Abstract
A relatively large number of protein tyrosine phosphatases (PTPs) are known to regulate signaling through the T cell receptor (TCR). Recent human genetics studies have shown that several of these PTPs are encoded by major autoimmunity genes. Here, we will focus on the lymphoid tyrosine phosphatase (LYP), a critical negative modulator of TCR signaling encoded by the PTPN22 gene. The functional analysis of autoimmune-associated PTPN22 genetic variants suggests that genetic variability of TCR signal transduction contributes to the pathogenesis of autoimmunity in humans.
Keywords: PTPN22, LYP, Pep, Csk, Lck, TCR signaling, Tyrosine phosphatase, Autoimmunity
Tyrosine phosphatases: from TCR signaling to human autoimmunity
A relatively large number of protein tyrosine phosphatases (PTPs) are able to either upregulate or downregulate signaling through the T cell receptor (TCR). For the current classification of PTPs and an overview of all the PTPs involved in T cell activation, we refer the reader to the following reviews [1–3]. There is well-established evidence that alteration of the expression/activity of PTPs involved in TCR signaling regulation causes immunopathology in mice. One of the first examples reported was the SH2-containing PTP SHP-1, which is encoded by the gene Ptpn6 and is a negative regulator of signaling through the TCR and other immune receptors. Functional inactivation of SHP-1 in mice gives rise to the motheaten phenotype, characterized by immune-deficiency, autoimmunity, and lymphoproliferation [4, 5]. Another well-known example is the receptor PTP CD45, encoded by the Ptprc gene, which is a positive regulator of TCR signaling. CD45 is currently considered a drug target for inflammatory diseases and autoimmunity, based upon several lines of in vivo evidence, including the fact that CD45 knockout (KO) mice show severe immunodeficiency [6], while mice carrying a gain-of-function E613R mutation develop autoimmune-like disease [7].
Studies on PTPs in human immunopathology have somehow lagged behind until recently, when genetic studies have shown that PTPs make up a significant fraction of the autoimmunity genes in humans. Initial studies using the candidate-gene approach and more recent genome-wide association (GWA) studies have shown that at least six major (i.e., showing genome-wide signal in association studies) autoimmunity genes encode known PTPs (PTPN22, PTPN2, PTPRC, UBASH3A, PTPN11, and PTPRT)[8–14]. Most of these enzymes are known regulators of T cell activation [3, 15]. Among the PTPs encoded by autoimmunity genes, only CD45 (encoded by PTPRC) has an established role in mouse autoimmunity, and excellent reviews have been published on the biochemistry and immunology of this enzyme [16, 17]. Little is known about the other PTPs and/or their mechanism of action in human autoimmunity. In this review, we will focus on the PTP encoded by PTPN22, which is known as lymphoid tyrosine phosphatase (LYP) and is a potent modulator of TCR signaling. The results of functional genetics studies of autoimmune-predisposing PTPN22 variants support the idea that genetic variability of TCR signaling predisposes a subset of individuals to development of autoimmune diseases. These same studies also suggest that inhibition of LYP might be beneficial in autoimmunity. If confirmed by further validation data, this will make LYP one of the first cases of a novel drug target emerging from human genetic studies.
LYP is a critical negative regulator of TCR signaling
LYP is a Class 1 cytosolic PTP [2], with expression restricted to hematopoietic cells. It belongs to a subfamily of “PEST-enriched PTPs” (corresponding to the NT4 subtype of classical PTPs according to another classification system [18]), which includes two additional enzymes: PTP-PEST (encoded by the PTPN12 gene) and BDP1 (encoded by the PTPN18 gene).
LYP and its mouse homolog pest-enriched phosphatase (Pep) are ∼105 kD proteins characterized by a ∼300-aa N-terminal tyrosine phosphatase domain and a ∼200-aa C-terminal domain which includes four putative polyproline (PEST-enriched) motifs (termed P1-P4) [19, 20]. The catalytic domain and the C-terminal domain are separated by a ∼300-aa region called “the interdomain.” A second shorter isoform of LYP called LYP2 has been identified in resting T cells [20]. Little is known about the expression/function of LYP2 or the possible existence of additional isoforms of LYP or Pep. However, by western blotting of lysates of resting or TCR-stimulated human T cells, we observed that full-length LYP is by far the predominant isoform in these cells and that LYP2 expression levels are much lower than full-length LYP both at the mRNA and protein levels (our unpublished observation).
In T cells, LYP and Pep are potent negative regulators of TCR signaling [21–26] through dephosphorylation of several key mediators of early TCR signal transduction [25, 27]. The activatory pTyr residues in the catalytic domain of the Src kinases Lck (Tyr394) and FynT (Tyr417) and of the Syk family kinase Zap70 (Tyr493) are considered to be physiological substrates of Pep and LYP. In addition to these kinases, TCRzeta, CD3epsilon, Vav, and Vcp were also identified in substrate-trapping experiments and are additional putative substrates of the phosphatase [27]. Importantly, in T cells, and presumably in other immune cells, Pep and LYP form a complex with the tyrosine kinase Csk, also a negative regulator of TCR signaling [8, 21, 22, 25]. The complex between the phosphatase and the kinase is dependent on the interaction between the most N-terminal P1 motif of Pep/LYP and the SH3 domain of Csk [21]. The molecular basis of the Pep/ Csk interaction has been elucidated by alanine scanning of the P1 domain [28] and by crystallography [29]. In T cells, the high stoichiometry of the complex (around 50% Pep co-precipitates with Csk [25]) points to an important physiological function. Early experiments carried out in mouse cell lines overexpressing Pep mutants [25] and in Jurkat cells transfected with Pep and/or Csk [26] suggested that the interaction between Pep and Csk leads to synergistic inhibition of TCR signaling. However, as discussed below, recent data emerging from the functional characterization of human genetic variants of LYP suggest that this model might not be universally valid and/or exhaustive. More work is needed in order to clarify the function of the LYP–Csk complex in TCR signaling. The recent finding that the phosphatase is excluded from lipid rafts in mouse T cells [30] adds complexity to the problem, since it is hard to reconcile with the high stoichiometry of the Pep–Csk complex and the main action of the phosphatase on Lck and Zap70. Also, there might be additional physiological interactors of the P1 or other domains of LYP/Pep beside Csk, and additional proteins might be recruited to the LYP/ Csk complex.
The regulation of LYP/Pep is currently an active area of research. Recently, the Zhang group showed that LYP can be phosphorylated on Ser35 by PKC, with consequent inhibition of the catalytic activity [31]. LYP and Pep contain multiple additional putative sites for Ser/Thr kinases. As mentioned below, we also recently observed phosphorylation of LYP on Tyr residues, which might contribute to regulate its activity. Additional interesting aspects of LYP/Pep regulation are emerging from molecular and structural studies. By studying truncation mutants of the recombinant phosphatase, we recently demonstrated that the activity of LYP is modulated by an intramolecular interaction between the proximal interdomain and the catalytic domain [32]. Crystallization of the catalytic domain of the phosphatase in our hands also showed that the enzyme might be regulated by a reversible oxidation mechanism involving a back-door Cys residue [33].
Relatively little is known about the role of LYP/Pep in the immune system in vivo. Andrew Chan’s group published in early 2004 the phenotype of a global Ptpn22 KO mouse [34]. Deletion of the phosphatase caused expansion of the T cell memory compartment and increased TCR signaling in effector T cells. At the thymic level, there was a phenotype of increased positive selection [34]. The phenotype of the mouse supports the idea (originally proposed by Andre Veillette [21]) that Pep is a key negative regulator of TCR signaling. The prominent effector/memory T cell phenotype is in line with the high expression of the phosphatase in these cells ([35] and our unpublished observation). Pep is highly expressed in thymocytes as well ([35] and our unpublished observation), and its role in thymic selection might help explain the effect of the phosphatase in human autoimmunity.
An open area of discussion is whether LYP and Pep play any role in regulation of B cell activation. The KO mouse shows no alterations of signaling through the B cell receptor (BCR) [34], and low expression levels of Pep have been found in mouse B cells [35]. Expansion of B cells in germinal centers occurs in aging mice, but the phenotype has been attributed to increased T cell help [34]. However, a recent study suggested that LYP might affect signaling through the BCR in primary human B cells ([36] and see below). Thus, it is possible that the KO mouse is a less faithful model of LYP function in human B cell physiology due to compensation phenomena or perhaps to real functional differences between the human and the mouse phosphatase. Besides T cells and B cells, little is known about the role of LYP/Pep in other immune cell subpopulations. For example, a simple search of the BioGPS database [37](http://biogps.gnf.org) suggests that dendritic cells (DC) and natural killer (NK) cells carry high mRNA levels of LYP/Pep. More immunological studies in the available KO mouse model are warranted. However, in order to fully understand the role of LYP/Pep in the immune system, we will also need to generate additional knockout and transgenic models, engineered in order to enable deletion/overexpression of the phosphatase in selected immune cell subpopulations.
Association of PTPN22 with human autoimmunity
Three reports in 2004 documented the association between a missense C1858T (R620W) single nucleotide polymorphism (SNP) in PTPN22 and type 1 diabetes (T1D) [8], rheumatoid arthritis (RA) [22], and systemic lupus erythematosus (SLE) [38]. The association between PTPN22 andT1D,RA,andSLEwassubsequentlywidely replicated by us and others (for example, see [39–41]) and extended to many additional autoimmune diseases, including Graves’ disease [39, 42, 43], Addison’s disease [44, 45], vitiligo [46, 47], myasthenia gravis [48–50], and systemic sclerosis [51]. PTPN22 *T1858 behaves as a dominant variant, conferring increased risk of disease alreadywhenpresentinsinglecopy[52, 53]. The risk conferred by PTPN22 is variable among diseases, but it is substantial in T1D and RA, with average reported odds ratios of 1.7–2.0 per single allele copy. Recent GWA studies detected a major signal on PTPN22 for T1D and RA, and PTPN22 currently ranks in third and second position, respectively, for single-gene contribution to the risk of T1D and RA in Caucasian populations [10]. In SLE [54]and T1D[55], the effect of PTPN22 seems to be even more prominent in the subgroup of patients who have familiarity for autoimmunity.
As mentioned, the association of PTPN22 C1858T with autoimmune diseases has been replicated across different populations. For example, the association with T1D has been replicated in the American [56, 57], Italian [58], German [59], Spanish [60], English [39], Estonian [61], and Ukrainian [62] populations. These studies also showed that there are significant differences in the frequency of the *T1858 allele among populations. The *T1858 allele is more frequent (around 12.5%) in Northern than in Southern Europeans, while it is almost absent in African–American and Asian populations [63–66]. Possible explanations for such dramatic geographic differences in allele frequency are that the *T1858 allele appeared recently during evolution and/or its frequency is severely affected by selection. By analyzing extended haplotype homozygosity, McPartland et al. showed evidence of positive selection at the PTPN22 locus [67]. One intriguing hypothesis is that the *T1858 allele confers protection toward some highly prevalent infectious disease. Only a few studies have been carried out on PTPN22 in infectious diseases so far. Chapman et al. found that the *T1858 allele might increase risk of complication in patients affected by common pneumococcal infections [68], while another study on immuno-compromised transplant recipients found an opposite protective effect of the *T1858 allele against infections [69]. In tuberculosis (TB), we found some evidence that the *T1858 allele might play a protective role [70, 71]. This is an interesting finding considering that TB might have been a powerful selective force in Northern Europe. However, these data need to be replicated and backed by more mechanistic studies before any conclusions can be drawn about the possible role of TB in shaping selection at the PTPN22 locus.
PTPN22 C1858T belongs to a growing family of “shared autoimmunity loci,” which are associated with multiple autoimmune diseases. Together with a subset of these genes, PTPN22 also has been shown to contribute to the recurrence of multiple different autoimmune diseases in certain families [72]. The identification and functional dissection of shared autoimmunity loci could potentially unravel major pathogenic mechanisms of autoimmunity. Association with certain but not other autoimmune diseases also can give important hints on the mechanism of action of shared autoimmunity loci. For PTPN22,someofthe negative findings might be due to variable combinations of small effect, low frequency of the allele, and sometime low statistical power of the study. However, well-powered studies found no association with multiple sclerosis and celiac disease [73–76]. The *T1858 allele was reported to have negligible effect on the risk of inflammatory bowel disease (for example, see [77] and [78]), but a recent large study suggests that in Crohn’s disease, it might be a protective factor [79]. A similar protective effect has been reported in Behcet’s disease [80]. Interestingly, PTPN22 *T1858 is associated with psoriatic arthritis [81, 82] but not with psoriasis. However, a psoriasis locus might be located in close proximity to PTPN22 [83, 84], suggesting that PTPN22 might lay within an autoimmunity hot spot on chromosome 1. The reason why PTPN22 associates with some but not other major autoimmune diseases is unclear at the moment. It has been suggested that the PTPN22 C1858T polymorphism preferentially associates with diseases characterized by a strong autoantibody (auto-Ab) component [85]. In favor of this hypothesis, PTPN22 associates with diseases which are exquisitely auto-Ab-mediated, like myasthenia gravis; also, it has been reported by some groups that the association between PTPN22 and RA is restricted to the anti-cyclic citrullinated peptide Ab positive subgroup [86]. However, it is unclear whether the association of PTPN22 with auto-Ab is a causal one, and some observations do not fit well with this theory, for example, PTPN22 does not associate with celiac disease, a disease characterized by auto-Ab production.
Genetic dissection and linkage studies in RA and T1D have determined that PTPN22 C1858T is a primary disease locus [87–91]. Studies are ongoing to clarify whether there are additional predisposition/protection loci within or close to PTPN22. We reported a rare missense variation R263Q within the catalytic domain, which is functional (see below) and segregates on different haplotypes than the R620W variant [92]. A recent comprehensive analysis of the T1D Genetic Consortium (T1DGC) collection has confirmed the primary association between T1D and C1858T and found evidence for an independent protective haplotype (Steck et al., manuscript in press). Also, there is some evidence that a variation G-1123C in the promoter region of PTPN22 associates with autoimmunity [93]. The data on this SNP have been difficult to interpret due to the strict linkage disequilibrium (LD) between the G-1123C and the C1858T SNPs in European populations [94]. In the Sardinian population, where LD between the two variants is lower, we could detect a strong signal at the C1858T locus only, suggesting that in the presence of C1858T, the effect of G-1123C might be a minor one [90]. However, the G-1123C polymorphism might be a significant risk factor in Asian populations, and more work is needed in order to establish the association of this SNP with autoimmunity and its functional effect.
Other important areas of genetic research concern possible interactions between PTPN22 and other genetic or environmental factors, and whether PTPN22 associates with prognosis and disease variables other than incidence. Some groups reported that the risk conferred by PTPN22 depends on HLA genotypes [95], while no PTPN22-HLA interaction has been found in other studies [60, 96, 97]. Few studies reported interactions with environmental factors, for example, smoking in RA [97, 98] and early exposure to cow milk in T1D [99]. In some populations, the association between PTPN22 and autoimmunity seems to depend on gender [59, 100]; however, this finding has not been universally confirmed, and it is of difficult interpretation at the moment. As for the clinical variability of disease, beside the mentioned data on the association with auto-Ab-positive subsets of RA, there are sporadic reports about a possible effect of PTPN22 on age of onset [56, 60]. The possible value of PTPN22 in early diagnosis/prognosis has been assessed in prospective studies in T1D and RA. An association with radiographic progression of disease has been reported in RA [101]. The data in T1D are less homogeneous, and association with progression to disease has been found in some studies [102] but not in others [103], including a notable one, which reported exclusive association with development of persistent auto-Abs [104].
Functional genetics of the PTPN22 C1858T polymorphism
Understanding the effect of the R620W polymorphism on the function of LYP/Pep is obviously of critical importance. Several functional genetics studies have focused on possible differences in signal transduction through the TCR in cells from carriers of the W620 variant of LYP when compared to subjects carrying exclusively the R620 variant. We reported that primary T cells from T1D patients carrying the W620 allele exhibited reduced IL-2 response to TCR engagement. These findings were replicated by Aarnisalo et al. in T1D [105]. The Buckner group reported decreased TCR-induced phosphorylation of early signaling intermediates and Ca++ mobilization in T cells from *T1858 homozygous RA patients [36]. Importantly, the same group also extended their studies to B cells and reported that BCR signaling in primary B cells from carriers of the *T1858 allele is also reduced [36]. Thus, the vast majority of reports points to decreased TCR-mediated T cell activation in primary T cells from carriers of the LYP-W620 variant. The only exception so far is a single report on myasthenia gravis, where the R620W polymorphism associated with increased TCR-induced IL-2 production [106]; however, this finding has not been replicated by another group [50]. Further functional genetics studies on large samples of primary T cells from healthy controls are ongoing in several laboratories and hopefully will help solidify even further our view of the effect of the polymorphism on T cell activation.
Despite most studies have focused so far on T cells and on readouts of TCR signaling, as mentioned, the polymorphism seems to affect signaling through the BCR as well [36]. It is currently unknown whether the polymorphism affects the function of other cell types which are critical for autoimmunity, for example, DC and NK/NKT cells.
Few studies have looked so far at possible effects of the polymorphism on the phenotype of primary T and B cells from genotyped patients and controls. For example, a study suggested that *T1858 carriers have increased numbers of memory T cells, and that T cells from these individuals might secrete less of the immunosuppressive IL-10 cytokine [36]. Much progress is expected in this area in the near future, which will shed light on the mechanism of action of the polymorphism in autoimmunity.
At the molecular level, the PTPN22 C1858T polymorphism leads to a substitution of a Trp for an Arg at position 620. This is a nonconservative variation of a residue within the P1 motif, which had been previously shown to be critical for the Pep-Csk binding. As a consequence, the autoimmune-associated LYP-W620 variant exhibits reduced interaction with Csk [8, 22]. Besides its effect on the complex between the kinase and the phosphatase, the mechanism of action of the polymorphism at the biochemical and molecular level is still poorly understood. By studying T cells overexpressing LYP-W620 or LYP-R620, we found that LYP-W620 expression induces a gain-of-function decrease of TCR-induced phosphorylation of early key signaling players and decreased TCR-induced Ca++ mobilization [23]. In our hands, also the autoimmune-associated LYP-W620 variant showed increased phosphatase activity when compared to LYP-R620 [23]. We concluded that gain-of-function inhibition of TCR signaling by LYP-W620, mediated at least in part by increased phosphatase activity, is responsible for the decreased T cell activation observed in carriers of the pathogenic phosphatase variant. These conclusions have been recently challenged by a report from Zikherman et al., claiming that the W620 mutant of LYP (and the homolog W619 mutant of Pep) is a hypomorph variant, and the gain-of-function phenotype is an artifact which arises when the phosphatase is overexpressed without parallel overexpression of Csk [35]. However, it is unclear whether the system used by Zikherman et al. (based on co-transfection of cells with LYP/Pep, Csk, and GFP) is more respectful of the physiological stoichiometry of the interaction between the phosphatase and the kinase. It also remains to be explained how an hypomorph allele of PTPN22 would give rise to the anomalies of T cell activation reported in primary T cells from LYP-W620 carriers. More studies in multiple systems including cells from mice carrying knock-in mutations of LYP/Pep (which are in preparation in several laboratories in the world) are needed in order to better define the effect of the mutation on cell signaling.
We and others are also actively investigating how the mutation affects the function of LYP at the molecular level. LYP mRNA levels seem to be unaffected by the polymorphism, suggesting that the mutation acts mainly at the protein level [107]. We recently found that LYP undergoes Csk-dependent phosphorylation on at least one tyrosine, which results in inhibition of the phosphatase activity. We suggest that anomalies in Csk-dependent posttranslational modification(s) of LYP mediate at least in part the molecular mechanism of the R620W variation (Fiorillo et al., manuscript submitted). Csk-independent mechanisms are also possible, including impaired binding to additional interactors and/or recruitment of LYP-W620 to an unknown protein through its P1-W620 motif. One of the challenges of the current studies at the molecular level is to reconcile the gain-of-function phenotype of LYP-W620 with previous results suggesting a model of synergistic inhibition of signaling by the complex between the kinase and the phosphatase [25, 26]. A possible explanation is that the regulation of LYP and Pep is not entirely conserved: indeed, the two phosphatases share relatively low (<60%) identity in the interdomain, which might be important for regulation.
(Putative) mechanisms of action of PTPN22 in autoimmunity
Our current working model is that gain-of-function inhibition of signaling in T cells (and perhaps other cell types) mediates the pathogenic action of the polymorphism and the increased risk of autoimmunity of carriers of LYP-W620. It might sound a little counter-intuitive that decreased immune cell activation leads to autoimmunity; however, our model is in line with the current view that decreased TCR signaling increases risk of at least a subset of autoimmune diseases. Decreased TCR signaling has been reported in T cells from NOD mice [108] and in peripheral T cells from T1D patients [109]. Mechanistic evidence of a link between decreased TCR signaling and autoimmunity was also provided by the description of the SKG mouse by Sakaguchi et al., in 2003 [110]. In this mouse, a loss-of-function mutation of Zap70 (one of the physiological substrates of LYP) causes a spontaneous autoimmune disease which is remarkably similar to human RA. Similar findings of autoimmune-like disease in mice carrying loss-of-function of Zap70 have been subsequently reported by two additional groups [111, 112]. These studies also are showing that decreased TCR signaling impinges on RA through multiple mechanisms [111]. In the case of PTPN22, we could also speculate that some mechanisms might be prominent over others in different diseases, or even in different subjects affected by the same disease.
One of the favored models is that gain-of-function inhibition of TCR signaling at the thymic level leads to decreased negative selection and elimination of potentially autoreactive T cells and/or decreased production of natural regulatory T cells (Treg). The observations that (1) Pep is highly expressed in the thymus, (2) the Ptpn22 KO mouse shows anomalies of thymic selection, and (3) normalization of TCR signaling in the thymus is able to rescue the phenotype of the SKG mouse [110] provide additional support to the “thymic selection” theory. In addition or alternative to thymic selection anomalies, other mechanisms have been envisioned. For example, an anomalous increase in the activity of LYP in effector T cells might negatively impact the activity/expansion of peripheral Treg in carriers of LYP-W620 [113]. Decreased TCR-induced production of IL-2 by effector T cells is known to impair expansion of Treg in mouse T1D, and neutralization of IL-2 in vivo induces autoimmunity in mice through a Treg-mediated mechanism [114–116]. Marson et al. also recently identified PTPN22 among a group of genes that are highly occupied by FoxP3 in Treg. The same study also showed that Treg carry lower levels of the phosphatase compared to effector T cells [113], suggesting a direct mechanism by which gain-of-function of LYP can affect Treg function. Since TCR signaling intensity is an important regulator of naïve T cell differentiation, it is also possible that decreased TCR signaling affects polarization of naïve T cells into specialized Thelper populations, for example, favoring the appearance of diabetogenic interfer-on-gamma-producing Th1 cells, in carriers of LYP-W620. The observation that peripheral T cells from LYP-W620 carriers have a different cytokine secretion pattern characterized by reduced production of IL-10 is in line with this hypothesis. Other mechanisms are possible and, considering the association between PTPN22 and the presence of auto-Ab, we cannot exclude that LYP-W620 also affects B cell differentiation and/or tolerization directly.
LYP as a possible drug target for autoimmunity
Several academic laboratories have begun developing small molecule inhibitors of LYP [31, 117, 118]. Given the increased activity of LYP-W620, it has been hypothesized that a selective inhibitor of LYP could revert the negative effects of the W620 variant on TCR signaling, and thus constitute an effective etiological therapy of autoimmunity in carriers of the autoimmune-predisposing variant. Because PTPN22 is implicated in many prevalent autoimmune diseases (and perhaps even in transplant rejection [119]), an anti-LYP drug could be of broader value for the treatment of multiple human conditions. Also, the phenotype of the KO mouse suggests that a specific inhibitor of PTPN22 is likely to have limited side effects.
Although it remains to be proven that the mechanism of action of LYP in autoimmunity is reversible, the success of trials targeting T1D with monoclonal antibodies against CD3 supports the idea that positive modulation of the TCR helps re-establishing tolerance in at least a subset of autoimmune patients [120, 121]. In support of the above-mentioned hypothesis, we also recently found a rare loss-of-function LYP-R263Q genetic variation, which segregates on haplotypes different from the R620W one and plays a protective role in SLE [92].
Further studies in animal models are needed in order to validate LYP as a drug target for autoimmunity. Importantly, therapeutic inhibition of LYP in autoimmunity might also carry some risks. For example, excessive inhibition of LYP activity might lead to parallel counterproductive increase in TCR signaling in effector T cells. Although the Ptpn22 KO mutation on the B6 background fails to induce autoimmunity [34], Zikherman et al. recently showed that crossing of the Ptpn22 KO mouse with the CD45 E613R (“wedge”) model led to more severe autoimmunity [35]. Since spontaneous autoimmunity on B6 background in the CD45 wedge model is driven by increased TCR [122] and BCR signaling [123, 124], the results of Zikherman et al. suggest that inhibition of LYP might be counterproductive in diseases or subsets of diseases with a strong component of TCR or BCR hyperactivity (which has been observed in large subsets of lupus patients [125–127]) and thus not easily extended to noncarriers of gain-of-function LYP mutation(s).
Acknowledgments
Acknowledgements This work was supported by NIH grant R01AI070544 to N.B. This is publication #1257 from the La Jolla Institute for Allergy and Immunology.
Contributor Information
Stephanie M. Stanford, Division of Cell Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA.
Tomas M. Mustelin, Sanford-Burnham Medical Research Institute, Infectious and Inflammatory Disease Center, La Jolla, CA 92037, USA
Nunzio Bottini, Division of Cell Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA. nunzio@liai.org.
References
- 1.Mustelin T, Tasken K. Positive and negative regulation of T-cell activation through kinases and phosphatases. Biochem J. 2003;371(Pt 1):15–27. doi: 10.1042/BJ20021637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Alonso A, Sasin J, Bottini N, et al. Protein tyrosine phosphatases in the human genome. Cell. 2004;117(6):699–711. doi: 10.1016/j.cell.2004.05.018. [DOI] [PubMed] [Google Scholar]
- 3.Mustelin T, Alonso A, Bottini N, et al. Protein tyrosine phosphatases in T cell physiology. Mol Immunol. 2004;41(6–7):687–700. doi: 10.1016/j.molimm.2004.04.015. [DOI] [PubMed] [Google Scholar]
- 4.Kozlowski M, Mlinaric-Rascan I, Feng GS, et al. Expression and catalytic activity of the tyrosine phosphatase PTP1C is severely impaired in motheaten and viable motheaten mice. J Exp Med. 1993;178(6):2157–2163. doi: 10.1084/jem.178.6.2157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Tsui FW, Tsui HW. Molecular basis of the motheaten phenotype. Immunol Rev. 1994;138:185–206. doi: 10.1111/j.1600-065x.1994.tb00852.x. [DOI] [PubMed] [Google Scholar]
- 6.Koretzky GA, Picus J, Schultz T, Weiss A. Tyrosine phosphatase CD45 is required for T-cell antigen receptor and CD2-mediated activation of a protein tyrosine kinase and interleukin 2 production. Proc Natl Acad Sci USA. 1991;88(6):2037–2041. doi: 10.1073/pnas.88.6.2037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Majeti R, Xu Z, Parslow TG, et al. An inactivating point mutation in the inhibitory wedge of CD45 causes lymphoproliferation and autoimmunity. Cell. 2000;103(7):1059–1070. doi: 10.1016/s0092-8674(00)00209-9. [DOI] [PubMed] [Google Scholar]
- 8.Bottini N, Musumeci L, Alonso A, et al. A functional variant of lymphoid tyrosine phosphatase is associated with type I diabetes. Nat Genet. 2004;36(4):337–338. doi: 10.1038/ng1323. [DOI] [PubMed] [Google Scholar]
- 9.The Wellcome Trust Case Control Consortium. Genome-wide association study of 14, 000 cases of seven common diseases and 3, 000 shared controls. Nature. 2007;447(7145):661–678. doi: 10.1038/nature05911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Todd JA, Walker NM, Cooper JD, et al. Robust associations of four new chromosome regions from genome-wide analyses of type 1 diabetes. Nat Genet. 2007;39(7):857–864. doi: 10.1038/ng2068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Hennig BJ, Fry AE, Hirai K, et al. PTPRC (CD45) variation and disease association studied using single nucleotide polymorphism tagging. Tissue antigens. 2008;71(5):458–463. doi: 10.1111/j.1399-0039.2008.01014.x. [DOI] [PubMed] [Google Scholar]
- 12.Julia A, Ballina J, Canete JD, et al. Genome-wide association study of rheumatoid arthritis in the Spanish population: KLF12 as a risk locus for rheumatoid arthritis susceptibility. Arthritis Rheum. 2008;58(8):2275–2286. doi: 10.1002/art.23623. [DOI] [PubMed] [Google Scholar]
- 13.Concannon P, Onengut-Gumuscu S, Todd JA, et al. A human type 1 diabetes susceptibility locus maps to chromosome 21q22.3. Diabetes. 2008;57(10):2858–2861. doi: 10.2337/db08-0753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Raychaudhuri S, Thomson BP, Remmers EF, et al. Genetic variants at CD28, PRDM1 and CD2/CD58 are associated with rheumatoid arthritis risk. Nat Genet. 2009;41(12):1313–1318. doi: 10.1038/ng.479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Carpino N, Turner S, Mekala D, et al. Regulation of ZAP-70 activation and TCR signaling by two related proteins, Sts-1 and Sts-2. Immunity. 2004;20(1):37–46. doi: 10.1016/s1074-7613(03)00351-0. [DOI] [PubMed] [Google Scholar]
- 16.Irie-Sasaki J, Sasaki T, Penninger JM. CD45 regulated signaling pathways. Curr Top Med Chem. 2003;3(7):783–796. doi: 10.2174/1568026033452339. [DOI] [PubMed] [Google Scholar]
- 17.Hermiston ML, Xu Z, Weiss A. CD45: a critical regulator of signaling thresholds in immune cells. Annu Rev Immunol. 2003;21:107–137. doi: 10.1146/annurev.immunol.21.120601.140946. [DOI] [PubMed] [Google Scholar]
- 18.Andersen JN, Jansen PG, Echwald SM, et al. A genomic perspective on protein tyrosine phosphatases: gene structure, pseudogenes, and genetic disease linkage. Faseb J. 2004;18(1):8–30. doi: 10.1096/fj.02-1212rev. [DOI] [PubMed] [Google Scholar]
- 19.Matthews RJ, Bowne DB, Flores E, Thomas ML. Characterization of hematopoietic intracellular protein tyrosine phosphatases: description of a phosphatase containing an SH2 domain and another enriched in proline-, glutamic acid-, serine-, and threonine-rich sequences. Mol Cell Biol. 1992;12(5):2396–2405. doi: 10.1128/mcb.12.5.2396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Cohen S, Dadi H, Shaoul E, Sharfe N, Roifman CM. Cloning and characterization of a lymphoid-specific, inducible human protein tyrosine phosphatase, Lyp. Blood. 1999;93(6):2013–2024. [PubMed] [Google Scholar]
- 21.Cloutier JF, Veillette A. Association of inhibitory tyrosine protein kinase p50csk with protein tyrosine phosphatase PEP in T cells and other hemopoietic cells. Embo J. 1996;15(18):4909–4918. [PMC free article] [PubMed] [Google Scholar]
- 22.Begovich AB, Carlton VE, Honigberg LA, et al. A missense single-nucleotide polymorphism in a gene encoding a protein tyrosine phosphatase (PTPN22) is associated with rheumatoid arthritis. Am J Hum Genet. 2004;75(2):330–337. doi: 10.1086/422827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Vang T, Congia M, Macis MD, et al. Autoimmuneassociated lymphoid tyrosine phosphatase is a gain-of-function variant. Nat Genet. 2005;37(12):1317–1319. doi: 10.1038/ng1673. [DOI] [PubMed] [Google Scholar]
- 24.Hill RJ, Zozulya S, Lu YL, et al. The lymphoid protein tyrosine phosphatase Lyp interacts with the adaptor molecule Grb2 and functions as a negative regulator of T-cell activation. Exp Hematol. 2002;30(3):237–244. doi: 10.1016/s0301-472x(01)00794-9. [DOI] [PubMed] [Google Scholar]
- 25.Cloutier JF, Veillette A. Cooperative inhibition of T-cell antigen receptor signaling by a complex between a kinase and a phosphatase. J Exp Med. 1999;189(1):111–121. doi: 10.1084/jem.189.1.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Gjorloff-Wingren A, Saxena M, Williams S, Hammi D, Mustelin T. Characterization of TCR-induced receptor-proximal signaling events negatively regulated by the protein tyrosine phosphatase PEP. Eur J Immunol. 1999;29(12):3845–3854. doi: 10.1002/(SICI)1521-4141(199912)29:12<3845::AID-IMMU3845>3.0.CO;2-U. [DOI] [PubMed] [Google Scholar]
- 27.Wu J, Katrekar A, Honigberg LA, et al. Identification of substrates of human protein-tyrosine phosphatase PTPN22. J Biol Chem. 2006;281(16):11002–11010. doi: 10.1074/jbc.M600498200. [DOI] [PubMed] [Google Scholar]
- 28.Gregorieff A, Cloutier JF, Veillette A. Sequence requirements for association of protein-tyrosine phosphatase PEP with the Src homology 3 domain of inhibitory tyrosine protein kinase p50(csk) J Biol Chem. 1998;273(21):13217–13222. doi: 10.1074/jbc.273.21.13217. [DOI] [PubMed] [Google Scholar]
- 29.Ghose R, Shekhtman A, Goger MJ, Ji H, Cowburn D. A novel, specific interaction involving the Csk SH3 domain and its natural ligand. Nat Struct Biol. 2001;8(11):998–1004. doi: 10.1038/nsb1101-998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Davidson D, Bakinowski M, Thomas ML, Horejsi V, Veillette A. Phosphorylation-dependent regulation of T-cell activation by PAG/Cbp, a lipid raft-associated transmembrane adaptor. Mol Cell Biol. 2003;23(6):2017–2028. doi: 10.1128/MCB.23.6.2017-2028.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Yu X, Sun JP, He Y, et al. Structure, inhibitor, and regulatory mechanism of Lyp, a lymphoid-specific tyrosine phosphatase implicated in autoimmune diseases. Proc Natl Acad Sci USA. 2007;104(50):19767–19772. doi: 10.1073/pnas.0706233104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Liu Y, Stanford SM, Jog SP, et al. Regulation of lymphoid tyrosine phosphatase activity: inhibition of the catalytic domain by the proximal interdomain. Biochemistry. 2009;48(31):7525–7532. doi: 10.1021/bi900332f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Tsai SJ, Sen U, Zhao L, et al. Crystal structure of the human lymphoid tyrosine phosphatase catalytic domain: insights into redox regulation. Biochemistry. 2009;48(22):4838–4845. doi: 10.1021/bi900166y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Hasegawa K, Martin F, Huang G, et al. PEST domainenriched tyrosine phosphatase (PEP) regulation of effector/memory T cells. Science (New York, NY) 2004;303(5658):685–689. doi: 10.1126/science.1092138. [DOI] [PubMed] [Google Scholar]
- 35.Zikherman J, Hermiston M, Steiner D, et al. PTPN22 deficiency cooperates with the CD45 E613R allele to break tolerance on a non-autoimmune background. J Immunol. 2009;182(7):4093–4106. doi: 10.4049/jimmunol.0803317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Rieck M, Arechiga A, Onengut-Gumuscu S, et al. Genetic variation in PTPN22 corresponds to altered function of T and B lymphocytes. J Immunol. 2007;179(7):4704–4710. doi: 10.4049/jimmunol.179.7.4704. [DOI] [PubMed] [Google Scholar]
- 37.Wu C, Orozco C, Boyer J, et al. BioGPS: an extensible and customizable portal for querying and organizing gene annotation resources. Genome Biol. 2009;10(11):R130. doi: 10.1186/gb-2009-10-11-r130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Kyogoku C, Langefeld CD, Ortmann WA, et al. Genetic association of the R620W polymorphism of protein tyrosine phosphatase PTPN22 with human SLE. Am J Hum Genet. 2004;75(3):504–507. doi: 10.1086/423790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Smyth D, Cooper JD, Collins JE, et al. Replication of an association between the lymphoid tyrosine phosphatase locus (LYP/PTPN22) with type 1 diabetes, and evidence for its role as a general autoimmunity locus. Diabetes. 2004;53(11):3020–3023. doi: 10.2337/diabetes.53.11.3020. [DOI] [PubMed] [Google Scholar]
- 40.Orozco G, Sanchez E, Gonzalez-Gay MA, et al. Association of a functional single-nucleotide polymorphism of PTPN22, encoding lymphoid protein phosphatase, with rheumatoid arthritis and systemic lupus erythematosus. Arthritis Rheum. 2005;52(1):219–224. doi: 10.1002/art.20771. [DOI] [PubMed] [Google Scholar]
- 41.Plenge RM, Padyukov L, Remmers EF, et al. Replication of putative candidate-gene associations with rheumatoid arthritis in >4, 000 samples from North America and Sweden: association of susceptibility with PTPN22, CTLA4, and PADI4. Am J Hum Genet. 2005;77(6):1044–1060. doi: 10.1086/498651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Skorka A, Bednarczuk T, Bar-Andziak E, Nauman J, Ploski R. Lymphoid tyrosine phosphatase (PTPN22/LYP) variant and Graves’ disease in a Polish population: association and gene dose-dependent correlation with age of onset. Clin Endocrinol. 2005;62(6):679–682. doi: 10.1111/j.1365-2265.2005.02279.x. [DOI] [PubMed] [Google Scholar]
- 43.Heward JM, Brand OJ, Barrett JC, et al. Association of PTPN22 haplotypes with Graves’ disease. J Clin Endocrinol Metab. 2007;92(2):685–690. doi: 10.1210/jc.2006-2064. [DOI] [PubMed] [Google Scholar]
- 44.Velaga MR, Wilson V, Jennings CE, et al. The codon 620 tryptophan allele of the lymphoid tyrosine phosphatase (LYP) gene is a major determinant of Graves’ disease. J Clin Endocrinol Metab. 2004;89(11):5862–5865. doi: 10.1210/jc.2004-1108. [DOI] [PubMed] [Google Scholar]
- 45.Skinningsrud B, Husebye ES, Gervin K, et al. Mutation screening of PTPN22: association of the 1858T-allele with Addison’s disease. Eur J Hum Genet. 2008;16(8):977–982. doi: 10.1038/ejhg.2008.33. [DOI] [PubMed] [Google Scholar]
- 46.Canton I, Akhtar S, Gavalas NG, et al. A single-nucleotide polymorphism in the gene encoding lymphoid protein tyrosine phosphatase (PTPN22) confers susceptibility to generalised vitiligo. Genes Immun. 2005;6(7):584–587. doi: 10.1038/sj.gene.6364243. [DOI] [PubMed] [Google Scholar]
- 47.LaBerge GS, Bennett DC, Fain PR, Spritz RA. PTPN22 is genetically associated with risk of generalized vitiligo, but CTLA4 is not. J Invest Dermatol. 2008;128(7):1757–1762. doi: 10.1038/sj.jid.5701233. [DOI] [PubMed] [Google Scholar]
- 48.Vandiedonck C, Capdevielle C, Giraud M, et al. Association of the PTPN22*R620W polymorphism with autoimmune myasthenia gravis. Ann Neurol. 2006;59(2):404–407. doi: 10.1002/ana.20751. [DOI] [PubMed] [Google Scholar]
- 49.Greve B, Hoffmann P, Illes Z, et al. The autoimmunity-related polymorphism PTPN22 1858C/T is associated with anti-titin antibody-positive myasthenia gravis. Hum Immunol. 2009;70(7):540–542. doi: 10.1016/j.humimm.2009.04.027. [DOI] [PubMed] [Google Scholar]
- 50.Chuang WY, Strobel P, Belharazem D, et al. The PTPN22 (gain-of-function)+1858T(+) genotypes correlate with low IL-2 expression in thymomas and predispose to myasthenia gravis. Genes Immun. 2009;10(8):667–672. doi: 10.1038/gene.2009.64. [DOI] [PubMed] [Google Scholar]
- 51.Gourh P, Tan FK, Assassi S, et al. Association of the PTPN22 R620W polymorphism with anti-topoisomerase I- and anticentromere antibody-positive systemic sclerosis. Arthritis Rheum. 2006;54(12):3945–3953. doi: 10.1002/art.22196. [DOI] [PubMed] [Google Scholar]
- 52.Bottini N, Vang T, Cucca F, Mustelin T. Role of PTPN22 in type 1 diabetes and other autoimmune diseases. Semin Immunol. 2006;18(4):207–213. doi: 10.1016/j.smim.2006.03.008. [DOI] [PubMed] [Google Scholar]
- 53.Gregersen PK, Lee HS, Batliwalla F, Begovich AB. PTPN22: setting thresholds for autoimmunity. Semin Immunol. 2006;18(4):214–223. doi: 10.1016/j.smim.2006.03.009. [DOI] [PubMed] [Google Scholar]
- 54.Kaufman KM, Kelly JA, Herring BJ, et al. Evaluation of the genetic association of the PTPN22 R620W polymorphism in familial and sporadic systemic lupus erythematosus. Arthritis Rheum. 2006;54(8):2533–2540. doi: 10.1002/art.21963. [DOI] [PubMed] [Google Scholar]
- 55.Chelala C, Duchatelet S, Joffret ML, et al. PTPN22 R620W functional variant in type 1 diabetes and autoimmunity related traits. Diabetes. 2007;56(2):522–526. doi: 10.2337/db06-0942. [DOI] [PubMed] [Google Scholar]
- 56.Ladner MB, Bottini N, Valdes AM, Noble JA. Association of the single nucleotide polymorphism C1858T of the PTPN22 gene with type 1 diabetes. Hum Immunol. 2005;66(1):60–64. doi: 10.1016/j.humimm.2004.09.016. [DOI] [PubMed] [Google Scholar]
- 57.Zheng W, She JX. Genetic association between a lymphoid tyrosine phosphatase (PTPN22) and type 1 diabetes. Diabetes. 2005;54(3):906–908. doi: 10.2337/diabetes.54.3.906. [DOI] [PubMed] [Google Scholar]
- 58.Saccucci P, Del Duca E, Rapini N, et al. Association between PTPN22 C1858T and type 1 diabetes: a replication in continental Italy. Tissue antigens. 2008;71(3):234–237. doi: 10.1111/j.1399-0039.2007.00987.x. [DOI] [PubMed] [Google Scholar]
- 59.Kahles H, Ramos-Lopez E, Lange B, et al. Sex-specific association of PTPN22 1858T with type 1 diabetes but not with Hashimoto’s thyroiditis or Addison’s disease in the German population. Eur J Endocrinol. 2005;153(6):895–899. doi: 10.1530/eje.1.02035. [DOI] [PubMed] [Google Scholar]
- 60.Santiago JL, Martinez A, de la Calle H, et al. Susceptibility to type 1 diabetes conferred by the PTPN22 C1858T polymorphism in the Spanish population. BMC Med Genet. 2007;8:54. doi: 10.1186/1471-2350-8-54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Douroudis K, Prans E, Haller K, et al. Protein tyrosine phosphatase non-receptor type 22 gene variants at position 1858 are associated with type 1 and type 2 diabetes in Estonian population. Tissue antigens. 2008;72(5):425–430. doi: 10.1111/j.1399-0039.2008.01115.x. [DOI] [PubMed] [Google Scholar]
- 62.Fedetz M, Matesanz F, Caro-Maldonado A, et al. The 1858T PTPN22 gene variant contributes to a genetic risk of type 1 diabetes in a Ukrainian population. Tissue antigens. 2006;67(5):430–433. doi: 10.1111/j.1399-0039.2006.00591.x. [DOI] [PubMed] [Google Scholar]
- 63.Mori M, Yamada R, Kobayashi K, Kawaida R, Yamamoto K. Ethnic differences in allele frequency of autoimmune-disease-associated SNPs. J Hum Genet. 2005;50(5):264–266. doi: 10.1007/s10038-005-0246-8. [DOI] [PubMed] [Google Scholar]
- 64.Ikegami H, Kawabata Y, Noso S, Fujisawa T, Ogihara T. Genetics of type 1 diabetes in Asian and Caucasian populations. Diabetes Res Clin Pract. 2007;77 Suppl 1:S116–S121. doi: 10.1016/j.diabres.2007.01.044. [DOI] [PubMed] [Google Scholar]
- 65.Zhang ZH, Chen F, Zhang XL, et al. PTPN22 allele polymorphisms in 15 Chinese populations. Int J Immunogenet. 2008;35(6):433–437. doi: 10.1111/j.1744-313X.2008.00803.x. [DOI] [PubMed] [Google Scholar]
- 66.Lee HS, Korman BD, Le JM, et al. Genetic risk factors for rheumatoid arthritis differ in Caucasian and Korean populations. Arthritis Rheum. 2009;60(2):364–371. doi: 10.1002/art.24245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.McPartland JM, Norris RW, Kilpatrick CW. Tempo and mode in the endocannaboinoid system. J Mol Evol. 2007;65(3):267–276. doi: 10.1007/s00239-007-9004-1. [DOI] [PubMed] [Google Scholar]
- 68.Chapman SJ, Khor CC, Vannberg FO, et al. PTPN22 and invasive bacterial disease. Nat Genet. 2006;38(5):499–500. doi: 10.1038/ng0506-499. [DOI] [PubMed] [Google Scholar]
- 69.Azarian M, Busson M, Rocha V, et al. The PTPN22 R620W polymorphism is associated with severe bacterial infections after human leukocyte antigen geno-identical haematopoietic stem-cell transplantations. Transplantation. 2008;85(12):1859–1862. doi: 10.1097/TP.0b013e31817729c4. [DOI] [PubMed] [Google Scholar]
- 70.Gomez LM, Anaya JM, Martin J. Genetic influence of PTPN22 R620W polymorphism in tuberculosis. Hum Immunol. 2005;66(12):1242–1247. doi: 10.1016/j.humimm.2005.11.008. [DOI] [PubMed] [Google Scholar]
- 71.Lamsyah H, Rueda B, Baassi L, et al. Association of PTPN22 gene functional variants with development of pulmonary tuberculosis in Moroccan population. Tissue antigens. 2009;74(3):228–232. doi: 10.1111/j.1399-0039.2009.01304.x. [DOI] [PubMed] [Google Scholar]
- 72.Criswell LA, Pfeiffer KA, Lum RF, et al. Analysis of families in the multiple autoimmune disease genetics consortium (MADGC) collection: the PTPN22 620 W allele associates with multiple autoimmune phenotypes. Am J Hum Genet. 2005;76(4):561–571. doi: 10.1086/429096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Rueda B, Nunez C, Orozco G, et al. C1858T functional variant of PTPN22 gene is not associated with celiac disease genetic predisposition. Hum Immunol. 2005;66(7):848–852. doi: 10.1016/j.humimm.2005.04.008. [DOI] [PubMed] [Google Scholar]
- 74.Zhernakova A, Eerligh P, Wijmenga C, et al. Differential association of the PTPN22 coding variant with autoimmune diseases in a Dutch population. Genes Immun. 2005;6(6):459–461. doi: 10.1038/sj.gene.6364220. [DOI] [PubMed] [Google Scholar]
- 75.Lee YH, Rho YH, Choi SJ, et al. The PTPN22 C1858T functional polymorphism and autoimmune diseases—a meta-analysis. Rheumatology (Oxford, England) 2007;46(1):49–56. doi: 10.1093/rheumatology/kel170. [DOI] [PubMed] [Google Scholar]
- 76.Smyth DJ, Plagnol V, Walker NM, et al. Shared and distinct genetic variants in type 1 diabetes and celiac disease. N Engl J Med. 2008;359(26):2767–2777. doi: 10.1056/NEJMoa0807917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.van Oene M, Wintle RF, Liu X, et al. Association of the lymphoid tyrosine phosphatase R620W variant with rheumatoid arthritis, but not Crohn’s disease, in Canadian populations. Arthritis Rheum. 2005;52(7):1993–1998. doi: 10.1002/art.21123. [DOI] [PubMed] [Google Scholar]
- 78.Prescott NJ, Fisher SA, Onnie C, et al. A general autoimmunity gene (PTPN22) is not associated with inflammatory bowel disease in a British population. Tissue antigens. 2005;66(4):318–320. doi: 10.1111/j.1399-0039.2005.00494.x. [DOI] [PubMed] [Google Scholar]
- 79.Barrett JC, Hansoul S, Nicolae DL, et al. Genome-wide association defines more than 30 distinct susceptibility loci for Crohn’s disease. Nat Genet. 2008;40(8):955–962. doi: 10.1038/NG.175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Baranathan V, Stanford MR, Vaughan RW, et al. The association of the PTPN22 620W polymorphism with Behcet’s disease. Ann Rheum Dis. 2007;66(11):1531–1533. doi: 10.1136/ard.2007.073866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Huffmeier U, Reis A, Steffens M, et al. Male restricted genetic association of variant R620W in PTPN22 with psoriatic arthritis. J Invest Dermatol. 2006;126(4):932–935. doi: 10.1038/sj.jid.5700179. [DOI] [PubMed] [Google Scholar]
- 82.Butt C, Peddle L, Greenwood C, et al. Association of functional variants of PTPN22 and tp53 in psoriatic arthritis: a case-control study. Arthritis Res Ther. 2006;8(1):R27. doi: 10.1186/ar1880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Huffmeier U, Steffens M, Burkhardt H, et al. Evidence for susceptibility determinant(s) to psoriasis vulgaris in or near PTPN22 in German patients. J Med Genet. 2006;43(6):517–522. doi: 10.1136/jmg.2005.037515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Smith RL, Warren RB, Eyre S, et al. Polymorphisms in the PTPN22 region are associated with psoriasis of early onset. Br J Dermatol. 2008;158(5):962–968. doi: 10.1111/j.1365-2133.2008.08482.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.McGonagle D, Aziz A, Dickie LJ, McDermott MF. An integrated classification of pediatric inflammatory diseases, based on the concepts of autoinflammation and the immunological disease continuum. Pediatr Res. 2009;65(5 Pt 2):38R–45R. doi: 10.1203/PDR.0b013e31819dbd0a. [DOI] [PubMed] [Google Scholar]
- 86.Kallberg H, Padyukov L, Plenge RM, et al. Gene-gene and gene-environment interactions involving HLA-DRB1, PTPN22, and smoking in two subsets of rheumatoid arthritis. Am J Hum Genet. 2007;80(5):867–875. doi: 10.1086/516736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Onengut-Gumuscu S, Ewens KG, Spielman RS, Concannon P. A functional polymorphism (1858C/T) in the PTPN22 gene is linked and associated with type I diabetes in multiplex families. Genes Immun. 2004;5(8):678–680. doi: 10.1038/sj.gene.6364138. [DOI] [PubMed] [Google Scholar]
- 88.Carlton VE, Hu X, Chokkalingam AP, et al. PTPN22 genetic variation: evidence for multiple variants associated with rheumatoid arthritis. Am J Hum Genet. 2005;77(4):567–581. doi: 10.1086/468189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Michou L, Lasbleiz S, Rat AC, et al. Linkage proof for PTPN22, a rheumatoid arthritis susceptibility gene and a human autoimmunity gene. Proc Natl Acad Sci USA. 2007;104(5):1649–1654. doi: 10.1073/pnas.0610250104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Zoledziewska M, Perra C, Orru V, et al. Further evidence of a primary, causal association of the PTPN22 620 W variant with type 1 diabetes. Diabetes. 2008;57(1):229–234. doi: 10.2337/db07-0289. [DOI] [PubMed] [Google Scholar]
- 91.Smyth DJ, Cooper JD, Howson JM, et al. PTPN22 Trp620 explains the association of chromosome 1p13 with type 1 diabetes and shows a statistical interaction with HLA class II genotypes. Diabetes. 2008;57(6):1730–1737. doi: 10.2337/db07-1131. [DOI] [PubMed] [Google Scholar]
- 92.Orru V, Tsai SJ, Rueda B, et al. A loss-of-function variant of PTPN22 is associated with reduced risk of systemic lupus erythematosus. Hum Mol Genet. 2009;18(3):569–579. doi: 10.1093/hmg/ddn363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Kawasaki E, Awata T, Ikegami H, et al. Systematic search for single nucleotide polymorphisms in a lymphoid tyrosine phosphatase gene (PTPN22): association between a promoter polymorphism and type 1 diabetes in Asian populations. Am J Med Genet. 2006;140(6):586–593. doi: 10.1002/ajmg.a.31124. [DOI] [PubMed] [Google Scholar]
- 94.Cinek O, Hradsky O, Ahmedov G, et al. No independent role of the −1123 G>C and +2740 A>G variants in the association of PTPN22 with type 1 diabetes and juvenile idiopathic arthritis in two Caucasian populations. Diabetes Res Clin Pract. 2007;76(2):297–303. doi: 10.1016/j.diabres.2006.09.009. [DOI] [PubMed] [Google Scholar]
- 95.Steck AK, Liu SY, McFann K, et al. Association of the PTPN22/LYP gene with type 1 diabetes. Pediatr Diabetes. 2006;7(5):274–278. doi: 10.1111/j.1399-5448.2006.00202.x. [DOI] [PubMed] [Google Scholar]
- 96.Morgan AW, Thomson W, Martin SG, et al. Reevaluation of the interaction between HLA-DRB1 shared epitope alleles, PTPN22, and smoking in determining susceptibility to autoantibody-positive and autoantibody-negative rheumatoid arthritis in a large UK Caucasian population. Arthritis Rheum. 2009;60(9):2565–2576. doi: 10.1002/art.24752. [DOI] [PubMed] [Google Scholar]
- 97.Costenbader KH, Chang SC, De Vivo I, Plenge R, Karlson EW. Genetic polymorphisms in PTPN22, PADI-4, and CTLA-4 and risk for rheumatoid arthritis in two longitudinal cohort studies: evidence of gene-environment interactions with heavy cigarette smoking. Arthritis Res Ther. 2008;10(3):R52. doi: 10.1186/ar2421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Mahdi H, Fisher BA, Kallberg H, et al. Specific interaction between genotype, smoking and autoimmunity to citrullinated alpha-enolase in the etiology of rheumatoid arthritis. Nat Genet. 2009;41(12):1319–1324. doi: 10.1038/ng.480. [DOI] [PubMed] [Google Scholar]
- 99.Lempainen J, Vaarala O, Makela M, et al. Interplay between PTPN22 C1858T polymorphism and cow’s milk formula exposure in type 1 diabetes. J Autoimmun. 2009;33(2):155–164. doi: 10.1016/j.jaut.2009.04.003. [DOI] [PubMed] [Google Scholar]
- 100.Pierer M, Kaltenhauser S, Arnold S, et al. Association of PTPN22 1858 single-nucleotide polymorphism with rheumatoid arthritis in a German cohort: higher frequency of the risk allele in male compared to female patients. Arthritis Res Ther. 2006;8(3):R75. doi: 10.1186/ar1945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Lie BA, Viken MK, Odegard S, et al. Associations between the PTPN22 1858C->T polymorphism and radiographic joint destruction in patients with rheumatoid arthritis: results from a 10-year longitudinal study. Ann Rheum Dis. 2007;66(12):1604–1609. doi: 10.1136/ard.2006.067892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Hermann R, Lipponen K, Kiviniemi M, et al. Lymphoid tyrosine phosphatase (LYP/PTPN22) Arg620Trp variant regulates insulin autoimmunity and progression to type 1 diabetes. Diabetologia. 2006;49(6):1198–1208. doi: 10.1007/s00125-006-0225-4. [DOI] [PubMed] [Google Scholar]
- 103.Butty V, Campbell C, Mathis D, Benoist C. Impact of diabetes susceptibility loci on progression from pre-diabetes to diabetes in at-risk individuals of the diabetes prevention trialtype 1 (DPT-1) Diabetes. 2008;57(9):2348–2359. doi: 10.2337/db07-1736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Steck AK, Baschal EE, Jasinski JM, Boehm BO, Bottini N, Concannon P, Julier C, Morahan G, Noble JA, Polychronakos C, She JX, Eisenbarth GS. Type I Diabetes Genetics Consortium (2009) Rs2476601 T allele (R620W) defines high-risk PTPN22 type I diabetesassociated haplotypes with preliminary evidence for an additional protective haplotype. Genes Immun Suppl. 1:S21–S26. doi: 10.1038/gene.2009.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Aarnisalo J, Treszl A, Svec P, et al. Reduced CD4(+)T cell activation in children with type 1 diabetes carrying the PTPN22/Lyp 620Trp variant. J Autoimmun. 2008;31(1):13–21. doi: 10.1016/j.jaut.2008.01.001. [DOI] [PubMed] [Google Scholar]
- 106.Lefvert AK, Zhao Y, Ramanujam R, et al. PTPN22 R620W promotes production of anti-AChR autoantibodies and IL-2 in myasthenia gravis. J Neuroimmunol. 2008;197(2):110–113. doi: 10.1016/j.jneuroim.2008.04.004. [DOI] [PubMed] [Google Scholar]
- 107.Nielsen C, Barington T, Husby S, Lillevang ST. Expression of human PTPN22 alleles. Genes Immun. 2007;8(2):131–137. doi: 10.1038/sj.gene.6364369. [DOI] [PubMed] [Google Scholar]
- 108.Zhang J, Salojin K, Delovitch TL. Sequestration of CD4-associated Lck from the TCR complex may elicit T cell hyporesponsiveness in nonobese diabetic mice. J Immunol. 1998;160(3):1148–1157. [PubMed] [Google Scholar]
- 109.Buchs AE, Rapoport MJ. T cell signaling and autoimmune diabetes. J Pediatr Endocrinol Metab. 2000;13(9):1549–1554. doi: 10.1515/jpem.2000.13.9.1549. [DOI] [PubMed] [Google Scholar]
- 110.Sakaguchi N, Takahashi T, Hata H, et al. Altered thymic T-cell selection due to a mutation of the ZAP-70 gene causes autoimmune arthritis in mice. Nature. 2003;426(6965):454–460. doi: 10.1038/nature02119. [DOI] [PubMed] [Google Scholar]
- 111.Siggs OM, Miosge LA, Yates AL, 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(6):912–926. doi: 10.1016/j.immuni.2007.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Hsu LY, Tan YX, Xiao Z, Malissen M, Weiss A. A hypomorphic allele of ZAP-70 reveals a distinct thymic threshold for autoimmune disease versus autoimmune reactivity. J Exp Med. 2009;206(11):2527–2541. doi: 10.1084/jem.20082902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Marson A, Kretschmer K, Frampton GM, et al. Foxp3 occupancy and regulation of key target genes during T-cell stimulation. Nature. 2007;445(7130):931–935. doi: 10.1038/nature05478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Setoguchi R, Hori S, Takahashi T, Sakaguchi S. Homeostatic maintenance of natural Foxp3(+) CD25(+) CD4 (+) regulatory T cells by interleukin (IL)-2 and induction of autoimmune disease by IL-2 neutralization. J Exp Med. 2005;201(5):723–735. doi: 10.1084/jem.20041982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Yamanouchi J, Rainbow D, Serra P, et al. Interleukin-2 gene variation impairs regulatory T cell function and causes autoimmunity. Nat Genet. 2007;39(3):329–337. doi: 10.1038/ng1958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Tang Q, Adams JY, Penaranda C, et al. Central role of defective interleukin-2 production in the triggering of islet autoimmune destruction. Immunity. 2008;28(5):687–697. doi: 10.1016/j.immuni.2008.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Xie Y, Liu Y, Gong G, et al. Discovery of a novel submicromolar inhibitor of the lymphoid specific tyrosine phosphatase. Bioorg Med Chem Lett. 2008;18(9):2840–2844. doi: 10.1016/j.bmcl.2008.03.079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Wu S, Bottini M, Rickert RC, Mustelin T, Tautz L. In silico screening for PTPN22 inhibitors: active hits from an inactive phosphatase conformation. ChemMedChem. 2009;4(3):440–444. doi: 10.1002/cmdc.200800375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Sfar I, Gorgi Y, Aouadi H, et al. The PTPN22 C1858T (R620W) functional polymorphism in kidney transplantation. Transplant Proc. 2009;41(2):657–659. doi: 10.1016/j.transproceed.2009.01.029. [DOI] [PubMed] [Google Scholar]
- 120.Chatenoud L, Thervet E, Primo J, Bach JF. Anti-CD3 antibody induces long-term remission of overt autoimmunity in nonobese diabetic mice. Proc Natl Acad Sci USA. 1994;91(1):123–127. doi: 10.1073/pnas.91.1.123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Chatenoud L. CD3-specifc antibodies as promising tools to aim at immune tolerance in the clinic. Int Rev Immunol. 2006;25(3–4):215–233. doi: 10.1080/08830180600743032. [DOI] [PubMed] [Google Scholar]
- 122.Hermiston ML, Zikherman J, Tan AL, et al. Differential impact of the CD45 juxtamembrane wedge on central and peripheral T cell receptor responses. Proc Natl Acad Sci USA. 2009;106(2):546–551. doi: 10.1073/pnas.0811647106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Hermiston ML, Tan AL, Gupta VA, Majeti R, Weiss A. The juxtamembrane wedge negatively regulates CD45 function in B cells. Immunity. 2005;23(6):635–647. doi: 10.1016/j.immuni.2005.11.001. [DOI] [PubMed] [Google Scholar]
- 124.Gupta VA, Hermiston ML, Cassafer G, Daikh DI, Weiss A. B cells drive lymphocyte activation and expansion in mice with the CD45 wedge mutation and Fas deficiency. J Exp Med. 2008;205(12):2755–2761. doi: 10.1084/jem.20081204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Liossis SN, Kovacs B, Dennis G, Kammer GM, Tsokos GC. B cells from patients with systemic lupus erythematosus display abnormal antigen receptor-mediated early signal transduction events. J Clin Invest. 1996;98(11):2549–2557. doi: 10.1172/JCI119073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Kammer GM, Perl A, Richardson BC, Tsokos GC. Abnormal T cell signal transduction in systemic lupus erythematosus. Arthritis Rheum. 2002;46(5):1139–1154. doi: 10.1002/art.10192. [DOI] [PubMed] [Google Scholar]
- 127.Khan IU, Tsokos GC, Kammer GM. Abnormal B cell signal transduction in systemic lupus erythematosus. Curr Dir Autoimmun. 2003;6:89–104. doi: 10.1159/000066857. [DOI] [PubMed] [Google Scholar]
