) that affected Lck protein expression and abrogated its kinase activity.
The immunological phenotype was characterized by severe CD4+ T cell lymphopenia with oligoclonal TCRαβ+ T cells, markedly reduced expression of both CD4 and CD8 molecules on the surface of CD3+ T cells, increase of central memory (CD45R0+ CCR7+) CD4+ cells and of CD45RA+ CCD27− CD62L− “exhausted” T effector memory (TEMRA) CD8+ cells. Upon in vitro activation with anti-CD3, early tyrosine-phosphorylation events during T cell activation were markedly reduced, and Ca2+ mobilization was abrogated, resulting in a severe proliferation defect. Serum IgM was elevated and autoantibodies to multiple self-antigens were present. The low number of regulatory T cells, and the reduced activation-induced cell death of the patient’s T cells may have contributed to the immune dysregulation.
Idiopathic CD4 lymphopenia due to UNC119 deficiency
Uncoordinated 119 (UNC119) is a chaperone involved in Lck-mediated signaling by transporting myrystoilated Lck to the cell membrane, and disrupting intramolecular interactions that keep Lck in a closed, inactive conformation31,32. A heterozygous dominant-negative missense mutation of UNC119, affecting the N-terminus of the molecule, has been identified in an adult with a history of recurrent upper and lower respiratory infections, frequent episodes of shingles, chronic cutaneous and nail fungal infections, and a diagnosis of idiopathic CD4 lymphopenia33. In vitro proliferative response to mitogens and antigens was markedly decreased. An abnormal intracellular distribution of Lck protein was demonstrated, with sequestration in the endosomal compartment, and reduced amount at the cell membrane. Expression of mutant UNC119 in primary CD4+ lymphocytes and in Jurkat cells blocked activation of Lck, confirming the dominant-negative effect of the mutant33.
RHOH deficiency
The Ras homology family member H (RhoH) is an atypical small GTPase. Mainly expressed in hematopoietic cells34, it plays an important role in T cell activation. Upon TCR stimulation, RhoH undergoes tyrosine phosphorylation and mediates recruitment of Zap70 and Lck to the TCR/LAT signalosome35 (Fig. 1).
Figure 1.
Schematic representation of signaling through the T cell receptor (TCR)/CD3 complex. Molecules whose mutations have been associated with partial defect of T cell development and impaired T cell function are indicated in red and highlighted in bold.
Créquer et al. have described two siblings who carried a homozygous nonsense mutation in the RHOH gene. Both patients suffered from persistent cutaneous human papillomavirus (HPV) infection resembling epidermodysplasia verruciformis (EV). The older sibling also developed Burkitt’s lymphoma in childhood, granulomatous lung disease, and psoriatic-like lesions, whereas the younger sibling had molluscum contagiosum, psoriatic lesions, and gingivostomatitis36. The immunological phenotype was characterized by reduced number of naïve CD4+ cells and of recent thymic emigrants, increased proportion of effector memory T cells and of TEMRA cells, restricted T cell repertoire diversity, and reduced in vitro proliferation to anti-CD3. Interestingly, the proportion of T cells expressing skin-homing receptors (cutaneous lymphocyte antigen, CCR4, CCR6, CCR10 and β7-integrin) was significantly reduced, possibly accounting for the EV-like cutaneous manifestations.
Severe T cell lymphopenia, with an incomplete block at double negative 3 (DN3) to double positive (DP) stage of differentiation, and abnormalities of peripheral T cells (in vivo activated phenotype, impaired proliferation and reduced cytokine release in response to CD3 engagement) have been reported in Rhoh−/− mice35,37,38.
ZAP70 deficiency
Following CD3/TCR activation, the tyrosine kinase ZAP70 binds to the phosphorylated ITAM motifs of CD3ζ39, and is tyrosine-phosphorylated by Lck, thereby enabling ZAP70 kinase activity, resulting in phosphorylation of the Linker of Activation in T cells (LAT) 40 and the SH2 domain-containing Leukocyte Phosphoprotein of 76 kDa (SLP-76) 41 (Fig. 1).
The essential role played by ZAP70 during T cell activation was unraveled in 1991, when ZAP70 mutations were identified in patients who lacked circulating CD8+ T cells and whose CD4+ lymphocytes failed to respond to CD3 stimulation42–44. By contrast, Zap70−/− mice have a more profound block in T cell development, with an arrest at DP stage45. This discordance may reflect species-specific differences in the pattern of expression of tyrosine kinases that control T cell differentiation. In particular, Syk expression is rapidly turned off during differentiation from DN to DP cells in mice, but is maintained in human DP cells46. This observation, along with the requirement of higher levels of Zap70 expression for the development of CD8+ than of CD4+ SP thymocytes47, may account for the generation of SP CD4+ lymphocytes in humans. However, peripheral human T cells do not express SYK, thus explaining the profound functional defect of circulating CD4+ cells in ZAP70-deficient patients, including impaired proliferation, aberrant gene expression profile and defective directional migration48,49.
Reduced levels of T cell receptor excision circles (TRECs) have been reported in ZAP70 deficiency48,50, indicating that this kinase plays an important role also in pre-TCR signaling.
Although most patients with ZAP70 deficiency present with selective lack of CD8+ lymphocytes and increased susceptibility to severe and recurrent infections, several patients have been reported in which residual expression of ZAP70 was associated with delayed onset and manifestations of immune dysregulation, including skin rash, wheezing and eosinophilia51–53. In mice, Zap70 mutations that reduce, but do not abrogate TCR signaling, are associated with arthritis54 and hyper-IgE55. Impaired TCR strength signaling in peripheral T cells may affect activation-induced cell death. However, recent data indicate that reduced number of AIRE+ thymic epithelial cells and of FOXP3+ Treg cells56 may also contribute to the immune dysregulation associated with hypomorphic ZAP70 mutations
MST1 deficiency
Macrophage stimulating 1 (MST1), also known as Serine Threonine Kinase 4 (STK4) is the homolog of the hippo protein, that in Drosophila controls cell proliferation and apoptosis57. Evidence that MST1 plays a critical role in T cell survival came from Mst1−/− mice, that show severe reduction in the number of naïve T cells due to increased apoptosis resulting from activation-induced oxidative stress58. Moreover, Mst1 regulates transcription of Foxo1, a gene that encodes for a transcription factor that controls expression of IL-7Rα in naïve T cells, thereby promoting IL-7-mediated survival59. Foxo1 is also involved in the expression of L-selectin and CCR7, two important homing receptors. Consistent with this, Mst1−/− mice show impaired thymocyte egress60 and defective homing of T lymphocytes to peripheral lymphoid organs61. Moreover, inefficient migration of Mst1-deficient thymocytes is associated with defective recognition of self-antigens in the thymic medulla and impaired negative selection, thus accounting for T cell-dependent inflammatory infiltrates in peripheral organs and autoimmunity62.
Three groups have reported that autosomal recessive MST1 deficiency in humans is associated with recurrent bacterial and viral infections (the latter including warts, molluscum contagiosum and EBV-driven lymphoproliferative disease), mucocutaneous candidiasis, skin rash, and autoimmunity. Congenital heart disease and moderate neutropenia have been also reported63–65. The immunological phenotype of MST1 deficiency recapitulates what observed in Mst1−/− mice, and includes severe reduction of naïve T cells with virtual absence of recent thymic emigrants, increased proportion of TEMRA cells, oligoclonal T cell repertoire, impaired proliferation in vitro to mitogens and antigens, and increased apoptosis of T lymphocytes, associated with reduced expression of IL-7Rα and increased expression of FAS63,64. Furthermore, the number of circulating B cells is often reduced, with predominance of transitional B lymphocytes and few CD27+ memory B cells63,64. Overall, these data identify MST1 as a molecule that plays a critical role in T cell development, selection, survival and migration, at the cross-road between immune surveillance and tolerance.
DOCK8 deficiency
The dedicator of cytokinesis 8 (DOCK8) is a member of the DOCK180 superfamily of atypical guanine nucleotide exchange factors (GEFs) that activate GTPases of the Rho/Rac/Cdc42 family66,67. The DOCK-homology region 1 (DHR-1) domain allows DOCK8 to bind phosphatidylinositol-(3,4,5)-trisphosphate (PIP3) at the cell membrane, while the DHR-2 domain mediates interaction with GTPases68 (Fig. 1). DOCK8 is encoded by a large gene that includes 48 exons, spanning 250 kb of genomic DNA; Although this gene is widely expressed, higher levels are detected in T and B lymphocytes69.
DOCK8 mutations have been identified in patients with autosomal recessive combined immunodeficiency and a hyper-IgE phenotype69,70. Most patients carry large intragenic deletions as the result of Alu-mediated recombination, and therefore lack DOCK8 protein expression.
Eczema and manifestations of food or environmental allergy, often with prominent eosinophilia, are present in >90% of the patients. Recurrent respiratory infections and mucocutaneous candidiasis are also very common. Viral cutaneous infections affect >80% of the patients in the form of recurrent orolabial, ocular, or genital herpes simplex infection, molluscum contagiosum, and warts due to papillomavirus69–72. Progressive multifocal leukoencephalopathy due to JC virus has been described70. Papillomavirus infections often degenerate into squamous epithelial cell carcinoma73, and the risk of other tumors (lymphoma, leiomyosarcoma) is also increased74. Autoimmune manifestations include cytopenia and central nervous system vasculitis. The disease has a severe prognosis, with a high mortality rate due to infections and tumors, but definitive cure may be achieved with hematopoietic cell transplantation (HCT) 75–78.
Immunological abnormalities include a variable degree of T cell lymphopenia69,70,74, with loss of naïve CD8+ cells and increased proportion of TEMRA CD8+ lymphocytes79. A low number of TRECs, indicating impaired generation of T cells in the thymus, has been reported80. In vitro lymphocyte proliferative responses are often decreased, especially for CD8+ T lymphocytes69,70,79. The reduced number of Th17 cells may contribute to candidiasis81. Several studies in mouse models have confirmed a critical role for Dock8 in T lymphocyte survival, activation and trafficking. The number of naïve T lymphocytes in peripheral lymphoid organs is markedly reduced in Dock8-deficient mice79,82, a finding that may reflect impaired survival. Moreover, defective CD8+ T cell memory has been demonstrated in upon in vivo challenge with vaccinia virus and influenza virus79,82. Defective trafficking of Dock8-deficient dendritic cells, and impaired homing of DCs to lymph nodes83 may also contribute to poor T cell priming, defective generation of memory T cell responses and increased occurrence of chronic viral infections.
Humoral immune responses are also affected in DOCK8 deficiency. IgG levels may be normal or elevated; in contrast, IgM are often low, and IgE are significantly increased69,70. Patients may mount specific antibody responses to antigens, however these are not sustained84. Similar findings have been observed in Dock8-deficient mice, in which impaired formation of immunological synapse between B cells and follicular dendritic cells is associated with poor survival of germinal center B cells and inability to establish high-affinity and memory B cell responses85. A recent study in humans has shown that TLR9 activation induces Pyk2-mediated DOCK8 tyrosine phosphorylation, allowing recruitment of Src and/or LYN and phosphorylation of SYK and STAT384. Consistent with these findings, CpG-induced proliferation of B lymphocytes and in vitro production of IgM and IgG are profoundly impaired in patients with DOCK8 deficiency84. Overall, these observations indicate that DOCK8 plays a critical role in immune surveillance and homeostasis by affecting the function of multiple cell types.
ITK deficiency
Interleukin-2-inducible tyrosine kinase (ITK) is a member of the Tec family of nonreceptor tyrosine kinases that is expressed in T lymphocytes and participates at TCR signaling (Fig. 1). In particular, ITK pleckstrin homology domain binds to phosphatidylinositol monophosphates, allowing ITK recruitment to the cell membrane86. Following TCR/CD3 cross-linking, ITK activation increases PLC-γ1 activation and Ca2+ influx87–89. It has been shown that ITK controls the spatiotemporal organization of T cell activation; in the absence of ITK, the distribution of several signaling molecules at the immunological synapse between T lymphocytes and antigen presenting cells is altered, causing impaired actin accumulation90.
In 2009, Huck et al. described two sisters with EBV-associated lymphoproliferative disease and autoimmune cytopenias due to ITK mutation. One of the siblings also had a history of Pneumocystis jiroveci pneumonia, candidiasis, BK polyoma infection, and EBV-related Hodgkin lymphoma91. The immunological phenotype was marked by lymphopenia and predominance of T cells with activated/memory phenotype. Since then, several other cases have been described, with increased susceptibility to EBV-mediated lymphoproliferation (often associated with development of pulmonary nodules and mediastinal involvement), but also to other herpes virus infections (varicella, CMV). Most cases have developed progressive hypogammaglobulinemia and T cell lymphopenia, with reduced proportion of naïve T cells and an increase of activated T cells92–94. Studies in mice have shown that Itk-deficient T cells have defective cytotoxic activity, fail to mount primary or memory T cell responses95,96, and are characterized by an aberrant transcriptional signature, with increased expression of eomesodermin97, a transcription factor that promotes development of CD44+ CD122+ CXCR3+ innate, memory-like CD8+ T cells98–100.
ITK controls also development and survival of NKT lymphocytes101. The lack of circulating NKT cells in ITK-deficient patients91 may contribute to EBV-associated lymphoproliferation, similarly to what observed in patients with X-linked lymphoproliferative disease102,103.
Coronin-1A deficiency
Coronin-1a is a member of the Coronin family of proteins that bind F-actin and the Arp2,3 complex, thereby regulating cytoskeleton organization104. In particular, coronin-1A inhibits F-actin formation, integrin outside-in signaling, and chemokine-induced migration in T lymphocytes105. Severe reduction of naïve T cells, and accumulation of apoptosis-prone effector memory T lymphocytes, has been observed in Coro1a−/− mice105,106. Moreover, Coro1a-deficient T cells fail to migrate in response to various chemokines and to proliferate upon CD3 stimulation105–107.
The only case of CORO1A deficiency so far reported in humans was a female child with a history of thrush, respiratory tract infections, severe varicella, protracted diarrhea, and developmental delay. The child was a compound heterozygote for a CORO1A dinucleotide deletion and a large deletion that encompassed also 24 other genes108,109. Similar to Coro1a−/− mice, the patient showed severe T cell lymphopenia, impaired in vitro proliferation to mitogens and antigens, and defective specific antibody response. These abnormalities were however less severe than in typical forms of SCID, and more typical of a combined immune deficiency. Consistent with this, the patient survived several years before she was treated with HCT. Identification of CORO1A mutations in other patients will be needed to better define the phenotype of the disease.
Defects of Calcium release-activated channels (CRAC): STIM1 and ORAI1 deficiencies
Ca2+ signaling plays a critical role in immune activation and homeostasis110,111. In T lymphocytes, engagement of the TCR leads to phosphorylation of PLC-γ1, that converts phosphatidylinositol 4,5-bisphosphate (PIP2) into inositol 1,4,5-trisphosphate (InsP3) and diacylglycerol (DAG). Binding of insP3 to its receptor promotes release of Ca2+ from the endoplasmic reticulum (ER) stores. Depletion of Ca2+ ER stores is sensed by the Stromal Interaction Molecule (STIM) proteins, that oligomerize and bind to the ORAI proteins (ORAI1, -2, and -3) that form the pore of CRAC channels on the cell membrane, thereby permitting Ca2+ influx (Fig. 1). The increase of intracellular Ca2+ concentration induces activation of NFAT and ultimately promotes cell proliferation, cytokine production, and induction of cell-mediated cytotoxicity111.
Biallelic loss-of-function mutations of STIM1 and ORAI1 have been identified in patients with bacterial, viral or fungal infections, non progressive myopathy, ectodermal dysplasia and a variable degree of immune dysregulation112–114. Herpes virus infections (CMV, EBV, HSV, VZV) are frequent; in one case with STIM1 deficiency, HHV-8 infection led to Kaposi sarcoma115. Autoimmune manifestations (cytopenias, lymphadenopathy, splenomegaly) are especially common in patients with STIM1 deficiency112,113,116. Although T cell development is not compromised, peripheral T lymphocytes from affected patients show markedly defective in vitro proliferation to mitogens and antigens and a severe impairment of cytokine production112,113,117,118. The function of NK lymphocytes is also affected in patients with STIM1 or ORAI1 mutations, as indicated by defects of degranulation and cytokine production, and impaired cytotolytic activity116,119. Furthermore, complete lack of circulating NKT cells was demonstrated in one patient with STIM1 deficiency116, suggesting that normal CRAC function may be required for their development.
The molecular mechanisms accounting for immune dysregulation in these patients are still unclear. A reduced number of Treg cells has been reported in some patients113, but not others116. Moreover, based on studies in Orai1−/− mice120, it is possible that resistance of CD4+ T cells to activation-induced cell death may also contribute to defective T cell homeostasis. Finally, impaired secretion of IL-10 by B cells may also be involved in autoimmunity associated with STIM1 deficiency121.
Defective degranulation of platelets, severe heart failure and impaired differentiation and function of osteoblasts have been observed in animal models of the disease122–124, but not in patients.
Overall, these data indicate that integrity of CRAC channels is dispensable for lymphoid development (with the possible exception of NKT lymphocytes), but is required for effector and regulatory functions of peripheral T and NK lymphocytes.
MAGT1 deficiency
Mg2+ plays an important role in intracellular signaling125. Several Mg2+ transporters have been identified, whose mutations lead to distinct clinical phenotypes126–128. MAGT1 encodes for a membrane-associated transporter with high selectivity for Mg2+.129 Recently, three male patients with mutations in the X-linked Magnesium Transporter protein 1 (MAGT1) gene have been reported to suffer from X-linked immunodeficiency with magnesium defect and EBV infection and neoplasia (XMEN syndrome) 128. Respiratory tract infections, chronic diarrhea, and increased susceptibility to other viral infections (HSV-1) were also present128. Immunological abnormalities of MAGT1 deficiency include CD4 lymphopenia, reduced number of recent thymic emigrants, and impaired T cell proliferation in response to CD3 stimulation. By contrast, T cell response to phorbol myristate acetate and ionomycin is intact, consistent with a role for Mg2 in proximal intracellular signaling. Female carriers of MAGT1 deficiency exhibit skewed X chromosome inactivation in circulating T lymphocytes, indicating that Mg2+ signaling plays an important role in T cell development and/or survival.
MAGT1 deficiency compromises not only Mg2+ flux, but also Ca2+ influx128. This reflects impaired activation of PLC-γ1 and decreased generation of InsP3. Early signaling events (such as phosphorylation of CD3ζ, ZAP-70 and LAT) in response to CD3 stimulation are intact, indicating that Mg2+ flux is activated downstream from these signals and upstream of PLC-γ1 phosphorylation128. However, the precise mechanisms by which TCR signaling activates MAGT1 and Mg2+ regulates PLC-γ1 phosphorylation remain unknown.
Conclusions
Advances in genome sequencing have rapidly led to the identification of a growing number of conditions characterized by impaired T cell development and function, whose clinical phenotype is marked not just by increased susceptibility to infections, but also immune dysregulation. This series of discoveries has been also facilitated by increased collaboration with clinicians and scientists from countries with a high consanguinity rate. From a public health perspective, now that newborn screening for SCID based on enumeration of TRECs is becoming widely available, it will be very important to carefully annotate which ones of these conditions are also characterized by reduced thymic output, and to develop diagnostic platforms that may lead to correct identification of these diseases in the second-tier phase of the diagnostic process.
From a scientific perspective, the study of these conditions is providing important insights into the mechanisms that govern immune homeostasis. In particular, patients with partial defects of T cell development and function may develop immune dysregulation because of abnormalities that may variably affect the strength of TCR signaling, T cell survival, activation-induced cell death, positive and negative selection, Treg development and function, and trafficking of immune cells. Although many of the phenotypic features observed in patients with these disorders are also recapitulated in corresponding animal models, important differences have emerged which may reflect species-specific properties, pressure from environmental factors, and genotype-specific effects. Overall, this emphasizes the importance of studying inborn errors of immunity to decipher the mechanisms that control human immune system development and function. It can be anticipated that the list of partial defects of T cell function will increase substantially in the next few years, thus broadening our understanding of normal and pathological immune system development.
Acknowledgments
This work was partially contributed by NIH grant 1PO1AI076210-01A1R01, the March of Dimes grant, the Jeffrey Modell Foundation and The Manton Foundation
Abbreviations
- AIRE
Autoimmune regulator
- CMV
Cytomegalovirus
- CRAC
Calcium Release Activated Channels
- DAG
Diacylglycerol
- DHR
Dock-homology region
- DN
double negative
- DOCK8
Dedicator of Cytokinesis 8
- DP
Double Positive
- EBV
Epstein-Barr Virus
- ER
Endoplasmic Reticulum
- EV
Epidermodysplasia Verruciformis
- GEF
Guanine Nucleotide Exchange Factor
- HHV
Human Herpes Virus
- HSV-1
Herpes Simplex Virus 1
- IL
Interleukin
- insP3
inositol 1,4,5-trisphosphate
- ITAM
Immunoreceptor Tyrosine Activation Motif
- ITK
Interleukin-2-inducible Tyrosine Kinase
- LAT
Linker of Activation in T cells
- Lck
Lymphocyte-specific protein tyrosine kinase
- MAGT1
Magnesium Transporter 1
- MHC
Major Histocompatibility Complex
- MST1
Macrophage stimulating 1
- NFAT
Nuclear Factor of Activated T cells
- NKT
Natural Killer T cells
- OS
Omenn syndrome
- PIP2
Phosphatidylinositol 4,5-bisphosphate
- PLC-γ
Phospholipase C-γ
- RAG
Recombinase Activating Gene
- RHOH
Ras homology family member H
- SCID
Severe combined Immune deficiency
- STAT
Signal Transducer and Activator of Transcription
- STIM1
Stromal Interaction Molecule 1
- STK4
Serine Threonine Kinase 4
- TCR
T Cell Receptor
- Th
T helper
- TEMRA
T effector memory CD45RA+ cells
- TRAC
T cell receptor α constant gene
- TREC
T cell receptor excision circles
- Treg
regulatory T cell
- UNC119
Uncoordinated 119
- VZV
Varicella Zoster Virus
- ZAP70
ζ-associated protein of 70 kDa
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errorsmaybe discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.van der Burg M, Gennery AR. Educational paper. The expanding clinical and immunological spectrum of severe combined immunodeficiency. Eur J Pediatr. 2011;170:561–71. doi: 10.1007/s00431-011-1452-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Hirschhorn R, Candotti F. Immunodeficiency due to defects of purine metabolism. In: Ochs HD, Smith CIE, Puck JM, editors. Primary Immunodeficiency Diseases. A Molecular and Genetic Approach. Oxford University Press; 2007. pp. 169–196. [Google Scholar]
- 3.Zimmer J, Andrès E, Donato L, Hanau D, Hentges F, de la Salle H. Clinical and immunological aspects of HLA class I deficiency. QJM. 2005;98:719–27. doi: 10.1093/qjmed/hci112. [DOI] [PubMed] [Google Scholar]
- 4.Picard C, Fischer A. Hematopoietic stem cell transplantation and other management strategies for MHC class II deficiency. Immunol Allergy Clin North Am. 2010;30:173–8. doi: 10.1016/j.iac.2010.01.001. [DOI] [PubMed] [Google Scholar]
- 5.de la Calle-Martin O, Hernandez M, Ordi J, Casamitjana N, Arostegui JI, Caragol I, et al. Familial CD8 deficiency due to a mutation in the CD8α gene. J Clin Invest. 2001;108:117–23. doi: 10.1172/JCI10993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Felgentreff K, Perez-Becker R, Speckmann C, Schwarz K, Kalwak K, Markelj G, et al. Clinical and immunological manifestations of patients with atypical severe combined immunodeficiency. Clin Immunol. 2011;141:73–82. doi: 10.1016/j.clim.2011.05.007. [DOI] [PubMed] [Google Scholar]
- 7.Marrella V, Maina V, Villa A. Omenn syndrome does not live by V(D)J recombination alone. Curr Opin Allergy Clin Immunol. 2011;11:525–31. doi: 10.1097/ACI.0b013e32834c311a. [DOI] [PubMed] [Google Scholar]
- 8.Poliani PL, Facchetti F, Ravanini M, Gennery AR, Villa A, Roifman CM, et al. Early defects in human T-cell development severely affect distribution and maturation of thymic stromal cells: possible implications for the pathophysiology of Omenn syndrome. Blood. 2009;114:105–8. doi: 10.1182/blood-2009-03-211029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Somech R, Simon AJ, Lev A, Dalal I, Spirer Z, Goldstein I, et al. Reduced central tolerance in Omenn syndrome leads to immature self-reactive oligoclonal T cells. J Allergy Clin Immunol. 2009;124:793–800. doi: 10.1016/j.jaci.2009.06.048. [DOI] [PubMed] [Google Scholar]
- 10.Cassani B, Poliani PL, Moratto D, Sobacchi C, Marrella V, Imperatori L, et al. Defect of regulatory T cells in patients with Omenn syndrome. J Allergy Clin Immunol. 2010;125:209–16. doi: 10.1016/j.jaci.2009.10.023. [DOI] [PubMed] [Google Scholar]
- 11.Schuetz C, Huck K, Gudowius S, Megahed M, Feyen O, Hubner B, et al. An immunodeficiency disease with RAG mutations and granulomas. N Engl J Med. 2008;358:2030–8. doi: 10.1056/NEJMoa073966. [DOI] [PubMed] [Google Scholar]
- 12.De Ravin SS, Cowen EW, Zarember KA, Whiting-Theobald NL, Kuhns DB, Sandler NG, et al. Hypomorphic Rag mutations can cause destructive midline granulomatous disease. Blood. 2010;116:1263–71. doi: 10.1182/blood-2010-02-267583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Avila EM, Uzel G, Hsu A, Milner JD, Turner ML, Pittaluga S, et al. Highly variable clinical phenotypes of hypomorphic RAG1 mutations. Pediatrics. 2010;126:e1248–52. doi: 10.1542/peds.2009-3171. [DOI] [PubMed] [Google Scholar]
- 14.Chou J, Hanna-Wakim R, Tirosh I, Kane J, Fraulino D, Lee YN, et al. A novel homozygous mutation in recombination activating gene 2 in 2 relatives with different clinical phenotypes: Omenn syndrome and hyper-IgM syndrome. J Allergy Clin Immunol. 2012;130:1414–6. doi: 10.1016/j.jaci.2012.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kuijpers TW, Ijspeert H, van Leeuwen EM, Jansen MH, Hazenberg MD, Weijer KC, et al. Idiopathic CD4+ T lymphopenia without autoimmunity or granulomatous disease in the slipstream of RAG mutations. Blood. 2011;117:5892–6. doi: 10.1182/blood-2011-01-329052. [DOI] [PubMed] [Google Scholar]
- 16.Hirschhorn R, Yang DR, Israni A, Huie ML, Ownby DR. Somatic mosaicism for a newly identified splice-site mutation in a patient with adenosine deaminase-deficient immunodeficiency and spontaneous clinical recovery. Am J Hum Genet. 1994;55:59–68. [PMC free article] [PubMed] [Google Scholar]
- 17.Wada T, Candotti F. Somatic mosaicism in primary immune deficiencies. Curr Opin Allergy Clin Immunol. 2008;8:510–4. doi: 10.1097/ACI.0b013e328314b651. [DOI] [PubMed] [Google Scholar]
- 18.Wada T, Toma T, Okamoto H, Kasahara Y, Koizumi S, Agematsu K, et al. Oligoclonal expansion of T lymphocytes with multiple second-site mutations leads to Omenn syndrome in a patient with RAG1-deficient severe combined immunodeficiency. Blood. 2005;106:2099–101. doi: 10.1182/blood-2005-03-0936. [DOI] [PubMed] [Google Scholar]
- 19.Wada T, Yasui M, Toma T, Nakayama Y, Nishida M, Shimizu M, et al. Detection of T lymphocytes with a second-site mutation in skin lesions of atypical X-linked severe combined immunodeficiency mimicking Omenn syndrome. Blood. 2008;112:1872–5. doi: 10.1182/blood-2008-04-149708. [DOI] [PubMed] [Google Scholar]
- 20.Stephan V, Wahn V, Le Deist F, Dirksen U, Broker B, Müller-Fleckenstein I, et al. Atypical X-linked severe combined immunodeficiency due to possible spontaneous reversion of the genetic defect in T cells. N Engl J Med. 1996;335:1563–7. doi: 10.1056/NEJM199611213352104. [DOI] [PubMed] [Google Scholar]
- 21.Kawai T, Saito M, Nishikomori R, Yasumi T, Izawa K, Murakami T, et al. Multiple reversions of an IL2RG mutation restore T cell function in an X-linked severe combined immunodeficiency patient. J Clin Immunol. 2012;32:690–7. doi: 10.1007/s10875-012-9684-1. [DOI] [PubMed] [Google Scholar]
- 22.de Villartay JP, Lim A, Al-Mousa H, Dupont S, Déchanet-Merville J, Coumau-Gatbois E, et al. A novel immunodeficiency associated with hypomorphic RAG1 mutations and CMV infection. J Clin Invest. 2005;115:3291–9. doi: 10.1172/JCI25178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ehl S, Schwarz K, Enders A, Duffner U, Pannicke U, Kühr J, et al. A variant of SCID with specific immune responses and predominance of gamma delta T cells. J Clin Invest. 2005;115:3140–8. doi: 10.1172/JCI25221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Dalal I, Tabori U, Bielorai B, Golan H, Rosenthal E, Amariglio N, et al. Evolution of a T-B- SCID into an Omenn syndrome phenotype following parainfluenza 3 virus infection. Clin Immunol. 2005;115:70–3. doi: 10.1016/j.clim.2004.08.016. [DOI] [PubMed] [Google Scholar]
- 25.Morgan NV, Goddard S, Cardno TS, McDonald D, Rahman F, Barge D, et al. Mutation in the TCRα subunit constant gene (TRAC) leads to a human immunodeficiency disorder characterized by a lack of TCRαβT cells. J Clin Invest. 2011;121:695–702. doi: 10.1172/JCI41931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Salmond RJ, Filby A, Qureshi I, Caserta S, Zamoyska R. T-cell receptor proximal signaling via the Src-family kinases, Lck and Fyn, influences T-cell activation, differentiation, and tolerance. Immunol Rev. 2009;228:9–22. doi: 10.1111/j.1600-065X.2008.00745.x. [DOI] [PubMed] [Google Scholar]
- 27.Goldman FD, Ballas ZK, Schutte BC, Kemp J, Hollenback C, Noraz N, et al. Defective expression of p56lck in an infant with severe combined immunodeficiency. J Clin Invest. 1998;102:421–9. doi: 10.1172/JCI3205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Hubert P, Bergeron F, Ferreira V, Seligmann M, Oksenhendler E, Debre P, et al. Defective p56Lck activity in T cells from an adult patient with idiopathic CD4 lymphocytopenia. Int Immunol. 2000;12:449–57. doi: 10.1093/intimm/12.4.449. [DOI] [PubMed] [Google Scholar]
- 29.Sawabe T, Horiuchi T, Nakamura M, Tsukamoto H, Nakahara K, Harashima SI, et al. Defect of lck in a patient with common variable immunodeficiency. Int J Mol Med. 2001;7:609–14. doi: 10.3892/ijmm.7.6.609. [DOI] [PubMed] [Google Scholar]
- 30.Hauck F, Randriamampita C, Martin E, Gerart S, Lambert N, Lim A, et al. Primary T-cell immunodeficiency with immunodysregulation caused by autosomal recessive LCK deficiency. J Allergy Clin Immunol. 2012;130:1144–1152. doi: 10.1016/j.jaci.2012.07.029. [DOI] [PubMed] [Google Scholar]
- 31.Gorska MM, Liang Q, Karim Z, Alam R. Uncoordinated 119 protein controls trafficking of Lck via the Rab11 endosome and is critical for immunological synapse formation. J Immunol. 2009;183:1675–84. doi: 10.4049/jimmunol.0900792. [DOI] [PubMed] [Google Scholar]
- 32.Gorska MM, Stafford SJ, Cen O, Sur S, Alam R. Unc119, a novel activator of Lck/Fyn, is essential for T cell activation. J Exp Med. 2004;199:369–79. doi: 10.1084/jem.20030589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Gorska MM, Alam R. A mutation in the human Uncoordinated 119 gene impairs TCR signaling and is associated with CD4 lymphopenia. Blood. 2012;119:1399–406. doi: 10.1182/blood-2011-04-350686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Gu Y, Jasti AC, Jansen M, Siefring JE. RhoH, a hematopoietic-specific Rho GTPase, regulates proliferation, survival, migration, and engraftment of hematopoietic progenitor cells. Blood. 2005;105:1467–75. doi: 10.1182/blood-2004-04-1604. [DOI] [PubMed] [Google Scholar]
- 35.Dorn T, Kuhn U, Bungartz G, Stiller S, Bauer M, Ellwart J, et al. RhoH is important for positive thymocyte selection and T-cell receptor signaling. Blood. 2007;109:2346–55. doi: 10.1182/blood-2006-04-019034. [DOI] [PubMed] [Google Scholar]
- 36.Créquer A, Troeger A, Patin E, Ma CS, Picard C, Pedergnana V, et al. Human RHOH deficiency causes T cell defects and susceptibility to EV-HPV infections. J Clin Invest. 2012;122:323–47. doi: 10.1172/JCI62949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Gu Y, Chae HD, Siefring JE, Jasti AC, Hildeman DA, Williams DA. RhoH GTPase recruits and activates Zap70 required for T cell receptor signaling and thymocyte development. Nat Immunol. 2006;7:1182–90. doi: 10.1038/ni1396. [DOI] [PubMed] [Google Scholar]
- 38.Porubsky S, Wang S, Kiss E, Dehmel S, Bonrouhi M, Dorn T, et al. Rhoh deficiency reduces peripheral T-cell function and attenuates allogenic transplant rejection. Eur J Immunol. 2011;41:76–88. doi: 10.1002/eji.201040420. [DOI] [PubMed] [Google Scholar]
- 39.Wange RL, Kong AN, Samelson LE. A tyrosine-phosphorylated 70-kDa protein binds a photoaffinity analogue of ATP and associates with both the α chain and CD3 components of the activated T cell antigen receptor. J Biol Chem. 1992;267:11685–88. [PubMed] [Google Scholar]
- 40.Zhang W, Sloan-Lancaster J, Kitchen J, Trible RP, Samelson LE. LAT: the ZAP-70 tyrosine kinase substrate that links T cell receptor to cellular activation. Cell. 1998;92:83–92. doi: 10.1016/s0092-8674(00)80901-0. [DOI] [PubMed] [Google Scholar]
- 41.Bubeck Wardenburg J, Fu C, Jackman JK, Flotow H, Wilkinson SE, Williams DH, et al. Phosphorylation of SLP-76 by the ZAP-70 protein-tyrosine kinase is required for T-cell receptor function. J Biol Chem. 1996;271:19641–44. doi: 10.1074/jbc.271.33.19641. [DOI] [PubMed] [Google Scholar]
- 42.Arpaia E, Shahar M, Dadi H, Cohen A, Roifman CM. Defective T cell receptor signaling and CD8+ thymic selection in humans lacking Zap-70 kinase. Cell. 1994;76:947–58. doi: 10.1016/0092-8674(94)90368-9. [DOI] [PubMed] [Google Scholar]
- 43.Chan AC, Kadlecek TA, Elder ME, Filipovich AH, Kuo WL, Iwashima M, et al. ZAP-70 deficiency in an autosomal recessive form of severe combined immunodeficiency. Science. 1994;264:1599–601. doi: 10.1126/science.8202713. [DOI] [PubMed] [Google Scholar]
- 44.Elder ME, Lin D, Clever J, Chan AC, Hope TJ, Weiss A, et al. Human severe combined immunodeficiency due to a defect in ZAP-70, a T cell tyrosine kinase. Science. 1994;264:1596–99. doi: 10.1126/science.8202712. [DOI] [PubMed] [Google Scholar]
- 45.Negishi I, Motoyama N, Nakayama K, Nakayama K, Senju S, Hatakeyama S, et al. Essential role for ZAP-70 in both positive and negative selection of thymocytes. Nature. 1995;376:435–38. doi: 10.1038/376435a0. [DOI] [PubMed] [Google Scholar]
- 46.Chu DH, van Oers NS, Malissen M, Harris J, Elder M, Weiss A. Pre-T cell receptor signals are responsible for the down-regulation of Syk protein tyrosine kinase expression. J Immunol. 1999;163:2610–20. [PubMed] [Google Scholar]
- 47.Saini M, Sinclair C, Marshall D, Tolaini M, Sakaguchi S, Seddon B. Regulation of Zap70 expression during thymocyte development enables temporal separation of CD4 and CD8 repertoire selection at different signaling thresholds. Sci Signal. 2010;3:ra23. doi: 10.1126/scisignal.2000702. [DOI] [PubMed] [Google Scholar]
- 48.Roifman CM, Dadi H, Somech R, Nahum A, Sharfe N. Characterization of ζ-associated protein, 70 kd (ZAP70)-deficient human lymphocytes. J Allergy Clin Immunol. 2010;126:1226–33. doi: 10.1016/j.jaci.2010.07.029. [DOI] [PubMed] [Google Scholar]
- 49.Lin YP, Cheng YJ, Huang JY, Lin HC, Yang BC. Zap70 controls the interaction of talin with integrin to regulate the chemotactic directionality of T-cell migration. Mol Immunol. 2010;47:2022–29. doi: 10.1016/j.molimm.2010.04.011. [DOI] [PubMed] [Google Scholar]
- 50.Roifman CM, Somech R, Kavadas F, Pires L, Nahum A, Dalal I, et al. Defining combined immunodeficiency. J Allergy Clin Immunol. 2012;130:177–83. doi: 10.1016/j.jaci.2012.04.029. [DOI] [PubMed] [Google Scholar]
- 51.Picard C, Dogniaux S, Chemin K, Maciorowski Z, Lim A, Mazerolles F, et al. Hypomorphic mutation of ZAP70 in human results in a late onset immunodeficiency and no autoimmunity. Eur J Immunol. 2009;39:1966–76. doi: 10.1002/eji.200939385. [DOI] [PubMed] [Google Scholar]
- 52.Katamura K, Tai G, Tachibana T, Yamabe H, Ohmori K, Mayumi M, et al. Existence of activated and memory CD4+ T cells in peripheral blood and their skin infiltration in CD8 deficiency. Clin Exp Immunol. 1999;115:124–30. doi: 10.1046/j.1365-2249.1999.00759.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Turul T, Tezcan I, Artac H, de Bruin-Versteeg S, Barendregt BH, Reisli I, et al. Clinical heterogeneity can hamper the diagnosis of patients with ZAP70 deficiency. Eur J Pediatr. 2009;168:87–93. doi: 10.1007/s00431-008-0718-x. [DOI] [PubMed] [Google Scholar]
- 54.Sakaguchi N, Takahashi T, Hata H, Nomura T, Tagami T, Yamazaki S, et al. Altered thymic T-cell selection due to a mutation of the ZAP-70 gene causes autoimmune arthritis in mice. Nature. 2003;426:454–60. doi: 10.1038/nature02119. [DOI] [PubMed] [Google Scholar]
- 55.Siggs OM, Miosge LA, Yates AL, Kucharska EM, Sheahan D, Brdicka T, et al. Opposing functions of the T cell receptor kinase ZAP-70 in immunity and tolerance differentially titrate in response to nucleotide substitutions. Immunity. 2007;27:912–26. doi: 10.1016/j.immuni.2007.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Poliani PL, Fontana E, Roifman CM, Notarangelo LD. ζ-chain-associated protein of 70 kDa (ZAP70) deficiency in human subjects is associated with abnormalities of thymic stromal cells: Implications for T-cell tolerance. J Allergy Clin Immunol. 2012 Dec 13; doi: 10.1016/j.jaci.2012.11.002. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
- 57.Wu S, Huang J, Dong J, Pan D. hippo encodes a Ste-20 family protein kinase that restricts cell proliferation and promotes apoptosis in conjunction with salvador and warts. Cell. 2003;114:445–56. doi: 10.1016/s0092-8674(03)00549-x. [DOI] [PubMed] [Google Scholar]
- 58.Zhou D, Medoff BD, Chen L, Li L, Zhang XF, Praskova M, et al. The Nore1B/Mst1 complex restrains antigen receptor-induced proliferation of naive T cells. Proc Natl Acad Sci USA. 2008;105:20321–26. doi: 10.1073/pnas.0810773105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Ouyang W, Beckett O, Flavell RA, Li MO. An essential role of the Forkhead-box transcription factor Foxo1 in control of T cell homeostasis and tolerance. Immunity. 2009;30:358–71. doi: 10.1016/j.immuni.2009.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Mou F, Praskova M, Xia F, Van Buren D, Hock H, Avruch J, et al. The Mst1 and Mst2 kinases control activation of rho family GTPases and thymic egress of mature thymocytes. J Exp Med. 2012;209:741–59. doi: 10.1084/jem.20111692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Katagiri K, Katakai T, Ebisuno Y, Ueda Y, Okada T, Kinashi T. Mst1 controls lymphocyte trafficking and interstitial motility within lymph nodes. EMBO J. 2009;28:1319–31. doi: 10.1038/emboj.2009.82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ueda Y, Katagiri K, Tomiyama T, Yasuda K, Habiro K, Katakai T, et al. Mst1 regulates integrin-dependent thymocyte trafficking and antigen recognition in the thymus. Nat Commun. 2012;3:1098. doi: 10.1038/ncomms2105. [DOI] [PubMed] [Google Scholar]
- 63.Nehme NT, Schmid JP, Debeurme F, André-Schmutz I, Lim A, Nitschke P, et al. MST1 mutations in autosomal recessive primary immunodeficiency characterized by defective naive T-cell survival. Blood. 2012;119:3458–68. doi: 10.1182/blood-2011-09-378364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Abdollahpour H, Appaswamy G, Kotlarz D, Diestelhorst J, Beier R, Schäffer AA, et al. The phenotype of human STK4 deficiency. Blood. 2012;119:3450–57. doi: 10.1182/blood-2011-09-378158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Crequer A, Picard C, Patin E, D’Amico A, Abhyankar A, Munzer M, et al. Inherited MST1 Deficiency Underlies Susceptibility to EV-HPV Infections. PLoS ONE. 2012;7:e44010. doi: 10.1371/journal.pone.0044010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Côté JF, Vuori K. Identification of an evolutionarily conserved superfamily of DOCK180-related proteins with guanine nucleotide exchange activity. J Cell Sci. 2002;115:4901–13. doi: 10.1242/jcs.00219. [DOI] [PubMed] [Google Scholar]
- 67.Ruusala A, Aspenström P. Isolation and characterisation of DOCK8, a member of the DOCK180-related regulators of cell morphology. FEBS Lett. 2004;572:159–66. doi: 10.1016/j.febslet.2004.06.095. [DOI] [PubMed] [Google Scholar]
- 68.Côté JF, Vuori K. In vitro guanine nucleotide exchange activity of DHR-2/DOCKER/CZH2 domains. Methods Enzymol. 2006;406:41–57. doi: 10.1016/S0076-6879(06)06004-6. [DOI] [PubMed] [Google Scholar]
- 69.Zhang Q, Davis JC, Lamborn IT, Freeman AF, Jing H, Favreau AJ, et al. Combined immunodeficiency associated with DOCK8 mutations. N Engl J Med. 2009;361:2046–55. doi: 10.1056/NEJMoa0905506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Engelhardt KR, McGhee S, Winkler S, Sassi A, Woellner C, Lopez-Herrera G, et al. Large deletions and point mutations involving the dedicator of cytokinesis 8 (DOCK8) in the autosomal-recessive form of hyper-IgE syndrome. J Allergy Clin Immunol. 2009;124:1289–302. e4. doi: 10.1016/j.jaci.2009.10.038. Erratum in: J Allergy Clin Immunol 2010, 125, 743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Alsum Z, Hawwari A, Alsmadi O, Al-Hissi S, Borrero E, Abu-Staiteh A, et al. Clinical, Immunological and Molecular Characterization of DOCK8 and DOCK8-like Deficient Patients: Single Center Experience of Twenty Five Patients. J Clin Immunol. 2012 Sep 12; doi: 10.1007/s10875-012-9769-x. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
- 72.Al-Herz W, Ragupathy R, Massaad MJ, Al-Attiyah R, Nanda A, Engelhardt KR, et al. Clinical, immunologic and genetic profiles of DOCK8-deficient patients in Kuwait. Clin Immunol. 2012;143:266–72. doi: 10.1016/j.clim.2012.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Chu EY, Freeman AF, Jing H, Cowen EW, Davis J, Su HC, et al. Cutaneous manifestations of DOCK8 deficiency syndrome. Arch Dermatol. 2012;148:79–84. doi: 10.1001/archdermatol.2011.262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Sanal O, Jing H, Ozgur T, Ayvaz D, Strauss-Albee DM, Ersoy-Evans S, et al. Additional diverse findings expand the clinical presentation of DOCK8 deficiency. J Clin Immunol. 2012;32:698–708. doi: 10.1007/s10875-012-9664-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Bittner TC, Pannicke U, Renner ED, Notheis G, Hoffmann F, Belohradsky BH, et al. Successful long-term correction of autosomal recessive hyper-IgE syndrome due to DOCK8 deficiency by hematopoietic stem cell transplantation. Klin Pädiatr. 2010;222:351–55. doi: 10.1055/s-0030-1265135. [DOI] [PubMed] [Google Scholar]
- 76.Gatz SA, Benninghoff U, Schütz C, Schulz A, Hönig M, Pannicke U, et al. Curative treatment of autosomal-recessive hyper-IgE syndrome by hematopoietic cell transplantation. Bone Marrow Transplant. 2011;46:552–56. doi: 10.1038/bmt.2010.169. [DOI] [PubMed] [Google Scholar]
- 77.Barlogis V, Galambrun C, Chambost H, Lamoureux-Toth S, Petit P, Stephan JL, et al. Successful allogeneic hematopoietic stem cell transplantation for DOCK8 deficiency. J Allergy Clin Immunol. 2011;128:420–22. doi: 10.1016/j.jaci.2011.03.025. [DOI] [PubMed] [Google Scholar]
- 78.Ghosh S, Schuster FR, Fuchs I, Laws HJ, Borkhardt A, Meisel R. Treosulfan-based conditioning in DOCK8 deficiency: Complete lympho-hematopoietic reconstitution with minimal toxicity. Clin Immunol. 2012;145:259–61. doi: 10.1016/j.clim.2012.10.003. [DOI] [PubMed] [Google Scholar]
- 79.Randall KL, Chan SS, Ma CS, Fung I, Mei Y, Yabas M, et al. DOCK8 deficiency impairs CD8 T cell survival and function in humans and mice. J Exp Med. 2011;208:2305–20. doi: 10.1084/jem.20110345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Dasouki M, Okonkwo KC, Ray A, Folmsbeel CK, Gozales D, Keles S, et al. Deficient T cell receptor excision circles (TRECs) in autosomal recessive hyper IgE syndrome caused by DOCK8 mutation: implications for pathogenesis and potential detection by newborn screening. Clin Immunol. 2011;141:128–32. doi: 10.1016/j.clim.2011.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Su HC, Jing H, Zhang Q. DOCK8 deficiency. Ann N Y Acad Sci. 2011;1246:26–33. doi: 10.1111/j.1749-6632.2011.06295.x. [DOI] [PubMed] [Google Scholar]
- 82.Lambe T, Crawford G, Johnson AL, Crockford TL, Bouriez-Jones T, Smyth AM, et al. DOCK8 is essential for T-cell survival and the maintenance of CD8+ T-cell memory. Eur J Immunol. 2011;41:3423–35. doi: 10.1002/eji.201141759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Harada Y, Tanaka Y, Terasawa M, Pieczyk M, Habiro K, Katakai T, et al. DOCK8 is a Cdc42 activator critical for interstitial dendritic cell migration during immune responses. Blood. 2012;119:4451–61. doi: 10.1182/blood-2012-01-407098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Jabara HH, McDonald DR, Janssen E, Massaad MJ, Ramesh N, Borzutzky A, et al. DOCK8 functions as an adaptor that links TLR-MyD88 signaling to B cell activation. Nat Immunol. 2012;13:612–20. doi: 10.1038/ni.2305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Randall KL, Lambe T, Johnson AL, Treanor B, Kucharska E, Domaschenz H, et al. Dock8 mutations cripple B cell immunological synapses, germinal centers and long-lived antibody production. Nat Immunol. 2009;10:1283–91. doi: 10.1038/ni.1820. Erratum in: Nat Immunol 2009, 11, 644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Hirve N, Levytskyy RM, Rigaud S, Guimond DM, Zal T, Sauer K, Tsoukas CD. A conserved motif in the ITK PH-domain is required for phosphoinositide binding and TCR signaling but dispensable for adaptor protein interactions. PLoS One. 2012;7:e45158. doi: 10.1371/journal.pone.0045158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Schaeffer EM, Debnath J, Yap G, McVicar D, Liao XC, Littman DR, et al. Requirement for Tec kinases Rlk and Itk in T cell receptor signaling and immunity. Science. 1999;284:638–41. doi: 10.1126/science.284.5414.638. [DOI] [PubMed] [Google Scholar]
- 88.Berg LJ, Finkelstein LD, Lucas JA, Schwartzberg PL. Tec family kinases in T lymphocyte development and function. Annu Rev Immunol. 2005;23:549–600. doi: 10.1146/annurev.immunol.22.012703.104743. [DOI] [PubMed] [Google Scholar]
- 89.Xie Q, Joseph RE, Fulton DB, Andreotti AH. Substrate Recognition of PLC-γ via a Specific Docking Surface on Itk. J Mol Biol. 2012 Dec 3; doi: 10.1016/j.jmb.2012.10.023. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Singleton KL, Gosh M, Dandekar RD, Au-Yeung BB, Ksionda O, Tybulewicz VL, et al. Itk controls the spatiotemporal organization of T cell activation. Sci Signal. 2011;4:ra66. doi: 10.1126/scisignal.2001821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Huck K, Feyen O, Niehues T, Rüschendorf F, Hübner N, Laws HJ, et al. Girls homozygous for an IL-2-inducible T cell kinase mutation that leads to protein deficiency develop fatal EBV-associated lymphoproliferation. J Clin Invest. 2009;119:1350–58. doi: 10.1172/JCI37901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Stepensky P, Weintraub M, Yanir A, Revel-Vilk S, Krux F, Huck K, et al. IL-2-inducible T-cell kinase deficiency: clinical presentation and therapeutic approach. Haematologica. 2011;96:472–76. doi: 10.3324/haematol.2010.033910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Linka RM, Risse SL, Bienemann K, Werner M, Linka Y, Krux F, et al. Loss-of-function mutations within the IL-2 inducible kinase ITK in patients with EBV-associated lymphoproliferative diseases. Leukemia. 2012;26:963–71. doi: 10.1038/leu.2011.371. [DOI] [PubMed] [Google Scholar]
- 94.Mansouri D, Mahdaviani SA, Khalilzadeh S, Mohajerani SA, Hasanzad M, Sadr S, et al. IL-2-inducible T-cell kinase deficiency with pulmonary manifestations due to disseminated Epstein-Barr virus infection. Int Arch Allergy Immunol. 2012;158:418–2. doi: 10.1159/000333472. [DOI] [PubMed] [Google Scholar]
- 95.Bachmann MF, Littman DR, Liao XC. Antiviral immune responses in Itk-deficient mice. J Virol. 2007;71:7253–57. doi: 10.1128/jvi.71.10.7253-7257.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Atherly LO, Brehm MA, Welsh RM, Berg LJ. Tec kinases Itk and Rlk are required for CD8+ T cell responses to virus infection independent of their role in CD4+ T cell help. J Immunol. 2006;176:1571–81. doi: 10.4049/jimmunol.176.3.1571. Erratum in: J Immunol 2006, 176, 3842. [DOI] [PubMed] [Google Scholar]
- 97.Blomberg KE, Boucheron N, Lindvall JM, Yu L, Raberger J, Berglöf A, et al. Transcriptional signatures of Itk-deficient CD3+, CD4+ and CD8+ T-cells. BMC Genomics. 2009;10:233. doi: 10.1186/1471-2164-10-233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Broussard C, Fleischacker C, Horai R, Chetana M, Venegas AM, Sharp LL, et al. Altered development of CD8+ T cell lineages in mice deficient for the Tec kinases Itk and Rlk. Immunity. 2006;25:93–104. doi: 10.1016/j.immuni.2006.05.011. Erratum in: Immunity 2006, 25, 849. [DOI] [PubMed] [Google Scholar]
- 99.Atherly LO, Lucas JA, Felices M, Yin CC, Reiner SL, Berg LJ. The Tec family tyrosine kinases Itk and Rlk regulate the development of conventional CD8+ T cells. Immunity. 2006;25:79–91. doi: 10.1016/j.immuni.2006.05.012. [DOI] [PubMed] [Google Scholar]
- 100.Horai R, Mueller KL, Handon RA, Cannons JL, Anderson SM, Kirby MR, et al. Requirements for selection of conventional and innate T lymphocyte lineages. Immunity. 2007;27:775–85. doi: 10.1016/j.immuni.2007.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Qi Q, Huang W, Bai Y, Balmus G, Weiss RS, August A. A unique role for ITK in survival of invariant NKT cells associated with the p53-dependent pathway in mice. J Immunol. 2012;188:3611–9. doi: 10.4049/jimmunol.1102475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Pasquier B, Yin L, Fondanèche MC, Relouzat F, Bloch-Queyrat C, Lambert N, et al. Defective NKT cell development in mice and humans lacking the adapter SAP, the X-linked lymphoproliferative syndrome gene product. J Exp Med. 2005;201:695–701. doi: 10.1084/jem.20042432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Nichols KE, Hom J, Gong SY, Ganguly A, Ma CS, Cannons JL, et al. Regulation of NKT cell development by SAP, the protein defective in XLP. Nat Med. 2005;11:340–45. doi: 10.1038/nm1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Rybakin V, Clemen CS. Coronin proteins as multifunctional regulators of the cytoskeleton and membrane trafficking. Bioessays. 2005;27:625–32. doi: 10.1002/bies.20235. [DOI] [PubMed] [Google Scholar]
- 105.Föger N, Rangell L, Danilenko DM, Chan AC. Requirement for coronin 1 in T lymphocyte trafficking and cellular homeostasis. Science. 2006;313:839–42. doi: 10.1126/science.1130563. [DOI] [PubMed] [Google Scholar]
- 106.Mugnier B, Nal B, Verthuy C, Boyer C, Lam D, Chasson L, et al. Coronin-1A links cytoskeleton dynamics to TCRαβ-induced cell signaling. PLoS ONE. 2008;3:e3467. doi: 10.1371/journal.pone.0003467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Mueller P, Massner J, Jayachandran R, Combaluzier B, Albrecht I, Gatfield J, et al. Regulation of T cell survival through coronin-1-mediated generation of inositol- 1,4,5-trisphosphate and calcium mobilization after T cell receptor triggering. Nat Immunol. 2008;9:424–31. doi: 10.1038/ni1570. [DOI] [PubMed] [Google Scholar]
- 108.Shiow LR, Roadcap DW, Paris K, Watson SR, Grigorova IL, Lebet T, et al. The actin regulator coronin 1A is mutant in a thymic egress-deficient mouse strain and in a patient with severe combined immunodeficiency. Nat Immunol. 2008;9:1307–15. doi: 10.1038/ni.1662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Shiow LR, Paris K, Akana MC, Cyster JG, Sorensen RU, Puck JM. Severe combined immunodeficiency (SCID) and attention deficit hyperactivity disorder (ADHD) associated with a Coronin-1A mutation and a chromosome 16p11. 2 deletion. Clin Immunol. 2009;131:24–30. doi: 10.1016/j.clim.2008.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Vig M, Kinet JP. Calcium signaling in immune cells. Nat Immunol. 2009;10(1):21–27. doi: 10.1038/ni.f.220. Erratum in: Nat Immunol 2009, 10, 223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Shaw PJ, Feske S. Regulation of lymphocyte function by ORAI and STIM proteins in infection and autoimmunity. J Physiol. 2012;590:4157–67. doi: 10.1113/jphysiol.2012.233221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Feske S, Gwack Y, Prakriya M, Srikanth S, Puppel SH, Tanasa B, et al. A mutation in Orai1 causes immune deficiency by abrogating CRAC channel function. Nature. 2006;441:179–85. doi: 10.1038/nature04702. [DOI] [PubMed] [Google Scholar]
- 113.Picard C, McCarl CA, Papolos A, Khalil S, Lüthy K, Hivroz C, et al. STIM1 mutation associated with a syndrome of immunodeficiency and autoimmunity. N Engl J Med. 2009;360:1971–80. doi: 10.1056/NEJMoa0900082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.McCarl CA, Picard C, Khalil S, Kawasaki T, Röther J, Papolos A, et al. ORAI1 deficiency and lack of store-operated Ca2+ entry cause immunodeficiency, myopathy, and ectodermal dysplasia. J Allergy Clin Immunol. 2009;124:1311–18. doi: 10.1016/j.jaci.2009.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Byun M, Abhyankar A, Lelarge V, Plancoulaine S, Palanduz A, Telhan L, et al. Whole-exome sequencing-based discovery of STIM1 deficiency in a child with fatal classic Kaposi sarcoma. J Exp Med. 2010;207:2307–12. doi: 10.1084/jem.20101597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Fuchs S, Rensing-Ehl A, Speckmann C, Bengsch B, Schmitt-Graeff A, Bondzio I, et al. Antiviral and regulatory T cell immunity in a patient with stromal interaction molecule 1 deficiency. J Immunol. 2012;188:1523–33. doi: 10.4049/jimmunol.1102507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Le Deist F, Hivroz C, Partiseti M, Thomas C, Buc HA, Oleastro M, et al. A primary T-cell immunodeficiency associated with defective transmembrane calcium influx. Blood. 1995;85:1053–62. [PubMed] [Google Scholar]
- 118.Feske S, Müller JM, Graf D, Kroczek RA, Dräger R, Niemeyer C, et al. Severe combined immunodeficiency due to defective binding of the nuclear factor of activated T cells in T lymphocytes of two male siblings. Eur J Immunol. 1996;26:2119–26. doi: 10.1002/eji.1830260924. [DOI] [PubMed] [Google Scholar]
- 119.Maul-Pavicic A, Chiang SC, Rensing-Ehl A, Jessen B, Fauriat C, Wood SM, et al. ORAI1-mediated calcium influx is required for human cytotoxic lymphocyte degranulation and target cell lysis. Proc Natl Acad Sci USA. 2011;108:3324–29. doi: 10.1073/pnas.1013285108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Kim KD, Srikanth S, Yee MK, Mock DC, Lawson GW, Gwack Y. ORAI1 deficiency impairs activated T cell death and enhances T cell survival. J Immunol. 2011;187:3620–30. doi: 10.4049/jimmunol.1100847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Matsumoto M, Fujii Y, Baba A, Hikida M, Kurosaki T, Baba Y. The calcium sensors STIM1 and STIM2 control B cell regulatory function through interleukin-10 production. Immunity. 2011;34:703–14. doi: 10.1016/j.immuni.2011.03.016. [DOI] [PubMed] [Google Scholar]
- 122.Bergmeier W, Oh-Hora M, McCarl CA, Roden RC, Bray PF, Feske S. R93W mutation in Orai1 causes impaired calcium influx in platelets. Blood. 2009;113:675–78. doi: 10.1182/blood-2008-08-174516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Völkers M, Dolatabadi N, Gude N, Most P, Sussman MA, Hassel D. Orai1 deficiency leads to heart failure and skeletal myopathy in zebrafish. J Cell Sci. 2012;125:287–94. doi: 10.1242/jcs.090464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Robinson LJ, Mancarella S, Songsawad D, Tourkova IL, Barnett JB, Gill DL, et al. Gene disruption of the calcium channel Orai1 results in inhibition of osteoclast and osteoblast differentiation and impairs skeletal development. Lab Invest. 2012;92:1071–83. doi: 10.1038/labinvest.2012.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Brandao K, Deason-Towne F, Perraud AL, Schmitz C. The role of Mg2+ in immune cells. Immunol Res. 2012 Sep 19; doi: 10.1007/s12026-012-8371-x. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
- 126.Schlingmann KP, Weber S, Peters M, Niemann Nejsum L, Vitzthum H, Klingel K, et al. Hypomagnesemia with secondary hypocalcemia is caused by mutations in TRPM6, a new member of the TRPM gene family. Nat Genet. 2002;31:166–70. doi: 10.1038/ng889. [DOI] [PubMed] [Google Scholar]
- 127.Walder RY, Landau D, Meyer P, Shalev H, Tsolia M, Borochowitz Z, et al. Mutation of TRPM6 causes familial hypomagnesemia with secondary hypocalcemia. Nat Genet. 2002;31:171–4. doi: 10.1038/ng901. [DOI] [PubMed] [Google Scholar]
- 128.Li FY, Chaigne-Delalande B, Kanellopoulou C, Davis JC, Matthews HF, Douek DC, et al. Second messenger role for Mg2+ revealed by human T-cell immunodeficiency. Nature. 2011;475:471–76. doi: 10.1038/nature10246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Goytain A, Quamme GA. Identification and characterization of a novel mammalian Mg2+ transporter with channel-like properties. BMC Genomics. 2005;6:48. doi: 10.1186/1471-2164-6-48. [DOI] [PMC free article] [PubMed] [Google Scholar]

