Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Blood Adv ; 2(5): 517-528, 2018 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-29507075

RESUMO

Practical methods are needed to increase the applicability and efficacy of chimeric antigen receptor (CAR) T-cell therapies. Using donor-derived CAR-T cells is attractive, but expression of endogenous T-cell receptors (TCRs) carries the risk for graft-versus-host-disease (GVHD). To remove surface TCRαß, we combined an antibody-derived single-chain variable fragment specific for CD3ε with 21 different amino acid sequences predicted to retain it intracellularly. After transduction in T cells, several of these protein expression blockers (PEBLs) colocalized intracellularly with CD3ε, blocking surface CD3 and TCRαß expression. In 25 experiments, median TCRαß expression in T lymphocytes was reduced from 95.7% to 25.0%; CD3/TCRαß cell depletion yielded virtually pure TCRαß-negative T cells. Anti-CD3ε PEBLs abrogated TCRαß-mediated signaling, without affecting immunophenotype or proliferation. In anti-CD3ε PEBL-T cells, expression of an anti-CD19-41BB-CD3ζ CAR induced cytokine secretion, long-term proliferation, and CD19+ leukemia cell killing, at rates meeting or exceeding those of CAR-T cells with normal CD3/TCRαß expression. In immunodeficient mice, anti-CD3ε PEBL-T cells had markedly reduced GVHD potential; when transduced with anti-CD19 CAR, these T cells killed engrafted leukemic cells. PEBL blockade of surface CD3/TCRαß expression is an effective tool to prepare allogeneic CAR-T cells. Combined PEBL and CAR expression can be achieved in a single-step procedure, is easily adaptable to current cell manufacturing protocols, and can be used to target other T-cell molecules to further enhance CAR-T-cell therapies.


Assuntos
Imunoterapia Adotiva/métodos , Receptores de Antígenos de Linfócitos T/deficiência , Receptores de Antígenos Quiméricos , Animais , Antígenos CD19/imunologia , Complexo CD3/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Engenharia Genética/métodos , Humanos , Métodos , Camundongos , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Linfócitos T/imunologia
2.
Infect Immun ; 85(9)2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28630061

RESUMO

The Suppressor of TCR signaling proteins (Sts-1 and Sts-2) are two homologous phosphatases that negatively regulate signaling pathways in a number of hematopoietic lineages, including T lymphocytes. Mice lacking Sts expression are characterized by enhanced T cell responses. Additionally, a recent study demonstrated that Sts-/- mice are profoundly resistant to systemic infection by Candida albicans, with resistance characterized by enhanced survival, more rapid fungal clearance in key peripheral organs, and an altered inflammatory response. To investigate the role of Sts in the primary host response to infection by a bacterial pathogen, we evaluated the response of Sts-/- mice to infection by a Gram-negative bacterial pathogen. Francisella tularensis is a facultative bacterial pathogen that replicates intracellularly within a variety of cell types and is the causative agent of tularemia. Francisella infections are characterized by a delayed immune response, followed by an intense inflammatory reaction that causes widespread tissue damage and septic shock. Herein, we demonstrate that mice lacking Sts expression are significantly resistant to infection by the live vaccine strain (LVS) of F. tularensis Resistance is characterized by reduced lethality following high-dose intradermal infection, an altered cytokine response in the spleen, and enhanced bacterial clearance in multiple peripheral organs. Sts-/- bone marrow-derived monocytes and neutrophils, infected with F. tularensis LVS ex vivo, display enhanced restriction of intracellular bacteria. These observations suggest the Sts proteins play an important regulatory role in the host response to bacterial infection, and they underscore a role for Sts in regulating functionally relevant immune response pathways.


Assuntos
Suscetibilidade a Doenças , Francisella tularensis/imunologia , Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Tularemia/imunologia , Estruturas Animais/microbiologia , Estruturas Animais/patologia , Animais , Carga Bacteriana , Citocinas/análise , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Knockout , Monoéster Fosfórico Hidrolases/deficiência , Proteínas Tirosina Fosfatases/deficiência , Receptores de Antígenos de Linfócitos T/deficiência , Análise de Sobrevida
3.
J Immunol ; 198(4): 1452-1459, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28062695

RESUMO

Invariant NKT (iNKT) cells in healthy people express iNKT-TCRs with widely varying affinities for CD1d, suggesting different roles for high- and low-affinity iNKT clones in immune regulation. However, the functional implications of this heterogeneity have not yet been determined. Functionally aberrant iNKT responses have been previously demonstrated in different autoimmune diseases, including human type 1 diabetes, but their relationship to changes in the iNKT clonal repertoire have not been addressed. In this study, we directly compared the clonal iNKT repertoire of people with recent onset type 1 diabetes and age- and gender-matched healthy controls with regard to iNKT-TCR affinity and cytokine production. Our results demonstrate a selective loss of clones expressing high-affinity iNKT-TCRs from the iNKT repertoire of people with type 1 diabetes. Furthermore, this bias in the clonal iNKT repertoire in type 1 diabetes was associated with increased GM-CSF, IL-4, and IL-13 cytokine secretion among Ag-stimulated low-affinity iNKT clones. Thus, qualitative changes of the clonal iNKT repertoire with the potential to affect the regulatory function of this highly conserved T cell population are already established at the early stages in type 1 diabetes. These findings may inform future rationales for the development of iNKT-based therapies aiming to restore immune tolerance in type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Células T Matadoras Naturais/imunologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Adolescente , Adulto , Antígenos CD1d/genética , Células Clonais , Diabetes Mellitus Tipo 1/fisiopatologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Interleucina-13/imunologia , Interleucina-4/imunologia , Receptores de Antígenos de Linfócitos T/deficiência , Adulto Jovem
4.
Clin Cancer Res ; 23(9): 2255-2266, 2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-27815355

RESUMO

Purpose: Using gene-disrupted allogeneic T cells as universal effector cells provides an alternative and potentially improves current chimeric antigen receptor (CAR) T-cell therapy against cancers and infectious diseases.Experimental Design: The CRISPR/Cas9 system has recently emerged as a simple and efficient way for multiplex genome engineering. By combining lentiviral delivery of CAR and electro-transfer of Cas9 mRNA and gRNAs targeting endogenous TCR, ß-2 microglobulin (B2M) and PD1 simultaneously, to generate gene-disrupted allogeneic CAR T cells deficient of TCR, HLA class I molecule and PD1.Results: The CRISPR gene-edited CAR T cells showed potent antitumor activities, both in vitro and in animal models and were as potent as non-gene-edited CAR T cells. In addition, the TCR and HLA class I double deficient T cells had reduced alloreactivity and did not cause graft-versus-host disease. Finally, simultaneous triple genome editing by adding the disruption of PD1 led to enhanced in vivo antitumor activity of the gene-disrupted CAR T cells.Conclusions: Gene-disrupted allogeneic CAR and TCR T cells could provide an alternative as a universal donor to autologous T cells, which carry difficulties and high production costs. Gene-disrupted CAR and TCR T cells with disabled checkpoint molecules may be potent effector cells against cancers and infectious diseases. Clin Cancer Res; 23(9); 2255-66. ©2016 AACR.


Assuntos
Edição de Genes , Neoplasias/terapia , Receptor de Morte Celular Programada 1/genética , Linfócitos T/imunologia , Sistemas CRISPR-Cas , Doença Enxerto-Hospedeiro , Humanos , Imunoterapia Adotiva , Ativação Linfocitária/imunologia , Neoplasias/genética , Neoplasias/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/deficiência , Receptores de Antígenos de Linfócitos T/deficiência , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/imunologia
5.
Mol Carcinog ; 55(7): 1187-95, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26153082

RESUMO

The association between inflammation and the risk of colorectal cancer (CRC) is well documented in animal models and in humans, but the mechanistic role of inflammation in CRC is less well understood. To address this question, the induction of colon tumors was evaluated in (i) wild type (WT) and athymic BALB/c mice treated with the colon carcinogen azoxymethane (AOM) as a single agent, and (ii) in an inflammation model of colon cancer employing AOM and dextran sodium sulfate (DSS) in WT, athymic, TCRß(-/-) , TCRδ(-/-) and TCRß(-/-) TCRδ(-/-) C57Bl/6 mice. The athymic BALB/c mice treated with only AOM developed 90% fewer tumors than the WT mice. The difference in response was not due to metabolic activation of AOM or repair of DNA adducts. In the inflammation model using a standard sequential exposure to AOM followed by DSS treatment, the tumor incidence in WT mice was 58% with 7 adenomas and 6 adenocarcinomas. In contrast, the TCRß(-/-) , TCRδ(-/-) and TCRß(-/-) TCRδ(-/-) C57Bl/6 mice showed adenoma incidences of 10, 33, and 11%, respectively, and none of the immune compromised mice developed adenocarcinomas. When the DSS exposure was increased and the AOM lowered, no difference was observed between WT and TCRß(-/-) mice due to an increase in the incidence in the TCR null mice without concomitant increase in the WT mice. No tumors were observed in mice treated with AOM or DSS alone. © 2015 Wiley Periodicals, Inc.


Assuntos
Azoximetano/efeitos adversos , Neoplasias do Colo/epidemiologia , Neoplasias do Colo/patologia , Sulfato de Dextrana/administração & dosagem , Animais , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/imunologia , Sulfato de Dextrana/farmacologia , Hospedeiro Imunocomprometido , Incidência , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Receptores de Antígenos de Linfócitos T/deficiência
6.
PLoS One ; 9(2): e90196, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24587276

RESUMO

The human gammaherpesviruses establish life-long infections that are associated with the development of lymphomas and neoplasms, especially in immunocompromised individuals. T cells play a crucial role in the control of gammaherpesvirus infection through multiple functions, including the direct killing of infected cells, production of cytokines such as interferon-γ (IFN-γ), and costimulation of B cells. Impaired T cell function in mice infected with murine gammaherpesvirus 68 (MHV68) leads to increased reactivation and pathologies, including a higher incidence of lymphoid hyperplasia. Here we report that the absence of Suppressor of TCR signaling -1 and -2 (Sts-1(-/-)/2(-/-)) during MHV68 infection leads to the generation of T cells with significantly heightened responses. Transient differences in the T and B cell response of infected Sts-1(-/-)/2(-/-) (Sts dKO) mice were also observed when compared to WT mice. However, these alterations in the immune response and the overall absence of Sts-1 and Sts-2 did not impact viral pathogenesis or lead to pathology. Acute lytic replication in the lungs, establishment of latency in the spleen and reactivation from latency in the spleen in the Sts dKO mice were comparable to WT mice. Our studies indicate that Sts-1 and Sts-2 are not required for the immune control of MHV68 in a normal course of gammaherpesvirus infection, but suggest that interference with negative regulators of T cell responses might be further explored as a safe and efficacious strategy to improve adoptive T cell therapy.


Assuntos
Infecções por Herpesviridae/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Rhadinovirus/imunologia , Linfócitos T/imunologia , Infecções Tumorais por Vírus/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/virologia , Expressão Gênica , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Imunidade Inata , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Knockout , Proteínas Tirosina Fosfatases , Receptores de Antígenos de Linfócitos T/deficiência , Receptores de Antígenos de Linfócitos T/genética , Transdução de Sinais , Baço/imunologia , Baço/patologia , Baço/virologia , Linfócitos T/virologia , Infecções Tumorais por Vírus/patologia , Infecções Tumorais por Vírus/virologia , Ativação Viral , Latência Viral , Replicação Viral
7.
Nature ; 491(7426): 774-8, 2012 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-23103862

RESUMO

The mammalian host has developed a long-standing symbiotic relationship with a considerable number of microbial species. These include the microbiota on environmental surfaces, such as the respiratory and gastrointestinal tracts, and also endogenous retroviruses (ERVs), comprising a substantial fraction of the mammalian genome. The long-term consequences for the host of interactions with these microbial species can range from mutualism to parasitism and are not always completely understood. The potential effect of one microbial symbiont on another is even less clear. Here we study the control of ERVs in the commonly used C57BL/6 (B6) mouse strain, which lacks endogenous murine leukaemia viruses (MLVs) able to replicate in murine cells. We demonstrate the spontaneous emergence of fully infectious ecotropic MLV in B6 mice with a range of distinct immune deficiencies affecting antibody production. These recombinant retroviruses establish infection of immunodeficient mouse colonies, and ultimately result in retrovirus-induced lymphomas. Notably, ERV activation in immunodeficient mice is prevented in husbandry conditions associated with reduced or absent intestinal microbiota. Our results shed light onto a previously unappreciated role for immunity in the control of ERVs and provide a potential mechanistic link between immune activation by microbial triggers and a range of pathologies associated with ERVs, including cancer.


Assuntos
Anticorpos Antivirais/biossíntese , Retrovirus Endógenos/fisiologia , Hospedeiro Imunocomprometido/imunologia , Ativação Viral , Criação de Animais Domésticos , Animais , Anticorpos Antivirais/imunologia , Transformação Celular Viral , Retrovirus Endógenos/genética , Retrovirus Endógenos/crescimento & desenvolvimento , Retrovirus Endógenos/imunologia , Feminino , Leucemia/virologia , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/crescimento & desenvolvimento , Vírus da Leucemia Murina/imunologia , Vírus da Leucemia Murina/fisiologia , Linfoma/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/deficiência , Receptores de Antígenos de Linfócitos T/genética , Recombinação Genética , Viremia/imunologia , Viremia/virologia
8.
J Immunol ; 189(8): 4005-13, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22984075

RESUMO

The ability of the transmembrane adaptor protein linker for activation of T cells (LAT) to regulate T cell development, activation, survival, and homeostasis depends upon phosphorylation of its multiple tyrosine residues. The mutation of tyrosine 136 on LAT abrogates its interaction with phospholipase C-γ1, causing severe ramifications on TCR-mediated signaling. Mice harboring this mutation, LATY136F mice, have significantly impaired thymocyte development; however, they rapidly develop a fatal lymphoproliferative disease marked by the uncontrolled expansion of Th2-skewed CD4(+) T cells, high levels of IgE and IgG1, and autoantibody production. In this study, we assessed the contribution of multiple signaling pathways in LATY136F disease development. The deletion of the critical signaling proteins Gads and RasGRP1 caused a further block in thymocyte development, but, over time, could not prevent CD4(+) T cell hyperproliferation. Also, restoring signaling through the NF-κB and NFAT pathways was unable to halt the development of disease. However, expression of a constitutively active Raf transgene enhanced lymphoproliferation, indicating a role for the Ras-MAPK pathway in LAT-mediated disease.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Doenças Autoimunes/enzimologia , Doenças Autoimunes/imunologia , Transtornos Linfoproliferativos/enzimologia , Transtornos Linfoproliferativos/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Proteínas de Membrana/fisiologia , Fosfoproteínas/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Doenças Autoimunes/genética , Fatores de Troca do Nucleotídeo Guanina/deficiência , Fatores de Troca do Nucleotídeo Guanina/genética , Transtornos Linfoproliferativos/genética , Sistema de Sinalização das MAP Quinases/genética , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Receptores de Antígenos de Linfócitos T/deficiência , Receptores de Antígenos de Linfócitos T/genética , Quinases raf/biossíntese , Quinases raf/genética
9.
J Immunol ; 187(2): 654-63, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21677135

RESUMO

CD8(+) T cells recognize immunogenic peptides presented at the cell surface bound to MHCI molecules. Ag recognition involves the binding of both TCR and CD8 coreceptor to the same peptide-MHCI (pMHCI) ligand. Specificity is determined by the TCR, whereas CD8 mediates effects on Ag sensitivity. Anti-CD8 Abs have been used extensively to examine the role of CD8 in CD8(+) T cell activation. However, as previous studies have yielded conflicting results, it is unclear from the literature whether anti-CD8 Abs per se are capable of inducing effector function. In this article, we report on the ability of seven monoclonal anti-human CD8 Abs to activate six human CD8(+) T cell clones with a total of five different specificities. Six of seven anti-human CD8 Abs tested did not activate CD8(+) T cells. In contrast, one anti-human CD8 Ab, OKT8, induced effector function in all CD8(+) T cells examined. Moreover, OKT8 was found to enhance TCR/pMHCI on-rates and, as a consequence, could be used to improve pMHCI tetramer staining and the visualization of Ag-specific CD8(+) T cells. The anti-mouse CD8 Abs, CT-CD8a and CT-CD8b, also activated CD8(+) T cells despite opposing effects on pMHCI tetramer staining. The observed heterogeneity in the ability of anti-CD8 Abs to trigger T cell effector function provides an explanation for the apparent incongruity observed in previous studies and should be taken into consideration when interpreting results generated with these reagents. Furthermore, the ability of Ab-mediated CD8 engagement to deliver an activation signal underscores the importance of CD8 in CD8(+) T cell signaling.


Assuntos
Anticorpos/fisiologia , Antígenos CD8/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Citotoxicidade Imunológica , Antígenos HLA-A/química , Peptídeos/química , Receptores de Antígenos de Linfócitos T/deficiência , Anticorpos/metabolismo , Linfócitos T CD8-Positivos/citologia , Células Clonais , Antígenos HLA-A/imunologia , Antígeno HLA-A2 , Humanos , Imunofenotipagem , Ligantes , Peptídeos/análise , Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/imunologia , Coloração e Rotulagem , Ressonância de Plasmônio de Superfície
10.
J Immunol ; 186(2): 733-44, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21148038

RESUMO

Retinoic acid (RA) imprints gut-homing specificity on T cells upon activation by inducing the expression of chemokine receptor CCR9 and integrin α4ß7. CCR9 expression seemed to be more highly dependent on RA than was the α4ß7 expression, but its molecular mechanism remained unclear. In this article, we show that NFAT isoforms NFATc1 and NFATc2 directly interact with RA receptor (RAR) and retinoid X receptor (RXR) but play differential roles in RA-induced CCR9 expression on murine naive CD4(+) T cells. TCR stimulation for 6-24 h was required for the acquisition of responsiveness to RA and induced activation of NFATc1 and NFATc2. However, RA failed to induce CCR9 expression as long as TCR stimulation continued. After terminating TCR stimulation or adding cyclosporin A to the culture, Ccr9 gene transcription was induced, accompanied by inactivation of NFATc1 and sustained activation of NFATc2. Reporter and DNA-affinity precipitation assays demonstrated that the binding of NFATc2 to two NFAT-binding sites and that of the RAR/RXR complex to an RA response element half-site in the 5'-flanking region of the mouse Ccr9 gene were critical for RA-induced promoter activity. NFATc2 directly bound to RARα and RXRα, and it enhanced the binding of RARα to the RA response element half-site. NFATc1 also bound to the NFAT-binding sites and directly to RARα and RXRα, but it inhibited the NFATc2-dependent promoter activity. These results suggest that the cooperativity between NFATc2 and the RAR/RXR complex is essential for CCR9 expression on T cells and that NFATc1 interferes with the action of NFATc2.


Assuntos
Fatores de Transcrição NFATC/fisiologia , Receptores de Antígenos de Linfócitos T/fisiologia , Receptores CCR/biossíntese , Receptores do Ácido Retinoico/fisiologia , Receptores X de Retinoides/fisiologia , Tretinoína/farmacologia , Animais , Sequência de Bases , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Células COS , Linhagem Celular Tumoral , Células Cultivadas , Chlorocebus aethiops , Técnicas de Cocultura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Fatores de Transcrição NFATC/antagonistas & inibidores , Fatores de Transcrição NFATC/metabolismo , Regiões Promotoras Genéticas/imunologia , Ligação Proteica/imunologia , Receptores de Antígenos de Linfócitos T/deficiência , Receptores de Antígenos de Linfócitos T/genética , Fatores de Tempo
11.
J Immunol ; 185(3): 1393-403, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20592278

RESUMO

X-linked SCID patients are deficient in functional IL-2Rgamma(c) leading to the loss of IL-2/IL-4/IL-7/IL-9/IL-15/IL-21 signaling and a lack of NK and mature T cells. Patients treated with IL-2Rgamma(c) gene therapy have T cells develop; however, their NK cell numbers remain low, suggesting antiviral responses may be compromised. Similarly, IL-2Rgamma(c)(-/-) mice reconstituted with IL-2Rgamma(c) developed few NK cells, and reconstituted T cells exhibited defective proliferative responses suggesting incomplete recovery of IL-2Rgamma(c) signaling. Given the shift toward self-inactivating long terminal repeats with weaker promoters to control the risk of leukemia, we assessed NK and T cell numbers and function in IL-2Rgamma(c)(-/-) mice reconstituted with limiting amounts of IL-2Rgamma(c). Reconstitution resulted in lower IL-2/-15-mediated STAT5 phosphorylation and proliferation in NK and T cells. However, TCR costimulation restored cytokine-driven T cell proliferation to wild-type levels. Vector modifications that improved IL-2Rgamma(c) levels increased cytokine-induced STAT5 phosphorylation in both populations and increased NK cell proliferation demonstrating that IL-2Rgamma(c) levels are limiting. In addition, although the half-lives of both NK and T cells expressing intermediate levels of IL-2Rgamma(c) are reduced compared with wild-type cells, the reduction in NK cell half-live is much more severe than in T cells. Collectively, these data indicate different IL-2Rgamma(c) signaling thresholds for lymphocyte development and proliferation making functional monitoring imperative during gene therapy. Further, our findings suggest that IL-2Rgamma(c) reconstituted T cells may persist more efficiently than NK cells due to compensation for suboptimal IL-2Rgamma(c) signaling by the TCR.


Assuntos
Diferenciação Celular/imunologia , Regulação da Expressão Gênica/imunologia , Terapia Genética/métodos , Subunidade gama Comum de Receptores de Interleucina/biossíntese , Subunidade gama Comum de Receptores de Interleucina/genética , Células Matadoras Naturais/imunologia , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Diferenciação Celular/genética , Proliferação de Células , Subunidade gama Comum de Receptores de Interleucina/deficiência , Interleucina-15/antagonistas & inibidores , Interleucina-15/fisiologia , Interleucina-2/antagonistas & inibidores , Interleucina-2/fisiologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Fosforilação/genética , Fosforilação/imunologia , Receptores de Antígenos de Linfócitos T/deficiência , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/fisiologia , Fator de Transcrição STAT5/antagonistas & inibidores , Fator de Transcrição STAT5/metabolismo , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/imunologia , Imunodeficiência Combinada Severa/terapia , Transdução de Sinais/genética , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/metabolismo , Transdução Genética
12.
Clin Infect Dis ; 50(6): e34-7, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20156061

RESUMO

We describe a unique case of fulminant infectious mononucleosis and recurrent Epstein-Barr virus reactivation presenting in an adolescent. Detailed assays of Epstein-Barr virus-specific T cell immunity revealed defects in the patient's T cell receptor signalling pathway characterized by a lack of interleukin-2 and CD25 expression, which may have contributed to her clinical course. Allogeneic stem cell transplantation reversed the clinical and laboratory phenotype.


Assuntos
Herpesvirus Humano 4/imunologia , Herpesvirus Humano 4/isolamento & purificação , Mononucleose Infecciosa/diagnóstico , Mononucleose Infecciosa/imunologia , Ativação Viral , Adolescente , Criança , Humanos , Mononucleose Infecciosa/terapia , Interleucina-2/deficiência , Subunidade alfa de Receptor de Interleucina-2/deficiência , Receptores de Antígenos de Linfócitos T/deficiência , Recidiva , Transplante de Células-Tronco , Linfócitos T/imunologia , Adulto Jovem
13.
Carcinogenesis ; 31(3): 334-41, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20028726

RESUMO

The receptor for advanced glycation end products (RAGE), firstly described in 1992, is a single-transmembrane and multiligand member of the immunoglobulin protein family. RAGE engagement produces activation of multiple intracellular signaling mechanisms involved in several inflammation-associated clinical entities, such as diabetes, cancer, renal and heart failures, as well as neurodegenerative diseases. Although RAGE expression has been extensively reported in many cancer types, it is now emerging as a relevant element that can continuously fuel an inflammatory milieu at the tumor microenvironment, thus changing our perception of its contribution to cancer biology. In this review, we will discuss the role of multiligand/RAGE axis, particularly at the multicellular cross talk established in the inflammatory tumor microenvironment. A better understanding of its contribution may provide new targets for tumor management and risk assessment.


Assuntos
Produtos Finais de Glicação Avançada/fisiologia , Inflamação/fisiopatologia , Proteínas de Neoplasias/fisiologia , Neoplasias/fisiopatologia , Receptores Imunológicos/fisiologia , Animais , Comunicação Autócrina , Hipóxia Celular , Dieta/efeitos adversos , Progressão da Doença , Glicólise , Humanos , Ligantes , Camundongos , Modelos Biológicos , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/fisiopatologia , Comunicação Parácrina , Receptor Cross-Talk , Receptor para Produtos Finais de Glicação Avançada , Receptores de Antígenos de Linfócitos T/deficiência , Receptores Imunológicos/química , Receptores Imunológicos/deficiência , Receptores Imunológicos/genética , Transdução de Sinais
14.
Cancer Res ; 69(10): 4175-83, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19435902

RESUMO

Restimulation of previously activated T cells via the T-cell receptor (TCR) leads to activation-induced cell death (AICD), which is, at least in part, dependent on the death receptor CD95 (APO-1, FAS) and its natural ligand (CD95L). Here, we characterize cutaneous T-cell lymphoma (CTCL) cells (CTCL tumor cell lines and primary CTCL tumor cells from CTCL patients) as AICD resistant. We show that CTCL cells have elevated levels of the CD95-inhibitory protein cFLIP. However, cFLIP is not responsible for CTCL AICD resistance. Instead, our data suggest that reduced TCR-proximal signaling in CTCL cells is responsible for the observed AICD resistance. CTCL cells exhibit no PLC-gamma1 activity, resulting in an impaired Ca(2+)release and reduced generation of reactive oxygen species upon TCR stimulation. Ca(2+) and ROS production are crucial for up-regulation of CD95L and reconstitution of both signals resulted in AICD sensitivity of CTCL cells. In accordance with these data, CTCL tumor cells from patients with Sézary syndrome do not up-regulate CD95L upon TCR-stimulation and are therefore resistant to AICD. These results show a novel mechanism of AICD resistance in CTCL that could have future therapeutic implications to overcome apoptosis resistance in CTCL patients.


Assuntos
Linfoma de Células T/patologia , Receptores de Antígenos de Linfócitos T/fisiologia , Receptor fas/genética , Apoptose , Morte Celular , Proteína Ligante Fas/fisiologia , Citometria de Fluxo , Humanos , Ativação Linfocitária , Linfoma de Células T/genética , Linfoma de Células T/imunologia , Receptores de Antígenos de Linfócitos T/deficiência , Síndrome de Sézary/imunologia , Síndrome de Sézary/patologia , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Linfócitos T/imunologia , Regulação para Cima
15.
Infect Immun ; 75(8): 4040-9, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17562776

RESUMO

Pertussis toxin (PTx) is an AB(5) toxin produced by the human pathogen Bordetella pertussis. Previous work demonstrates that the five binding (B) subunits of PTx can have profound effects on T lymphocytes independent of the enzymatic activity of the A subunit. Stimulation of T cells with holotoxin (PTx) or the B subunit alone (PTxB) rapidly induces signaling events resulting in inositol phosphate accumulation, Ca(2+) mobilization, interleukin-2 (IL-2) production, and mitogenic cell growth. Although previous reports suggest the presence of PTx signaling receptors expressed on T cells, to date, the receptor(s) and membrane proximal signaling events utilized by PTx remain unknown. Here we genetically and biochemically define the membrane proximal components utilized by PTx to initiate signal transduction in T cells. Using mutants of the Jurkat T-cell line deficient for key components of the T-cell receptor (TCR) pathway, we have compared stimulation with PTx to that of anti-CD3 monoclonal antibody (MAb), which directly interacts with and activates the TCR complex. Our genetic data in combination with biochemical analysis show that PTx (via the B subunit) activates TCR signaling similar to that of anti-CD3 MAb, including activation of key signaling intermediates such as Lck, ZAP-70, and phospholipase C-gamma1. Moreover, the data indicate that costimulatory activity, as provided by CD28 ligation, is required for PTx to fully stimulate downstream indicators of T-cell activation such as IL-2 gene expression. By illuminating the signaling pathways that PTx activates in T cells, we provide a mechanistic understanding for how these signals deregulate immune system functions during B. pertussis infection.


Assuntos
Toxina Pertussis/imunologia , Toxina Pertussis/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Anticorpos Monoclonais/imunologia , Bordetella pertussis/imunologia , Antígenos CD28/imunologia , Complexo CD3/imunologia , Cálcio/metabolismo , Linhagem Celular , Proliferação de Células , Humanos , Fosfatos de Inositol/metabolismo , Interleucina-2/biossíntese , Isoenzimas/análise , Células Jurkat , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/análise , Modelos Biológicos , Fosfolipase C beta , Fosfolipase C gama/metabolismo , Ligação Proteica , Receptores de Antígenos de Linfócitos T/deficiência , Linfócitos T/química , Linfócitos T/metabolismo , Fosfolipases Tipo C/análise , Proteína-Tirosina Quinase ZAP-70/análise
16.
Blood ; 110(7): 2610-9, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17507663

RESUMO

Inactivation of the CDKN2 genes that encode the p16(INK4A) and p14(ARF) proteins occurs in the majority of human T-cell acute lymphoblastic leukemias (T-ALLs). Ectopic expression of TAL1 and LMO1 genes is linked to the development of T-ALL in humans. In TAL1xLMO1 mice, leukemia develops in 100% of mice at 5 months. To identify the molecular events crucial to leukemic transformation, we produced several mouse models. We report here that expression of P16(INK4A) in developing TAL1xLMO1 thymocytes blocks leukemogenesis in the majority of the mice, and the leukemias that eventually develop show P16(INK4A) loss of expression. Events related to the T-cell receptor beta selection process are thought to be important for leukemic transformation. We show here that the absence of the pTalpha chain only slightly delays the appearance of TAL1xLMO1-induced T-ALL, which indicates a minor role of the pTalpha chain. We also show that the CD3epsilon-mediated signal transduction pathway is essential for this transformation process, since the TAL1xLMO1xCD3epsilon-deficient mice do not develop T-ALL for up to 1 year.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Complexo CD3/metabolismo , Transformação Celular Neoplásica/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Leucemia/metabolismo , Leucemia/patologia , Proteínas Proto-Oncogênicas/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Complexo CD3/genética , Diferenciação Celular , Linhagem da Célula , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Ciclina D3 , Inibidor p16 de Quinase Dependente de Ciclina/genética , Ciclinas/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas com Domínio LIM , Leucemia/genética , Camundongos , Camundongos Transgênicos , Mutação/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/genética , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptores de Antígenos de Linfócitos T/deficiência , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Taxa de Sobrevida , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Timo/citologia , Timo/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
17.
J Immunol ; 175(2): 829-38, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16002680

RESUMO

Oral tolerance is systemic immune hyporesponsiveness induced by the oral administration of soluble Ags. Hyporesponsiveness of Ag-specific CD4 T cells is responsible for this phenomenon. However, the molecular mechanisms underlying the hyporesponsive state of these T cells are not fully understood. In the present study, we investigated the ability of orally tolerized T cells to form conjugates with Ag-bearing APCs and to translocate TCR, protein kinase C-theta (PKC-theta), and lipid rafts into the interface between T cells and APCs. Orally tolerized T cells were prepared from the spleens of OVA-fed DO11.10 mice. Interestingly, the orally tolerized T cells did not show any impairment in the formation of conjugates with APCs. The conjugates were formed in a LFA-1-dependent manner. Upon antigenic stimulation, the tolerized T cells could indeed activate Rap1, which is critical for LFA-1 activation and thus cell adhesion. However, orally tolerized T cells showed defects in the translocation of TCR, PKC-theta, and lipid rafts into the interface between T cells and APCs. Translocation of TCR and PKC-theta to lipid raft fractions upon antigenic stimulation was also impaired in the tolerized T cells. Ag-induced activation of Vav, Rac1, and cdc42, which are essential for immunological synapse and raft aggregation, were down-regulated in orally tolerized T cells. These results demonstrate that orally tolerized T cells can respond to specific Ags in terms of conjugate formation but not with appropriate immunological synapse formation. This may account for the hyporesponsive state of orally tolerized T cells.


Assuntos
Apresentação de Antígeno , Células Apresentadoras de Antígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/transplante , Comunicação Celular/imunologia , Tolerância Imunológica , Administração Oral , Sequência de Aminoácidos , Animais , Apresentação de Antígeno/genética , Células Apresentadoras de Antígenos/enzimologia , Células Apresentadoras de Antígenos/metabolismo , Linfócitos T CD4-Positivos/enzimologia , Comunicação Celular/genética , Proteínas de Ciclo Celular/metabolismo , Feminino , Tolerância Imunológica/genética , Isoenzimas/deficiência , Isoenzimas/metabolismo , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Dados de Sequência Molecular , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Proteína Quinase C/deficiência , Proteína Quinase C/metabolismo , Proteína Quinase C-theta , Transporte Proteico/genética , Transporte Proteico/imunologia , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-vav , Receptores de Antígenos de Linfócitos T/deficiência , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Proteína cdc42 de Ligação ao GTP/deficiência , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/deficiência , Proteínas rac1 de Ligação ao GTP/metabolismo
18.
J Immunol ; 174(4): 1830-40, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15699109

RESUMO

CD8+ tumor-infiltrating lymphocytes (TIL) are severely deficient in cytolysis, a defect that may permit tumor escape from immune-mediated destruction. Because lytic function is dependent upon TCR signaling, we have tested the hypothesis that primary TIL have defective signaling by analysis of the localization and activation status of TIL proteins important in TCR-mediated signaling. Upon conjugate formation with cognate target cells in vitro, TIL do not recruit granzyme B+ granules, the microtubule-organizing center, F-actin, Wiskott-Aldrich syndrome protein, nor proline rich tyrosine kinase-2 to the target cell contact site. In addition, TIL do not flux calcium nor demonstrate proximal tyrosine kinase activity, deficiencies likely to underlie failure to fully activate the lytic machinery. Confocal microscopy and fluorescence resonance energy transfer analyses demonstrate that TIL are triggered by conjugate formation in that the TCR, p56lck, CD3zeta, LFA-1, lipid rafts, ZAP70, and linker for activation of T cells localize at the TIL:tumor cell contact site, and CD43 and CD45 are excluded. However, proximal TCR signaling is blocked upon conjugate formation because the inhibitory motif of p56lck is rapidly phosphorylated (Y505) and COOH-terminal Src kinase is recruited to the contact site, while Src homology 2 domain-containing protein phosphatase 2 is cytoplasmic. Our data support a novel mechanism explaining how tumor-induced inactivation of proximal TCR signaling regulates lytic function of antitumor T cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Citotoxicidade Imunológica , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Receptores de Antígenos de Linfócitos T/fisiologia , Transdução de Sinais/imunologia , Actinas/deficiência , Actinas/metabolismo , Animais , Antígenos CD2/metabolismo , Complexo CD3/metabolismo , Antígenos CD8/metabolismo , Linfócitos T CD8-Positivos/patologia , Cálcio/metabolismo , Linhagem Celular Tumoral , Separação Celular , Grânulos Citoplasmáticos/imunologia , Grânulos Citoplasmáticos/metabolismo , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/antagonistas & inibidores , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Linfócitos do Interstício Tumoral/enzimologia , Linfócitos do Interstício Tumoral/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Fosfotirosina/metabolismo , Transporte Proteico/imunologia , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/metabolismo , Complexo Receptor-CD3 de Antígeno de Linfócitos T/metabolismo , Receptores de Antígenos de Linfócitos T/deficiência , Proteína-Tirosina Quinase ZAP-70
19.
Verh Dtsch Ges Pathol ; 89: 261-6, 2005.
Artigo em Alemão | MEDLINE | ID: mdl-18035700

RESUMO

Anaplastic large cell lymphoma (ALCL) designates a heterogeneous group of CD30+ (systemic or primary cutaneous) peripheral T-cell lymphomas (PTCLs). A subgroup of systemic ALCL is transformed by anaplastic lymphoma kinase (ALK). We compared 46 ALCL with 22 PTCLs in terms of T-cell receptor (TCR) rearrangements, expression of TCRs and TCR-associated molecules [CD3, ZAP-70 (zeta-associated protein 70)]. Despite their frequent clonal rearrangement for TCRbeta, only 4% of ALCLs expressed TCRbeta protein, whereas TCRs were detected in 86% of PTCLs. Moreover, both TCRbeta+ ALCLs lacked CD3 and ZAP-70 (ie, molecules indispensable for the transduction of cognate TCR signals). Defective expression of TCRs is a common characteristic of all types of ALCL, which may contribute to the dysregulation of intracellular signaling pathways controlling T-cell activation and survival. This molecular hallmark of ALCL is analogous to defective immunoglobulin expression distinguishing Hodgkin lymphoma from other B-cell lymphomas.


Assuntos
Linfoma Anaplásico de Células Grandes/genética , Linfoma Anaplásico de Células Grandes/patologia , Receptores de Antígenos de Linfócitos T/deficiência , Receptores de Antígenos de Linfócitos T/genética , Antígenos CD/análise , Humanos , Antígeno Ki-1/análise , Linfoma Anaplásico de Células Grandes/imunologia , Linfócitos T/imunologia
20.
J Clin Invest ; 114(10): 1409-11, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15545990

RESUMO

SCID, a syndrome characterized by the absence of T cells and adaptive immunity, can result from mutations in multiple genes that encode components of the immune system. Three such components are cytokine receptor chains or signaling molecules, five are needed for antigen receptor development, one is adenosine deaminase--a purine salvage pathway enzyme, and the last is a phosphatase, CD45. In this issue of the JCI, a report describes how complete deficiency of the CD3epsilon chain of the T cell antigen receptor/CD3 complex causes human SCID.


Assuntos
Adenosina Desaminase/deficiência , Complexo CD3/química , Subunidades Proteicas/química , Complexo Receptor-CD3 de Antígeno de Linfócitos T/deficiência , Receptores de Antígenos de Linfócitos T/deficiência , Imunodeficiência Combinada Severa/etiologia , Adenosina Desaminase/genética , Complexo CD3/genética , Cromossomos Humanos Par 11 , Cromossomos Humanos X , Ligação Genética , Marcadores Genéticos , Humanos , Antígenos Comuns de Leucócito/genética , Modelos Biológicos , Mutação , Linhagem , Subunidades Proteicas/genética , Complexo Receptor-CD3 de Antígeno de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/genética , Receptores de Citocinas/metabolismo , Imunodeficiência Combinada Severa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA