Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Allergy Clin Immunol ; 153(1): 230-242, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37769878

RESUMEN

BACKGROUND: Pathogenic variants of phospholipase C gamma 2 (PLCG2) cause 2 related forms of autosomal-dominant immune dysregulation (ID), PLCγ2-associated antibody deficiency and immune dysregulation (PLAID) and autoinflammatory PLAID (APLAID). Since describing these conditions, many PLCG2 variants of uncertain significance have been identified by clinical sequencing of patients with diverse features of ID. OBJECTIVE: We sought to functionally classify PLCG2 variants and explore known and novel genotype-function-phenotype relationships. METHODS: Clinical data from patients with PLCG2 variants were obtained via standardized questionnaire. PLCG2 variants were generated by mutagenesis of enhanced green fluorescent protein (EGFP)-PLCG2 plasmid, which was overexpressed in Plcg2-deficient DT-40 B cells. B-cell receptor-induced calcium flux and extracellular signal-regulated kinase phosphorylation were assayed by flow cytometry. In some cases, stimulation-induced calcium flux was also measured in primary patient cells. RESULTS: Three-fourths of PLCG2 variants produced functional alteration of B-cell activation, in vitro. Thirteen variants led to gain of function (GOF); however, most functional variants defined a new class of PLCG2 mutation, monoallelic loss of function (LOF). Susceptibility to infection and autoinflammation were common with both GOF and LOF variants, whereas a new phenotypic cluster consisting of humoral immune deficiency, autoinflammation, susceptibility to herpesvirus infection, and natural killer cell dysfunction was observed in association with multiple heterozygous LOF variants detected in both familial and sporadic cases. In some cases, PLCG2 variants produced greater effects in natural killer cells than in B cells. CONCLUSIONS: This work expands the genotypic and phenotypic associations with functional variation in PLCG2, including a novel form of ID in carriers of heterozygous loss of PLCG2 function. It also demonstrates the need for more diverse assays for assessing the impact of PLCG2 variants on human disease.


Asunto(s)
Síndromes de Inmunodeficiencia , Fosfolipasa C gamma , Humanos , Enfermedades Autoinmunes , Calcio/metabolismo , Síndromes de Inmunodeficiencia/genética , Mutación , Fosfolipasa C gamma/genética
2.
Cells ; 11(12)2022 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-35741037

RESUMEN

Signaling through the TNF-family receptor Fas/CD95 can trigger apoptosis or non-apoptotic cellular responses and is essential for protection from autoimmunity. Receptor clustering has been observed following interaction with Fas ligand (FasL), but the stoichiometry of Fas, particularly when triggered by membrane-bound FasL, the only form of FasL competent at inducing programmed cell death, is not known. Here we used super-resolution microscopy to study the behavior of single molecules of Fas/CD95 on the plasma membrane after interaction of Fas with FasL on planar lipid bilayers. We observed rapid formation of Fas protein superclusters containing more than 20 receptors after interactions with membrane-bound FasL. Fluorescence correlation imaging demonstrated recruitment of FADD dependent on an intact Fas death domain, with lipid raft association playing a secondary role. Flow-cytometric FRET analysis confirmed these results, and also showed that some Fas clustering can occur in the absence of FADD and caspase-8. Point mutations in the Fas death domain associated with autoimmune lymphoproliferative syndrome (ALPS) completely disrupted Fas reorganization and FADD recruitment, confirming structure-based predictions of the critical role that these residues play in Fas-Fas and Fas-FADD interactions. Finally, we showed that induction of apoptosis correlated with the ability to form superclusters and recruit FADD.


Asunto(s)
Apoptosis , Receptor fas , Apoptosis/fisiología , Análisis por Conglomerados , Proteína Ligando Fas/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptor fas/metabolismo
3.
J Clin Invest ; 129(4): 1551-1565, 2019 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-30694219

RESUMEN

Across clinical trials, T cell expansion and persistence following adoptive cell transfer (ACT) have correlated with superior patient outcomes. Herein, we undertook a pan-cancer analysis to identify actionable ligand-receptor pairs capable of compromising T cell durability following ACT. We discovered that FASLG, the gene encoding the apoptosis-inducing ligand FasL, is overexpressed within the majority of human tumor microenvironments (TMEs). Further, we uncovered that Fas, the receptor for FasL, is highly expressed on patient-derived T cells used for clinical ACT. We hypothesized that a cognate Fas-FasL interaction within the TME might limit both T cell persistence and antitumor efficacy. We discovered that genetic engineering of Fas variants impaired in the ability to bind FADD functioned as dominant negative receptors (DNRs), preventing FasL-induced apoptosis in Fas-competent T cells. T cells coengineered with a Fas DNR and either a T cell receptor or chimeric antigen receptor exhibited enhanced persistence following ACT, resulting in superior antitumor efficacy against established solid and hematologic cancers. Despite increased longevity, Fas DNR-engineered T cells did not undergo aberrant expansion or mediate autoimmunity. Thus, T cell-intrinsic disruption of Fas signaling through genetic engineering represents a potentially universal strategy to enhance ACT efficacy across a broad range of human malignancies.


Asunto(s)
Traslado Adoptivo , Ingeniería Genética , Neoplasias Experimentales/terapia , Receptores Quiméricos de Antígenos , Transducción de Señal/inmunología , Microambiente Tumoral/inmunología , Animales , Proteína Ligando Fas/genética , Proteína Ligando Fas/inmunología , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/uso terapéutico , Transducción de Señal/genética , Microambiente Tumoral/genética , Receptor fas/genética , Receptor fas/inmunología
4.
Trends Mol Med ; 24(7): 642-653, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29880309

RESUMEN

Originally discovered as an inducer of apoptosis, the TNF-family receptor Fas (CD95, APO-1, TNFRSF6) has more recently been found to have functions beyond cell death, including T cell co-stimulation and promoting terminal differentiation of CD4+ and CD8+ T cells. Other TNF family members also discovered as apoptosis inducers, such as TRAIL (APO-2L, TNFSF10), can promote inflammation through caspase-8. Surprisingly, non-apoptotic signaling through Fas can protect from the autoimmunity seen in Fas deficiency independently from the cell death inducing functions of the receptor. Non-apoptotic Fas signaling can induce tumor cell growth and migration, and impair the efficacy of T cell adoptive immunotherapy. Blocking of non-apoptotic functions of these receptors may be a novel strategy to regulate autoimmunity and inflammation, and enhance antitumor immunity.


Asunto(s)
Autoinmunidad/fisiología , Muerte Celular/fisiología , Citocinas/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis/fisiología , Proliferación Celular/fisiología , Humanos , Inmunoterapia/métodos , Neoplasias/inmunología
5.
J Allergy Clin Immunol ; 139(4): 1293-1301.e4, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27609654

RESUMEN

BACKGROUND: The B-cell receptor transmembrane activator and calcium modulator ligand interactor (TACI) is important for T-independent antibody responses. One in 200 blood donors are heterozygous for the TACI A181E mutation. OBJECTIVE: We sought to investigate the effect on B-cell function of TACI A181E heterozygosity in reportedly healthy subjects and of the corresponding TACI A144E mutation in mice. METHODS: Nuclear factor κB (NF-κB) activation was measured by using the luciferase assay in 293T cells cotransfected with wild-type and mutant TACI. TACI-driven proliferation, isotype switching, and antibody responses were measured in B cells from heterozygous TACI A144E knock-in mice. Mouse mortality was monitored after intranasal pneumococcal challenge. RESULTS: Levels of natural antibodies to the pneumococcal polysaccharide component phosphocholine were significantly lower in A181E-heterozygous than TACI-sufficient Swedish blood donors never immunized with pneumococcal antigens. Although overexpressed hTACI A181E and mTACI A144E acted as dominant-negative mutations in transfectants, homozygosity for A144E in mice resulted in absent TACI expression in B cells, indicating that the mutant protein is unstable when naturally expressed. A144E heterozygous mice, such as TACI+/- mice, expressed half the normal level of TACI on their B cells and exhibited similar defects in a proliferation-inducing ligand-driven B-cell activation, antibody responses to TNP-Ficoll, production of natural antibodies to phosphocholine, and survival after intranasal pneumococcal challenge. CONCLUSION: These results suggest that TACI A181E heterozygosity results in TACI haploinsufficiency with increased susceptibility to pneumococcal infection. This has important implications for asymptomatic TACI A181E carriers.


Asunto(s)
Neumonía Neumocócica/genética , Proteína Activadora Transmembrana y Interactiva del CAML/genética , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Técnicas de Sustitución del Gen , Células HEK293 , Haploinsuficiencia , Heterocigoto , Humanos , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Neumonía Neumocócica/inmunología , Reacción en Cadena de la Polimerasa , Proteína Activadora Transmembrana y Interactiva del CAML/inmunología
6.
Nat Commun ; 7: 13895, 2016 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-28008916

RESUMEN

Mutations affecting the apoptosis-inducing function of the Fas/CD95 TNF-family receptor result in autoimmune and lymphoproliferative disease. However, Fas can also costimulate T-cell activation and promote tumour cell growth and metastasis. Palmitoylation at a membrane proximal cysteine residue enables Fas to localize to lipid raft microdomains and induce apoptosis in cell lines. Here, we show that a palmitoylation-defective Fas C194V mutant is defective in inducing apoptosis in primary mouse T cells, B cells and dendritic cells, while retaining the ability to enhance naive T-cell differentiation. Despite inability to efficiently induce cell death, the Fas C194V receptor prevents the lymphoaccumulation and autoimmunity that develops in Fas-deficient mice. These findings indicate that induction of apoptosis through Fas is dependent on receptor palmitoylation in primary immune cells, and Fas may prevent autoimmunity by mechanisms other than inducing apoptosis.


Asunto(s)
Apoptosis/inmunología , Autoinmunidad/inmunología , Microdominios de Membrana/inmunología , Ratones Transgénicos , Receptor fas/inmunología , Animales , Apoptosis/genética , Autoinmunidad/genética , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Células HEK293 , Humanos , Lipoilación/inmunología , Microdominios de Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Linfocitos T/inmunología , Linfocitos T/metabolismo , Receptor fas/genética , Receptor fas/metabolismo
7.
Mol Cell ; 64(2): 236-250, 2016 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-27746017

RESUMEN

Caspase-8 activation can be triggered by death receptor-mediated formation of the death-inducing signaling complex (DISC) and by the inflammasome adaptor ASC. Caspase-8 assembles with FADD at the DISC and with ASC at the inflammasome through its tandem death effector domain (tDED), which is regulated by the tDED-containing cellular inhibitor cFLIP and the viral inhibitor MC159. Here we present the caspase-8 tDED filament structure determined by cryoelectron microscopy. Extensive assembly interfaces not predicted by the previously proposed linear DED chain model were uncovered, and were further confirmed by structure-based mutagenesis in filament formation in vitro and Fas-induced apoptosis and ASC-mediated caspase-8 recruitment in cells. Structurally, the two DEDs in caspase-8 use quasi-equivalent contacts to enable assembly. Using the tDED filament structure as a template, structural analyses reveal the interaction surfaces between FADD and caspase-8 and the distinct mechanisms of regulation by cFLIP and MC159 through comingling and capping, respectively.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/química , Caspasa 8/química , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/química , Proteína de Dominio de Muerte Asociada a Fas/química , Proteínas Virales/química , Secuencia de Aminoácidos , Apoptosis/efectos de los fármacos , Sitios de Unión , Proteínas Adaptadoras de Señalización CARD , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Caspasa 8/genética , Caspasa 8/metabolismo , Microscopía por Crioelectrón , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/genética , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Dominio Efector de Muerte , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Expresión Génica , Humanos , Células Jurkat , Plásmidos/química , Plásmidos/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transfección , Proteínas Virales/genética , Proteínas Virales/metabolismo , Receptor fas/farmacología
8.
Mol Cell ; 61(4): 602-613, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26853147

RESUMEN

Fas (CD95, Apo-1, or TNFRSF6) is a prototypical apoptosis-inducing death receptor in the tumor necrosis factor receptor (TNFR) superfamily. While the extracellular domains of TNFRs form trimeric complexes with their ligands and the intracellular domains engage in higher-order oligomerization, the role of the transmembrane (TM) domains is unknown. We determined the NMR structures of mouse and human Fas TM domains in bicelles that mimic lipid bilayers. Surprisingly, these domains use proline motifs to create optimal packing in homotrimer assembly distinct from classical trimeric coiled-coils in solution. Cancer-associated and structure-based mutations in Fas TM disrupt trimerization in vitro and reduce apoptosis induction in vivo, indicating the essential role of intramembrane trimerization in receptor activity. Our data suggest that the structures represent the signaling-active conformation of Fas TM, which appears to be different from the pre-ligand conformation. Analysis of other TNFR sequences suggests proline-containing sequences as common motifs for receptor TM trimerization.


Asunto(s)
Membrana Dobles de Lípidos/metabolismo , Prolina/metabolismo , Receptor fas/química , Receptor fas/metabolismo , Animales , Apoptosis , Células HEK293 , Células HeLa , Humanos , Imagen por Resonancia Magnética , Ratones , Modelos Moleculares , Mutación , Multimerización de Proteína , Estructura Terciaria de Proteína , Transducción de Señal , Receptor fas/genética
9.
J Clin Invest ; 126(1): 318-34, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26657860

RESUMEN

Adoptive cell transfer (ACT) of purified naive, stem cell memory, and central memory T cell subsets results in superior persistence and antitumor immunity compared with ACT of populations containing more-differentiated effector memory and effector T cells. Despite a clear advantage of the less-differentiated populations, the majority of ACT trials utilize unfractionated T cell subsets. Here, we have challenged the notion that the mere presence of less-differentiated T cells in starting populations used to generate therapeutic T cells is sufficient to convey their desirable attributes. Using both mouse and human cells, we identified a T cell-T cell interaction whereby antigen-experienced subsets directly promote the phenotypic, functional, and metabolic differentiation of naive T cells. This process led to the loss of less-differentiated T cell subsets and resulted in impaired cellular persistence and tumor regression in mouse models following ACT. The T memory-induced conversion of naive T cells was mediated by a nonapoptotic Fas signal, resulting in Akt-driven cellular differentiation. Thus, induction of Fas signaling enhanced T cell differentiation and impaired antitumor immunity, while Fas signaling blockade preserved the antitumor efficacy of naive cells within mixed populations. These findings reveal that T cell subsets can synchronize their differentiation state in a process similar to quorum sensing in unicellular organisms and suggest that disruption of this quorum-like behavior among T cells has potential to enhance T cell-based immunotherapies.


Asunto(s)
Memoria Inmunológica , Inmunoterapia Adoptiva , Linfocitos T/inmunología , Animales , Diferenciación Celular , Proteína Ligando Fas/fisiología , Femenino , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/fisiología , Linfocitos T/citología , Receptor fas/fisiología
11.
J Immunol ; 194(8): 3567-82, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25786692

RESUMEN

The TNF family cytokine TL1A (Tnfsf15) costimulates T cells and type 2 innate lymphocytes (ILC2) through its receptor DR3 (Tnfrsf25). DR3-deficient mice have reduced T cell accumulation at the site of inflammation and reduced ILC2-dependent immune responses in a number of models of autoimmune and allergic diseases. In allergic lung disease models, immunopathology and local Th2 and ILC2 accumulation is reduced in DR3-deficient mice despite normal systemic priming of Th2 responses and generation of T cells secreting IL-13 and IL-4, prompting the question of whether TL1A promotes the development of other T cell subsets that secrete cytokines to drive allergic disease. In this study, we find that TL1A potently promotes generation of murine T cells producing IL-9 (Th9) by signaling through DR3 in a cell-intrinsic manner. TL1A enhances Th9 differentiation through an IL-2 and STAT5-dependent mechanism, unlike the TNF-family member OX40, which promotes Th9 through IL-4 and STAT6. Th9 differentiated in the presence of TL1A are more pathogenic, and endogenous TL1A signaling through DR3 on T cells is required for maximal pathology and IL-9 production in allergic lung inflammation. Taken together, these data identify TL1A-DR3 interactions as a novel pathway that promotes Th9 differentiation and pathogenicity. TL1A may be a potential therapeutic target in diseases dependent on IL-9.


Asunto(s)
Asma/inmunología , Diferenciación Celular/inmunología , Interleucina-9/inmunología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Animales , Asma/genética , Asma/patología , Diferenciación Celular/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Interleucina-9/genética , Ratones , Ratones Noqueados , Miembro 25 de Receptores de Factores de Necrosis Tumoral/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T Colaboradores-Inductores/patología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
12.
Nat Struct Mol Biol ; 17(11): 1324-9, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20935634

RESUMEN

The death-inducing signaling complex (DISC) formed by the death receptor Fas, the adaptor protein FADD and caspase-8 mediates the extrinsic apoptotic program. Mutations in Fas that disrupt the DISC cause autoimmune lymphoproliferative syndrome (ALPS). Here we show that the Fas-FADD death domain (DD) complex forms an asymmetric oligomeric structure composed of 5-7 Fas DD and 5 FADD DD, whose interfaces harbor ALPS-associated mutations. Structure-based mutations disrupt the Fas-FADD interaction in vitro and in living cells; the severity of a mutation correlates with the number of occurrences of a particular interaction in the structure. The highly oligomeric structure explains the requirement for hexameric or membrane-bound FasL in Fas signaling. It also predicts strong dominant negative effects from Fas mutations, which are confirmed by signaling assays. The structure optimally positions the FADD death effector domain (DED) to interact with the caspase-8 DED for caspase recruitment and higher-order aggregation.


Asunto(s)
Proteína de Dominio de Muerte Asociada a Fas/química , Mutación , Receptor fas/química , Secuencia de Aminoácidos , Animales , Apoptosis/fisiología , Síndrome Linfoproliferativo Autoinmune/genética , Caspasa 8/química , Línea Celular , Proteína de Dominio de Muerte Asociada a Fas/genética , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Alineación de Secuencia , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem , Receptor fas/genética
13.
Proc Natl Acad Sci U S A ; 107(21): 9801-6, 2010 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-20457915

RESUMEN

TNF, acting through p55 tumor necrosis factor receptor 1 (TNFR1), contributes to the pathogenesis of many inflammatory diseases. TNFR-associated periodic syndrome (TRAPS, OMIM 142680) is an autosomal dominant autoinflammatory disorder characterized by prolonged attacks of fevers, peritonitis, and soft tissue inflammation. TRAPS is caused by missense mutations in the extracellular domain of TNFR1 that affect receptor folding and trafficking. These mutations lead to loss of normal function rather than gain of function, and thus the pathogenesis of TRAPS is an enigma. Here we show that mutant TNFR1 accumulates intracellularly in peripheral blood mononuclear cells of TRAPS patients and in multiple cell types from two independent lines of knockin mice harboring TRAPS-associated TNFR1 mutations. Mutant TNFR1 did not function as a surface receptor for TNF but rather enhanced activation of MAPKs and secretion of proinflammatory cytokines upon stimulation with LPS. Enhanced inflammation depended on autocrine TNF secretion and WT TNFR1 in mouse and human myeloid cells but not in fibroblasts. Heterozygous TNFR1-mutant mice were hypersensitive to LPS-induced septic shock, whereas homozygous TNFR1-mutant mice resembled TNFR1-deficient mice and were resistant to septic shock. Thus WT and mutant TNFR1 act in concert from distinct cellular locations to potentiate inflammation in TRAPS. These findings establish a mechanism of pathogenesis in autosomal dominant diseases where full expression of the disease phenotype depends on functional cooperation between WT and mutant proteins and also may explain partial responses of TRAPS patients to TNF blockade.


Asunto(s)
Enfermedades Autoinflamatorias Hereditarias/inmunología , Enfermedades Autoinflamatorias Hereditarias/metabolismo , Mutación , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Animales , Enfermedades Autoinflamatorias Hereditarias/genética , Humanos , Lipopolisacáridos/inmunología , MAP Quinasa Quinasa 4/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Blood ; 114(11): 2254-62, 2009 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-19605846

RESUMEN

TNFRSF13B, which encodes TACI (transmembrane activator and calcium-modulator and cyclophilin ligand interactor), is mutated in 10% of patients with common variable immune deficiency (CVID). One of the 2 most common TACI mutations in CVID, A181E, introduces a negative charge into the transmembrane domain. To define the consequence of the A181E mutation on TACI function, we studied the effect of its murine equivalent, mTACI A144E, on TACI signaling in transfected cells and on TACI function in transgenic mice. The mTACI A144E mutant, like its human TACI A181E counterpart, was expressed on the surface of 293T transfectants and was able to bind ligand, but exhibited impaired constitutive and ligand-induced NF kappaB signaling. In addition, constitutive and ligand-induced clustering of the intracellular domain was deficient for A144E as measured by fluorescence resonance energy transfer. Transgenic mice expressing the A144E mutant on TACI(-/-) background had low serum IgA levels and significantly impaired antibody responses to the type II T-independent antigen TNP-Ficoll. B cells from A144E transgenic mice were impaired in their capacity to proliferate and secrete IgG1 and IgA in response to TACI ligation. These results suggest that mTACI A144E mutation and its human counterpart, A181E, disrupt TACI signaling and impair TACI-dependent B-cell functions.


Asunto(s)
Linfocitos B/inmunología , Inmunodeficiencia Variable Común/inmunología , Mutación Missense , Transducción de Señal/inmunología , Proteína Activadora Transmembrana y Interactiva del CAML/inmunología , Sustitución de Aminoácidos , Animales , Inmunodeficiencia Variable Común/genética , Inmunodeficiencia Variable Común/metabolismo , Ficoll/análogos & derivados , Ficoll/farmacología , Transferencia Resonante de Energía de Fluorescencia , Humanos , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/inmunología , FN-kappa B/metabolismo , Estructura Terciaria de Proteína/genética , Transducción de Señal/genética , Proteína Activadora Transmembrana y Interactiva del CAML/genética , Proteína Activadora Transmembrana y Interactiva del CAML/metabolismo , Trinitrobencenos/farmacología
15.
Results Probl Cell Differ ; 49: 17-47, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19132321

RESUMEN

Interactions between the TNF-family receptor Fas (CD95) and Fas Ligand (FasL, CD178) can efficiently induce apoptosis and are critical for the maintenance of immunological self-tolerance. FasL is kept under strict control by transcriptional and posttranslational regulation. Surface FasL can be cleaved by metalloproteases, resulting in shed extracellular domains, and FasL can also traffic to secretory lysosomes. Each form of FasL has distinct biological functions. Fas is more ubiquitously expressed, but its apoptosis-inducing function is regulated by a number of mechanisms including submembrane localization, efficiency of receptor signaling complex assembly and activation, and bcl-2 family members in some circumstances. When apoptosis is not induced, Fas-FasL interactions can also trigger a number of activating and proinflammatory signals. Harnessing the apoptosis-inducing potential of Fas for therapy of cancer and autoimmune disease has been actively pursued, and despite a number of unexpected side-effects that result from manipulating Fas-FasL interactions, this remains a worthy goal.


Asunto(s)
Apoptosis/inmunología , Proteína Ligando Fas/inmunología , Transducción de Señal/inmunología , Receptor fas/inmunología , Animales , Apoptosis/efectos de los fármacos , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/inmunología , Proteína Ligando Fas/biosíntesis , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptor fas/biosíntesis , Receptor fas/uso terapéutico
16.
J Immunol ; 179(10): 6384-8, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17982024

RESUMEN

In activated CD4(+) T cells, TCR restimulation triggers apoptosis that depends on interactions between the death receptor Fas and its ligand, FasL. This process, termed restimulation-induced cell death (RICD), is a mechanism of peripheral immune tolerance. TCR signaling sensitizes activated T cells to Fas-mediated apoptosis, but what pathways mediate this process are not known. In this study we identify the Rho GTPases Rac1 and Rac2 as essential components in restimulation-induced cell death. RNA interference-mediated knockdown of Rac GTPases greatly reduced Fas-dependent, TCR-induced apoptosis. The ability of Rac1 to sensitize T cells to Fas-induced apoptosis correlated with Rac-mediated cytoskeletal reorganization, dephosphorylation of the ERM (ezrin/radixin/moesin) family of cytoskeletal linker proteins, and the translocation of Fas to lipid raft microdomains. In primary activated CD4(+) T cells, Rac1 and Rac2 were independently required for maximal TCR-induced apoptosis. Activating Rac signaling may be a novel way to sensitize chronically stimulated lymphocytes to Fas-induced apoptosis, an important goal in the treatment of autoimmune diseases.


Asunto(s)
Apoptosis/inmunología , Linfocitos T CD4-Positivos/inmunología , Tolerancia Inmunológica , Activación de Linfocitos/inmunología , Receptor fas/inmunología , Proteínas de Unión al GTP rac/inmunología , Proteína de Unión al GTP rac1/inmunología , Enfermedades Autoinmunes/enzimología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/enzimología , Muerte Celular/inmunología , Proteínas del Citoesqueleto/inmunología , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/inmunología , Citoesqueleto/metabolismo , Activación Enzimática/inmunología , Proteína Ligando Fas , Humanos , Células Jurkat , Microdominios de Membrana/inmunología , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/inmunología , Proteínas de Microfilamentos/metabolismo , Transporte de Proteínas/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/inmunología , Receptor fas/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína RCA2 de Unión a GTP
17.
J Biol Chem ; 281(3): 1489-94, 2006 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-16303761

RESUMEN

Hepatocyte growth factor (HGF) is a plasminogen-like protein with an alpha chain linked to a trypsin-like beta chain without peptidase activity. The interaction of HGF with c-met, a receptor tyrosine kinase expressed by many cells, is important in cell growth, migration, and formation of endothelial and epithelial tubes. Stimulation of c-met requires two-chain, disulfide-linked HGF. Portions of an alpha chain containing an N-terminal segment and four kringle domains (NK4) antagonize HGF activity. Until now, no physiological pathway for generating NK4 was known. Here we show that chymases, which are chymotryptic peptidases secreted by mast cells, hydrolyze HGF, thereby abolishing scatter factor activity while generating an NK4-like antagonist of HGF scatter factor activity. Thus, chymase interferes with HGF directly by destroying active protein and indirectly by generating an antagonist. The site of hydrolysis, Leu480, lies in the alpha chain on the N-terminal side of the cysteine linking the alpha and beta chains. This site appears to be specific for HGF because chymase does not hydrolyze other plasminogen-like proteins, such as macrophage-stimulating protein and plasminogen itself. Mast cell/neutrophil cathepsin G and neutrophil elastase generate similar fragments of HGF by cleaving near the chymase site. Mast cell and neutrophil peptidases are secreted during tissue injury, infection, ischemia, and allergic inflammation, where they may oppose HGF effects on epithelial repair. Thus, HGF possesses an "inactivation segment" that serves as an Achilles' heel attacked by inflammatory proteases. This work reveals a potential physiological pathway for inactivation of HGF and generation of NK4-like antagonists.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Mastocitos/enzimología , Neutrófilos/enzimología , Péptido Hidrolasas/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Factor de Crecimiento de Hepatocito/química , Humanos , Hidrólisis , Cinética , Datos de Secuencia Molecular , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido
18.
J Pathol ; 206(3): 279-90, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15887294

RESUMEN

Diverse interstitial lung diseases (ILD) demonstrate mesenchymal infiltration by an abundance of activated mast cells whose role in parenchymal fibrogenesis remains unclear. Since mast cells differentiate in a dynamic, tissue-specific manner via signals transduced by c-Kit receptor, we examined the effect of ILD microenvironments on c-Kit expression and metalloproteinase phenotypes of mesenchymal mast cell populations. Immunohistochemical and flow cytometric analyses characterized surface expression of c-Kit on mast cells in tissues obtained from patients with idiopathic pulmonary fibrosis, systemic sclerosis, sarcoidosis, and lymphangioleiomyomatosis, thus identifying a unique immunophenotype not shared by normal lung mast cells. Isolation of c-Kit+/FcepsilonRI+/CD34- mast cells via immunocytometric sorting of heterogeneous cell populations from mechanically disaggregated lung tissues permitted analysis of gene expression patterns by two-step real-time polymerase chain reaction. Transcriptional profiling identified expression of c-Kit and the neutral serine proteases, tryptase and chymase, establishing the identity of sorted populations as mature mast cells. Mast cells harvested from ILD tissues demonstrated characteristic metalloproteinase phenotypes which included expression of matrix metalloproteinase (MMP)-1 and a disintegrin and metalloproteinase (ADAM)-9, -10, and -17. Immunohistochemical co-localization guided by gene profiling data confirmed expression of chymase, MMP-1, and ADAM-17 protein in subpopulations of mast cells in remodelling interstitium. Gene profiling of harvested mast cells also showed increased transcript copy numbers for TNFalpha and CC chemokine receptor 2, which play critical roles in lung injury. We conclude that ILD microenvironments induce unique c-Kit receptor and metalloproteinase mast cell phenotypes.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Enfermedades Pulmonares Intersticiales/genética , Mastocitos/química , Metaloproteasas/genética , Proteínas Proto-Oncogénicas c-kit/análisis , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Línea Celular , Citocinas/análisis , Citometría de Flujo/métodos , Humanos , Inmunohistoquímica/métodos , Inmunofenotipificación/métodos , Pulmón/inmunología , Pulmón/patología , Enfermedades Pulmonares Intersticiales/inmunología , Enfermedades Pulmonares Intersticiales/patología , Linfangioleiomiomatosis/genética , Linfangioleiomiomatosis/inmunología , Linfangioleiomiomatosis/patología , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Sarcoidosis/genética , Sarcoidosis/inmunología , Sarcoidosis/patología , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/inmunología , Esclerodermia Sistémica/patología , Transducción de Señal/genética , Transducción de Señal/inmunología , Transcripción Genética/genética , Transcripción Genética/inmunología
19.
Am J Respir Cell Mol Biol ; 31(2): 154-61, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15059785

RESUMEN

Activated mast cells release stored and newly synthesized mediators that influence the caliber and responsiveness of inflamed airways. In this work, we show that alloimmune-mediated mechanisms induce mast cell activation and expression of CC chemokines in remodeling rat tracheal allografts. Decreased expression of rat mast cell protease (RMCP) I and II, in concert with tryptase release in tracheal allografts, identified degranulation of stored serine proteases as an early mast cell response to allotransplantation. Transient upregulation of c-Kit expression occurred in a synchronous manner, suggesting that c-Kit receptor signaling controls mast cell responses. Increased expression of CC chemokine ligand (CCL) 2 and CCL3 by RMCP I-positive cells identified mast cells as epithelial and mesenchymal sources of chemoattractant chemokines in allograft airways. Cyclosporin A immunosuppression both attenuated and delayed these changes in mast cell phenotypes. Incubation of rat basophil leukemia 2H3 cells with CCL2 or CCL3 decreased surface c-Kit expression, an effect blocked by protease inhibitors. By controlling surface receptor availability, CC chemokines may regulate c-Kit signaling via a novel proteolytic mechanism. These data suggest that targeting alloimmune responses and restoring quiescence of mast cells may attenuate the development of fibroproliferative and obstructive distortions of bronchiolar architecture in lung allografts.


Asunto(s)
Quimiocinas CC/inmunología , Mastocitos/inmunología , Tráquea/inmunología , Animales , Degranulación de la Célula , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Masculino , Ratas , Ratas Endogámicas Lew , Especificidad de la Especie
20.
J Biol Chem ; 279(7): 5612-20, 2004 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-14625290

RESUMEN

Transmembrane metalloproteinases of the disintegrin and metalloproteinase (ADAM) family control cell signaling interactions via hydrolysis of protein extracellular domains. Prior work has shown that the receptor tyrosine kinase, c-Kit (CD117), is essential for mast cell survival and that serum levels of c-Kit increase in proliferative mast cell disorders, suggesting the existence of c-Kit shedding pathways in mast cells. In the present work, we report that tumor necrosis factor alpha-converting enzyme (TACE; ADAM-17) mediates shedding of c-Kit. Stimulation of transfected cells with phorbol 12-myristate 13-acetate (PMA) induced metalloproteinase-mediated release of c-Kit ectodomain, which increased further upon TACE overexpression. By contrast, TACE-deficient fibroblasts did not demonstrate inducible release, thus identifying TACE as the metalloproteinase primarily responsible for PMA-induced c-Kit shedding. Surface expression of c-Kit by the human mast cell-1 line decreased upon phorbol-induced shedding, which involved metalloproteinase activity susceptible to inhibition by tissue inhibitor of metalloproteinase (TIMP)-3. To further explore the role of TACE in shedding of c-Kit from mast cells, we compared the behavior of mast cells derived from murine embryonic stem cells. In these studies, PMA decreased surface c-Kit levels on mast cells expressing wild-type (+/+) TACE but not on those expressing an inactive mutant (DeltaZn/DeltaZn), confirming the role of TACE in PMA-induced c-Kit shedding. Compared with TACE(+/+) cells, TACE(DeltaZn/DeltaZn) mast cells also demonstrated decreased constitutive shedding and increased basal surface expression of c-Kit, with diminished apoptosis in response to c-Kit ligand deprivation. These data suggest that TACE controls mast cell survival by regulating shedding and surface expression of c-Kit.


Asunto(s)
Embrión de Mamíferos/citología , Mastocitos/metabolismo , Metaloendopeptidasas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Células Madre/metabolismo , Proteínas ADAM , Proteína ADAM17 , Animales , Apoptosis , Diferenciación Celular , División Celular , Línea Celular , Separación Celular , Supervivencia Celular , Citoplasma/metabolismo , Detergentes/farmacología , Relación Dosis-Respuesta a Droga , Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Immunoblotting , Ligandos , Ratones , Ratones Transgénicos , Mutagénesis Sitio-Dirigida , Octoxinol , Polietilenglicoles/farmacología , Pruebas de Precipitina , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Factor de Células Madre/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA