392 resultados para TRANSGENIC SQUASH
Resumo:
The superantigen (SAg) expressed by mouse mammary tumor virus (MMTV) has been shown to play an essential role in the course of the viral life cycle. In the present study, we describe a V beta 4-specific SAg encoded by a new exogenous MMTV carried by the SIM mouse strain. This is the first report of a viral or bacterial SAg reacting with mouse V beta 4+ T cells. Injection of MMTV(SIM) into adult BALB/c mice leads to a rapid and strong stimulation of V beta 4+ CD4+ T cells, followed by a slow deletion of these cells. Neonatal exposure to the virus also leads to a progressive deletion of V beta 4+ T cells. In contrast to other strong MMTV SAg, this new SAg requires the presence of major histocompatibility complex class II I-E molecules to be presented efficiently to T cells. Sequence analysis revealed a new predicted amino acid sequence in the C-terminal polymorphic region of this SAg. Furthermore, sequence comparisons to the most closely related SAg with different V beta specificities hint at the specific residues involved in the interaction with the T cell receptor.
Resumo:
T-cell receptor affinity for self-antigen has an important role in establishing self-tolerance. Three transgenic mouse strains expressing antigens of variable affinity for the OVA transgenic-I T-cell receptor were generated to address how TCR affinity affects the efficiency of negative selection, the ability to prime an autoimmune response, and the elimination of the relevant target cell. Mice expressing antigens with an affinity just above the negative selection threshold exhibited the highest risk of developing experimental autoimmune diabetes. The data demonstrate that close to the affinity threshold for negative selection, sufficient numbers of self-reactive T cells escape deletion and create an increased risk for the development of autoimmunity.
Resumo:
CD8(+) CTLs play a critical role in antitumor immunity. However, vaccination with synthetic peptide containing CTL epitopes has not been generally effective in inducing protective antitumor immunity. In this study, we addressed the detailed mechanism(s) involved in this failure using a new tumor model of BALB/c transplanted tumors expressing NY-ESO-1, an extensively studied human cancer/testis Ag. Whereas peptide immunization with an H2-D(d)-restricted CTL epitope derived from NY-ESO-1 (NY-ESO-1 p81-88) induced NY-ESO-1(81-88)-specific CD8(+) T cells in draining lymph nodes and spleens, tumor growth was significantly enhanced. Single-cell analysis of specific CD8(+) T cells revealed that peptide immunization caused apoptosis of >80% of NY-ESO-1(81-88)-specific CD8(+) T cells at tumor sites and repetitive immunization further diminished the number of specific CD8(+) T cells. This phenomenon was associated with elevated surface expression of Fas and programmed death-1. When peptide vaccination was combined with an adjuvant, a TLR9 ligand CpG, the elevated Fas and programmed death-1 expression and apoptosis induction were not observed, and vaccine with peptide and CpG was associated with strong tumor growth inhibition. Selection of appropriate adjuvants is essential for development of effective cancer vaccines, with protection of effector T cells from peptide vaccine-induced apoptosis being a prime objective.
Resumo:
Central and peripheral tolerance prevent autoimmunity by deleting the most aggressive CD8(+) T cells but they spare cells that react weakly to tissue-restricted antigen (TRA). To reveal the functional characteristics of these spared cells, we generated a transgenic mouse expressing the TCR of a TRA-specific T cell that had escaped negative selection. Interestingly, the isolated TCR matches the affinity/avidity threshold for negatively selecting T cells, and when developing transgenic cells are exposed to their TRA in the thymus, only a fraction of them are eliminated but significant numbers enter the periphery. In contrast to high avidity cells, low avidity T cells persist in the antigen-positive periphery with no signs of anergy, unresponsiveness, or prior activation. Upon activation during an infection they cause autoimmunity and form memory cells. Unexpectedly, peptide ligands that are weaker in stimulating the transgenic T cells than the thymic threshold ligand also induce profound activation in the periphery. Thus, the peripheral T cell activation threshold during an infection is below that of negative selection for TRA. These results demonstrate the existence of a level of self-reactivity to TRA to which the thymus confers no protection and illustrate that organ damage can occur without genetic predisposition to autoimmunity.
Resumo:
The caspase 8 inhibitor c-FLIP(L) can act in vitro as a molecular switch between cell death and growth signals transmitted by the death receptor Fas (CD95). To elucidate its function in vivo, transgenic mice were generated that overexpress c-FLIP(L) in the T-cell compartment (c-FLIP(L) Tg mice). As anticipated, FasL-induced apoptosis was inhibited in T cells from the c-FLIP(L) Tg mice. In contrast, activation-induced cell death of T cells in c-FLIP(L) Tg mice was unaffected, suggesting that this deletion process can proceed in the absence of active caspase 8. Accordingly, c-FLIP(L) Tg mice differed from Fas-deficient mice by showing no accumulation of B220(+) CD4(-) CD8(-) T cells. However, stimulation of T lymphocytes with suboptimal doses of anti-CD3 or antigen revealed increased proliferative responses in T cells from c-FLIP(L) Tg mice. Thus, a major role of c-FLIP(L) in vivo is the modulation of T-cell proliferation by decreasing the T-cell receptor signaling threshold.
Resumo:
Fas-deficient mice (Fas(lpr/lpr)) and humans have profoundly dysregulated T lymphocyte homeostasis, which manifests as an accumulation of CD4(+) and CD8(+) T cells as well as an unusual population of CD4(-)CD8(-)TCRαβ(+) T cells. To date, no unifying model has explained both the increased T-cell numbers and the origin of the CD4(-)CD8(-)TCRαβ(+) T cells. As Fas(lpr/lpr) mice raised in a germ-free environment still manifest lymphadenopathy, we considered that this process is primarily driven by recurrent low-avidity TCR signaling in response to self-peptide/MHC as occurs during homeostatic proliferation. In these studies, we developed two independent systems to decrease the number of self-peptide/MHC contacts. First, expression of MHC class I was reduced in OT-I TCR transgenic mice. Although OT-I Fas(lpr/lpr) mice did not develop lymphadenopathy characteristic of Fas(lpr/lpr) mice, in the absence of MHC class I, OT-I Fas(lpr/lpr) T cells accumulated as both CD8(+) and CD4(-)CD8(-) T cells. In the second system, re-expression of β(2)m limited to thymic cortical epithelial cells of Fas(lpr/lpr) β(2)m-deficient mice yielded a model in which polyclonal CD8(+) thymocytes entered a peripheral environment devoid of MHC class I. These mice accumulated significantly greater numbers of CD4(-)CD8(-)TCRαβ(+) T cells than conventional Fas(lpr/lpr) mice. Thus, Fas shapes the peripheral T-cell repertoire by regulating the survival of a subset of T cells proliferating in response to limited self-peptide/MHC contacts.
Resumo:
RESUME DESTINE AUX NON SCIENTIFIQUESLe diabète est une maladie associée à un excès de glucose (sucre) dans le sang. Le taux de glucose sanguin augmente lorsque l'action d'une hormone, l'insuline, responsable du transport du glucose du sang vers les tissus de l'organisme diminue, ou lorsque les quantités d'insuline à disposition sont inadéquates.L'une des causes communes entre les deux grands types de diabète connus, le type 1 et le type 2, est la disparition des cellules beta du pancréas, spécialisées dans la sécrétion d'insuline, par mort cellulaire programmée aussi appelée apoptose. Alors que dans le diabète de type 1, la destruction des cellules beta est causée par notre propre système immunitaire, dans le diabète de type 2, la mort de ces cellules, est principalement causée par des concentrations élevées de graisses saturés ou de molécules impliquées dans l'inflammation que l'on rencontre en quantités augmentées chez les personnes obèses. Etant donné l'augmentation épidémique du nombre de personnes obèses de par le monde, on estime que le nombre de personnes diabétiques (dont une majorité sont des diabétiques de type 2), va passer de 171 million en l'an 2000, à 366 million en l'an 2030, expliquant la nécessité absolue de mettre au point de nouvelles stratégies thérapeutique pour combattre cette maladie.L'apoptose est un processus complexe dont la dérégulation induit de nombreuses affections allant du cancer jusqu'au diabète. L'activation de caspase 3, une protéine clé contrôlant la mort cellulaire, était connue pour systématiquement mener à la mort cellulaire programmée. Ces dernières années, notre laboratoire a décrit des mécanismes de survie qui sont activés par caspase 3 et qui expliquent sans doute pourquoi son activation ne mène pas systématiquement à la mort cellulaire. Lorsqu'elle est faiblement activée, caspase 3 clive une autre protéine appelée RasGAP en deux protéines plus courtes dont l'une, appelée le fragment Ν a la particularité de protéger les cellules contre l'apoptose.Durant ma thèse, j'ai été impliqué dans divers projets destinés à mieux comprendre comment le fragment Ν protégeait les cellules contre l'apoptose et à savoir s'il pouvait être utilisé comme outil thérapeutique dans les conditions de survenue d'un diabète expérimental. C'est dans ce but que nous avons créé une souris transgénique, appelée RIP-N, exprimant le fragment Ν spécifiquement dans les cellules beta. Comme attendu, les cellules beta de ces souris étaient plus résistantes à la mort induite par des composés connus pour induire le diabète, comme certaines molécules induisant l'inflammation ou les graisses saturées. Nous avons ensuite pu montrer que les souris RIP-N étaient plus résistantes à la survenue d'un diabète expérimental que ce soit par l'injection d'une drogue induisant l'apoptose des cellules beta, que ce soit dans un fond génétique caractérisé par une attaque spontanée des cellules beta par le système immunitaire ou dans le contexte d'un diabète de type 2 induit par l'obésité. Dans plusieurs des modèles animaux étudiés, nous avons pu montrer que le fragment Ν protégeait les cellules en activant une voie protectrice bien connue impliquant successivement les protéines Ras, PI3K et Akt ainsi qu'en bloquant la capacité d'Akt d'activer le facteur NFKB, connu pour être délétère pour la survie de la cellule beta. La capacité qu'a le fragment Ν d'activer Akt tout en prévenant l'activation de NFKB par Akt est par conséquent particulièrement intéressante dans l'intégration des signaux régulant la mort cellulaire dans le contexte de la survenue d'un diabète.La perspective d'utiliser le fragment Ν comme outil thérapeutique dépendra de notre capacité à activer les signaux protecteurs induits par le fragment Ν depuis l'extérieur de la cellule ou de dériver des peptides perméables aux cellules possédant les propriétés du fragment N.2 SUMMARYDiabetes mellitus is an illness associated with excess blood glucose. Blood glucose levels raise when the action of insulin decreases or when insulin is provided in inappropriate amounts. In type 1 diabetes (T1D) as well as in type 2 diabetes (T2D), the insulin secreting beta cells in the pancreas undergo controlled cell death also called apoptosis. Whereas in T1D, beta cells are killed by the immune system, in T2D, they are killed by several factors, among which are increased blood glucose levels, increased levels of harmful lipids or pro-inflammatory cytokines that are released by the dysfunctional fat tissue of obese people. Given the epidemic increase in the number of obese people throughout the world, the number of diabetic people (a majority of which are type 2 diabetes) is estimated to rise from 171 million affected people in the year 2000 to 366 million in 2030 explaining the absolute requirement for new therapies to fight the disease.Apoptosis is a very complex process whose deregulation leads to a wide range of diseases going from cancer to diabetes. Caspase 3 although known as a key molecule controlling apoptosis, has been shown to have various other functions. In the past few years, our laboratory has described a survival mechanism, that takes place at low caspase activity and that might explain how cells that activate their caspases for reasons other than apoptosis survive. In such conditions, caspase 3 cleaves another protein called RasGAP into two shorter proteins, one of which, called fragment N, protects cells from apoptosis.We decided to check whether fragment Ν could be used as a therapeutical tool in the context of diabetes inducing conditions. We thus derived a transgenic mouse line, called RIP-N, in which the expression of fragment Ν is restricted to beta cells. As expected, the beta cells of these mice were more resistant ex-vivo to cell death induced by diabetes inducing factors. We then showed that the RIP-N transgenic mice were resistant to streptozotocin induced diabetes, a mouse model mimicking type 1 diabetes, which correlated to fewer number of apoptotic beta cells in the pancreas of the transgenic mice compared to their controls. The RIP-N transgene also delayed overt diabetes development in the NOD background, a mouse model of autoimmune type 1 diabetes, and delayed the occurrence of obesity induced hyperglycemia in a mouse model of type 2-like diabetes. Interestingly, fragment Ν was mediating its protection by activating the protective Akt kinase, and by blocking the detrimental NFKB factor. Our future ability to activate the protective signals elicited by fragment Ν from the outside of cells or to derive cell permeable peptides bearing the protective properties of fragment Ν might condition our ability to use this protein as a therapeutic tool.3 RESUMELe diabète est une maladie associée à un excès de glucose plasmatique. La glycémie augmente lorsque l'action de l'insuline diminue ou lorsque les quantités d'insuline à disposition sont inadéquates. Dans le diabète de type 1 (D1) comme dans le diabète de type 2 (D2), les cellules beta du pancréas subissent la mort cellulaire programmée aussi appelée apoptose. Alors que dans le D1 les cellules beta sont tuées par le système immunitaire, dans le D2 elles sont tuées par divers facteurs parmi lesquels on trouve des concentrations élevées de glucose, d'acides gras saturés ou de cytokines pro-inflammatoires qui sont sécrétées en concentrations augmentées par le tissu adipeux dysfonctionnel des personnes obèses. Etant donné l'augmentation épidémique du nombre de personnes obèses de par le monde, on estime que le nombre de personnes diabétiques (dont une majorité sont des diabétiques de type 2), va passer de 171 million en l'an 2000, à 366 million en l'an 2030, justifiant la nécessité absolue de mettre au point de nouvelles stratégies thérapeutique pour combattre cette maladie.L'apoptose est un processus complexe dont la dérégulation induit de nombreuses affections allant du cancer jusqu'au diabète. Caspase 3, bien que connue comme étant une protéine clé contrôlant l'apoptose a bien d'autres fonctions démontrées. Ces dernières années, notre laboratoire a décrit un mécanisme de survie qui est activé lorsque caspase 3 est faiblement activée et qui explique probablement comment des cellules qui ont activé leurs caspases pour une autre raison que l'apoptose peuvent survivre. Dans ces conditions, caspase 3 clive une autre protéine appelée RasGAP en deux protéines plus courtes dont l'une, appelée le fragment Ν a la particularité de protéger les cellules contre l'apoptose.Nous avons donc décidé de vérifier si le fragment Ν pouvait être utilisé comme outil thérapeutique dans les conditions de survenue d'un diabète expérimental. Pour se faire, nous avons créé une souris transgénique, appelée RIP-N, exprimant le fragment Ν spécifiquement dans les cellules beta. Comme attendu, les cellules beta de ces souris étaient plus résistantes ex-vivo à la mort induite par des facteurs pro-diabétogènes. Nous avons ensuite pu montrer que les souris RIP-N étaient plus résistantes à la survenue d'un diabète induit par la streptozotocine, un drogue mimant la survenue d'un D1 et que ceci était corrélée à une diminution du nombre de cellules en apoptose dans le pancréas des souris transgéniques comparé à leurs contrôles. L'expression du transgène a aussi eu pour effet de retarder la survenue d'un diabète franc dans le fond génétique NOD, un modèle génétique de diabète de type 1 auto-immun, ainsi que de retarder la survenue d'une hyperglycémie dans un modèle murin de diabète de type 2 induit par l'obésité. Dans plusieurs des modèles animaux étudiés, nous avons pu montrer que le fragment Ν protégeait les cellules en activant la kinase protectrice Akt ainsi qu'en bloquant le facteur délétère NFKB. La perspective d'utiliser le fragment Ν comme outil thérapeutique dépendra de notre capacité à activer les signaux protecteurs induits par le fragment Ν depuis l'extérieur de la cellule ou de dériver des peptides perméables aux cellules possédant les propriétés du fragment
Resumo:
Beta-catenin is a component of the intercalated disc in cardiomyocytes, but can also be involved in signalling and activation of gene transcription. We wanted to determine how long-term changes in beta-catenin expression levels would affect mature cardiomyocytes. Conditional transgenic mice that either lacked beta-catenin or that expressed a non-degradable form of beta-catenin in the adult ventricle were created. While mice lacking beta-catenin in the ventricle do not have an overt phenotype, mice expressing a non-degradable form develop dilated cardiomyopathy and do not survive beyond 5 months. A detailed analysis could reveal that this phenotype is correlated with a distinct localisation of beta-catenin in adult cardiomyocytes, which cannot be detected in the nucleus, no matter how much protein is present. Our report is the first study that addresses long-term effects of either the absence of beta-catenin or its stabilisation on ventricular cardiomyocytes and it suggests that beta-catenin's role in the nucleus may be of little significance in the healthy adult heart.
Resumo:
Corneal integrity and transparency are indispensable for good vision. Cornea homeostasis is entirely dependent upon corneal stem cells, which are required for complex wound-healing processes that restore corneal integrity following epithelial damage. Here, we found that leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) is highly expressed in the human holoclone-type corneal epithelial stem cell population and sporadically expressed in the basal cells of ocular-surface epithelium. In murine models, LRIG1 regulated corneal epithelial cell fate during wound repair. Deletion of Lrig1 resulted in impaired stem cell recruitment following injury and promoted a cell-fate switch from transparent epithelium to keratinized skin-like epidermis, which led to corneal blindness. In addition, we determined that LRIG1 is a negative regulator of the STAT3-dependent inflammatory pathway. Inhibition of STAT3 in corneas of Lrig1-/- mice rescued pathological phenotypes and prevented corneal opacity. Additionally, transgenic mice that expressed a constitutively active form of STAT3 in the corneal epithelium had abnormal features, including corneal plaques and neovascularization similar to that found in Lrig1-/- mice. Bone marrow chimera experiments indicated that LRIG1 also coordinates the function of bone marrow-derived inflammatory cells. Together, our data indicate that LRIG1 orchestrates corneal-tissue transparency and cell fate during repair, and identify LRIG1 as a key regulator of tissue homeostasis.
Resumo:
The identification of CTL-defined tumor-associated Ags has allowed the development of new strategies for cancer immunotherapy. To potentiate the CTL responses, peptide-based vaccines require the coadministration of adjuvants. Because oligodeoxynucleotides (ODN) containing CpG motifs are strong immunostimulators, we analyzed the ability of CpG ODN to act as adjuvant of the CTL response against tumor-derived synthetic peptide in the absence or presence of IFA. Mice transgenic for a chimeric MHC class I molecule were immunized with a peptide analog of MART-1/Melan-A(26-35) in the presence of CpG ODN alone or CpG ODN emulsified in IFA. The CTL response was monitored ex vivo by tetramer staining of lymphocytes. In blood, spleen, and lymph nodes, peptide mixed with CpG ODN alone was able to elicit a stronger systemic CTL response as compared with peptide emulsified in IFA. Moreover, CpG ODN in combination with IFA further enhanced the CTL response in terms of the frequency of tetramer+CD8+ T cells ex vivo. The CTL induced in vivo against peptide analog in the presence of CpG ODN are functional, as they were able to recognize and kill melanoma cells in vitro. Overall, these results indicate that CpG ODN by itself is a good candidate adjuvant of CTL response and can also enhance the effect of classical adjuvant.
Resumo:
Phytic acid (PA) is poorly digested by humans and monogastric animals and negatively affects human/animal nutrition and the environment. Rice mutants with reduced PA content have been developed but are often associated with reduced seed weight and viability, lacking breeding value. In the present study, a new approach was explored to reduce seed PA while attaining competitive yield. The OsMRP5 gene, of which mutations are known to reduce seed PA as well as seed yield and viability, was down-regulated specifically in rice seeds by using an artificial microRNA driven by the rice seed specific promoter Ole18. Seed PA contents were reduced by 35.8-71.9% in brown rice grains of transgenic plants compared to their respective null plants (non-transgenic plants derived from the same event). No consistent significant differences of plant height or number of tillers per plant were observed, but significantly lower seed weights (up to 17.8% reduction) were detected in all transgenic lines compared to null plants, accompanied by reductions of seed germination and seedling emergence. It was observed that the silencing of the OsMRP5 gene increased the inorganic P (Pi) levels (up to 7.5 times) in amounts more than the reduction of PA-P in brown rice. This indicates a reduction in P content in other cellular compounds, such as lipids and nucleic acids, which may affect overall seed development. Put together, the present study demonstrated that seed specific silencing of OsMRP5 could significantly reduce the PA content and increase Pi levels in seeds; however, it also significantly lowers seed weight in rice. Discussions were made regarding future directions towards producing agronomically competitive and nutritionally valuable low PA rice.
Resumo:
Cone photoreceptors mediate visual acuity under daylight conditions, so loss of cone-mediated central vision of course dramatically affects the quality of life of patients suffering from retinal degeneration. Therefore, promoting cone survival has become the goal of many ocular therapies and defining the stage of degeneration that still allows cell rescue is of prime importance. Using the Rpe65(R91W/R91W) mouse, which carries a mutation in the Rpe65 gene leading to progressive photoreceptor degeneration in both patients and mice, we defined stages of retinal degeneration that still allow cone rescue. We evaluated the therapeutic window within which cones can be rescued, using a subretinal injection of a lentiviral vector driving expression of RPE65 in the Rpe65(R91W/R91W) mice. Surprisingly, when applied to adult mice (1 month) this treatment not only stalls or slows cone degeneration but, actually, induces cone-specific protein expression that was previously absent. Before the intervention only part of the cones (40% of the number found in wild-type animals) in the Rpe65(R91W/R91W) mice expressed cone transducin (GNAT2); this fraction increased to 64% after treatment. Correct S-opsin localization is also recovered in the transduced region. In consequence these results represent an extended therapeutic window compared to the Rpe65(-/-) mice, implying that patients suffering from missense mutations might also benefit from a prolonged therapeutic window. Moreover, cones are not only rescued during the course of the degeneration, but can actually recover their initial status, meaning that a proportion of altered cones in chromophore deficiency-related disease can be rehabilitated even though they are severely affected.
Resumo:
Proteins secreted from adipose tissue are increasingly recognized to play an important role in the regulation of glucose metabolism. However, much less is known about their effect on lipid metabolism. The fasting-induced adipose factor (FIAF/angiopoietin-like protein 4/peroxisome proliferator-activated receptor gamma angiopoietin-related protein) was previously identified as a target of hypolipidemic fibrate drugs and insulin-sensitizing thiazolidinediones. Using transgenic mice that mildly overexpress FIAF in peripheral tissues we show that FIAF is an extremely powerful regulator of lipid metabolism and adiposity. FIAF overexpression caused a 50% reduction in adipose tissue weight, partly by stimulating fatty acid oxidation and uncoupling in fat. In addition, FIAF overexpression increased plasma levels of triglycerides, free fatty acids, glycerol, total cholesterol, and high density lipoprotein (HDL)-cholesterol. Functional tests indicated that FIAF overexpression severely impaired plasma triglyceride clearance but had no effect on very low density lipoprotein production. The effects of FIAF overexpression were amplified by a high fat diet, resulting in markedly elevated plasma and liver triglycerides, plasma free fatty acids, and plasma glycerol levels, and impaired glucose tolerance in FIAF transgenic mice fed a high fat diet. Remarkably, in mice the full-length form of FIAF was physically associated with HDL, whereas truncated FIAF was associated with low density lipoprotein. In human both full-length and truncated FIAF were associated with HDL. The composite data suggest that via physical association with plasma lipoproteins, FIAF acts as a powerful signal from fat and other tissues to prevent fat storage and stimulate fat mobilization. Our data indicate that disturbances in FIAF signaling might be involved in dyslipidemia.
Resumo:
Peroxisome proliferator-activated receptor γ (PPARγ) is a nuclear receptor involved in diverse biological processes including adipocyte differentiation, glucose homeostasis, and inflammatory responses. Analyses of PPARγ knockout animals have been so far preempted by the early embryonic death of PPARγ-/- embryos as a consequence of the severe alteration of their placental vasculature. Using Sox2Cre/PPARγL2/L2 mice, we obtained fully viable PPARγ-null mice through specific and total epiblastic gene deletion, thereby demonstrating that the placental defect is the unique cause of PPARγ-/- embryonic lethality. The vasculature defects observed in PPARγ-/- placentas at embryonic d 9.5 correlated with an unsettled balance of pro- and antiangiogenic factors as demonstrated by increased levels of proliferin (Prl2c2, PLF) and decreased levels of proliferin-related protein (Prl7d1, PRP), respectively. To analyze the role of PPARγ in the later stage of placental development, when its expression peaks, we treated pregnant wild-type mice with the PPARγ agonist rosiglitazone. This treatment resulted in a disorganization of the placental layers and an altered placental microvasculature, accompanied by the decreased expression of proangiogenic genes such as Prl2c2, vascular endothelial growth factor, and Pecam1. Together our data demonstrate that PPARγ plays a pivotal role in controlling placental vascular proliferation and contributes to its termination in late pregnancy.
Resumo:
A C1858T (R620W) variation in the PTPN22 gene encoding the tyrosine phosphatase LYP is a major risk factor for human autoimmunity. LYP is a known negative regulator of signaling through the T cell receptor (TCR), and murine Ptpn22 plays a role in thymic selection. However, the mechanism of action of the R620W variant in autoimmunity remains unclear. One model holds that LYP-W620 is a gain-of-function phosphatase that causes alterations in thymic negative selection and/or thymic output of regulatory T cells (Treg) through inhibition of thymic TCR signaling. To test this model, we generated mice in which the human LYP-W620 variant or its phosphatase-inactive mutant are expressed in developing thymocytes under control of the proximal Lck promoter. We found that LYP-W620 expression results in diminished thymocyte TCR signaling, thus modeling a "gain-of-function" of LYP at the signaling level. However, LYP-W620 transgenic mice display no alterations of thymic negative selection and no anomalies in thymic output of CD4(+)Foxp3(+) Treg were detected in these mice. Lck promoter-directed expression of the human transgene also causes no alteration in thymic repertoire or increase in disease severity in a model of rheumatoid arthritis, which depends on skewed thymic selection of CD4(+) T cells. Our data suggest that a gain-of-function of LYP is unlikely to increase risk of autoimmunity through alterations of thymic selection and that LYP likely acts in the periphery perhaps selectively in regulatory T cells or in another cell type to increase risk of autoimmunity.