Highlights
▶ Autosomal recessive complete loss-of-function STAT1 deficiency predisposes to mycobacterial and viral diseases. ▶ Autosomal recessive partial loss-of-function STAT1 deficiency predisposes to mild mycobacterial and viral diseases. ▶ Autosomal dominant loss-of-function STAT1 deficiency predisposes to mycobacterial disease. ▶ Autosomal dominant gain-of-function STAT1 disorder predisposes to chronic mucocutaneous disease.
Abstract
The genetic dissection of various human infectious diseases has led to the definition of inborn errors of human STAT1 immunity of four types, including (i) autosomal recessive (AR) complete STAT1 deficiency, (ii) AR partial STAT1 deficiency, (iii) autosomal dominant (AD) STAT1 deficiency, and (iv) AD gain of STAT1 activity. The two types of AR STAT1 defect give rise to a broad infectious phenotype with susceptibility to intramacrophagic bacteria (mostly mycobacteria) and viruses (herpes viruses at least), due principally to the impairment of IFN-γ-mediated and IFN-α/β-mediated immunity, respectively. Clinical outcome depends on the extent to which the STAT1 defect decreases responsiveness to these cytokines. AD STAT1 deficiency selectively predisposes individuals to mycobacterial disease, owing to the impairment of IFN-γ-mediated immunity, as IFN-α/β-mediated immunity is maintained. Finally, AD gain of STAT1 activity is associated with autoimmunity, probably owing to an enhancement of IFN-α/β-mediated immunity. More surprisingly, it is also associated with chronic mucocutaneous candidiasis, through as yet undetermined mechanisms involving an inhibition of the development of IL-17-producing T cells. Thus, germline mutations in human STAT1 define four distinct clinical disorders. Various combinations of viral, mycobacterial and fungal infections are therefore allelic at the human STAT1 locus. These experiments of Nature neatly highlight the clinical and immunological impact of the human genetic dissection of infectious phenotypes.
Current Opinion in Immunology 2012, 24:364–378
This review comes from a themed issue on Host pathogens
Edited by Anne O’Garra and Eric Pamer
For a complete overview see the Issue and the Editorial
Available online 30th May 2012
0952-7915/$ – see front matter, © 2012 Elsevier Ltd. All rights reserved.
Introduction: structure and function of STAT1
STAT1 was the first STAT to be identified, in 1989, and is a key transcription factor mediating IFN-α/β signaling [1, 2, 3, 4, 5]. In mice and humans, the STAT1 gene encodes two STAT1 isoforms, generated by alternative splicing. STAT1-α is transcriptionally active and encodes a protein of 750 amino acids (91 kDa), whereas STAT1-β (84 kDa) acts as its dominant-negative inhibitor as it lacks part of the transactivation domain and the Ser phosphorylation site (Ser 727) [6]. STAT1-α (hereafter referred to as STAT1) is involved in various signaling pathways, including the IFN-α/β, IFN-γ, IFN-λ, IL-2, IL-3, IL-6, IL-9, IL-10, IL-11, IL-12, IL-15, IL-21, IL-22, IL-26, IL-27, EGF, VEGF, FGF, HGF, GH, angiotensin and OSM pathways [7]. STAT1 activation conforms to the same general pattern as for other STATs (Figure 1 )[8, 9, 10]. Briefly, the binding of the soluble extracellular agonist to its specific cell surface receptor leads to the activation of specific JAKs constitutively associated with the receptor chains. The JAKs phosphorylate the intracellular domains of the receptors, thereby creating a docking site for latent cytoplasmic STAT1, which is recruited, undergoes Tyr 701 phosphorylation and is then released from the receptor complex. Phosphorylated STAT1 can form homodimers and heterodimers and multimers, which are then translocated to the nucleus, where they bind to conserved genetic boxes to activate or modulate the transcription of specific target genes [10]. STAT1 is subsequently dephosphorylated in the nucleus [11, 12, 13] and exported back to the cytoplasm, where it remains as a monomer or antiparallel unphosphorylated dimer [14, 15]. A role for unphosphorylated STAT1 (U-STAT1) in the mediation of some transcriptional activity has been reported [16]. Other posttranslational modifications of STAT1, in addition to the phosphorylation of Tyr and Ser residues, have been reported to contribute the activity of this protein. These modifications include acetylation, methylation and sumoylation [17, 18, 19, 20]. However, the specific roles of posttranslational modifications other than Tyr 701 and Ser 727 phosphorylation remain to be clarified.
The N-terminal domain (NTD) of STAT1 is known to be involved in the interaction of the protein with its surface receptor [21], and in phosphorylation [21], nuclear translocation [22] and transcriptional activity, through the facilitation of tetramerization [23, 24] and dephosphorylation (Figure 1, Figure 2 ) [22, 25, 26]. The coiled-coil domain (CC) has considerable potential for involvement in protein-protein interactions and plays a key role in the dimerization of unphosphorylated STAT1 and nuclear STAT1 dephosphorylation [10, 14, 26, 27, 28, 29, 30]. The DNA-binding domain (DNA-B) includes residues such as Asn 460 and Lys 336, which come into contact with the DNA major groove, and Glu 421, which comes into contact with the minor groove [28]. In addition, some residues contribute to the nuclear import of phosphorylated STAT1 dimers by binding to importin-α5 [31, 32, 33, 34], whereas others are involved in the nuclear export of the protein [12, 35, 36]. The linker domain (L) is involved in IFN-γ-driven transcription [37] and in the stability of DNA binding [38]. The SH2 domain (SH2) plays an essential role in binding to the phosphorylated surface receptor and to the phosphorylated tail of other STATs [28]. The tail segment (TS) includes the crucial Tyr 701 residue, which is phosphorylated by JAKs upon activation, thereby facilitating dimerization through interaction with the SH2 domain of another STAT [28]. The transactivation domain (TA) enables the protein to induce or modulate the transcription of target genes. It also contains the Ser 727 residue, the phosphorylation of which increases the transcriptional activity of STAT1 [10, 39]. The structure-function relationship of various domains and residues of STAT1 has been characterized in detail.
Immunological and infectious phenotypes of STAT1-deficient mice
STAT1 knockout mice were generated in 1996 by two different groups [40, 41]. Despite the demonstration of STAT1 activation by diverse cytokines and hormones in vitro [3, 5, 7], the knockout mice were born in Mendelian proportions, displayed no overt developmental abnormalities and their leukocyte counts were not low for the populations of cells tested (T cells, B cells, monocytes, macrophages and granulocytes), with the exception of CD4+CD25+ regulatory T cells [42]. However, these mice responded very poorly to IFN-α/β and IFN-γ, as demonstrated by the absence of detectable gamma activating factor (GAF; STAT1 homodimer) and interferon-stimulated gene factor 3 (ISGF3; STAT1/STAT2/IRF9 heterotrimer). The transcription of most of the genes usually induced by IFN-α/β and IFN-γ was abolished, highlighting a non-redundant role of STAT1 in the two corresponding pathways. Interestingly, residual STAT1-independent responses were subsequently reported [43, 44]. It was not until 2004 and 2010 that STAT1 was also shown to be non-redundant for cellular responses to IFN-λ and IL-27, respectively [45, 46, 47, 48, 49]. Overall, correct cellular responses to at least four cytokines in the mouse model seem to be highly dependent on STAT1. Mouse STAT1 appears to be redundant for the responses to at least 11 other cytokines (IL-2, IL-3, IL-6, IL-9, IL-10, IL-11, IL-12, IL-15, IL-21, IL-22, IL-26), and seven hormones (EGF, VEGF, FGF, HGF, GH, angiotensin, OSM), at least in the experimental conditions tested.
STAT1 knockout mice have been challenged with at least 27 pathogens, including 17 viruses, five bacteria, four parasites and one fungus (Table 1 ). Unsurprisingly, STAT1-deficient mice are highly susceptible to most viruses, including (+) single-strand RNA viruses, such as Sindbis [43], chikungunya virus (CHIKV) [50, 51], mouse hepatitis virus (MHV) [40], severe acute respiratory syndrome coronavirus (SARS-CoV) [52] and murine norovirus 1 [53, 54], (−) single-strand RNA viruses, such as vesicular stomatitis virus (VSV) [40, 41], influenza virus A [48, 55], filoviruses (Ebola, Marburg, Reston Ebola) [56•, 57•], Machupo virus (MACV) [58•], Crimean-Congo hemorrhagic fever virus (CCHFV) [59•], and Sendai virus (SeV) [60], double-strand DNA viruses, such as herpes simplex virus (HSV) [61, 62•] and mouse cytomegalovirus (MCMV) [43]. However, they seem to be normally resistant to the (+) single-strand RNA dengue virus [63, 64•] and to the (−) single-strand RNA measles viruses [65, 66]. STAT1 knockout mice are less susceptible to some infections (MCMV and Sindbis viruses) than mice with a double knockout for IFN-γR and IFN-α/βR, presumably owing to STAT1-independent responses [43]. STAT1 knockout mice are also susceptible to infection with intracellular Gram-positive bacteria, such as Listeria monocytogenes [40] and Mycobacterium tuberculosis [67], and intracellular Gram-negative bacteria, such as Francisella novicida [68] and Chlamydia pneumoniae [69], but not in the largely extracellular Borrelia burgdorferi [70]. STAT1-deficient mice are highly susceptible to the parasites Toxoplasma gondii [71, 72], Cryptosporidium parvum [73] and Leishmania major [74], but are normally resistant to infection with Leishmania donovani [75]. Most of these bacteria and parasites infect macrophages, and the susceptibility of STAT1-deficient mice probably closely resembles that of IFN-γR-deficient mice. STAT1-deficient mice have also been challenged with the fungus Candida albicans; their response to this challenge was similar to that of wild-type mice [76]. To our knowledge, mice heterozygous for a null STAT1 allele have not been tested. Transgenic mice overexpressing wild-type STAT1 and mice carrying hypomorphic or hypermorphic STAT1 alleles do not appear to have been described either.
Table 1.
Pathogens | Classification | Additional information | S or N | Reference |
---|---|---|---|---|
Sindbis (SINV) | Virus | Togaviridae ss RNA (+) | S | [43] |
Chikungunya virus (CHIKV) | Virus | Togaviridae ss RNA (+) | S | [50, 51] |
Mouse hepatitis virus (MHV) | Virus | Coronaviridae ss RNA (+) | S | [40] |
Severe acute respiratory syndrome coronavirus (SARS-CoV) | Virus | Coronaviridae ss RNA (+) | S | [52] |
Murine norovirus 1 (MNO1) | Virus | Caliciviridae ss RNA (+) | S | [53, 54] |
Dengue virus (DENV) | Virus | Flaviviridae ss RNA (+) | N | [63, 64•] |
Vesicular stomatitis virus (VSV) | Virus | Rhabdoviridae ss RNA (−) | S | [40, 41] |
Influenza A virus | Virus | Orthomyxoviridae ss RNA (−) | S | [48] |
Filoviruses (Ebola, Marburg, Reston Ebola) | Virus | Filoviridae ss RNA (−) | S | [56•, 57•] |
Machupo Virus (MACV) | Virus | Arenaviridae ss RNA (−) | S | [58•] |
Crimean-Congo hemorrhagic fever virus (CCHFV) | Virus | Bunyaviridae ss RNA (−) | S | [59•] |
Sendai virus (SeV) | Virus | Paramyxoviridae ss RNA (−) | S | [60] |
Measles virus | Virus | Paramyxoviridae ss RNA (−) | N | [65, 66] |
Herpes simplex virus (HSV) | Virus | Herpesviridae ds DNA | S | [61, 62•] |
Mouse cytomegalovirus (MCMV) | Virus | Herpesviridae ds DNA | S | [43] |
Listeria monocytogenes | Bacteria | Gram + | S | [40] |
Mycobacterium tuberculosis | Bacteria | Acid fast Gram + | S | [67] |
Francisella novicida | Bacteria | Gram − | S | [68] |
Chlamydia pneumoniae | Bacteria | Gram − | S | [69] |
Borrelia burgdorferi | Bacteria | Gram − | N | [70] |
Toxoplasma gondii | Parasite | S | [71, 72] | |
Cryptosporidium parvum | Parasite | S | [73] | |
Leishmania major | Parasite | S | [74] | |
Leishmania donovani | Parasite | N | [75] | |
Candida albicans | Fungus | N | [76] |
S: susceptible. N: normal.
Human AR complete STAT1 deficiency: life-threatening intramacrophagic bacterial and viral diseases
A new syndrome characterized by the association of infectious diseases caused by intracellular bacteria (typically mycobacteria) and viruses (typically herpes viruses) was clinically described and genetically elucidated in 2003 (Table 2 , Figure 1, Figure 2, Figure 3, Figure 4, Figure 5 ) [77]. Some infections resemble those seen in patients with T-cell combined immunodeficiencies, except that the patients have normal T-lymphocyte counts and function [78]. The known patients carry biallelic loss-of-expression and loss-of-function (LOF) STAT1 alleles, leading to a complete absence of the wild-type protein [77]. They have missense mutations (L600P and Q123H, leading to the in-frame splicing out of coding exon 3), small frameshift deletions (1758_1759delAG), and small frameshift insertions (1928insA). Only six patients from four unrelated families have been identified to date (Table 2) (Figure 2) [77, 79, 80•] (unpublished data). This is a purely recessive disorder and no haplo-insufficiency at the STAT1 locus has been reported for any of the known cellular or clinical phenotypes. The patients are susceptible to weakly virulent mycobacteria, such as BCG vaccines and Mycobacterium kansasii, and to herpes viruses, such as HSV-1 and CMV. The control of attenuated poliovirus type III vaccine, rhinovirus and parainfluenza virus type II in one patient suggests that STAT-1-independent innate antiviral mechanisms may be sufficient in humans [79, 81]. Nevertheless, five patients died: two at 3 months of age from mycobacterial disease, two at 12 and 16 months of age from viral disease (including one from HSV-1 encephalitis, HSE) [82•] and another at 11 months of age from severe hepatitis in the course of hematopoietic stem cell transplantation (HSCT) [79]. The sixth patient is still alive one year after HSCT [80•]. Some key infectious phenotypes of STAT1-deficient patients have been seen in patients with other inborn errors of cytokine immunity. The TLR3-dependent induction of IFN-α/β and IFN-λ was shown to be impaired in other children with HSE [82•]. Moreover, mycobacterial disease in some other children has been shown to result from impaired IFN-γ-mediated immunity [83]. Both TLR3 and IFN-γR deficiencies are also life-threatening disorders.
Table 2.
Number of patients described | Number of kindreds | STAT1 domain affected | Main cellular phenotype | Main clinical phenotype | |
---|---|---|---|---|---|
AR complete STAT1 deficiency | 6 | 4 | NTD, SH2 | No STAT1-dependent response to IFN-γ, IFN-α/β, IFN-λ and IL-27 | Lethal intracellular bacteria (mainly mycobacteria) and viral (mainly herpes) diseases |
AR partial STAT1 deficiency | 5 | 3 | NTD, CC, TS | Impaired STAT1-dependent response to IFN-γ, IFN-α/β, IFN-λ and IL-27 | Curable intracellular bacteria (mainly mycobacteria) and viral (mainly herpes) diseases |
AD LOF STAT1 deficiency | 16 | 8 | DNA-B, SH2, TS | Impaired STAT1-dependent response to IFN-γ and IL-27 | Mycobacterial diseases |
AD GOF STAT1 disorder | 68 | 27 | CC | Enhanced STAT1-dependent response to at least IFN-γ, IFN-α/β, IL-27, IL-6 and IL-21. Impaired IL-17 T-cell immunity | CMCD and autoimmunity |
AR: autosomal recessive. AD: autosomal dominant. LOF: loss-of-function. GOF: gain-of-function. NTD: N-terminal domain. SH2: SH2 domain. CC: coiled-coil domain. DNA-B: DNA-binding domain. TS: tail segment domain. To date, incomplete clinical penetrance is only observed for AD LOF STAT1 deficiency. The unpublished identification of defects in AD LOF STAT1 deficiency (mutation Y701F in one kindred) and AD GOF STAT1 disorder (24 and 18 CMCD kindreds with mutations in the STAT1 DNA-B and CCD, respectively) are not taking into account in the table.
EBV-transformed B cells (EBV-B cells) and SV40-transformed fibroblasts (SV40-fibroblasts) from these patients were unresponsive to IFN-γ and IFN-α in terms of both GAF and ISGF3 activity [77, 79, 80•]. They were also unresponsive to IL-27 and IFN-λ stimulation, in terms of GAF-DNA binding activity and the induction of target genes, such as IFIT1, respectively [84]. EBV-B cells and SV-40 fibroblasts from patients with complete STAT1 deficiency cannot control the replication of viruses, such as VSV, EMCV, or HSV-1, even after treatment with IFN-α [77, 79]. In addition, induction of MxA after Influenza A infection was shown to require IFN-α/β-dependent or IFN-λ STAT-1-dependent signaling [85]. Moreover, leukocytes from these patients have an impaired response to IFN-γ in terms of the induction of IL-12 and other target genes. The STAT1-independent induction of IFN-γ has been documented in IFN-α-stimulated NK cells from a patient [80•]. In addition, STAT1 has been shown to be dispensable for T follicular helper (Tfh) cell differentiation [86•], memory B-cell formation and IL-21-induced Ig secretion in patients’ cells in vitro [87•]. The overall susceptibility of patients to mycobacteria and other intracellular bacteria may be largely explained by the impairment of IFN-γ-GAF immunity. Indeed, IFN-γ has already been shown to be essential for human antimycobacterial immunity (Figure 3, Figure 4, Figure 5) (see below [83]). A role for impaired IL-27 immunity, although unlikely [88], is possible, at least if we consider IL-27 as an IFN-γ-inducing cytokine (through a decrease in STAT1-independent IFN-γ responses). By contrast, the observed susceptibility to viruses is largely owing to the impairment of IFN-α/β responses, as inferred from the susceptibility of IFN-α/βR-deficient mice (Figure 3). A contribution of impaired IFN-λ immunity is plausible, at least for some viral infections, as inferred from both the mouse model [89•, 90] and the role of human IFN-λ in the clearance of hepatitis C virus [91••, 92••, 93••, 94••]. Patients lacking IL-10R2 (one of the two chains of the receptor for IFN-λ) have not been reported to be particularly susceptible to viral diseases [95]. However, these rare patients died or underwent HSCT early in life, possibly preventing the expression of a viral phenotype. In any case, the cellular and clinical penetrance of complete STAT1 deficiency appears to be complete, as all homozygous patients have an equally severe cellular and clinical phenotype. HSCT appears to be the only curative treatment for these patients. The identification and investigation of a larger number of patients is required to improve delineation of the redundant and non-redundant roles of human STAT1 in cell signaling, host defense and beyond.
AR partial STAT1 deficiency: milder intramacrophagic bacterial and viral diseases
From 2009 onwards, five patients from three unrelated kindreds were identified with a partial, as opposed to complete form of AR STAT1 deficiency (Table 2) (Figure 2) [96, 97•, 98]. The patients harbor biallelic STAT1 mutations (homozygous in two kindreds and compound heterozygous in the third), which are hypomorphic (as opposed to amorphic). The four mutations are missense and all reduce but do not abolish STAT1 production. One mutation affects the NTD (A46T), two affect the CCD (K201N and K211R) and one affects the TSD (P696S). No precise functional data are available for the A46T mutation, but the other three mutations are unusual in being not only missense mutations, but also, primarily, splice mutations. Indeed, the two mutations in the CCD region cause the splicing-out of exon 8 [97•, 98], whereas that in the TSD region splices out exon 23 [96]. The P696S and K201N mutations have no effect on the function of the residual full-length STAT1 produced, accounting for the partial nature of the defect [96, 97•]. The K211R mutation has not been characterized in this respect [98]. By comparison with complete STAT1 deficiency, this disorder is thus associated with milder intramacrophagic bacterial (Salmonella, M. avium, BCG) and viral (CMV, VZV, HSV-1, RSV) diseases, most of which are cured by antibacterial or antiviral drugs [96, 97•, 98]. Four of the five patients are alive and were aged 12, 14, 14 and 16 years at last follow-up. The fifth patient died of sepsis at the age three years, after an episode of disseminated mycobacterial disease. Two patients are seropositive for EBV and CMV and another is seropositive for EBV, CMV and VZV, so these patients were able to mount an effective immune response against at least these viruses, leading to their control [81, 97•, 98]. As they are now in their teens, they probably have been exposed to and controlled other viruses as well.
Unlike EBV-B cells and SV40-fibroblasts from patients with complete STAT1 deficiency, cells from patients with a partial deficiency produce residual non-null amounts of functional STAT1, corresponding to between 10% and 25% normal levels (depending on the mutation). Consequently, their cells have an impaired, but not abolished STAT1-dependent response to IFN-γ and IFN-α treatment, in terms of both GAF and ISGF3 activity [96, 97•]. They also display an impaired response to stimulation with IL-27 and IFN-λ. The ability of the patients’ cells to control viral infection (VSV and HSV) in vitro after treatment with IFN-α seems to depend on the mutation and, possibly, on the level of residual STAT1 activation after IFN-α stimulation. In any case, the impaired, but not abolished IFN-α/β and/or IFN-λ responses probably account for the viral infection phenotype being less severe than that in patients with complete STAT1 deficiency (Figure 3, Figure 4, Figure 5). Only a small number of patients with partial STAT1 deficiency have been identified, but it may be significant that none has had HSE [96, 97•, 98]. Similarly, the residual response to IFN-γ (and perhaps IL-27) may well account for their mycobacterial infections being curable. A thorough investigation of the cellular phenotype conferred by one of these STAT1 mutations (K201N/K201N) suggested that the expression of genes induced by IFN-γ during the early response phase was particularly impaired, at least in SV-40 fibroblasts, identifying this group of genes as important for antimycobacterial immunity [97•]. Overall, the defect being partial, these patients display less severe clinical diseases than patients with a complete defect. However, the degree of severity of the disease has been shown to vary between patients, even within a given family. The amount of functional residual STAT1 produced may differ between siblings with the same mutation (a splicing mutation) [96]. Nevertheless, the clinical penetrance of partial STAT1 deficiency also appears to be complete. Overall, levels of STAT1 production and activity are correlated with the severity of bacterial and viral infections in patients with AR STAT1 deficiency. There are clearly two different forms of AR STAT1 deficiency, studies of the natural history of which provide different and complementary information about the role of STAT1 in host defense.
AD STAT1 deficiency: Mendelian susceptibility to mycobacterial disease
Mendelian susceptibility to mycobacterial disease (MSMD) is a rare syndrome characterized by infections with weakly pathogenic mycobacteria (such as the BCG vaccine and environmental mycobacteria) in otherwise healthy individuals [83, 99]. The patients are also susceptible to the more virulent Mycobacterium tuberculosis and half of them also suffer from non-typhoidal salmonellosis [100, 101]. Since the discovery in 1996 of its first genetic etiology, eight morbid genes (IFNGR1, IFNGR2, STAT1, IRF8, IL12B, IL12RB1, NEMO and CYBB) have been found (Figure 4) [99, 102••, 103••]. They collectively define up to 15 genetic disorders affecting IL-12-dependent, IFNγ-mediated immunity. In this context, the first human patients with any form of inherited STAT1 deficiency were discovered in 2001, with the identification of two patients from unrelated kindreds, both heterozygous for the L706S LOF dominant-negative STAT1 allele [104]. These two patients suffered only from relatively mild MSMD (disseminated BCG disease and M. avium, both curable without recurrence). Two children of the first patient were also heterozygous and, in the absence of BCG vaccination, have remained healthy [84, 104]. None of the four patients identified had ever displayed any unusually severe viral illness at least until their most recent check-up visits at the ages of 10, 14, 21 and 44 years (unpublished data). Since 2001, up to six other kindreds have been found to carry dominant-negative STAT1 mutations associated with MSMD (Figure 2). All the mutations are missense, two mutations affect the DNA-B (E320Q and Q463H) [84], three others the SH2D (K637E, M654K and K673R) [105, 106] and a fifth mutation affects Tyr 701 itself (Y701C) (Hirata et al., in preparation). Most patients from the eight kindreds suffered from MSMD (BCG-osis or M. avium disease). One patient suffered only from bona fide tuberculosis, caused by M. tuberculosis [84]. For several mutations, clinical penetrance is incomplete, as up to five genetically affected relatives of index cases, two of whom are known not to have been vaccinated with BCG, have remained clinically healthy.
The transfection of fibrosarcoma cells lacking STAT1 owing to a somatic event with MSMD-causing STAT1 alleles has shown some of these alleles to be LOF (in a broad sense, including amorphic and hypomorphic alleles) owing to loss-of-phosphorylation (L706S, M654K and K673R) for both the IFN-γ-GAF and IFN-α-ISRE signaling pathways [104, 105, 106]. Others (E320Q and Q463H) were found to be deleterious owing to impaired DNA-binding [84]. One mutation (K637E) affects both Tyr 701 phosphorylation and DNA-binding activity [105]. However, in the cells of heterozygous patients, the mutant alleles were shown to be dominant for IFN-γ-GAF activity but recessive for IFN-α-ISRE activity. In other words, the response to IFN-γ was impaired in heterozygous cells, whereas the response to IFN-α/β was unaffected. This provided the first example of a human allele intrinsically null for two cellular phenotypes but recessive for one and dominant for the other in heterozygous cells [104]. We also know that this dichotomy applies at the level of the whole organism, as patients with AD STAT1 deficiency have MSMD, whereas those with AR STAT1 deficiency are susceptible to both mycobacterial and viral diseases. The mechanisms of dominance and recessiveness, for GAF and ISGF3 activation, respectively, have been deciphered [84]. There is no haploinsufficiency for STAT1, as assessed by GAF and ISGF3 DNA-binding activity, in cells heterozygous for a loss-of-expression STAT1 allele [84]. The mutant proteins are also intrinsically able to associate with phosphorylated IFN-γ-R1 and to form dimers with mutant or WT STAT1. The lack of phosphorylation of the mutant proteins or their inability to bind DNA therefore destroys the activity of dimers containing them. As a result, only dimers combining two phosphorylated wild-type STAT1 molecules are functional, accounting for the detection of about 25% the normal level of GAF activity (for null alleles) in response to IFN-γ in the patients’ cells. Conversely, some mutated STAT1 molecules (like L706S) do not bind to phosphorylated STAT2 recruited to and activated at the IFN-α/β receptor complex. Only WT STAT1 molecules are therefore recruited in heterozygous cells. Other mutated STAT1 molecules (in the DNA-B) bind phosphorylated STAT2 but do not impair the binding of ISGF3 with its DNA target. This results, in a normal response to IFN-α and the formation of ISGF3, albeit in sufficient quantities because there is no haploinsufficiency for the STAT1 locus. The ability of the patients’ cells to respond correctly to IFN-α/β and to control viral infections in vitro accounts for the lack of a clinical phenotype in vivo. The patients’ cells also respond well to IFN-λ in terms of IFIT1 mRNA induction [105]. As discussed above, the poor cellular response to IFN-γ accounts for the patients’ susceptibility to mycobacteria (Figure 3, Figure 4, Figure 5). In STAT1-deficient recipient cells, the transfected mutant STAT1 alleles are null for GAF activation in response to IL-27 [105]. Moreover, the patients’ cells also respond poorly to IL-27, potentially contributing to their clinical phenotype, given that IL-27 has been implicated in IFN-γ-dependent antibacterial immunity in the mouse [88]. The identification of patients carrying mutations in the genes encoding IL-27 or its receptor would facilitate assessments of the contribution of this molecule to antimycobacterial immunity. In any event, the dominant-negative STAT1 alleles identified do not confer a predisposition to viral disease, but they may underlie AD MSMD in rare patients, because although null or severely hypomorphic for both signaling pathways, these alleles impair IFN-γ responses, but not IFNα/β responses in heterozygous cells.
AD gain of STAT1 activity: CMCD and autoimmunity
Heterozygous gain-of-function STAT1 alleles were discovered in 2011, in patients with chronic mucocutaneous candidiasis (CMC) [107••, 108•]. Candida albicans is a fungus that causes invasive or chronic mucocutaneous disease in immunocompromised patients [109]. CMC is common in patients with other clinical signs carrying various inborn errors of immunity, including mutations in STAT3 [110, 111, 112, 113, 114], IL12B [111], IL12RB1 [111] and AIRE [115•, 116•]. All these defects are associated with impaired IL-17 immunity, whether owing to the impaired development of IL-17-producing T cells (mutations in STAT3 and, to a lesser extent, IL12RB1 and IL12B) or to high titers of neutralizing auto-Abs against IL-17 cytokines (AIRE) [115•]. This suggests that IL-17A, IL-17F and/or IL-22 play an important role in mucocutaneous immunity to C. albicans (Figure 4) [117]. Consistent with this hypothesis, genetic predisposition to CMC disease (CMCD) in rare patients without the clinical features associated with the aforementioned defects was attributed to a specific hypomorphic heterozygous mutation of IL17F in a kindred with AD CMCD, and to biallelic amorphic mutations of IL17RA in a kindred with AR CMCD [117, 118••]. Surprisingly, heterozygous mutations of STAT1 were discovered in this context by whole-exome sequencing [107••] or genome-wide linkage analysis [108•] in patients with CMCD, including some patients with autoimmune signs (Figure 2, Figure 5) [107••, 108•]. A first report described 12 mutations in 47 patients from 20 kindreds [107••]. Another report identified two mutations in 14 patients from five families [108•]. A third report subsequently identified two other families and seven patients with CMCD carrying a known mutation [119]. Altogether, 68 patients with CMCD from 27 kindreds have been reported to be heterozygous for STAT1 mutant alleles (Table 2). Remarkably, all these mutations affect the CCD of STAT1. Moreover, all are missense mutations, including six found in several unrelated kindreds (R274Q, M202V, R274W, T288A, M202I, and A267V), and they appear to be due to mutational hotspots [107••] rather than a founder effect [108•]. The mutations segregate with clinical phenotype, and clinical penetrance appears to be complete, although clinical severity varies between and even within families [107••, 108•]. In addition, we recently identified 24 and 18 new CMCD patients from 15 and 13 families heterozygous for mutations affecting the DNA-B and CCD of STAT1, respectively (Okada S. et al., in preparation). Heterozygous STAT1 mutations account for approximately half the patients enrolled in our CMC cohort.
These CMCD-causing mutant STAT1 alleles were shown to be gain-of-function (GOF) [107••]. At the cellular level, this gain of function results from a gain of phosphorylation [107••]. Following the transfection of STAT1-deficient fibrosarcoma cells with these alleles, the response to IFN-γ, IFN-α, IL-27, IL-6 and IL-21 was reproducibly two to three times stronger than that in control cells, in terms of STAT1 phosphorylation, GAS-binding activity, reporter gene induction, and the induction of some endogeneous target genes. The mechanism underlying the GOF has been analyzed with inhibitors of kinases and phosphatases, and has been shown to be largely, if not exclusively, associated with an impairment of nuclear STAT1 dephosphorylation [107••] (Okada S. et al., in preparation). Several residues close to those affected by CMCD-causing mutations in the CCD and DNA-B have been shown, by site-directed mutagenesis, to be essential for STAT1 dephosphorylation [10, 12, 14, 26, 27, 28, 29, 30, 35, 36]. Heterozygous cells from patients also display a stronger response to IFN-γ, IFN-α and IL-27, demonstrating that the GOF alleles are dominant at the cellular level. These cells also displayed defective nuclear STAT1 dephosphorylation. Heterozygosity for GOF STAT1 alleles therefore accounts for AD CMCD with autoimmunity. The autoimmunity seen in these patients probably reflects an enhancement of the cellular response to IFN-α/β [120••]. What is the immunological mechanism underlying CMC? The analysis of IL-17-producing T cells ex vivo and in vitro highlighted a profound defect in the development of these cells in the patients [107••]. This observation provided a cellular basis for CMC in patients heterozygous for GOF STAT1 alleles, but also raised questions about the underlying mechanism. It is possible that an excessive response to one or several of the cytokines known to inhibit IL-17 T-cell development, such as IFN-γ, IFN-α and IL-27 (Figure 4, Figure 5) [121, 122, 123] (whether leading to a decrease or an increase in the transcription of key target genes with respect to controls) impairs the development of these cells in the patients, rendering them susceptible to CMC. Alternatively, enhanced STAT1 activation may divert the response to IL-6, IL-21 and IL-23 from the normal preferential use of STAT3 for signaling, which favors the development of IL-17 T cells [124, 125, 126, 127, 128] (Figure 4, Figure 5). These two hypotheses are not mutually exclusive. Other mechanisms may also operate. Further work is required to investigate the mechanisms by which the heterozygous STAT1 GOF alleles impair IL-17 T-cell development, at both the molecular and cellular levels. However, recent studies have clearly established that CMC develops in patients heterozygous for GOF STAT1 alleles because of the impaired development of IL-17 T cells.
Conclusion and perspectives
STAT1 is, to our knowledge, the first human gene identified for which LOF and GOF heterozygous alleles are apparently associated with a relatively narrow set of distinct infectious diseases. Heterozygotes for LOF dominant-negative alleles display MSMD, whereas heterozygotes for GOF dominant alleles display CMCD. The mutations may even affect the same domain of STAT1. STAT1 is not the first host defense gene to harbor LOF or GOF mutations in different patients, as this situation has already been demonstrated for the X-linked WASP gene, for which LOF mutations underlie XR Wiskott–Aldrich syndrome whereas GOF mutations underlie XR severe congenital neutropenia [129, 130]. The range of infections seen in these two X-linked traits is, however, broader and there is some overlap. Moreover, there are not two, but four distinct disorders of STAT1, each of which is apparently associated with a specific set of infectious diseases. Heterozygous morbid alleles of STAT1 are associated with AD MSMD or AD CMCD, whereas biallelic LOF mutations (whether amorphic or hypomorphic) are associated with AR predisposition to both mycobacterial and viral diseases. Intriguingly however, two patients heterozygous for GOF STAT1 alleles were recently shown to display recurrent herpes virus infections [131], which is reminiscent of STAT3-deficient patients and might involve the same mechanism of impaired T cell memory [132]. Finally, the severity of the infectious phenotype is more pronounced in patients with AR complete STAT1 deficiency than in patients with a partial form of STAT1 deficiency. The set of bacterial and viral diseases may be broader in patients with complete STAT1 deficiency, although the better prognosis of patients with a partial defect may reveal infections not seen in those with complete deficiency, who die in early childhood unless treated by HSCT.
The discovery and characterization of the various human STAT1 morbid alleles has built on elegant studies on human and mouse cell lines in vitro and the mouse model in vivo. Human studies have helped to elucidate the function of STAT1 in host defense in natura, in the context of a natural ecosystem governed by natural selection [133, 134]. The range and nature of viral infections controlled by IFNα/β-dependent and IFN-λ-dependent STAT1 immunity are gradually being deciphered. Clearly, human STAT1-dependent IFN-γ immunity is crucial for protection against mycobacteria and a relatively limited set of intramacrophagic microbes. More surprisingly, a gain of STAT1 activity impairs the development of IL-17-producing T cells and impairs mucocutaneous immunity to C. albicans. Too little STAT1 immunity, whether owing to monoallelic dominant-negative or biallelic LOF mutations, is associated with a predisposition to viral and/or mycobacterial diseases. Conversely, too much STAT1 immunity, owing to monoallelic GOF mutations, is associated with CMC and autoimmunity (Figure 5). There is therefore probably a strong evolutionary pressure in favor of the maintenance of optimal STAT1 activity [135••], as even heterozygous mutations increasing or decreasing the activity of this protein outside a particular range would be subject to strong counterselection during the evolution of the population. Evolutionary genetic studies in human populations would probably reveal a strong purifying selection operating at the STAT1 locus, as recently shown for the human genes encoding several IFNs, including IFN-α/β and IFN-γ [136••].
References and recommended reading
Papers of particular interest, published within the period of review, have been highlighted as:
• of special interest
•• of outstanding interest
Acknowledgements
We thank Yelena Nemirovskaya and Eric Anderson for secretarial assistance, Shen-Ying Zhang for the HSE figure, Emmanuelle Jouanguy and Amandine Crequer for critical reading and all the Laboratory of Human Genetics of Infectious Diseases for helpful discussions. The Laboratory of Human Genetics of Infectious Diseases is supported partly by grants from the St. Giles Foundation, the Jeffrey Modell Foundation, The Rockefeller University Center for Clinical and Translational Science grant number 5UL1RR024143, the National Institute of Allergy and Infectious Diseases grant numbers 5R01AI089970-02 and 1R37AI095983-01, The Rockefeller University, the Agence Nationale de la Recherche (ANR), and the European Research Council (ERC). X.F.K. was supported by a Choh-Hao Li Memorial Fund Scholar Award and the Shanghai Educational Development Foundation, and S.C. is supported by the AXA Research Fund.
References
- 1.Dale T.C., Imam A.M., Kerr I.M., Stark G.R. Rapid activation by interferon alpha of a latent DNA-binding protein present in the cytoplasm of untreated cells. Proc Natl Acad Sci USA. 1989;86:1203–1207. doi: 10.1073/pnas.86.4.1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Levy D.E., Kessler D.S., Pine R., Darnell J.E., Jr. Cytoplasmic activation of ISGF3, the positive regulator of interferon-alpha-stimulated transcription, reconstituted in vitro. Genes Dev. 1989;3:1362–1371. doi: 10.1101/gad.3.9.1362. [DOI] [PubMed] [Google Scholar]
- 3.Casanova J.L., Holland S.M., Notarangelo L.D. Inborn errors of human JAKs & STATs. Immunity. 2012;36:515–528. doi: 10.1016/j.immuni.2012.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Schindler C., Fu X.Y., Improta T., Aebersold R., Darnell J.E., Jr. Proteins of transcription factor ISGF-3: one gene encodes the 91-and 84-kDa ISGF-3 proteins that are activated by interferon alpha. Proc Natl Acad Sci USA. 1992;89:7836–7839. doi: 10.1073/pnas.89.16.7836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Darnell J.E., Jr., Kerr I.M., Stark G.R. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science. 1994;264:1415–1421. doi: 10.1126/science.8197455. [DOI] [PubMed] [Google Scholar]
- 6.Zakharova N., Lymar E.S., Yang E., Malik S., Zhang J.J., Roeder R.G., Darnell J.E., Jr. Distinct transcriptional activation functions of STAT1alpha and STAT1beta on DNA and chromatin templates. J Biol Chem. 2003;278:43067–43073. doi: 10.1074/jbc.M308166200. [DOI] [PubMed] [Google Scholar]
- 7.Najjar I., Fagard R. STAT1 and pathogens, not a friendly relationship. Biochimie. 2010;92:425–444. doi: 10.1016/j.biochi.2010.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Leonard W.J., O'Shea J.J. Jaks and STATs: biological implications. Annu Rev Immunol. 1998;16:293–322. doi: 10.1146/annurev.immunol.16.1.293. [DOI] [PubMed] [Google Scholar]
- 9.Bach E., Aguet M., Schreiber R.D. The interferon gamma receptor: a paradigm for cytokine receptor signaling. Annu Rev Immunol. 1997;15:563–591. doi: 10.1146/annurev.immunol.15.1.563. [DOI] [PubMed] [Google Scholar]
- 10.Levy D.E., Darnell J.E., Jr. Stats: transcriptional control and biological impact. Nat Rev Mol Cell Biol. 2002;3:651–662. doi: 10.1038/nrm909. [DOI] [PubMed] [Google Scholar]
- 11.Meyer T., Marg A., Lemke P., Wiesner B., Vinkemeier U. DNA binding controls inactivation and nuclear accumulation of the transcription factor Stat1. Genes Dev. 2003;17:1992–2005. doi: 10.1101/gad.268003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Haspel R.L., Darnell J.E., Jr. A nuclear protein tyrosine phosphatase is required for the inactivation of Stat1. Proc Natl Acad Sci USA. 1999;96:10188–10193. doi: 10.1073/pnas.96.18.10188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.ten Hoeve J., de Jesus Ibarra-Sanchez M., Fu Y., Zhu W., Tremblay M., David M., Shuai K. Identification of a nuclear Stat1 protein tyrosine phosphatase. Mol Cell Biol. 2002;22:5662–5668. doi: 10.1128/MCB.22.16.5662-5668.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zhong M., Henriksen M.A., Takeuchi K., Schaefer O., Liu B., ten Hoeve J., Ren Z., Mao X., Chen X., Shuai K. Implications of an antiparallel dimeric structure of nonphosphorylated STAT1 for the activation-inactivation cycle. Proc Natl Acad Sci USA. 2005;102:3966–3971. doi: 10.1073/pnas.0501063102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wenta N., Strauss H., Meyer S., Vinkemeier U. Tyrosine phosphorylation regulates the partitioning of STAT1 between different dimer conformations. Proc Natl Acad Sci USA. 2008;105:9238–9243. doi: 10.1073/pnas.0802130105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Cheon H., Yang J., Stark G.R. The functions of signal transducers and activators of transcriptions 1 and 3 as cytokine-inducible proteins. J Interferon Cytokine Res. 2011;31:33–40. doi: 10.1089/jir.2010.0100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Mowen K.A., Tang J., Zhu W., Schurter B.T., Shuai K., Herschman H.R., David M. Arginine methylation of STAT1 modulates IFNalpha/beta-induced transcription. Cell. 2001;104:731–741. doi: 10.1016/s0092-8674(01)00269-0. [DOI] [PubMed] [Google Scholar]
- 18.Meissner T., Krause E., Lodige I., Vinkemeier U. Arginine methylation of STAT1: a reassessment. Cell. 2004;119:587–589. doi: 10.1016/j.cell.2004.11.024. discussion 589–590. [DOI] [PubMed] [Google Scholar]
- 19.Kramer O.H., Knauer S.K., Greiner G., Jandt E., Reichardt S., Guhrs K.H., Stauber R.H., Bohmer F.D., Heinzel T. A phosphorylation-acetylation switch regulates STAT1 signaling. Genes Dev. 2009;23:223–235. doi: 10.1101/gad.479209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Begitt A., Droescher M., Knobeloch K.P., Vinkemeier U. SUMO conjugation of STAT1 protects cells from hyperresponsiveness to IFNgamma. Blood. 2011;118:1002–1007. doi: 10.1182/blood-2011-04-347930. [DOI] [PubMed] [Google Scholar]
- 21.Murphy K.M., Ouyang W., Farrar J.D., Yang J., Ranganath S., Asnagli H., Afkarian M., Murphy T.L. Signaling and transcription in T helper development. Annu Rev Immunol. 2000;18:451–494. doi: 10.1146/annurev.immunol.18.1.451. [DOI] [PubMed] [Google Scholar]
- 22.Strehlow I., Schindler C. Amino-terminal signal transducer and activator of transcription (STAT) domains regulate nuclear translocation and STAT deactivation. J Biol Chem. 1998;273:28049–28056. doi: 10.1074/jbc.273.43.28049. [DOI] [PubMed] [Google Scholar]
- 23.Vinkemeier U., Cohen S.L., Moarefi I., Chait B.T., Kuriyan J., Darnell J.E., Jr. DNA binding of in vitro activated Stat1 alpha, Stat1 beta and truncated Stat1: interaction between NH2-terminal domains stabilizes binding of two dimers to tandem DNA sites. EMBO J. 1996;15:5616–5626. [PMC free article] [PubMed] [Google Scholar]
- 24.Xu X., Sun Y.L., Hoey T. Cooperative DNA, binding and sequence-selective recognition conferred by the STAT amino-terminal domain. Science. 1996;273:794–797. doi: 10.1126/science.273.5276.794. [DOI] [PubMed] [Google Scholar]
- 25.Shuai K., Liao J., Song M.M. Enhancement of antiproliferative activity of gamma interferon by the specific inhibition of tyrosine dephosphorylation of Stat1. Mol Cell Biol. 1996;16:4932–4941. doi: 10.1128/mcb.16.9.4932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mertens C., Zhong M., Krishnaraj R., Zou W., Chen X., Darnell J.E., Jr. Dephosphorylation of phosphotyrosine on STAT1 dimers requires extensive spatial reorientation of the monomers facilitated by the N-terminal domain. Genes Dev. 2006;20:3372–3381. doi: 10.1101/gad.1485406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Braunstein J., Brutsaert S., Olson R., Schindler C. STATs dimerize in the absence of phosphorylation. J Biol Chem. 2003;278:34133–34140. doi: 10.1074/jbc.M304531200. [DOI] [PubMed] [Google Scholar]
- 28.Chen X., Vinkemeier U., Zhao Y., Jeruzalmi D., Darnell J.E., Jr., Kuriyan J. Crystal structure of a tyrosine phosphorylated STAT-1 dimer bound to DNA. Cell. 1998;93:827–839. doi: 10.1016/s0092-8674(00)81443-9. [DOI] [PubMed] [Google Scholar]
- 29.Hoshino A., Saint Fleur S., Fujii H. Regulation of Stat1 protein expression by phenylalanine 172 in the coiled-coil domain. Biochem Biophys Res Commun. 2006;346:1062–1066. doi: 10.1016/j.bbrc.2006.06.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Mao X., Ren Z., Parker G.N., Sondermann H., Pastorello M.A., Wang W., McMurray J.S., Demeler B., Darnell J.E., Jr., Chen X. Structural bases of unphosphorylated STAT1 association and receptor binding. Mol Cell. 2005;17:761–771. doi: 10.1016/j.molcel.2005.02.021. [DOI] [PubMed] [Google Scholar]
- 31.McBride K.M., Banninger G., McDonald C., Reich N.C. Regulated nuclear import of the STAT1 transcription factor by direct binding of importin-alpha. EMBO J. 2002;21:1754–1763. doi: 10.1093/emboj/21.7.1754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Fagerlund R., Melen K., Kinnunen L., Julkunen I. Arginine/lysine-rich nuclear localization signals mediate interactions between dimeric STATs and importin alpha 5. J Biol Chem. 2002;277:30072–30078. doi: 10.1074/jbc.M202943200. [DOI] [PubMed] [Google Scholar]
- 33.Meyer T., Begitt A., Lodige I., van Rossum M., Vinkemeier U. Constitutive and IFN-gamma-induced nuclear import of STAT1 proceed through independent pathways. EMBO J. 2002;21:344–354. doi: 10.1093/emboj/21.3.344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Melen K., Kinnunen L., Julkunen I. Arginine/lysine-rich structural element is involved in interferon-induced nuclear import of STATs. J Biol Chem. 2001;276:16447–16455. doi: 10.1074/jbc.M008821200. [DOI] [PubMed] [Google Scholar]
- 35.McBride K.M., McDonald C., Reich N.C. Nuclear export signal located within theDNA-binding domain of the STAT1transcription factor. EMBO J. 2000;19:6196–6206. doi: 10.1093/emboj/19.22.6196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Begitt A., Meyer T., van Rossum M., Vinkemeier U. Nucleocytoplasmic translocation of Stat1 is regulated by a leucine-rich export signal in the coiled-coil domain. Proc Natl Acad Sci USA. 2000;97:10418–10423. doi: 10.1073/pnas.190318397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Yang E., Wen Z., Haspel R.L., Zhang J.J., Darnell J.E., Jr. The linker domain of Stat1 is required for gamma interferon-driven transcription. Mol Cell Biol. 1999;19:5106–5112. doi: 10.1128/mcb.19.7.5106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Yang E., Henriksen M.A., Schaefer O., Zakharova N., Darnell J.E., Jr. Dissociation time from DNA determines transcriptional function in a STAT1 linker mutant. J Biol Chem. 2002;277:13455–13462. doi: 10.1074/jbc.M112038200. [DOI] [PubMed] [Google Scholar]
- 39.Decker T., Kovarik P. Serine phosphorylation of STATs. Oncogene. 2000;19:2628–2637. doi: 10.1038/sj.onc.1203481. [DOI] [PubMed] [Google Scholar]
- 40.Durbin J.E., Hackenmiller R., Simon M.C., Levy D.E. Targeted disruption of the mouse Stat1 gene results in compromised innate immunity to viral disease. Cell. 1996;84:443–450. doi: 10.1016/s0092-8674(00)81289-1. [DOI] [PubMed] [Google Scholar]
- 41.Meraz M.A., White J.M., Sheehan K.C., Bach E.A., Rodig S.J., Dighe A.S., Kaplan D.H., Riley J.K., Greenlund A.C., Campbell D. Targeted disruption of the Stat1 gene in mice reveals unexpected physiologic specificity in the JAK-STAT signaling pathway. Cell. 1996;84:431–442. doi: 10.1016/s0092-8674(00)81288-x. [DOI] [PubMed] [Google Scholar]
- 42.Nishibori T., Tanabe Y., Su L., David M. Impaired development of CD4+ CD25+ regulatory T cells in the absence of STAT1: increased susceptibility to autoimmune disease. J Exp Med. 2004;199:25–34. doi: 10.1084/jem.20020509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gil M.P., Bohn E., O’Guin A.K., Ramana C.V., Levine B., Stark G.R., Virgin H.W., Schreiber R.D. Biologic consequences of Stat1-independent IFN signaling. Proc Natl Acad Sci USA. 2001;98:6680–6685. doi: 10.1073/pnas.111163898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ramana C.V., Gil M.P., Han Y., Ransohoff R.M., Schreiber R.D., Stark G.R. Stat1-independent regulation of gene expression in response to IFN-gamma. Proc Natl Acad Sci USA. 2001;98:6674–6679. doi: 10.1073/pnas.111164198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kotenko S.V., Gallagher G., Baurin V.V., Lewis-Antes A., Shen M., Shah N.K., Langer J.A., Sheikh F., Dickensheets H., Donnelly R.P. IFN-lambdas mediate antiviral protection through a distinct class II cytokine receptor complex. Nat Immunol. 2003;4:69–77. doi: 10.1038/ni875. [DOI] [PubMed] [Google Scholar]
- 46.Takeda A., Hamano S., Yamanaka A., Hanada T., Ishibashi T., Mak T.W., Yoshimura A., Yoshida H. Cutting edge: role of IL-27/WSX-1 signaling for induction of T-bet through activation of STAT1 during initial Th1 commitment. J Immunol. 2003;170:4886–4890. doi: 10.4049/jimmunol.170.10.4886. [DOI] [PubMed] [Google Scholar]
- 47.Kamiya S., Owaki T., Morishima N., Fukai F., Mizuguchi J., Yoshimoto T. An indispensable role for STAT1 in IL-27-induced T-bet expression but not proliferation of naive CD4+ T cells. J Immunol. 2004;173:3871–3877. doi: 10.4049/jimmunol.173.6.3871. [DOI] [PubMed] [Google Scholar]
- 48.Jewell N.A., Cline T., Mertz S.E., Smirnov S.V., Flano E., Schindler C., Grieves J.L., Durbin R.K., Kotenko S.V., Durbin J.E. Lambda interferon is the predominant interferon induced by influenza A virus infection in vivo. J Virol. 2010;84:11515–11522. doi: 10.1128/JVI.01703-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Batten M., Li J., Yi S., Kljavin N.M., Danilenko D.M., Lucas S., Lee J., de Sauvage F.J., Ghilardi N. Interleukin 27 limits autoimmune encephalomyelitis by suppressing the development of interleukin 17-producing T cells. Nat Immunol. 2006;7:929–936. doi: 10.1038/ni1375. [DOI] [PubMed] [Google Scholar]
- 50.Schilte C., Couderc T., Chretien F., Sourisseau M., Gangneux N., Guivel-Benhassine F., Kraxner A., Tschopp J., Higgs S., Michault A. Type I, IFN controls chikungunya virus via its action on nonhematopoietic cells. J Exp Med. 2010;207:429–442. doi: 10.1084/jem.20090851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Gardner C.L., Burke C.W., Higgs S.T., Klimstra W.B., Ryman K.D. Interferon-alpha/beta deficiency greatly exacerbates arthritogenic disease in mice infected with wild-type chikungunya virus but not with the cell culture-adapted live-attenuated 181/25 vaccine candidate. Virology. 2012;425:103–112. doi: 10.1016/j.virol.2011.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Frieman M.B., Chen J., Morrison T.E., Whitmore A., Funkhouser W., Ward J.M., Lamirande E.W., Roberts A., Heise M., Subbarao K. SARS-CoV pathogenesis is regulated by a STAT1 dependent but a type I, II and III interferon receptor independent mechanism. PLoS Pathog. 2010;6:e1000849. doi: 10.1371/journal.ppat.1000849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Karst S.M., Wobus C.E., Lay M., Davidson J., Virgin HWt: STAT1-dependent innate immunity to a Norwalk-like virus. Science. 2003;299:1575–1578. doi: 10.1126/science.1077905. [DOI] [PubMed] [Google Scholar]
- 54.Strong D.W., Thackray L.B., Smith T.J., Virgin H.W. Protruding domain of capsid protein is necessary and sufficient to determine murine norovirus replication and pathogenesis in vivo. J Virol. 2012;86:2950–2958. doi: 10.1128/JVI.07038-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Garcia-Sastre A., Durbin R.K., Zheng H., Palese P., Gertner R., Levy D.E., Durbin J.E. The role of interferon in influenza virus tissue tropism. J Virol. 1998;72:8550–8558. doi: 10.1128/jvi.72.11.8550-8558.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56•.Raymond J., Bradfute S., Bray M. Filovirus infection of STAT-1 knockout mice. J Infect Dis. 2011;204(Suppl. 3):S986–S990. doi: 10.1093/infdis/jir335. [DOI] [PubMed] [Google Scholar]; See ref. [59•].
- 57•.de Wit E., Munster V.J., Metwally S.A., Feldmann H. Assessment of rodents as animal models for Reston ebolavirus. J Infect Dis. 2011;204(Suppl. 3):S968–S972. doi: 10.1093/infdis/jir330. [DOI] [PMC free article] [PubMed] [Google Scholar]; See ref. [59•].
- 58•.Bradfute S.B., Stuthman K.S., Shurtleff A.C., Bavari S. A STAT-1 knockout mouse model for Machupo virus pathogenesis. Virol J. 2011;8:300. doi: 10.1186/1743-422X-8-300. [DOI] [PMC free article] [PubMed] [Google Scholar]; See ref. [59•].
- 59•.Bente D.A., Alimonti J.B., Shieh W.J., Camus G., Stroher U., Zaki S., Jones S.M. Pathogenesis and immune response of Crimean-Congo hemorrhagic fever virus in a STAT-1 knockout mouse model. J Virol. 2010;84:11089–11100. doi: 10.1128/JVI.01383-10. [DOI] [PMC free article] [PubMed] [Google Scholar]; These papers (refs. [56•, 57•, 58•, 59•]) establish clearly the non-redundant role of mouse STAT1 during infections by different viruses.
- 60.Shornick L.P., Wells A.G., Zhang Y., Patel A.C., Huang G., Takami K., Sosa M., Shukla N.A., Agapov E., Holtzman M.J. Airway epithelial versus immune cell Stat1 function for innate defense against respiratory viral infection. J Immunol. 2008;180:3319–3328. doi: 10.4049/jimmunol.180.5.3319. [DOI] [PubMed] [Google Scholar]
- 61.Leib D.A., Harrison T.E., Laslo K.M., Machalek M.A., Moorman N.J., Virgin H.W. Interferons regulate the phenotype of wild-type and mutant herpes simplex viruses in vivo. J Exp Med. 1999;189:663–672. doi: 10.1084/jem.189.4.663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62•.Pasieka T.J., Cilloniz C., Carter V.S., Rosato P., Katze M.G., Leib D.A. Functional genomics reveals an essential and specific role for Stat1 in protection of the central nervous system following herpes simplex virus corneal infection. J Virol. 2011;85:12972–12981. doi: 10.1128/JVI.06032-11. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper describes the use of microarrays on brain stem cells derived from wild-type or STAT1-deficient mice infected with HSV-1 and the identification of a signature.
- 63.Shresta S., Sharar K.L., Prigozhin D.M., Snider H.M., Beatty P.R., Harris E. Critical roles for both STAT1-dependent and STAT1-independent pathways in the control of primary dengue virus infection in mice. J Immunol. 2005;175:3946–3954. doi: 10.4049/jimmunol.175.6.3946. [DOI] [PubMed] [Google Scholar]
- 64•.Perry S.T., Buck M.D., Lada S.M., Schindler C., Shresta S. STAT2 mediates innate immunity to Dengue virus in the absence of STAT1 via the type I interferon receptor. PLoS Pathog. 2011;7:e1001297. doi: 10.1371/journal.ppat.1001297. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper establishes for the first time a specific and crucial role of mouse STAT2, independently of STAT1 and mediated by IFNAR, in antiviral immunity.
- 65.O’Donnell L.A., Conway S., Rose R.W., Nicolas E., Slifker M., Balachandran S., Rall G.F. STAT1-independent control of a neurotropic measles virus challenge in primary neurons and infected mice. J Immunol. 2012;188:1915–1923. doi: 10.4049/jimmunol.1101356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Hahm B., Trifilo M.J., Zuniga E.I., Oldstone M.B. Viruses evade the immune system through type I interferon-mediated STAT2-dependent, but STAT1-independent, signaling. Immunity. 2005;22:247–257. doi: 10.1016/j.immuni.2005.01.005. [DOI] [PubMed] [Google Scholar]
- 67.Sugawara I., Yamada H., Mizuno S. STAT1 knockout mice are highly susceptible to pulmonary mycobacterial infection. Tohoku J Exp Med. 2004;202:41–50. doi: 10.1620/tjem.202.41. [DOI] [PubMed] [Google Scholar]
- 68.Roth K.M., Gunn J.S., Lafuse W., Satoskar A.R. Francisella inhibits STAT1-mediated signaling in macrophages and prevents activation of antigen-specific T cells. Int Immunol. 2009;21:19–28. doi: 10.1093/intimm/dxn119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Rothfuchs A.G., Trumstedt C., Mattei F., Schiavoni G., Hidmark A., Wigzell H., Rottenberg M.E. STAT1 regulates IFN-alpha beta- and IFN-gamma-dependent control of infection with Chlamydia pneumoniae by nonhemopoietic cells. J Immunol. 2006;176:6982–6990. doi: 10.4049/jimmunol.176.11.6982. [DOI] [PubMed] [Google Scholar]
- 70.Brown C.R., Blaho V.A., Fritsche K.L., Loiacono C.M. Stat1 deficiency exacerbates carditis but not arthritis during experimental lyme borreliosis. J Interferon Cytokine Res. 2006;26:390–399. doi: 10.1089/jir.2006.26.390. [DOI] [PubMed] [Google Scholar]
- 71.Gavrilescu L.C., Butcher B.A., Del Rio L., Taylor G.A., Denkers E.Y. STAT1 is essential for antimicrobial effector function but dispensable for gamma interferon production during Toxoplasma gondii infection. Infect Immun. 2004;72:1257–1264. doi: 10.1128/IAI.72.3.1257-1264.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lieberman L.A., Banica M., Reiner S.L., Hunter C.A. STAT1 plays a critical role in the regulation of antimicrobial effector mechanisms, but not in the development of Th1-type responses during toxoplasmosis. J Immunol. 2004;172:457–463. doi: 10.4049/jimmunol.172.1.457. [DOI] [PubMed] [Google Scholar]
- 73.Ehigiator H.N., McNair N., Mead J.R. Cryptosporidium parvum: the contribution of Th1-inducing pathways to the resolution of infection in mice. Exp Parasitol. 2007;115:107–113. doi: 10.1016/j.exppara.2006.07.001. [DOI] [PubMed] [Google Scholar]
- 74.Rosas L.E., Keiser T., Pyles R., Durbin J., Satoskar A.R. Development of protective immunity against cutaneous leishmaniasis is dependent on STAT1-mediated IFN signaling pathway. Eur J Immunol. 2003;33:1799–1805. doi: 10.1002/eji.200323163. [DOI] [PubMed] [Google Scholar]
- 75.Rosas L.E., Snider H.M., Barbi J., Satoskar A.A., Lugo-Villarino G., Keiser T., Papenfuss T., Durbin J.E., Radzioch D., Glimcher L.H. Cutting edge: STAT1 and T-bet play distinct roles in determining outcome of visceral leishmaniasis caused by Leishmania donovani. J Immunol. 2006;177:22–25. doi: 10.4049/jimmunol.177.1.22. [DOI] [PubMed] [Google Scholar]
- 76.Guarda G., Braun M., Staehli F., Tardivel A., Mattmann C., Forster I., Farlik M., Decker T., Du Pasquier R.A., Romero P. Type I interferon inhibits interleukin-1 production and inflammasome activation. Immunity. 2011;34:213–223. doi: 10.1016/j.immuni.2011.02.006. [DOI] [PubMed] [Google Scholar]
- 77.Dupuis S., Jouanguy E., Al-Hajjar S., Fieschi C., Al-Mohsen I.Z., Al-Jumaah S., Yang K., Chapgier A., Eidenschenk C., Eid P. Impaired response to interferon-alpha/beta and lethal viral disease in human STAT1 deficiency. Nat Genet. 2003;33:388–391. doi: 10.1038/ng1097. [DOI] [PubMed] [Google Scholar]
- 78.Al-Herz W., Bousfiha A., Casanova J.L., Chapel H., Conley M.E., Cunningham-Rundles C., Etzioni A., Fischer A., Franco J.L., Geha R.S. Primary immunodeficiency diseases: an update on the classification from the international union of immunological societies expert committee for primary immunodeficiency. Front Immunol. 2011;2:00054. doi: 10.3389/fimmu.2011.00054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Chapgier A., Wynn R.F., Jouanguy E., Filipe-Santos O., Zhang S., Feinberg J., Hawkins K., Casanova J.L., Arkwright P.D. Human complete Stat-1 deficiency is associated with defective type I and II IFN responses in vitro but immunity to some low virulence viruses in vivo. J Immunol. 2006;176:5078–5083. doi: 10.4049/jimmunol.176.8.5078. [DOI] [PubMed] [Google Scholar]
- 80•.Vairo D., Tassone L., Tabellini G., Tamassia N., Gasperini S., Bazzoni F., Plebani A., Porta F., Notarangelo L.D., Parolini S. Severe impairment of IFN-gamma and IFN-alpha responses in cells of a patient with a novel STAT1 splicing mutation. Blood. 2011;118:1806–1817. doi: 10.1182/blood-2011-01-330571. [DOI] [PubMed] [Google Scholar]; The authors describe a new patient with AR complete STAT1 deficiency who had suffered from severe mycobacterial and viral diseases. In contrast to the fatal outcome reported for the other patients described, this patient was successfully treated by HSCT. In addition, the authors highlight a STAT1-independent response to IFN-α in NK cells.
- 81.Averbuch D., Chapgier A., Boisson-Dupuis S., Casanova J.L., Engelhard D. The clinical spectrum of patients with deficiency of signal transducer and activator of transcription-1. Pediatr Infect Dis J. 2011;30:352–355. doi: 10.1097/INF.0b013e3181fdff4a. [DOI] [PubMed] [Google Scholar]
- 82•.Sancho-Shimizu V., de Diego R.P., Jouanguy E., Zhang S.Y., Casanova J.L. Inborn errors of anti-viral interferon immunity in humans. Curr Opin Virol. 2011;1:487–496. doi: 10.1016/j.coviro.2011.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]; Clear and comprehensive review on inborn errors of immunity affecting IFNs production and function.
- 83.Casanova J.L., Abel L. Genetic dissection of immunity to mycobacteria: the human model. Annu Rev Immunol. 2002;20:581–620. doi: 10.1146/annurev.immunol.20.081501.125851. [DOI] [PubMed] [Google Scholar]
- 84.Chapgier A., Boisson-Dupuis S., Jouanguy E., Vogt G., Feinberg J., Prochnicka-Chalufour A., Casrouge A., Yang K., Soudais C., Fieschi C. Novel STAT1 alleles in otherwise healthy patients with mycobacterial disease. PLoS Genet. 2006;2:e131. doi: 10.1371/journal.pgen.0020131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Holzinger D., Jorns C., Stertz S., Boisson-Dupuis S., Thimme R., Weidmann M., Casanova J.L., Haller O., Kochs G. Induction of MxA gene expression by influenza A virus requires type I or type III interferon signaling. J Virol. 2007;81:7776–7785. doi: 10.1128/JVI.00546-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86•.Ma C.S., Avery D.T., Chan A., Batten M., Bustamante J., Boisson-Dupuis S., Arkwright P.D., Kreins A.Y., Averbuch D., Engelhard D. Functional STAT3 deficiency compromises the generation of human T follicular helper cells. Blood. 2012;119:3997–4008. doi: 10.1182/blood-2011-11-392985. [DOI] [PMC free article] [PubMed] [Google Scholar]; See ref. [87•].
- 87•.Avery D.T., Deenick E.K., Ma C.S., Suryani S., Simpson N., Chew G.Y., Chan T.D., Palendira U., Bustamante J., Boisson-Dupuis S. B cell-intrinsic signaling through IL-21 receptor and STAT3 is required for establishing long-lived antibody responses in humans. J Exp Med. 2010;207:155–171. doi: 10.1084/jem.20091706. [DOI] [PMC free article] [PubMed] [Google Scholar]; Through the use of cells from patients with inborn errors of immunity, the authors of papers [86•] and [87•] identified the importance of STAT3 for the development of Tfh and the generation of effector B cells from naive precusrors, respectively.
- 88.Chen Q., Ghilardi N., Wang H., Baker T., Xie M.H., Gurney A., Grewal I.S., de Sauvage F.J. Development of Th1-type immune responses requires the type I cytokine receptor TCCR. Nature. 2000;407:916–920. doi: 10.1038/35038103. [DOI] [PubMed] [Google Scholar]
- 89•.Donnelly R.P., Kotenko S.V. Interferon-lambda: a new addition to an old family. J Interferon Cytokine Res. 2010;30:555–564. doi: 10.1089/jir.2010.0078. [DOI] [PMC free article] [PubMed] [Google Scholar]; Clear and comprehensive review on type III IFNs.
- 90.Uze G., Monneron D. IL-28 and IL-29: newcomers to the interferon family. Biochimie. 2007;89:729–734. doi: 10.1016/j.biochi.2007.01.008. [DOI] [PubMed] [Google Scholar]
- 91••.Ge D., Fellay J., Thompson A.J., Simon J.S., Shianna K.V., Urban T.J., Heinzen E.L., Qiu P., Bertelsen A.H., Muir A.J. Genetic variation in IL28B predicts hepatitis C treatment-induced viral clearance. Nature. 2009;461:399–401. doi: 10.1038/nature08309. [DOI] [PubMed] [Google Scholar]; See ref. [94••].
- 92••.Thomas D.L., Thio C.L., Martin M.P., Qi Y., Ge D., O’Huigin C., Kidd J., Kidd K., Khakoo S.I., Alexander G. Genetic variation in IL28B and spontaneous clearance of hepatitis C virus. Nature. 2009;461:798–801. doi: 10.1038/nature08463. [DOI] [PMC free article] [PubMed] [Google Scholar]; See ref. [94••].
- 93••.Suppiah V., Moldovan M., Ahlenstiel G., Berg T., Weltman M., Abate M.L., Bassendine M., Spengler U., Dore G.J., Powell E. IL28B is associated with response to chronic hepatitis C interferon-alpha and ribavirin therapy. Nat Genet. 2009;41:1100–1104. doi: 10.1038/ng.447. [DOI] [PubMed] [Google Scholar]; See ref. [94••].
- 94••.Tanaka Y., Nishida N., Sugiyama M., Kurosaki M., Matsuura K., Sakamoto N., Nakagawa M., Korenaga M., Hino K., Hige S. Genome-wide association of IL28B with response to pegylated interferon-alpha and ribavirin therapy for chronic hepatitis C. Nat Genet. 2009;41:1105–1109. doi: 10.1038/ng.449. [DOI] [PubMed] [Google Scholar]; The papers from refs. [91••, 92••, 93••, 94••] clearly identify genetic variants in IL28B associated with Hepatitis C virus clearance both under treatment and spontaneously.
- 95.Glocker E.O., Kotlarz D., Boztug K., Gertz E.M., Schaffer A.A., Noyan F., Perro M., Diestelhorst J., Allroth A., Murugan D. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N Engl J Med. 2009;361:2033–2045. doi: 10.1056/NEJMoa0907206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Chapgier A., Kong X.F., Boisson-Dupuis S., Jouanguy E., Averbuch D., Feinberg J., Zhang S.Y., Bustamante J., Vogt G., Lejeune J. A partial form of recessive STAT1 deficiency in humans. J Clin Invest. 2009;119:1502–1514. doi: 10.1172/JCI37083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97•.Kong X.F., Ciancanelli M., Al-Hajjar S., Alsina L., Zumwalt T., Bustamante J., Feinberg J., Audry M., Prando C., Bryant V. A novel form of human STAT1 deficiency impairing early but not late responses to interferons. Blood. 2010;116:5895–5906. doi: 10.1182/blood-2010-04-280586. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this paper, the authors identify and characterize in detail, at the cellular level, a new partial recessive STAT1 deficiency associated with mild intracellular bacterial and viral diseases, and they highlight a set of early IFN-γ responsive genes probably involved in antimycobacterial immunity.
- 98.Kristensen I.A., Veirum J.E., Moller B.K., Christiansen M. Novel STAT1 alleles in a patient with impaired resistance to mycobacteria. J Clin Immunol. 2011;31:265–271. doi: 10.1007/s10875-010-9480-8. [DOI] [PubMed] [Google Scholar]
- 99.Filipe-Santos O., Bustamante J., Chapgier A., Vogt G., de Beaucoudrey L., Feinberg J., Jouanguy E., Boisson-Dupuis S., Fieschi C., Picard C. Inborn errors of IL-12/23- and IFN-gamma-mediated immunity: molecular, cellular, and clinical features. Semin Immunol. 2006;18:347–361. doi: 10.1016/j.smim.2006.07.010. [DOI] [PubMed] [Google Scholar]
- 100.de Beaucoudrey L., Samarina A., Bustamante J., Cobat A., Boisson-Dupuis S., Feinberg J., Al-Muhsen S., Janniere L., Rose Y., de Suremain M. Revisiting human IL-12Rbeta1 deficiency: a survey of 141 patients from 30 countries. Medicine (Baltimore) 2010;89:381–402. doi: 10.1097/MD.0b013e3181fdd832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Boisson-Dupuis S., El Baghdadi J., Parvaneh N., Bousfiha A., Bustamante J., Feinberg J., Samarina A., Grant A.V., Janniere L., El Hafidi N. IL-12Rb1 deficiency in two of fifty children with severe tuberculosis from Iran, Morocco, and Turkey. PLoS ONE. 2011;6:e18524. doi: 10.1371/journal.pone.0018524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102••.Hambleton S., Salem S., Bustamante J., Bigley V., Boisson-Dupuis S., Azevedo J., Fortin A., Haniffa M., Ceron-Gutierrez L., Bacon C.M. IRF8 mutations and human dendritic-cell immunodeficiency. New Engl J Med. 2011;365:127–138. doi: 10.1056/NEJMoa1100066. [DOI] [PMC free article] [PubMed] [Google Scholar]; This is the first description of IRF8 mutations in humans. The authors describe AR and AD IRF8 deficiency associated with a lack of circulating monocytes and DCs and of CD11c+CD1c+ circulating DCs, respectively. They highlight a specific role for IRF8 in the development of monocytes and DCs and in antimycobacterial immunity.
- 103••.Bustamante J., Arias A.A., Vogt G., Picard C., Galicia L.B., Prando C., Grant A.V., Marchal C.C., Hubeau M., Chapgier A. Germline CYBB mutations that selectively affect macrophages in kindreds with X-linked predisposition to tuberculous mycobacterial disease. Nat Immunol. 2011;12:213–221. doi: 10.1038/ni.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper describes two new mutations of CYBB in two unrelated kindreds suffering from MSMD. The authors elegantly show that the mutations are specifically deleterious in EBV-B cells and macrophages from the patients and not in monocytes or neutrophils, accounting for the lack of pyogenic bacterial infections, typical of chronic granulomatous disease, and the specific susceptibility to mycobacterial disease of these patients.
- 104.Dupuis S., Dargemont C., Fieschi C., Thomassin N., Rosenzweig S., Harris J., Holland S.M., Schreiber R.D., Casanova J.L. Impairment of mycobacterial but not viral immunity by a germline human STAT1 mutation. Science. 2001;293:300–303. doi: 10.1126/science.1061154. [DOI] [PubMed] [Google Scholar]
- 105.Tsumura M, Okada S, Sakai H, Yasunaga S, Ohtsubo M, Murata T, Obata H, Yasumi T, Kong XF, Abhyankar A, et al: Dominant-negative STAT1 SH2 domain mutations in unrelated patients with Mendelian susceptibility to mycobacterial disease. Hum Mutat 2012, May 9. [DOI] [PMC free article] [PubMed]
- 106.Sampaio E.P., Bax H.I., Hsu A.P., Kristosturyan E., Pechacek J., Chandrasekaran P., Paulson M.L., Dias D.L., Spalding C., Uzel G. A novel STAT1 mutation associated with disseminated mycobacterial disease. J Clin Immunol. 2012 doi: 10.1007/s10875-012-9659-2. Feb 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107••.Liu L., Okada S., Kong X.F., Kreins A.Y., Cypowyj S., Abhyankar A., Toubiana J., Itan Y., Audry M., Nitschke P. Gain-of-function human STAT1 mutations impair IL-17 immunity and underlie chronic mucocutaneous candidiasis. J Exp Med. 2011;208:1635–1648. doi: 10.1084/jem.20110958. [DOI] [PMC free article] [PubMed] [Google Scholar]; The authors describe a new and surprising genetic etiology of CMCD, with dominant mutations in STAT1 in a series of 47 patients. The authors also decipher the mechanism of dominance, showing that the mutant alleles are gain-of-function mostly through a loss-of-dephosphorylation of STAT1. These mutations lead, in patients, to the impairment of IL-17-producing T-cell development, ultimately leading to CMCD.
- 108•.van de Veerdonk F.L., Plantinga T.S., Hoischen A., Smeekens S.P., Joosten L.A., Gilissen C., Arts P., Rosentul D.C., Carmichael A.J., Smits-van der Graaf C.A. STAT1 mutations in autosomal dominant chronic mucocutaneous candidiasis. N Engl J Med. 2011;365:54–61. doi: 10.1056/NEJMoa1100102. [DOI] [PubMed] [Google Scholar]; This paper, together with ref. [107••], identifies heterozygous STAT1 mutations associated with AD CMC in a series of 14 patients from 5 unrelated families.
- 109.Spellberg B.J., Filler S.G., Edwards J.E., Jr. Current treatment strategies for disseminated candidiasis. Clin Infect Dis. 2006;42:244–251. doi: 10.1086/499057. [DOI] [PubMed] [Google Scholar]
- 110.Milner J.D., Brenchley J.M., Laurence A., Freeman A.F., Hill B.J., Elias K.M., Kanno Y., Spalding C., Elloumi H.Z., Paulson M.L. Impaired T(H)17 cell differentiation in subjects with autosomal dominant hyper-IgE syndrome. Nature. 2008;452:773–776. doi: 10.1038/nature06764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.de Beaucoudrey L., Puel A., Filipe-Santos O., Cobat A., Ghandil P., Chrabieh M., Feinberg J., von Bernuth H., Samarina A., Janniere L. Mutations in STAT3 and IL12RB1 impair the development of human IL-17-producing T cells. J Exp Med. 2008;205:1543–1550. doi: 10.1084/jem.20080321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Ma C.S., Chew G.Y., Simpson N., Priyadarshi A., Wong M., Grimbacher B., Fulcher D.A., Tangye S.G., Cook M.C. Deficiency of Th17 cells in hyper IgE syndrome due to mutations in STAT3. J Exp Med. 2008;205:1551–1557. doi: 10.1084/jem.20080218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Renner E.D., Rylaarsdam S., Anover-Sombke S., Rack A.L., Reichenbach J., Carey J.C., Zhu Q., Jansson A.F., Barboza J., Schimke L.F. Novel signal transducer and activator of transcription 3 (STAT3) mutations, reduced T(H)17 cell numbers, and variably defective STAT3 phosphorylation in hyper-IgE syndrome. J Allergy Clin Immunol. 2008;122:181–187. doi: 10.1016/j.jaci.2008.04.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Minegishi Y., Saito M., Nagasawa M., Takada H., Hara T., Tsuchiya S., Agematsu K., Yamada M., Kawamura N., Ariga T. Molecular explanation for the contradiction between systemic Th17 defect and localized bacterial infection in hyper-IgE syndrome. J Exp Med. 2009;206:1291–1301. doi: 10.1084/jem.20082767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115•.Puel A., Doffinger R., Natividad A., Chrabieh M., Barcenas-Morales G., Picard C., Cobat A., Ouachee-Chardin M., Toulon A., Bustamante J. Autoantibodies against IL-17A, IL-17F, and IL-22 in patients with chronic mucocutaneous candidiasis and autoimmune polyendocrine syndrome type I. J Exp Med. 2010;207:291–297. doi: 10.1084/jem.20091983. [DOI] [PMC free article] [PubMed] [Google Scholar]; See ref. [116•].
- 116•.Kisand K., Boe Wolff A.S., Podkrajsek K.T., Tserel L., Link M., Kisand K.V., Ersvaer E., Perheentupa J., Erichsen M.M., Bratanic N. Chronic mucocutaneous candidiasis in APECED or thymoma patients correlates with autoimmunity to Th17-associated cytokines. J Exp Med. 2010;207:299–308. doi: 10.1084/jem.20091669. [DOI] [PMC free article] [PubMed] [Google Scholar]; In these reports (refs. [115•] and [116•]), the authors identify the cellular cause of CMC in patients with autoimmune polyendocrine type I syndrome owing to AIRE mutations. The presence of autoantibobies against IL-17A, IL-17F and IL-22 in these patients highlights the specific role of these cytokines in protection against CMC in humans.
- 117.Puel A., Picard C., Cypowyj S., Lilic D., Abel L., Casanova J.L. Inborn errors of mucocutaneous immunity to Candida albicans in humans: a role for IL-17 cytokines? Curr Opin Immunol. 2010;22:467–474. doi: 10.1016/j.coi.2010.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118••.Puel A., Cypowyj S., Bustamante J., Wright J.F., Liu L., Lim H.K., Migaud M., Israel L., Chrabieh M., Audry M. Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin-17 immunity. Science. 2011;332:65–68. doi: 10.1126/science.1200439. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this paper, the authors identify two genetic etiologies of CMCD: AR deficiency of the IL17 receptor (IL-17RA) and AD deficiency of the cytokine IL-17F, thereby demonstrating an essential role of IL-17A and/or IL-17F in mucocutaneous immunity to C. albicans in humans in natura.
- 119.Smeekens S.P., Plantinga T.S., van de Veerdonk F.L., Heinhuis B., Hoischen A., Joosten L.A., Arkwright P.D., Gennery A., Kullberg B.J., Veltman J.A. STAT1 hyperphosphorylation and defective IL12R/IL23R signaling underlie defective immunity in autosomal dominant chronic mucocutaneous candidiasis. PLoS ONE. 2011;6:e29248. doi: 10.1371/journal.pone.0029248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120••.Crow Y.J. Type I interferonopathies: a novel set of inborn errors of immunity. Ann N Y Acad Sci. 2011;1238:91–98. doi: 10.1111/j.1749-6632.2011.06220.x. [DOI] [PubMed] [Google Scholar]; Clear and comprehensive review on type I interferonopathies.
- 121.Ramgolam V.S., Sha Y., Jin J., Zhang X., Markovic-Plese S. IFN-beta inhibits human Th17 cell differentiation. J Immunol. 2009;183:5418–5427. doi: 10.4049/jimmunol.0803227. [DOI] [PubMed] [Google Scholar]
- 122.Liu H., Rohowsky-Kochan C. Interleukin-27-mediated suppression of human Th17 cells is associated with activation of STAT1 and suppressor of cytokine signaling protein 1. J Interferon Cytokine Res. 2011;31:459–469. doi: 10.1089/jir.2010.0115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Feng G., Gao W., Strom T.B., Oukka M., Francis R.S., Wood K.J., Bushell A. Exogenous IFN-gamma ex vivo shapes the alloreactive T-cell repertoire by inhibition of Th17 responses and generation of functional Foxp3+ regulatory T cells. Eur J Immunol. 2008;38:2512–2527. doi: 10.1002/eji.200838411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Hirahara K., Ghoreschi K., Laurence A., Yang X.P., Kanno Y., O'Shea J.J. Signal transduction pathways and transcriptional regulation in Th17 cell differentiation. Cytokine Growth Factor Rev. 2010;21:425–434. doi: 10.1016/j.cytogfr.2010.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Hunter C.A. New IL-12-family members: IL-23 and IL-27, cytokines with divergent functions. Nat Rev Immunol. 2005;5:521–531. doi: 10.1038/nri1648. [DOI] [PubMed] [Google Scholar]
- 126.Kishimoto T. Interleukin-6: from basic science to medicine – 40 years in immunology. Annu Rev Immunol. 2005;23:1–21. doi: 10.1146/annurev.immunol.23.021704.115806. [DOI] [PubMed] [Google Scholar]
- 127.Spolski R., Leonard W.J. Interleukin-21: basic biology and implications for cancer and autoimmunity. Annu Rev Immunol. 2008;26:57–79. doi: 10.1146/annurev.immunol.26.021607.090316. [DOI] [PubMed] [Google Scholar]
- 128.Kastelein R.A., Hunter C.A., Cua D.J. Discovery and biology of IL-23 and IL-27: related but functionally distinct regulators of inflammation. Annu Rev Immunol. 2007;25:221–242. doi: 10.1146/annurev.immunol.22.012703.104758. [DOI] [PubMed] [Google Scholar]
- 129.Derry J.M., Ochs H.D., Francke U. Isolation of a novel gene mutated in Wiskott-Aldrich syndrome. Cell. 1994;79 doi: 10.1016/0092-8674(94)90528-2. following 922. [DOI] [PubMed] [Google Scholar]
- 130.Devriendt K., Kim A.S., Mathijs G., Frints S.G., Schwartz M., Van Den Oord J.J., Verhoef G.E., Boogaerts M.A., Fryns J.P., You D. Constitutively activating mutation in WASP causes X-linked severe congenital neutropenia. Nat Genet. 2001;27:313–317. doi: 10.1038/85886. [DOI] [PubMed] [Google Scholar]
- 131.Toth B, Mehes L, Tasko S, Szalai Z, Tulassay Z, Cypowyj S, Casanova JL, Puel A, Marodi L: Reactivation herpes virus disease in a kindred with STAT1 mutation. Lancet 2012, in press. [DOI] [PubMed]
- 132.Siegel A.M., Heimall J., Freeman A.F., Hsu A.P., Brittain E., Brenchley J.M., Douek D.C., Fahle G.H., Cohen J.I., Holland S.M. A critical role for STAT3 transcription factor signaling in the development and maintenance of human T cell memory. Immunity. 2011;35:806–818. doi: 10.1016/j.immuni.2011.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Casanova J.L., Abel L. Primary immunodeficiencies: a field in its infancy. Science. 2007;317:617–619. doi: 10.1126/science.1142963. [DOI] [PubMed] [Google Scholar]
- 134.Quintana-Murci L., Alcais A., Abel L., Casanova J.L. Immunology in natura: clinical, epidemiological and evolutionary genetics of infectious diseases. Nat Immunol. 2007;8:1165–1171. doi: 10.1038/ni1535. [DOI] [PubMed] [Google Scholar]
- 135••.Barreiro L.B., Quintana-Murci L. From evolutionary genetics to human immunology: how selection shapes host defence genes. Nat Rev Genet. 2010;11:17–30. doi: 10.1038/nrg2698. [DOI] [PubMed] [Google Scholar]; Clear and comprehensive review on evolutionary genetics related to host defense genes.
- 136••.Manry J., Laval G., Patin E., Fornarino S., Itan Y., Fumagalli M., Sironi M., Tichit M., Bouchier C., Casanova J.L. Evolutionary genetic dissection of human interferons. J Exp Med. 2011;208:2747–2759. doi: 10.1084/jem.20111680. [DOI] [PMC free article] [PubMed] [Google Scholar]; The authors demonstrate that all IFNs have evolved differently, highlighting some of them under strong purifying selection, suggesting a potent role in antiviral immunity.