Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cytokine ; 92: 24-32, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28088614

RESUMO

The CCAAT/Enhancer Binding Protein ß (C/EBPß) transcription factor is activated by multiple inflammatory stimuli, including IL-17 and LPS, and C/EBPß itself regulates numerous genes involved in inflammation. However, the role of C/EBPß in driving autoimmunity is not well understood. Here, we demonstrate that Cebpb-/- mice are resistant to EAE. Cebpb-/- mice exhibited reduced lymphocyte and APC infiltration into CNS following EAE induction. Furthermore, MOG-induced Th17 cytokine production was impaired in draining LN, indicating defects in Th17 cell priming. In vitro Th17 polarization studies indicated that T cell responses are not inherently defective, instead supporting the known roles for C/EBPß in myeloid lineage cell activation as the likely mechanism for defective Th17 priming in vivo. However, we did uncover an unexpected role for C/EBPß in regulating ll23r expression in APCs. ChIP assays confirmed that C/EBPß binds directly to the Il23r gene promoter in dendritic cells and Th17 cells. These data establish C/EBPß as a key driver of autoimmune inflammation in EAE, and propose a novel role for C/EBPß in regulation of IL-23R expression.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/imunologia , Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/imunologia , Regulação da Expressão Gênica/imunologia , Células Th17/imunologia , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Citocinas/genética , Citocinas/imunologia , Células Dendríticas/patologia , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Knockout , Receptores de Interleucina/genética , Receptores de Interleucina/imunologia , Células Th17/patologia
2.
PLoS One ; 10(8): e0136538, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26317211

RESUMO

Humans or mice subjected to immunosuppression, such as corticosteroids or anti-cytokine biologic therapies, are susceptible to mucosal infections by the commensal fungus Candida albicans. Recently it has become evident that the Th17/IL-17 axis is essential for immunity to candidiasis, but the downstream events that control immunity to this fungus are poorly understood. The CCAAT/Enhancer Binding Protein-ß (C/EBPß) transcription factor is important for signaling by multiple inflammatory stimuli, including IL-17. C/EBPß is regulated in a variety of ways by IL-17, and controls several downstream IL-17 target genes. However, the role of C/EBPß in vivo is poorly understood, in part because C/EBPß-deficient mice are challenging to breed and work with. In this study, we sought to understand the role of C/EBPß in the context of an IL-17-dependent immune response, using C. albicans infection as a model system. Confirming prior findings, we found that C/EBPß is required for immunity to systemic candidiasis. In contrast, C/EBPß(-/-) mice were resistant to oropharyngeal candidiasis (OPC), in a manner indistinguishable from immunocompetent WT mice. However, C/EBPß(-/-) mice experienced more severe OPC than WT mice in the context of cortisone-induced immunosuppression. Expression of the antimicrobial peptide ß-defensin (BD)-3 correlated strongly with susceptibility in C/EBPß(-/-) mice, but no other IL-17-dependent genes were associated with susceptibility. Therefore, C/EBPß contributes to immunity to mucosal candidiasis during cortisone immunosuppression in a manner linked to ß-defensin 3 expression, but is apparently dispensable for the IL-17-dependent response.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/imunologia , Candida albicans/imunologia , Candidíase Bucal/imunologia , Regulação da Expressão Gênica/imunologia , beta-Defensinas/imunologia , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Candidíase Bucal/genética , Candidíase Bucal/patologia , Interleucina-17/genética , Interleucina-17/imunologia , Camundongos , Camundongos Knockout , beta-Defensinas/genética
3.
J Exp Med ; 211(10): 2075-84, 2014 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-25200028

RESUMO

Oropharyngeal candidiasis (OPC) is an opportunistic fungal infection caused by Candida albicans. OPC is frequent in HIV/AIDS, implicating adaptive immunity. Mice are naive to Candida, yet IL-17 is induced within 24 h of infection, and susceptibility is strongly dependent on IL-17R signaling. We sought to identify the source of IL-17 during the early innate response to candidiasis. We show that innate responses to Candida require an intact TCR, as SCID, IL-7Rα(-/-), and Rag1(-/-) mice were susceptible to OPC, and blockade of TCR signaling by cyclosporine induced susceptibility. Using fate-tracking IL-17 reporter mice, we found that IL-17 is produced within 1-2 d by tongue-resident populations of γδ T cells and CD3(+)CD4(+)CD44(hi)TCRß(+)CCR6(+) natural Th17 (nTh17) cells, but not by TCR-deficient innate lymphoid cells (ILCs) or NK cells. These cells function redundantly, as TCR-ß(-/-) and TCR-δ(-/-) mice were both resistant to OPC. Whereas γδ T cells were previously shown to produce IL-17 during dermal candidiasis and are known to mediate host defense at mucosal surfaces, nTh17 cells are poorly understood. The oral nTh17 population expanded rapidly after OPC, exhibited high TCR-ß clonal diversity, and was absent in Rag1(-/-), IL-7Rα(-/-), and germ-free mice. These findings indicate that nTh17 and γδ T cells, but not ILCs, are key mucosal sentinels that control oral pathogens.


Assuntos
Candida albicans/imunologia , Candidíase/prevenção & controle , Imunidade Inata/imunologia , Boca/imunologia , Células Th17/imunologia , Animais , Candidíase/imunologia , Citometria de Fluxo , Interleucina-23/deficiência , Camundongos , Camundongos Knockout , Microscopia Confocal , Boca/citologia , Boca/microbiologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Receptores de Interleucina-17/deficiência , Receptores de Interleucina-17/metabolismo
4.
Infect Immun ; 82(3): 1173-80, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24379290

RESUMO

Oropharyngeal candidiasis (OPC [thrush]) is an opportunistic infection caused by the commensal fungus Candida albicans. OPC is common in individuals with HIV/AIDS, infants, patients on chemotherapy, and individuals with congenital immune defects. Immunity to OPC is strongly dependent on the interleukin-23 (IL-23)/IL-17R axis, as mice and humans with defects in IL-17R signaling (IL17F, ACT1, IL-17RA) or in genes that direct Th17 differentiation (STAT3, STAT1, CARD9) are prone to mucocutaneous candidiasis. Conventional Th17 cells are induced in response to C. albicans infection via signals from C-type lectin receptors, which signal through the adaptor CARD9, leading to production of Th17-inducing cytokines such as IL-6, IL-1ß, and IL-23. Recent data indicate that IL-17 can also be made by numerous innate cell subsets. These innate "type 17" cells resemble conventional Th17 cells, but they can be activated without need for prior antigen exposure. Because C. albicans is not a commensal organism in rodents and mice are thus naive to this fungus, we had the opportunity to assess the role of CARD9 in innate versus adaptive responses using an OPC infection model. As expected, CARD9(-/-) mice failed to mount an adaptive Th17 response following oral Candida infection. Surprisingly, however, CARD9(-/-) mice had preserved innate IL-17-dependent responses to Candida and were almost fully resistant to OPC. Thus, CARD9 is important primarily for adaptive immunity to C. albicans, whereas alternate recognition systems appear to be needed for effective innate responses.


Assuntos
Imunidade Adaptativa/imunologia , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Candida albicans/imunologia , Candidíase/imunologia , Imunidade Inata/imunologia , Mucosa Bucal/imunologia , Animais , Proteínas Adaptadoras de Sinalização CARD , Candidíase/microbiologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Mucosa Bucal/microbiologia , Células Th17/imunologia , Células Th17/microbiologia
5.
PLoS One ; 8(7): e70168, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23922952

RESUMO

IL-17 is the founding member of a family of cytokines and receptors with unique structures and signaling properties. IL-17 is the signature cytokine of Th17 cells, a relatively new T cell population that promotes inflammation in settings of infection and autoimmunity. Despite advances in understanding Th17 cells, mechanisms of IL-17-mediated signal transduction are less well defined. IL-17 signaling requires contributions from two receptor subunits, IL-17RA and IL-17RC. Mutants of IL-17RC lacking the cytoplasmic domain are nonfunctional, indicating that IL-17RC provides essential but poorly understood signaling contributions to IL-17-mediated signaling. To better understand the role of IL-17RC in signaling, we performed a yeast 2-hybrid screen to identify novel proteins associated with the IL-17RC cytoplasmic tail. One of the most frequent candidates was the anaphase promoting complex protein 7 (APC7 or AnapC7), which interacted with both IL-17RC and IL-17RA. Knockdown of AnapC7 by siRNA silencing exerted no detectable impact on IL-17 signaling. However, AnapC5, which associates with AnapC7, was also able to bind IL-17RA and IL-17RC. Moreover, AnapC5 silencing enhanced IL-17-induced gene expression, suggesting an inhibitory activity. Strikingly, AnapC5 also associated with A20 (TNFAIP3), a recently-identified negative feedback regulator of IL-17 signal transduction. IL-17 signaling was not impacted by knockdown of Itch or TAXBP1, scaffolding proteins that mediate A20 inhibition in the TNFα and IL-1 signaling pathways. These data suggest a model in which AnapC5, rather than TAX1BP1 and Itch, is a novel adaptor and negative regulator of IL-17 signaling pathways.


Assuntos
Subunidade Apc5 do Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Proteínas de Ligação a DNA/metabolismo , Interleucina-17/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/metabolismo , Motivos de Aminoácidos , Animais , Subunidade Apc7 do Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular , Fator D do Complemento/metabolismo , Cisteína Endopeptidases , Camundongos , Modelos Biológicos , Ligação Proteica , Mapeamento de Interação de Proteínas , Subunidades Proteicas/metabolismo , Receptores de Interleucina-17/química , Receptores de Interleucina-17/metabolismo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Técnicas do Sistema de Duplo-Híbrido , Proteínas ras/metabolismo
6.
Cancer Immun ; 13: 14, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23882159

RESUMO

Human memory T cells present in ovarian tumor ascites fluids fail to respond normally to stimulation via the T cell receptor (TCR). This immunosuppression is manifested by decreases in NF-κB and NFAT activation, IFN-γ production, and cell proliferation in response to TCR stimulation with immobilized antibodies to CD3 and CD28. The anergy of the tumor-associated T cells (TATs) is mediated by soluble factors present in ovarian tumor ascites fluids. The non-responsiveness of the T cells is quickly reversed when the cells are assayed in the absence of the ascites fluid, and is rapidly reestablished when a cell-free ascites fluid is added back to the T cells. Based upon the observed normal phosphorylation patterns of the TCR proximal signaling molecules, the inhibition of NF-κB, and NFAT activation in response to TCR stimulation, as well as the ability of the diacylglycerol analog PMA and the ionophore ionomycin to bypass the ascites fluid-induced TCR signaling arrest, the site of the arrest in the activation cascade appears to be at or just upstream of PLC-γ. An identical TCR signaling arrest pattern was observed when T cells derived from normal donor peripheral blood were incubated with either malignant or nonmalignant (cirrhotic) ascites fluids. The immunosuppressive activity of ascites fluids reported here suggests that soluble factors acting directly or indirectly upon T cells present within tumors contribute to the anergy that has previously been observed in T cells derived from malignant and nonmalignant inflammatory microenvironments. The soluble immunosuppressive factors represent potential therapeutic targets for ovarian cancer.


Assuntos
NF-kappa B/imunologia , Fatores de Transcrição NFATC/imunologia , Neoplasias Ovarianas/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Ascite/imunologia , Ascite/patologia , Feminino , Humanos , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais
7.
J Assoc Res Otolaryngol ; 13(3): 423-35, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22310933

RESUMO

A majority of T cells from chronic inflammatory tissues derived from patients with nasal polyposis were found to express an effector memory phenotype. We report here that these memory T cells failed to activate NF-κB in response to TCR stimulation but responded normally when the proximal TCR signaling molecules were bypassed with PMA and ionomycin. The dysfunction of these cells was associated with a decrease in the phosphorylation of several TCR proximal signaling molecules including ZAP70, Lck and SLP-76. In addition to the disruption in the TCR signaling pathway, the nasal polyp-associated T cells were shown to have a defect in their ability to translocate LAMP-1 to the cell surface. The results presented here establish that the phenotype and anergy of the T cells in the nasal polyp are similar to those which is seen in memory T cells derived from human tumors and other sites of chronic inflammation.


Assuntos
Antígenos CD28/metabolismo , Complexo CD3/metabolismo , Pólipos Nasais/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/fisiologia , Ionóforos de Cálcio , Estudos de Casos e Controles , Citometria de Fluxo , Humanos , Memória Imunológica , Imunofenotipagem , Ionomicina , Ativação Linfocitária , Proteínas de Membrana Lisossomal/metabolismo , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/metabolismo , Fenótipo , Fosforilação , Transdução de Sinais , Linfócitos T/citologia
8.
J Immunol ; 185(5): 2681-92, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20686130

RESUMO

Fibroblasts are a dominant cell type in most human solid tumors. The possibility that fibroblasts have the capacity to interact with and modulate the function of tumor-associated T lymphocytes makes them a potential therapeutic target. To address this question, primary cultures of fibroblasts derived from human lung tumors were established and cultured with T cells derived from the same tumor. The tumor fibroblasts significantly enhance the production of IFN-gamma and IL-17A by the tumor-associated T cells following a CD3/CD28-induced activation of the T cells. This enhancement was fibroblast cell dose-dependent and did not require direct contact between the two cell types. Tumor-associated fibroblast-conditioned media similarly enhanced both IFN-gamma and IL-17A in activated T cells, and this enhancement was significantly reduced by Abs to IL-6. Conditioned media derived from activated lymphocyte cultures significantly enhanced IL-6 production by tumor fibroblasts. A similar enhancement of IFN-gamma and IL-17A was observed when activated T cells from a normal donor were cultivated with skin fibroblasts derived from the same donor. These results establish that fibroblasts and autologous lymphocytes, whether derived from the tumor microenvironment or from nonmalignant tissues, have the capacity to reciprocally interact and modulate function. In contrast to other reports, fibroblasts are shown to have an immunostimulatory effect upon activated T lymphocytes. The ability of fibroblasts to enhance two T cell cytokines known to have an impact upon tumor progression suggests that fibroblasts play an important role in tumor pathogenesis that could be exploited therapeutically.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/imunologia , Comunicação Celular/imunologia , Fibroblastos/imunologia , Fibroblastos/patologia , Neoplasias Pulmonares/imunologia , Ativação Linfocitária/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Separação Celular , Técnicas de Cocultura , Relação Dose-Resposta Imunológica , Fibroblastos/metabolismo , Humanos , Interferon gama/biossíntese , Interleucina-17/biossíntese , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Auxiliares-Indutores/patologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/patologia , Células Tumorais Cultivadas
9.
Cancer Microenviron ; 3(1): 29-47, 2010 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-21209773

RESUMO

The immune system of cancer patients recognizes tumor-associated antigens expressed on solid tumors and these antigens are able to induce tumor-specific humoral and cellular immune responses. Diverse immunotherapeutic strategies have been used in an attempt to enhance both antibody and T cell responses to tumors. While several tumor vaccination strategies significantly increase the number of tumor-specific lymphocytes in the blood of cancer patients, most vaccinated patients ultimately experience tumor progression. CD4+ and CD8+ T cells with an effector memory phenotype infiltrate human tumor microenvironments, but most are hyporesponsive to stimulation via the T cell receptor (TCR) and CD28 under conditions that activate memory T cells derived from the peripheral blood of the cancer patients or normal donors. Attempts to identify cells and molecules responsible for the TCR signaling arrest of tumor-infiltrating T cells have focused largely upon the immunosuppressive effects of tumor cells, tolerogenic dendritic cells and regulatory T cells. Here we review potential mechanisms by which human T cell function is arrested in the tumor microenvironment with a focus on the immunomodulatory effects of stromal fibroblasts. Determining in vivo which cells and molecules are responsible for the TCR arrest in human tumor-infiltrating T cells will be necessary to formulate and test strategies to prevent or reverse the signaling arrest of the human T cells in situ for a more effective design of tumor vaccines. These questions are now addressable using novel human xenograft models of tumor microenvironments.

10.
Clin Immunol ; 132(1): 71-82, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19395317

RESUMO

Using a novel loading technique, IL-12 is reported here to be efficiently encapsulated within large multilamellar liposomes. The preclinical efficacy of the cytokine loaded liposomes to deliver IL-12 into human tumors and to reactive tumor-associated T cells in situ is tested using a human tumor xenograft model. IL-12 is released in vivo from these liposomes in a biologically active form when injected into tumor xenografts that are established by the subcutaneous implantation of non-disrupted pieces of human lung, breast or ovarian tumors into immunodeficient mice. The histological architecture of the original tumor tissue, including tumor-associated leukocytes, tumor cells and stromal cells is preserved anatomically and the cells remain functionally responsive to cytokines in these xenografts. The local and sustained release of IL-12 into the tumor microenvironment reactivates tumor-associated quiescent effector memory T cells to proliferate, produce and release IFN-gamma resulting in the killing of tumor cells in situ. Very little IL-12 is detected in the serum of mice for up to 5 days after an intratumoral injection of the IL-12 liposomes. We conclude that IL-12 loaded large multilamellar liposomes provide a safe method for the local and sustained delivery of IL-12 to tumors and a therapeutically effective way of reactivating existing tumor-associated T cells in human solid tumor microenvironments. The potential of this local in situ T cell re-stimulation to induce a systemic anti-tumor immunity is discussed.


Assuntos
Interleucina-12/imunologia , Lipossomos/química , Neoplasias Experimentais/imunologia , Linfócitos T/imunologia , Animais , Dicroísmo Circular , Sistemas de Liberação de Medicamentos , Humanos , Imuno-Histoquímica , Memória Imunológica/imunologia , Interleucina-12/administração & dosagem , Interleucina-12/química , Antígeno Ki-67/análise , Camundongos , Camundongos SCID , Neoplasias Experimentais/sangue , Neoplasias Experimentais/terapia , Espectrometria de Fluorescência , Linfócitos T/citologia , Linfócitos T/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Immunol ; 180(10): 7009-18, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18453623

RESUMO

Non-disrupted pieces of primary human lung tumor implanted into NOD-scid IL2Rgamma(null) mice consistently result in successful xenografts in which tissue architecture, including tumor-associated leukocytes, stromal fibroblasts, and tumor cells are preserved for prolonged periods with limited host-vs-graft interference. Human CD45(+) tumor-associated leukocytes within the xenograft are predominantly CD3(+) T cells with fewer CD138(+) plasma cells. The effector memory T cells that had been shown to be quiescent in human lung tumor microenvironments can be activated in situ as determined by the production of human IFN-gamma in response to exogenous IL-12. Plasma cells remain functional as evidenced by production of human Ig. Significant levels of human IFN-gamma and Ig were detected in sera from xenograft-bearing mice for up to 9 wk postengraftment. Tumor-associated T cells were found to migrate from the microenvironment of the xenograft to the lung, liver, and primarily the spleen. At 8 wk postengraftment, a significant portion of cells isolated from the mouse spleens were found to be human CD45(+) cells. The majority of CD45(+) cells were CD3(+) and expressed a phenotype consistent with an effector memory T cell, consisting of CD4(+) or CD8(+) T cells that were CD45RO(+), CD44(+), CD62L(-), and CD25(-). Following adoptive transfer into non-tumor bearing NOD-scid IL2Rgamma(null) mice, these human T cells were found to expand in the spleen, produce IFN-gamma, and maintain an effector memory phenotype. We conclude that the NOD-scid IL2Rgamma(null) tumor xenograft model provides an opportunity to study tumor and tumor-stromal cell interactions in situ for prolonged periods.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/imunologia , Memória Imunológica , Neoplasias Pulmonares/imunologia , Linfócitos do Interstício Tumoral/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T/imunologia , Transferência Adotiva , Animais , Carcinoma Pulmonar de Células não Pequenas/patologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Imunofenotipagem , Interferon gama/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , Receptores de Interleucina-2/metabolismo , Transplante Heterólogo
12.
J Immunol ; 178(9): 5552-62, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17442937

RESUMO

The tumor microenvironment of human non-small cell lung cancer (NSCLC) is composed largely of stromal cells, including fibroblasts, yet these cells have been the focus of few studies. In this study, we established stromal cell cultures from primary NSCLC through isolation of adherent cells. Characterization of these cells by flow cytometry demonstrated a population which expressed a human fibroblast-specific 112-kDa surface molecule, Thy1, alpha-smooth muscle actin, and fibroblast activation protein, but failed to express CD45 and CD11b, a phenotype consistent with that of an activated myofibroblast. A subset of the tumor-associated fibroblasts (TAF) was found to express B7H1 (PD-L1) and B7DC (PD-L2) constitutively, and this expression was up-regulated by IFN-gamma. Production of cytokines and chemokines, including IFN-gamma, monokine induced by IFN-gamma, IFN-gamma-inducible protein-10, RANTES, and TGF-beta1 was also demonstrated in these cells. Together, these characteristics provide multiple opportunities for the TAF to influence cellular interactions within the tumor microenvironment. To evaluate the ability of TAF to modulate tumor-associated T cell (TAT) activation, we conducted coculture experiments between autologous TAF and TAT. In five of eight tumors, TAF elicited a contact-dependent enhancement of TAT activation, even in the presence of a TGF-beta1-mediated suppressive effect. In the three other tumors, TAF had a net suppressive effect upon TAT activation, and, in one of these cases, blockade of B7H1 or B7DC was able to completely abrogate the TAF-mediated suppression. We conclude that TAF in human NSCLC are functionally and phenotypically heterogeneous and provide multiple complex regulatory signals that have the potential to enhance or suppress TAT function in the tumor microenvironment.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/imunologia , Fibroblastos/imunologia , Neoplasias Pulmonares/imunologia , Linfócitos T/imunologia , Antígenos CD/análise , Antígenos CD/metabolismo , Antígeno B7-1/análise , Antígeno B7-1/metabolismo , Antígeno B7-H1 , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Quimiocinas/metabolismo , Técnicas de Cocultura , Citocinas/metabolismo , Fibroblastos/patologia , Humanos , Interferon gama/metabolismo , Neoplasias Pulmonares/patologia , Ativação Linfocitária , Proteína 2 Ligante de Morte Celular Programada 1 , Receptores de Antígenos de Linfócitos T/agonistas , Receptores de Antígenos de Linfócitos T/metabolismo , Fator de Crescimento Transformador beta/análise , Fator de Crescimento Transformador beta/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA