Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Autoimmun ; 137: 102946, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36402602

RESUMEN

BACKGROUND: Genetic aberrations in the NFκB pathway lead to primary immunodeficiencies with various degrees of severity. We previously demonstrated that complete ablation of the RelB transcription factor, a key component of the alternative pathway, results in an early manifested combined immunodeficiency requiring stem cell transplantation. OBJECTIVE: To study the molecular basis of a progressive severe autoimmunity and immunodeficiency in three patients. METHODS: Whole exome sequencing was performed to identify the genetic defect. Molecular and cellular techniques were utilized to assess the variant impact on NFκB signaling, canonical and alternative pathway crosstalk, as well as the resultant effects on immune function. RESULTS: Patients presented with multiple autoimmune progressive severe manifestations encompassing the liver, gut, lung, and skin, becoming debilitating in the second decade of life. This was accompanied by a deterioration of the immune system, demonstrating an age-related decline in naïve T cells and responses to mitogens, accompanied by a gradual loss of all circulating CD19+ cells. Whole exome sequencing identified a novel homozygous c. C1091T (P364L) transition in RELB. The P364L RelB protein was unstable, with extremely low expression, but retained some function and could be transiently and partially upregulated following Toll-like receptor stimulation. Stimulation of P364L patient fibroblasts resulted in a marked rise in a cluster of pro-inflammatory hyper-expressed transcripts consistent with the removal of RelB inhibitory effect on RelA function. This is likely the main driver of autoimmune manifestations in these patients. CONCLUSION: Incomplete loss of RelB provided a unique opportunity to gain insights into NFκB's pathway interactions as well as the pathogenesis of autoimmunity. The P364L RelB mutation leads to gradual decline in immune function with progression of severe debilitating autoimmunity.


Asunto(s)
Enfermedades Autoinmunes , Factor de Transcripción ReIB , Humanos , Factor de Transcripción ReIB/genética , Factor de Transcripción ReIB/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Regulación de la Expresión Génica , Enfermedades Autoinmunes/genética
2.
Pediatr Allergy Immunol ; 33(1): e13694, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34738677

RESUMEN

BACKGROUND: STAT1 gain-of-function (GOF) is an immune dysregulatory disorder with poorly studied genotype-phenotype correlation, impeding prognostication and early intervention. Given previous mechanistic studies, as well as anecdotal clinical reports, we sought to systematically determine whether DNA-binding domain (DBD) mutations in STAT1 result in a different phenotype than mutations in other gene domains. METHODS: Negative prognostic features previously identified by the International STAT1 GOF Study Group (invasive infections, intracranial aneurysms, and malignancy), as well as other clinical features and mortality, were compared within a cohort of 30 patients with STAT1 GOF diagnosed at our center, consisting of 9 patients with DBD mutations and 21 patients with non-DBD mutations. We subsequently re-analyzed mortality data from a large, previously-published 274-patient cohort by the International STAT1 GOF Study Group. RESULTS: While no differences were noted with respect to malignancy or symptomatic aneurysms, invasive /opportunistic infections were substantially more common among DBD patients, as were sinopulmonary infections, bronchiectasis, enteropathy, endocrinopathies, lymphoproliferative manifestations, and recurrent fevers/HLH. DBD patients also had a lower probability of survival and younger age of mortality compared with non-DBD patients. Our re-evaluation of the published data from the International STAT1 GOF Study Group revealed a similar finding of earlier mortality among patients harboring DBD mutations. CONCLUSION: We report that STAT1 GOF patients with DBD mutations may be regarded as a unique subgroup, impacted more by early-onset profound combined immunodeficiency and with earlier mortality. These findings may impact clinical decision making with respect to early intervention, and in particular hematopoietic stem cell transplant considerations, in such patients.


Asunto(s)
ADN , Mutación con Ganancia de Función , Estudios de Asociación Genética , Humanos , Mutación , Fenotipo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo
3.
J Allergy Clin Immunol ; 147(2): 727-733.e2, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32980423

RESUMEN

BACKGROUND: Genetic faults in several components of the nuclear factor-κB pathway cause immunodeficiency. Most defects lead to combined immunodeficiency with a range of severity. Heterozygous mutations in NFKB1 were associated with common variable immunodeficiency, however, homozygous mutations have not been described. OBJECTIVE: We studied the molecular basis of combined immunodeficiency in a patient who presented with failure to thrive, persistent EBV viremia and hepatitis, pneumocystis jirovecii pneumonitis, and generalized lymphadenopathy. METHODS: Whole genome and exome sequencing followed by Sanger confirmation were performed to identify the genetic defect. Molecular and cellular techniques were used to assess the variant impact on the nuclear factor-κB pathway and lymphocyte function. RESULTS: Genetic analysis revealed a novel homozygous mutation in NFKB1, c.2878G>A, p.Gly960Arg (G960R). This affected p105 phosphorylation and p50 formation on antigen and cytokine stimulation, as well as attenuating nuclear signal transmission. As a result, both T- and B-cell maturation and function were perturbed. The number of memory CD4+ T cells were reduced, while CD8+ T cells consisted predominately of expanded differentiated populations. The function of T cells were diminished as shown by reduced responses to mitogens as well as diminished cytokine secretion. B-cell maturation was also affected, with decreased IgD+CD27+ memory B cells while transitional B cells were increased, likely contributing to the reduced ability to produce specific antibodies. CONCLUSION: Homozygous G960R mutation in NFKB1 leads to a severe clinical presentation of combined immunodeficiency. This was associated with blockade of nuclear factor-κB pathway signaling, resulting in aberrations in T- and B-cell maturation and function.


Asunto(s)
Subunidad p50 de NF-kappa B/genética , Inmunodeficiencia Combinada Grave/genética , Homocigoto , Humanos , Lactante , Masculino , Mutación , Linaje
5.
J Allergy Clin Immunol ; 142(2): 618-629, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29180244

RESUMEN

BACKGROUND: We previously reported a novel syndrome characterized by combined immunodeficiency associated with severe developmental defects-subsequently known as Roifman-Chitayat syndrome (RCS; OMIM 613328). Linkage analysis identified 2 disease-associated loci. OBJECTIVES: We sought to identify the genetic defect in these patients and characterize their immunologic cellular abnormalities. METHODS: Genetic, immunologic, protein, and cellular functional analyses were used to identify and characterize patient genetic deficiencies and aberrant patient cell behavior. RESULTS: Deleterious variants were found at both loci identified by linkage analysis: a homozygous stop codon in PI3-kinase p110δ (PIK3CD) and a homozygous frame shift mutation in SKAP (KNSTRN), both ablating protein expression. Patients with RCS display aberrant B-cell development, similar to p110δ-deficient mice, but also aberrant T-cell spreading, cell-cell interaction, and migration. Patients also display significant developmental abnormalities not seen in p110δ knockouts (eg, optic nerve atrophy and skeletal anomalies) that we ascribe to loss of SKAP. Aberrant SKAP expression can prolong anaphase and this may contribute to developmental defects. However, we also identified microtubule-associated protein 4 microtubule-binding protein as a novel SKAP-binding partner and show that it undergoes relocalization in patient T cells, with associated areas of aberrant microtubule hyperstabilization, likely contributing not only to the altered properties of RCS lymphoid cells but also to developmental defects. CONCLUSIONS: The complex RCS presentation, with combined developmental and immunologic defects, is associated with a combined deficiency of 2 genes products, PI3-kinase p110δ and SKAP, both of which appear to play a significant role in the disease.


Asunto(s)
Linfocitos B/inmunología , Enfermedades del Desarrollo Óseo/genética , Linfocitos T CD4-Positivos/inmunología , Proteínas de Ciclo Celular/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Síndromes de Inmunodeficiencia/genética , Proteínas Asociadas a Microtúbulos/genética , Enfermedades del Nervio Óptico/genética , Eliminación de Secuencia/genética , Adolescente , Ciclo Celular/genética , Diferenciación Celular/genética , Línea Celular , Preescolar , Discapacidades del Desarrollo/genética , Facies , Femenino , Humanos , Inmunofenotipificación , Lactante , Linaje , Enfermedades de Inmunodeficiencia Primaria , Hermanos , Secuenciación Completa del Genoma , Adulto Joven
6.
J Allergy Clin Immunol ; 141(5): 1818-1830.e2, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28826773

RESUMEN

BACKGROUND: Combined immunodeficiency (CID) is a T-cell defect frequently presenting with recurrent infections, as well as associated immune dysregulation manifesting as autoimmunity or allergic inflammation. OBJECTIVE: We sought to identify the genetic aberration in 4 related patients with CID, early-onset asthma, eczema, and food allergies, as well as autoimmunity. METHODS: We performed whole-exome sequencing, followed by Sanger confirmation, assessment of the genetic variant effect on cell signaling, and evaluation of the resultant immune function. RESULTS: A heterozygous novel c.C88T 1-bp substitution resulting in amino acid change R30W in caspase activation and recruitment domain family member 11 (CARD11) was identified by using whole-exome sequencing and segregated perfectly to family members with severe atopy only but was not found in healthy subjects. We demonstrate that the R30W mutation results in loss of function while also exerting a dominant negative effect on wild-type CARD11. The CARD11 defect altered the classical nuclear factor κB pathway, resulting in poor in vitro T-cell responses to mitogens and antigens caused by reduced secretion of IFN-γ and IL-2. CONCLUSION: Unlike patients with biallelic mutations in CARD11 causing severe CID, the R30W defect results in a less profound yet prominent susceptibility to infections, as well as multiorgan atopy and autoimmunity.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/inmunología , Guanilato Ciclasa/genética , Guanilato Ciclasa/inmunología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Adulto , Proteínas Adaptadoras de Señalización CARD/deficiencia , Preescolar , Femenino , Guanilato Ciclasa/deficiencia , Humanos , Interferón gamma/genética , Interleucina-2/genética , Masculino , Mutación , FN-kappa B/genética , Estudios Prospectivos , Estudios Retrospectivos , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/inmunología , Secuenciación del Exoma/métodos
8.
J Autoimmun ; 65: 90-100, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26385063

RESUMEN

Multiple receptors that control cell growth and inflammation activate the NFκB pathway that comprises of two pathways. Dysfunction of the classical pathway leads to impaired adaptive and innate immunity in humans. In contrast the exact role of the alternative NFκB pathway mediated by RelB in humans remains largely elusive. We have recently identified deleterious mutations in RelB in patients with combined immunodeficiency and autoimmunity. We studied here the biological effects of RelB deficiency on the immune system. We show that the thymus in this patient is dysplastic and consequently new thymus emigrants are rare and there is an accumulation of CD45 RO(+) T cells with an increase in CD62L(+) central memory cells. The TCR repertoire of these cells appears skewed with selective clonal expansion. In vitro responses to T cell mitogens were markedly depressed and so were PHA induced IL2 and IFNγ production. In addition, the TH1 promoting T bet and STAT1 were reduced. In contrast, hyper-activation was seen in response to anti-CD3 and CD28. T cell dependent antibody responses were low to absent in all patients. We found that BAFF-R was reduced and CD40 signaling aberrant. Critically, CD27(+) memory cells were absent. We have shown here for the first time the role of RelB on lymphocyte development in humans. In the absence of RelB, B cells development is arrested, resulting in poor production of immunoglobulins and specific antibodies. T cell maturation in the thymus appears altered with reduced output and production of a skewed T cell repertoire with expansion of clones which are likely the cause of the autoimmune features observed in these patients.


Asunto(s)
Linfocitos B/fisiología , Subgrupos de Linfocitos T/fisiología , Timo/anomalías , Factor de Transcripción ReIB/inmunología , Autoinmunidad , Diferenciación Celular/genética , Proliferación Celular , Preescolar , Humanos , Lactante , Recién Nacido , Interleucina-2/inmunología , Masculino , Subunidad p50 de NF-kappa B/inmunología , Transducción de Señal/inmunología , Timo/inmunología , Timo/patología , Factor de Transcripción ReIB/genética
9.
Blood ; 122(24): 3940-50, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24159173

RESUMEN

B-cell responses are guided by the integration of signals through the B-cell receptor (BCR), CD40, and cytokine receptors. The common γ chain (γc)-binding cytokine interleukin (IL)-21 drives humoral immune responses via STAT3-dependent induction of transcription factors required for plasma cell generation. We investigated additional mechanisms by which IL-21/STAT3 signaling modulates human B-cell responses by studying patients with STAT3 mutations. IL-21 strongly induced CD25 (IL-2Rα) in normal, but not STAT3-deficient, CD40L-stimulated naïve B cells. Chromatin immunoprecipitation confirmed IL2RA as a direct target of STAT3. IL-21-induced CD25 expression was also impaired on B cells from patients with IL2RG or IL21R mutations, confirming a requirement for intact IL-21R signaling in this process. IL-2 increased plasmablast generation and immunoglobulin secretion from normal, but not CD25-deficient, naïve B cells stimulated with CD40L/IL-21. IL-2 and IL-21 were produced by T follicular helper cells, and neutralizing both cytokines abolished the B-cell helper capacity of these cells. Our results demonstrate that IL-21, via STAT3, sensitizes B cells to the stimulatory effects of IL-2. Thus, IL-2 may play an adjunctive role in IL-21-induced B-cell differentiation. Lack of this secondary effect of IL-21 may amplify the humoral immunodeficiency in patients with mutations in STAT3, IL2RG, or IL21R due to impaired responsiveness to IL-21.


Asunto(s)
Linfocitos B/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Interleucina-2/farmacología , Interleucinas/farmacología , Células Plasmáticas/efectos de los fármacos , Factor de Transcripción STAT3/genética , Linfocitos B/citología , Linfocitos B/metabolismo , Ligando de CD40/farmacología , Diferenciación Celular/genética , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Expresión Génica/efectos de los fármacos , Humanos , Interleucina-10/farmacología , Subunidad alfa del Receptor de Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Plasmáticas/citología , Células Plasmáticas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transcriptoma/efectos de los fármacos
10.
J Allergy Clin Immunol ; 133(3): 807-17, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24239102

RESUMEN

BACKGROUND: Mutations in the gene for the signal transducer and activator of transcription 1, STAT1, have been shown to be associated with death at an early age due to overwhelming viral infection (complete STAT1 deficiency) or, more commonly, selective deficiencies to mycobacterial or fungal infection (typically heterozygous STAT1 mutations). OBJECTIVES: To define the molecular basis of progressive combined immunodeficiency in a group of patients with fatal infections. METHODS: We studied a group of unrelated patients who displayed an unusual progressive form of combined immunodeficiency. Whole exome sequencing assisted in confirming a common genetic defect in this group, which consisted of a heterozygous mutation of the STAT1 gene. STAT1 protein level as well as function was assessed, and a detailed evaluation of the immune system, including analysis of thymus tissue, was performed. RESULTS: Patients were found to carry de novo heterozygous mutations in STAT1 encoding T385A, I294T, or C284R amino acid substitutions. STAT1 expression appeared significantly decreased as a result of these changes but not completely absent, with diminished signaling responses. This group display progressive loss in lymphocyte number and function accompanied by increasing autoimmune features as well as severe, fatal infections. CONCLUSIONS: These findings show that some heterozygous aberrations of STAT1 can be associated with progressive combined immunodeficiency, quite distinct from the limited susceptibilities to infection previously reported for heterozygous STAT1 mutations. These mutations were not inherited, rather, arose de novo in each case. Accompanied by significant patient mortality, this finding suggests that this class of STAT1 mutation is ultimately fatal due to overwhelming infection.


Asunto(s)
Síndromes de Inmunodeficiencia/genética , Mutación , Factor de Transcripción STAT1/genética , Adolescente , Niño , Preescolar , Femenino , Heterocigoto , Humanos , Síndromes de Inmunodeficiencia/mortalidad , Masculino , Factor de Transcripción STAT1/análisis , Factor de Transcripción STAT1/fisiología , Receptor fas/análisis
11.
Front Immunol ; 13: 801832, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35126392

RESUMEN

Background: STAT1 gain-of-function (GOF) is a primary immune dysregulatory disorder marked by wide infectious predisposition (most notably chronic mucocutaneous Candidiasis), autoimmunity, vascular disease and malignant predisposition. While atopic features have been described in some STAT1 GOF patients, they are not considered a predominant feature of the disease. Additionally, while eosinophilic gastrointestinal infiltration has been reported in some cases, this has always been described in the context of pre-existing oropharyngeal and/or esophageal Candidiasis. Clinical cases: Herein, we report 3 members of a multi-generational family diagnosed with STAT1 GOF caused by a novel mutation in the N-terminal domain, c.194A>C (p.D65A). The proband presented initially with a long-standing history of treatment-refractory eosinophilic esophagitis (EoE) without preceding gastrointestinal tract fungal infections, and her mother was diagnosed with esophagitis as well. Conclusion: EoE has been previously associated with alterations to STAT6 and STAT3 signaling pathways. The current report expands the possible association between JAK/STAT-related disorders and EoE, suggesting that EoE could be a primary disease manifestation of STAT1 GOF, even in the absence of oropharyngeal and/or esophageal Candidiasis.


Asunto(s)
Esofagitis Eosinofílica/diagnóstico , Esofagitis Eosinofílica/etiología , Mutación con Ganancia de Función , Factor de Transcripción STAT1/genética , Alelos , Biomarcadores , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Factor de Transcripción STAT1/metabolismo , Transducción de Señal
12.
J Allergy Clin Immunol ; 126(6): 1226-33.e1, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20864151

RESUMEN

BACKGROUND: ζ-associated protein, 70 kd (ZAP70), deficiency in human subjects results in a combined immunodeficiency characterized by normal numbers of circulating CD4 T cells and CD8 lymphocytopenia. Patients who live beyond infancy can also experience autoimmune manifestations. OBJECTIVES: We sought to further characterize the nature of the T-cell populations found in ZAP70-deficient patients and explored the mechanisms that might predispose them to autoimmunity. METHODS: T-cell development was assessed by examining T-cell receptor (TCR) gene rearrangements and thymopoiesis by measuring TCR exclusion circle levels. TCR repertoire on CD4 and CD8 T-cell populations was assessed by means of flow cytometry. T-cell gene expression patterns were examined by means of exonic microarray analysis and apoptotic responses by means of Annexin V binding and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling. RESULTS: Cells displaying recombination events from all stages of TCR gene rearrangement were present in the peripheral blood of ZAP70-deficient patients; however, the late TCRD-deleting rearrangement was significantly reduced. TCR exclusion circle levels were also found to be low. Surprisingly, all Vß families were detected in both CD4(+) and CD8(+) circulating T cells. Several Vß families were significantly overrepresented, which is reminiscent of autoimmune disorders. Levels of mRNA for cytotoxic T lymphocyte-associated antigen 4, TGF-ß, and IL-10 were found to be low, a signature of autoimmunity. Finally, Fas-mediated CD4 T-cell apoptosis was found to be reduced in vitro, and staining of thymus biopsy specimens revealed reduced thymocyte apoptosis. CONCLUSION: We show that in the absence of ZAP70, thymopoiesis is altered and differentiation to double-positive cells is hampered. Circulating T cells appear poorly regulated, do not differentiate into T(H)2 T cells, lack a number of inhibitory growth controls, and display reduced apoptosis, all predisposing patients to exaggerated inflammation and autoimmunity.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Timo/metabolismo , Proteína Tirosina Quinasa ZAP-70/metabolismo , Apoptosis/genética , Autoinmunidad , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Procesos de Crecimiento Celular/genética , Procesos de Crecimiento Celular/inmunología , Genes Codificadores de los Receptores de Linfocitos T/inmunología , Humanos , Inflamación , Linfopenia , Análisis por Micromatrices , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/patología , Inmunodeficiencia Combinada Grave/fisiopatología , Timo/inmunología , Timo/patología , Proteína Tirosina Quinasa ZAP-70/deficiencia , Proteína Tirosina Quinasa ZAP-70/genética , Proteína Tirosina Quinasa ZAP-70/inmunología
16.
Front Immunol ; 8: 576, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28603521

RESUMEN

With the advent of high-throughput genomic sequencing techniques, novel genetic etiologies are being uncovered for previously unexplained Mendelian phenotypes, and the underlying genetic architecture of disease is being unraveled. Although most of these "mendelizing" disease traits represent phenotypes caused by single-gene defects, a percentage of patients have blended phenotypes caused by pathogenic variants in multiple genes. We describe an adult patient with susceptibility to bacterial, herpesviral, and fungal infections. Immunologic defects included CD8+ T cell lymphopenia, decreased T cell proliferative responses to mitogens, hypogammaglobulinemia, and radiation sensitivity. Whole-exome sequencing revealed compound heterozygous variants in ZAP70. Biallelic mutations in ZAP70 are known to produce a spectrum of immune deficiency that includes the T cell abnormalities observed in this patient. Analyses for variants in genes associated with radiation sensitivity identified the presence of a homozygous RNF168 variant of unknown significance. RNF168 deficiency causes radiosensitivity, immunodeficiency, dysmorphic features, and learning difficulties syndrome and may account for the radiation sensitivity. Thus, the patient was found to have a novel blended phenotype associated with multilocus genomic variation: i.e., separate and distinct genetic defects. These findings further illustrate the clinical utility of applying genomic testing in patients with primary immunodeficiency diseases.

19.
Autoimmun Rev ; 11(5): 341-7, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22024499

RESUMEN

OBJECTIVE: Toll-like receptors, a major component of the innate immune system, play an important role in the initial response against pathogens. Genetic abnormalities in some receptors like TLR2, TLR3 and TLR4 have been associated with susceptibility to fungal and viral infections while other aberrations in TLR genes such as TLR3, TLR7 and TLR9 may predispose to autoimmunity. Recently we have shown an association of a TLR3 receptor variant, L412F, to susceptibility to chronic candidiasis, recurrent viral and bacterial infections and autoimmunity. We investigated here the biological implications of this TLR3 mutant. METHODS: To study the functional impact of the L412F variant of TLR3 we tested patients' peripheral blood mononuclear cells (PBMCs) as well as fibroblasts for secretion of cytokines in response to TLR3 ligand, candida or cytomegalovirus (CMV). In addition, the P2.1 cell line was used as a model for the TLR3 WT and L412F variant receptors function. RESULTS: Patient's cells carrying the L412F variant showed reduced IFNγ as well as TNFα secretion in response to stimulation with the TLR3 ligand, CMV or Candida albicans. Fibroblasts with the L412F variant showed decreased secretion of IFNλ in response to stimulation with both polyinosine ploycytidylic acid (Poly I:C) and CMV and P2.1 cells transfected with the L412F variant showed reduced secretion of IFN-ß in comparison to cells transfected with the wild type receptor. CONCLUSION: We have shown here aberrant biological responses mediated by the TLR3 variant receptor, L412F, which may explain in part susceptibility of patients to chronic candidiasis, viral infections and autoimmunity.


Asunto(s)
Autoinmunidad/genética , Candidiasis Cutánea/genética , Infecciones por Citomegalovirus/genética , Predisposición Genética a la Enfermedad , Mutación , Enfermedades Cutáneas Virales/genética , Receptor Toll-Like 3/genética , Anticuerpos Monoclonales/inmunología , Autoinmunidad/inmunología , Complejo CD3/inmunología , Candida albicans/inmunología , Candidiasis Cutánea/inmunología , Citocinas/inmunología , Citomegalovirus/inmunología , Infecciones por Citomegalovirus/inmunología , Células Dendríticas/inmunología , Fibroblastos/inmunología , Fibroblastos/metabolismo , Genotipo , Humanos , Ligandos , Activación de Linfocitos/inmunología , Linfocitos/inmunología , Poli I-C/inmunología , Enfermedades Cutáneas Virales/inmunología , Receptor Toll-Like 3/inmunología
20.
Eur J Immunol ; 32(12): 3745-55, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12516569

RESUMEN

Eph receptor tyrosine kinases and their ligands, the ephrins, are known to play an important role in regulating cell migration and targeting in neuronal and endothelial cells. Recently, it has been shown that lymphoid cells also express Eph receptors, raising the possibility that Eph receptors may similarly regulate lymphocyte migration. Chemotaxis in response to soluble chemokine factors is an essential facet of T cell biology. We demonstrate here that T cell chemotaxis in response to both the stromal cell-derived factor (SDF)-1alpha and macrophage inflammatory protein 3beta chemokines is modulated by costimulation with ephrins. Both ephrin-A and ephrin-B ligands were found to modify the chemotactic responses of a T cell line and primary T cells. Ephrin-A1, in particular, strongly inhibited chemotaxis. In accordance with the tyrosine kinase activity of EphA receptors, ephrin-A1 stimulation induced rapid intracellular tyrosine phosphorylation in T cells. Although strongly inhibiting chemotaxis, ephrin-A1 costimulus did not affect many of the signaling events downstream of the SDF-1alpha receptor CXCR4, including calcium flux and activation of MAPK. Rather, ephrin-A1 altered the balance of small G protein activity in T cells. Ephrin-A1 stimulation prevented SDF-1alpha-induced activation of cdc42, while simultaneously inducing rho activation. Ultimately, ephrin-A1 was found to inhibit chemokine-induced actin polymerization, thereby blocking migration. Ubiquitous ephrin expression in vivo creates enormous potential for T cells to encounter these ligands, suggesting that Eph receptors and ephrins may be important regulators of T cell migration.


Asunto(s)
Quimiotaxis de Leucocito/efectos de los fármacos , Efrina-A1/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Actinas/metabolismo , Quimiocina CCL19 , Quimiocina CXCL12 , Quimiocinas CC/farmacología , Quimiocinas CXC/farmacología , Niño , Humanos , Técnicas In Vitro , Células Jurkat , Fosforilación , Receptores CXCR4/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T/metabolismo , Tirosina/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA