Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 212(12): 1904-1912, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38668728

RESUMO

NK cells have been shown to exhibit inflammatory and immunoregulatory functions in a variety of healthy and diseased settings. In the context of chronic viral infection and cancer, distinct NK cell populations that inhibit adaptive immune responses have been observed. To understand how these cells arise and further characterize their immunosuppressive role, we examined in vitro conditions that could polarize human NK cells into an inhibitory subset. TGF-ß1 has been shown to induce regulatory T cells in vitro and in vivo; we therefore investigated if TGF-ß1 could also induce immunosuppressive NK-like cells. First, we found that TGF-ß1/IL-15, but not IL-15 alone, induced CD103+CD49a+ NK-like cells from peripheral blood NK cells, which expressed markers previously associated with inhibitory CD56+ innate lymphoid cells, including high expression of GITR and CD101. Moreover, supernatant from ascites collected from patients with ovarian carcinoma also induced CD103+CD49a+ NK-like cells in vitro in a TGF-ß-dependent manner. Interestingly, TGF-ß1/IL-15-induced CD103+CD56+ NK-like cells suppressed autologous CD4+ T cells in vitro by reducing absolute number, proliferation, and expression of activation marker CD25. Collectively, these findings provide new insight into how NK cells may acquire an inhibitory phenotype in TGF-ß1-rich environments.


Assuntos
Interleucina-15 , Células Matadoras Naturais , Fator de Crescimento Transformador beta1 , Humanos , Células Matadoras Naturais/imunologia , Interleucina-15/imunologia , Interleucina-15/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Feminino , Antígenos CD/metabolismo , Antígenos CD/imunologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Cadeias alfa de Integrinas/metabolismo , Cadeias alfa de Integrinas/imunologia , Antígeno CD56/metabolismo , Células Cultivadas , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/metabolismo , Ativação Linfocitária/imunologia
2.
Cell Rep Med ; 5(3): 101465, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38460518

RESUMO

The manipulation of T cell metabolism to enhance anti-tumor activity is an area of active investigation. Here, we report that activating the amino acid starvation response in effector CD8+ T cells ex vivo using the general control non-depressible 2 (GCN2) agonist halofuginone (halo) enhances oxidative metabolism and effector function. Mechanistically, we identified autophagy coupled with the CD98-mTOR axis as key downstream mediators of the phenotype induced by halo treatment. The adoptive transfer of halo-treated CD8+ T cells into tumor-bearing mice led to robust tumor control and curative responses. Halo-treated T cells synergized in vivo with a 4-1BB agonistic antibody to control tumor growth in a mouse model resistant to immunotherapy. Importantly, treatment of human CD8+ T cells with halo resulted in similar metabolic and functional reprogramming. These findings demonstrate that activating the amino acid starvation response with the GCN2 agonist halo can enhance T cell metabolism and anti-tumor activity.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias , Humanos , Animais , Camundongos , Imunoterapia Adotiva/métodos , Neoplasias/patologia , Imunoterapia , Aminoácidos
3.
Nat Commun ; 15(1): 1094, 2024 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-38321065

RESUMO

Immunotherapies targeting PD-1/PD-L1 are now widely used in the clinic to treat a variety of malignancies. While most of the research on T cell exhaustion and PD-1 blockade has been focused on conventional αß T cells, the contribution of innate-like T cells such as γδ T cells to anti-PD-1/PD-L1 mediated therapy is limited. Here we show that tumor reactive γδ T cells respond to PD-1 blockade in a Merkel cell carcinoma (MCC) patient experiencing a complete response to therapy. We find clonally expanded γδ T cells in the blood and tumor after pembrolizumab treatment, and this Vγ2Vδ1 clonotype recognizes Merkel cancer cells in a TCR-dependent manner. Notably, the intra-tumoral γδ T cells in the MCC patient are characterized by higher expression of PD-1 and TIGIT, relative to conventional CD4 and CD8 T cells. Our results demonstrate that innate-like T cells could also contribute to an anti-tumor response after PD-1 blockade.


Assuntos
Carcinoma de Célula de Merkel , Neoplasias Cutâneas , Humanos , Receptor de Morte Celular Programada 1/metabolismo , Antígeno B7-H1 , Linfócitos T CD8-Positivos/metabolismo , Neoplasias Cutâneas/patologia
4.
Cancer Immunol Res ; 10(4): 437-452, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35181779

RESUMO

Regulatory T cells (Treg) are an integral component of the adaptive immune system that negatively affect antitumor immunity. Here, we investigated the role of the E3 ubiquitin ligase casitas B-lineage lymphoma-b (Cbl-b) in establishing CD8+ T-cell resistance to Treg-mediated suppression to enhance antitumor immunity. Transcriptomic analyses suggested that Cbl-b regulates pathways associated with cytokine signaling and cellular proliferation. We showed that the hypersecretion of IFNγ by Cbl-b-deficient CD8+ T cells selectively attenuated CD8+ T-cell suppression by Tregs. Although IFNγ production by Cbl-b-deficient T cells contributed to phenotypic alterations in Tregs, the cytokine did not attenuate the suppressive function of Tregs. Instead, IFNγ had a profound effect on CD8+ T cells by directly upregulating interferon-stimulated genes and modulating T-cell activation. In murine models of adoptive T-cell therapy, Cbl-b-deficient T cells elicited superior antitumor immune response. Furthermore, Cbl-b-deficient CD8+ T cells were less susceptible to suppression by Tregs in the tumor through the effects of IFNγ. Collectively, this study demonstrates that the hypersecretion of IFNγ serves as a key mechanism by which Cbl-b-deficient CD8+ T cells are rendered resistant to Tregs. See related Spotlight by Wolf and Baier, p. 370.


Assuntos
Linfócitos T Reguladores , Proteínas Adaptadoras de Transdução de Sinal , Animais , Linfócitos T CD8-Positivos , Interferon gama/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-cbl/genética , Proteínas Proto-Oncogênicas c-cbl/metabolismo
5.
Cell Metab ; 33(12): 2415-2427.e6, 2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34879240

RESUMO

Metabolic programming is intricately linked to the anti-tumor properties of T cells. To study the metabolic pathways associated with increased anti-tumor T cell function, we utilized a metabolomics approach to characterize three different CD8+ T cell subsets with varying degrees of anti-tumor activity in murine models, of which IL-22-producing Tc22 cells displayed the most robust anti-tumor activity. Tc22s demonstrated upregulation of the pantothenate/coenzyme A (CoA) pathway and a requirement for oxidative phosphorylation (OXPHOS) for differentiation. Exogenous administration of CoA reprogrammed T cells to increase OXPHOS and adopt the CD8+ Tc22 phenotype independent of polarizing conditions via the transcription factors HIF-1α and the aryl hydrocarbon receptor (AhR). In murine tumor models, treatment of mice with the CoA precursor pantothenate enhanced the efficacy of anti-PDL1 antibody therapy. In patients with melanoma, pre-treatment plasma pantothenic acid levels were positively correlated with the response to anti-PD1 therapy. Collectively, our data demonstrate that pantothenate and its metabolite CoA drive T cell polarization, bioenergetics, and anti-tumor immunity.


Assuntos
Coenzima A , Subpopulações de Linfócitos T , Animais , Linfócitos T CD8-Positivos , Diferenciação Celular , Coenzima A/metabolismo , Humanos , Ativação Linfocitária , Camundongos , Subpopulações de Linfócitos T/metabolismo
6.
PLoS One ; 15(12): e0244366, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33382742

RESUMO

Dendritic cells are sentinels of the immune system and represent a key cell in the activation of the adaptive immune response. Hypoxia-inducible factor 1 alpha (HIF-1α)-a crucial oxygen sensor stabilized during hypoxic conditions-has been shown to have both activating and inhibitory effects in immune cells in a context- and cell-dependent manner. Previous studies have demonstrated that in some immune cell types, HIF-1α serves a pro-inflammatory role. Genetic deletion of HIF-1α in macrophages has been reported to reduce their pro-inflammatory function. In contrast, loss of HIF-1α enhanced the pro-inflammatory activity of dendritic cells in a bacterial infection model. In this study, we aimed to further clarify the effects of HIF-1α in dendritic cells. Constitutive expression of HIF-1α resulted in diminished immunostimulatory capacity of dendritic cells in vivo, while conditional deletion of HIF-1α in dendritic cells enhanced their ability to induce a cytotoxic T cell response. HIF-1α-expressing dendritic cells demonstrated increased production of inhibitory mediators including IL-10, iNOS and VEGF, which correlated with their reduced capacity to drive effector CD8+ T cell function. Altogether, these data reveal that HIF-1α can promote the anti-inflammatory functions of dendritic cells and provides insight into dysfunctional immune responses in the context of HIF-1α activation.


Assuntos
Biomarcadores/metabolismo , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/citologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Animais , Células Cultivadas , Células Dendríticas/metabolismo , Técnicas de Inativação de Genes , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Interleucina-10/metabolismo , Camundongos , Óxido Nítrico Sintase Tipo II/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Oncoimmunology ; 9(1): 1737368, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32313719

RESUMO

Regulatory T cells are integral to the regulation of autoimmune and anti-tumor immune responses. However, several studies have suggested that changes in T cell signaling networks can result in T cells that are resistant to the suppressive effects of regulatory T cells. Here, we investigated the role of Cbl-b, an E3 ubiquitin ligase, in establishing resistance to Treg-mediated suppression. We found that the absence of Cbl-b, a negative regulator of multiple TCR signaling pathways, rendered T cells impartial to Treg suppression by regulating cytokine networks leading to improved anti-tumor immunity despite the presence of Treg cells in the tumor. Specifically, Cbl-b KO CD4+FoxP3- T cells hyper-produced IL-2 and together with IL-2 Rα upregulation served as an essential mechanism to escape suppression by Treg cells. Furthermore, we report that IL-2 serves as the central molecule required for cytokine-induced Treg resistance. Collectively our data emphasize the role of IL-2 as a key mechanism that renders CD4+ T cells resistant to the inhibitory effects of Treg cells.


Assuntos
Interleucina-2 , Linfócitos T Reguladores , Animais , Interleucina-2/genética , Subunidade alfa de Receptor de Interleucina-2 , Camundongos Endogâmicos C57BL , Transdução de Sinais , Linfócitos T Reguladores/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
8.
Cancer Immunol Res ; 8(3): 321-333, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31964625

RESUMO

CD8+ T cells can be polarized into several different subsets as defined by the cytokines they produce and the transcription factors that govern their differentiation. Here, we identified the polarizing conditions to induce an IL22-producing CD8+ Tc22 subset, which is dependent on IL6 and the aryl hydrocarbon receptor transcription factor. Further characterization showed that this subset was highly cytolytic and expressed a distinct cytokine profile and transcriptome relative to other subsets. In addition, polarized Tc22 were able to control tumor growth as well as, if not better than, the traditional IFNγ-producing Tc1 subset. Tc22s were also found to infiltrate the tumors of human patients with ovarian cancer, comprising up to approximately 30% of expanded CD8+ tumor-infiltrating lymphocytes (TIL). Importantly, IL22 production in these CD8+ TILs correlated with improved recurrence-free survival. Given the antitumor properties of Tc22 cells, it may be prudent to polarize T cells to the Tc22 lineage when using chimeric antigen receptor (CAR)-T or T-cell receptor (TCR) transduction-based immunotherapies.


Assuntos
Imunoterapia Adotiva/métodos , Interleucina-6/farmacologia , Interleucinas/imunologia , Linfócitos do Interstício Tumoral/imunologia , Neoplasias Ovarianas/terapia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/imunologia , Polaridade Celular/imunologia , Feminino , Humanos , Interleucina-6/biossíntese , Interleucina-6/genética , Interleucina-6/imunologia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Hidrocarboneto Arílico/imunologia , Proteínas com Domínio T/imunologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/imunologia , Transcriptoma , Células Tumorais Cultivadas , Interleucina 22
9.
Oncoimmunology ; 8(12): e1665460, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31741762

RESUMO

B7-H4, an immune suppressive member of the B7 family, is highly expressed in a wide variety of human malignancies making it an attractive immunotherapeutic target. However, the association between B7-H4 expression in the tumor microenvironment and the immune infiltrate has not been comprehensively examined. To evaluate the immune tumor microenvironment, we analyzed epithelial ovarian tumors from 28 patients using flow cytometry, immunohistochemistry, functional, and genomic analyses. We determined B7-H4 expression patterns and compared the immune infiltrates of tumors with high and low surface expression of B7-H4. Frequencies and phenotypes of tumor and immune cells were determined using multiple flow cytometry panels. Immunohistochemistry was used to analyze cellular infiltration and location. Publicly available datasets were interrogated to determine intratumoral cytokine and chemokine expression. We found that B7-H4 was predominantly expressed by tumor cells in the epithelial ovarian tumor microenvironment. Surface expression of B7-H4 on tumor cells was correlated with higher levels of infiltrating mature antigen-presenting cells. Further, expression of CXCL17, a monocyte and dendritic cell chemoattractant, correlated strongly with B7-H4 expression. T cells expressed activation markers, but T cells expressing a combination of markers associated with T cell activation/exhaustion phenotype were not prevalent. Overall, our data suggest that B7-H4 is associated with a pro-inflammatory tumor microenvironment.

10.
Cancer Res ; 79(3): 445-451, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30573521

RESUMO

Memory CD8+ T cells (Tmem) are superior mediators of adoptive cell therapy (ACT) compared with effector CD8+ T cells (Teff) due to increased persistence in vivo. Underpinning Tmem survival is a shift in cellular metabolism away from aerobic glycolysis towards fatty acid oxidation (FAO). Here we investigated the impact of the peroxisome proliferator-activated receptor (PPAR) agonist GW501516 (GW), an agent known to boost FAO in other tissues, on CD8+ T-cell metabolism, function, and efficacy in a murine ACT model. Via activation of both PPARα and PPARδ/ß, GW treatment increased expression of carnitine palmitoyl transferase 1a, the rate-limiting enzyme of FAO, in activated CD8+ T cells. Using a metabolomics approach, we demonstrated that GW increased the abundance of multiple different acylcarnitines, consistent with enhanced FAO. T cells activated in the presence of GW and inflammatory signals, either mature dendritic cells or IL12, also demonstrated enhanced production of IFNγ and expression of T-bet. Despite high expression of T-bet, a characteristic of short-lived effector cells, GW-treated cells demonstrated enhanced persistence in vivo and superior efficacy in a model of ACT. Collectively, these data identify combined PPARα and PPARδ/ß agonists as attractive candidates for further studies and rapid translation into clinical trials of ACT. SIGNIFICANCE: Dual activation of peroxisome proliferator-activated receptors α and δ improves the efficacy of adoptive cell therapy by reprogramming T-cell metabolism and cytokine expression.


Assuntos
Imunoterapia Adotiva , Inflamação/genética , Neoplasias/genética , PPAR alfa/genética , PPAR delta/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Ácidos Graxos/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Inflamação/imunologia , Inflamação/patologia , Inflamação/terapia , Interferon gama/genética , Interleucina-12/genética , Interleucina-12/imunologia , Metabolismo dos Lipídeos/genética , Camundongos , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Oxirredução , PPAR alfa/agonistas , PPAR delta/agonistas , PPAR beta/agonistas , PPAR beta/genética , Tiazóis/uso terapêutico
11.
Cancer Immunol Res ; 3(2): 184-95, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25527357

RESUMO

The B7 family plays a critical role in both positive and negative regulation of immune responses by engaging a variety of receptors on lymphocytes. Importantly, blocking coinhibitory molecules using antibodies specific for CTLA-4 and PD-1 enhances tumor immunity in a subset of patients. Therefore, it is critical to understand the role of different B7 family members since they may be suitable therapeutic targets. B7-H4 is another member that inhibits T-cell function, and it is also upregulated on a variety of tumors and has been proposed to promote tumor growth. Here, we investigate the role of B7-H4 in tumor development and show that B7-H4 expression inhibits tumor growth in two mouse models. Furthermore, we show that B7-H4 expression is required for antitumor immune responses in a mouse model of mammary tumorigenesis. We found that the expression levels of B7-H4 correlate with MHC class I expression in both mouse and human samples. We show that IFNγ upregulates B7-H4 expression on mouse embryo fibroblasts and that the upregulation of B7-H4 on tumors is dependent on T cells. Notably, patients with breast cancer with increased B7-H4 expression show a prolonged time to recurrence. These studies demonstrate a positive role for B7-H4 in promoting antitumor immunity.


Assuntos
Neoplasias Mamárias Experimentais/imunologia , Microambiente Tumoral/imunologia , Inibidor 1 da Ativação de Células T com Domínio V-Set/imunologia , Animais , Biomarcadores Tumorais/metabolismo , Citotoxicidade Imunológica/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica/imunologia , Granzimas/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Imunidade Celular , Interferon gama/biossíntese , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/prevenção & controle , Camundongos Transgênicos , Proteínas de Neoplasias/imunologia , Linfócitos T Citotóxicos/enzimologia , Linfócitos T Citotóxicos/imunologia , Regulação para Cima/imunologia , Inibidor 1 da Ativação de Células T com Domínio V-Set/deficiência , Inibidor 1 da Ativação de Células T com Domínio V-Set/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA