Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Nat Immunol ; 15(9): 875-883, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25108421

RESUMO

T cells must be tolerant of self antigens to avoid autoimmunity but responsive to foreign antigens to provide protection against infection. We found that in both naive T cells and effector T cells, the tyrosine phosphatase PTPN22 limited signaling via the T cell antigen receptor (TCR) by weak agonists and self antigens while not impeding responses to strong agonist antigens. T cells lacking PTPN22 showed enhanced formation of conjugates with antigen-presenting cells pulsed with weak peptides, which led to activation of the T cells and their production of inflammatory cytokines. This effect was exacerbated under conditions of lymphopenia, with the formation of potent memory T cells in the absence of PTPN22. Our data address how loss-of-function PTPN22 alleles can lead to the population expansion of effector and/or memory T cells and a predisposition to human autoimmunity.


Assuntos
Autoimunidade/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 22/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Alelos , Animais , Células Apresentadoras de Antígenos/imunologia , Ativação Linfocitária/imunologia , Camundongos
2.
Immunol Cell Biol ; 100(1): 18-20, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34936141

RESUMO

In a new study, researchers identified protein tyrosine phosphatase PTP1B as an inhibitor of cytokine receptor signalling and demonstrated that blocking activity or expression of this enzyme unleashes T cell responses to cancer.


Assuntos
Neoplasias , Linfócitos T , Humanos , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Transdução de Sinais/fisiologia , Linfócitos T/metabolismo
3.
Proc Natl Acad Sci U S A ; 115(7): E1540-E1549, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29386395

RESUMO

Inhibition of immune checkpoints programmed death 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) on T cells results in durable antitumor activity in melanoma patients. Despite high frequency of melanoma brain metastases (BrM) and associated poor prognosis, the activity and mechanisms of immune checkpoint inhibitors (ICI) in metastatic tumors that develop within the "immune specialized" brain microenvironment, remain elusive. We established a melanoma tumor transplantation model with intracranial plus extracranial (subcutaneous) tumor, mimicking the clinically observed coexistence of metastases inside and outside the brain. Strikingly, intracranial ICI efficacy was observed only when extracranial tumor was present. Extracranial tumor was also required for ICI-induced increase in CD8+ T cells, macrophages, and microglia in brain tumors, and for up-regulation of immune-regulatory genes. Combined PD-1/CTLA-4 blockade had a superior intracranial efficacy over the two monotherapies. Cell depletion studies revealed that NK cells and CD8+ T cells were required for intracranial anti-PD-1/anti-CTLA-4 efficacy. Rather than enhancing CD8+ T cell activation and expansion within intracranial tumors, PD-1/CTLA-4 blockade dramatically (∼14-fold) increased the trafficking of CD8+ T cells to the brain. This was mainly through the peripheral expansion of homing-competent effector CD8+ T cells and potentially further enhanced through up-regulation of T cell entry receptors intercellular adhesion molecule 1 and vascular adhesion molecule 1 on tumor vasculature. Our study indicates that extracranial activation/release of CD8+ T cells from PD-1/CTLA-4 inhibition and potentiation of their recruitment to the brain are paramount to the intracranial anti-PD-1/anti-CTLA-4 activity, suggesting augmentation of these processes as an immune therapy-enhancing strategy in metastatic brain cancer.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias Encefálicas/terapia , Linfócitos T CD8-Positivos/imunologia , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/terapia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/imunologia , Animais , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/secundário , Feminino , Granzimas/imunologia , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/imunologia , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/secundário , Neoplasias Cutâneas/terapia , Carga Tumoral , Células Tumorais Cultivadas
4.
Eur J Immunol ; 49(8): 1147-1152, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31270810

RESUMO

The successful implementation of immunotherapies has provided new impetus in the fight against cancer. Antibody-mediated blockade of immune checkpoint molecules PD-1/PD-L1 and CTLA-4 has had a dramatic impact upon the treatment of previously intractable cancers such as malignant melanoma, while adoptive cell therapies using chimeric antigen receptor-bearing T cells have proven highly efficacious in B cell leukemia. Furthermore, significant progress has been made in understanding the mechanisms by which tumors evade or become resistant to these immunotherapies. In this regard, approaches to broaden the applicability and enhance the efficacy of immunotherapies increasingly include modulation of tumor and immune cell metabolism. In this mini-review, we highlight the most recent studies describing novel approaches and targets for the manipulation of the tumor microenvironment and T cell metabolism and describe how these approaches are being combined with current immunotherapies in preclinical studies.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Imunoterapia/métodos , Neoplasias/terapia , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/metabolismo , Animais , Antígeno B7-H1/antagonistas & inibidores , Antígeno CTLA-4/antagonistas & inibidores , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Humanos , Neoplasias/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptores de Antígenos Quiméricos/genética , Microambiente Tumoral
5.
Immunology ; 154(3): 377-382, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29512901

RESUMO

A number of polymorphisms in immune-regulatory genes have been identified as risk factors for the development of autoimmune disease. PTPN22 (that encodes a tyrosine phosphatase) has been associated with the development of several autoimmune diseases, including type 1 diabetes, rheumatoid arthritis and systemic lupus erythematosus. PTPN22 regulates the activity and effector functions of multiple important immune cell types, including lymphocytes, granulocytes and myeloid cells. In this review, we describe the role of PTPN22 in regulating T-cell activation and effector responses. We discuss progress in our understanding of the impact of PTPN22 in autoimmune disease in humans and mouse models, as well as recent evidence suggesting that genetic manipulation of PTPN22 expression might enhance the efficacy of anti-tumour T-cell responses.


Assuntos
Autoimunidade/genética , Imunomodulação , Neoplasias/etiologia , Neoplasias/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 22/genética , Proteína Tirosina Fosfatase não Receptora Tipo 22/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Modelos Animais de Doenças , Suscetibilidade a Doenças , Regulação da Expressão Gênica , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Neoplasias/patologia , Polimorfismo de Nucleotídeo Único
6.
J Immunol ; 197(2): 429-40, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27288531

RESUMO

The cytoplasmic phosphatase, protein tyrosine phosphatase nonreceptor type 22 (PTPN22), is a negative regulator of T cell signaling. Genome-wide association studies have shown that single-nucleotide polymorphisms in PTPN22 confer an increased risk of developing multiple autoimmune diseases in humans. The precise function of PTPN22 and how the variant protein contributes to autoimmunity is not well understood. To address this issue, we investigated the effect of PTPN22 deficiency on disease susceptibility in a mouse model of autoimmune arthritis. The SKG mouse expresses a hypomorphic mutant allele of ZAP70, which, upon exposure to fungal Ags, predisposes the mice to a CD4(+) T cell-mediated autoimmune arthritis that closely resembles rheumatoid arthritis in humans. Surprisingly, SKG Ptpn22(-/-) mice developed less severe mannan-induced arthritis compared with SKG mice. Diminution of disease was not due to significant alterations in thymocyte development or repertoire selection in SKG Ptpn22(-/-) mice, even though T cell-mediated signal transduction was improved. Instead, Ptpn22 deficiency appeared to bias CD4 Th cell differentiation away from the Th17 lineage, which is pathogenic in this setting, to a more Th1/T regulatory-focused response. These data show that even small perturbations in TCR signal transduction pathways can have profound consequences on the differentiation of T cell lineages and thus for the development of autoimmune diseases.


Assuntos
Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Linfócitos T CD4-Positivos/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 22/imunologia , Animais , Western Blotting , Diferenciação Celular/imunologia , Citometria de Fluxo , Mananas/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Mutantes , Reação em Cadeia da Polimerase , Proteína Tirosina Fosfatase não Receptora Tipo 22/deficiência , Receptores de Antígenos de Linfócitos T/imunologia , Células Th17/imunologia
7.
J Immunol ; 195(10): 4615-22, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26453749

RESUMO

Ag-dependent activation of naive T cells induces dramatic changes in cellular metabolism that are essential for cell growth, division, and differentiation. In recent years, the serine/threonine kinase mechanistic target of rapamycin (mTOR) has emerged as a key integrator of signaling pathways that regulate these metabolic processes. However, the role of specific downstream effectors of mTOR function in T cells is poorly understood. Ribosomal protein S6 (rpS6) is an essential component of the ribosome and is inducibly phosphorylated following mTOR activation in eukaryotic cells. In the current work, we addressed the role of phosphorylation of rpS6 as an effector of mTOR function in T cell development, growth, proliferation, and differentiation using knockin and TCR transgenic mice. Surprisingly, we demonstrate that rpS6 phosphorylation is not required for any of these processes either in vitro or in vivo. Indeed, rpS6 knockin mice are completely sensitive to the inhibitory effects of rapamycin and an S6 kinase 1 (S6K1)-specific inhibitor on T cell activation and proliferation. These results place the mTOR complex 1-S6K1 axis as a crucial determinant of T cell activation independently of its ability to regulate rpS6 phosphorylation.


Assuntos
Ativação Linfocitária/imunologia , Complexos Multiproteicos/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Proteína S6 Ribossômica/metabolismo , Linfócitos T/imunologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Ciclo Celular/genética , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Proteínas Quinases S6 Ribossômicas/antagonistas & inibidores , Transdução de Sinais/imunologia , Sirolimo/farmacologia , Linfócitos T/citologia
8.
J Immunol ; 192(5): 2442-8, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24470502

RESUMO

CD4(+) T cells have long been grouped into distinct helper subsets on the basis of their cytokine-secretion profile. In recent years, several subsets of innate lymphoid cell have been described as key producers of these same Th-associated cytokines. However, the functional relationship between Th cells and innate lymphoid cells (ILCs) remains unclear. We show in this study that lineage-negative ST2(+)ICOS(+)CD45(+) type 2 ILCs and CD4(+) T cells can potently stimulate each other's function via distinct mechanisms. CD4(+) T cell provision of IL-2 stimulates type 2 cytokine production by type 2 ILCs. By contrast, type 2 ILCs modulate naive T cell activation in a cell contact-dependent manner, favoring Th2 while suppressing Th1 differentiation. Furthermore, a proportion of type 2 ILCs express MHC class II and can present peptide Ag in vitro. Importantly, cotransfer experiments show that type 2 ILCs also can boost CD4(+) T cell responses to Ag in vivo.


Assuntos
Antígenos de Diferenciação/imunologia , Diferenciação Celular/imunologia , Citocinas/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Imunidade Inata/fisiologia , Células Th2/imunologia , Animais , Antígenos de Diferenciação/genética , Diferenciação Celular/genética , Citocinas/genética , Antígenos de Histocompatibilidade Classe II/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Células Th1/citologia , Células Th1/imunologia , Células Th2/citologia
9.
Trends Immunol ; 33(8): 389-96, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22609147

RESUMO

Interleukin (IL)-33 is a member of the IL-1 cytokine family that has been shown to play an important role in the induction and effector phases of type 2 immune responses. Both innate and adaptive immunity are regulated by IL-33, and many studies have shown disease-associated functions for this cytokine. Recently, IL-33 has been implicated in the function of novel innate lymphocyte populations that regulate both protective responses in parasitic infections and allergic airway inflammation. Here, we discuss recent data highlighting the dual roles of IL-33 in protective and deleterious immune responses.


Assuntos
Imunidade Inata , Interleucinas/imunologia , Tecido Linfoide/imunologia , Animais , Humanos , Transdução de Sinais
10.
Cells ; 13(3)2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38334623

RESUMO

Advances in immunotherapy have brought significant therapeutic benefits to many cancer patients. Nonetheless, many cancer types are refractory to current immunotherapeutic approaches, meaning that further targets are required to increase the number of patients who benefit from these technologies. Protein tyrosine phosphatases (PTPs) have long been recognised to play a vital role in the regulation of cancer cell biology and the immune response. In this review, we summarize the evidence for both the pro-tumorigenic and tumour-suppressor function of non-receptor PTPs in cancer cells and discuss recent data showing that several of these enzymes act as intracellular immune checkpoints that suppress effective tumour immunity. We highlight new data showing that the deletion of inhibitory PTPs is a rational approach to improve the outcomes of adoptive T cell-based cancer immunotherapies and describe recent progress in the development of PTP inhibitors as anti-cancer drugs.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Proteínas Tirosina Fosfatases/metabolismo , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Imunoterapia
11.
Methods Mol Biol ; 2743: 81-92, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38147209

RESUMO

Phosphotyrosine phosphatase non-receptor type 22 (PTPN22) is a key regulator of immune cell activation and responses. Genetic polymorphisms of PTPN22 have been strongly linked with an increased risk of developing autoimmune diseases, while analysis of PTPN22-deficient mouse strains has determined that PTPN22 serves as a negative regulator of T cell antigen receptor signaling. As well as these key roles in maintaining immune tolerance, PTPN22 acts as an intracellular checkpoint for T cell responses to cancer, suggesting that PTPN22 might be a useful target to improve T cell immunotherapies. To assess the potential for targeting PTPN22, we have crossed Ptpn22-deficient mice to an OT-I TCR transgenic background and used adoptive T cell transfer approaches in mouse cancer models. We provide basic methods for the in vitro expansion of effector OT-I cytotoxic T lymphocytes, in vitro phenotypic analysis, and in vivo adoptive T cell transfer models to assess the role of PTPN22 in anti-cancer immunity.


Assuntos
Neoplasias , Receptores de Antígenos de Linfócitos T , Animais , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/genética , Neoplasias/genética , Neoplasias/terapia , Transdução de Sinais , Modelos Animais de Doenças , Proteína Tirosina Fosfatase não Receptora Tipo 22/genética
12.
J Invest Dermatol ; 2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38583742

RESUMO

Peripheral blood transcriptomes from 383 patients with newly diagnosed melanoma were subjected to differential gene expression analysis. The hypotheses were that impaired systemic immunity is associated with poorer prognosis (thicker tumors and fewer tumor-infiltrating lymphocytes) and evidence of systemic inflammation (high-sensitivity CRP and fibrinogen levels). Higher fibrinogen levels were associated with thicker primary tumors. In single-gene analysis, high-sensitivity CRP levels were significantly associated with higher blood CD274 expression (coding for PD-L1), but each was independently prognostic, with high-sensitivity CRP associated with increased mortality and higher CD274 protective, independent of age. Pathway analysis identified downregulation of immune cell signaling pathways in the blood of people with thicker tumors and notable upregulation of signal transducer and activator of transcription 1 gene STAT1 in people with brisk tumor-infiltrating lymphocytes. Transcriptomic data provided evidence for increased NF-kB signaling with higher inflammatory markers but with reduction in expression of HLA class II molecules and higher CD274, suggesting that aberrant systemic inflammation is a significant mediator of reduced immune function in melanoma. In summary, transcriptomic data revealed evidence of reduced immune function in patients with thicker tumors and fewer tumor-infiltrating lymphocytes at diagnosis. Inflammatory markers were associated with thicker primaries and independently with death from melanoma, suggesting that systemic inflammation contributes to that reduced immune function.

13.
Blood ; 117(1): 108-17, 2011 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-20876849

RESUMO

T-cell development is critically dependent on the activities of the Src-family kinases p56(lck) and p59(fyn). While Lck plays a dominant role in the initiation of T-cell receptor (TCR) signaling and in thymocyte differentiation, Fyn plays a more subtle regulatory role. We sought to determine the role of intracellular localization in the differing functions of Lck and Fyn in T cells. By generating transgenic mice that express chimeric Lck-Fyn proteins, we showed that the N-terminal unique domain determines the intracellular localization and function of Lck in pre-TCR and mature αßTCR signaling in vivo. Furthermore, coexpression of a "domain-swap" Lck protein containing the Fyn unique domain with an inducible Lck transgene resulted in the development of thymomas. In contrast to previous reports of Lck-driven thymomas, tumor development was dependent on either pre-TCR or mature TCR signals, and was completely ablated when mice were crossed to a recombination activating gene 1 (Rag1)-deficient background. These data provide a mechanistic basis for the differing roles of Lck and Fyn in T-cell development, and show that intracellular localization as determined by the N-terminal unique domains is critical for Src-family kinase function in vivo.


Assuntos
Diferenciação Celular , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/fisiologia , Proteínas Proto-Oncogênicas c-fyn/fisiologia , Timoma/patologia , Timo/citologia , Animais , Western Blotting , Antígenos CD2/genética , Feminino , Citometria de Fluxo , Humanos , Imunoprecipitação , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , RNA Mensageiro/genética , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Linfócitos T/metabolismo , Timoma/metabolismo , Timo/metabolismo
14.
J Allergy Clin Immunol ; 130(5): 1159-1166.e6, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22738676

RESUMO

BACKGROUND: The IL-1 family cytokine IL-33 is involved in the induction of airway inflammation in allergic patients and after viral infection. Several cell types, including CD4(+) T(H)2 cells and the recently described type 2 innate lymphoid cells (ILCs), are targets for IL-33, yet the mechanisms by which this cytokine modulates their activation are not clear. OBJECTIVES: Our goal was to investigate a role for mammalian target of rapamycin (mTOR) signaling in the activation of T(H)2 and ILC responses and the induction of airway inflammation by IL-33. METHODS: We biochemically determined the effect of IL-33 on mTOR activation in T(H)2 cells and ILCs and examined the effect of this signaling pathway in vivo using a murine model of IL-33-induced lung inflammation. RESULTS: We found that IL-33 induces mTOR activation through p110δ phosphoinositide 3-kinase and that blockade of the mTOR pathway inhibited IL-33-induced IL-5 and IL-13 production by T(H)2 cells and ILCs. Furthermore, use of a ribosomal protein S6 kinase 1 inhibitor implicated a role for ribosomal protein S6 kinase 1 in IL-33-induced mTOR-dependent cytokine production. Intranasal administration of IL-33 to wild-type mice induced airway inflammation, whereas adoptive transfer of wild-type ILCs to IL-33 receptor-deficient (St2(-/-)) mice recapitulated this response. Importantly, coadministration of the mTOR inhibitor rapamycin reduced IL-33-dependent ILC, macrophage, and eosinophil accumulation; cytokine secretion; and mucus deposition in the airways. CONCLUSION: These data reveal a hitherto unrecognized role of mTOR signaling in IL-33-driven, ILC-dependent inflammation in vivo and suggest that manipulation of this pathway might represent a target for therapeutic intervention for airway inflammation.


Assuntos
Interleucinas/administração & dosagem , Pneumonia/tratamento farmacológico , Pneumonia/imunologia , Serina-Treonina Quinases TOR/metabolismo , Células Th2/efeitos dos fármacos , Animais , Células Cultivadas , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Humanos , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/genética , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/genética , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-13/metabolismo , Interleucina-33 , Interleucina-5/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Pneumonia/induzido quimicamente , Receptores de Interleucina/genética , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Th2/imunologia , Células Th2/transplante
15.
Immunol Rev ; 228(1): 9-22, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19290918

RESUMO

T-cell development in the thymus and activation of mature T cells in secondary lymphoid organs requires the ability of cells to respond appropriately to environmental signals at multiple stages of their development. The process of thymocyte selection insures a functional T-cell repertoire, while activation of naive peripheral T cells induces proliferation, gain of effector function, and, ultimately, long-lived T-cell memory. The T-cell immune response is initiated upon engagement of the T-cell receptor (TCR) and coreceptor, CD4 or CD8, by cognate antigen/major histocompatibility complexes presented by antigen-presenting cells. TCR/coreceptor engagement induces the activation of biochemical signaling pathways that, in combination with signals from costimulator molecules and cytokine receptors, direct the outcome of the response. Activation of the src-family kinases p56(lck) (Lck) and p59(fyn) (Fyn) is central to the initiation of TCR signaling pathways. This review focuses on our current understanding of the mechanisms by which these two proteins orchestrate T-cell function.


Assuntos
Tolerância Imunológica , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Linfócitos T/citologia , Diferenciação Celular , Humanos , Ativação Linfocitária
16.
Biology (Basel) ; 12(2)2023 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-36829573

RESUMO

Transforming growth factor beta (TGFß) receptor signalling regulates T cell development, differentiation and effector function. Expression of the immune-associated isoform of this cytokine, TGFß1, is absolutely required for the maintenance of immunological tolerance in both mice and humans, whilst context-dependent TGFß1 signalling regulates the differentiation of both anti- and pro-inflammatory T cell effector populations. Thus, distinct TGFß-dependent T cell responses are implicated in the suppression or initiation of inflammatory and autoimmune diseases. In cancer settings, TGFß signals contribute to the blockade of anti-tumour immune responses and disease progression. Given the key functions of TGFß in the regulation of immune responses and the potential for therapeutic targeting of TGFß-dependent pathways, the mechanisms underpinning these pleiotropic effects have been the subject of much investigation. This review focuses on accumulating evidence suggesting that modulation of T cell metabolism represents a major mechanism by which TGFß influences T cell immunity.

17.
J Immunother Cancer ; 11(12)2023 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-38056892

RESUMO

BACKGROUND: Adoptive cell therapy (ACT) is a promising strategy for treating cancer, yet it faces several challenges such as lack of long-term protection due to T cell exhaustion induced by chronic TCR stimulation in the tumor microenvironment. One benefit of ACT, however, is that it allows for cellular manipulations, such as deletion of the phosphotyrosine phosphatase non-receptor type 22 (PTPN22), which improves CD8+ T cell antitumor efficacy in ACT. We tested whether Ptpn22KO cytolytic T cells (CTLs) were also more effective than Ptpn22WT CTL in controlling tumors in scenarios that favor T cell exhaustion. METHODS: Tumor control by Ptpn22WT and Ptpn22KO CTL was assessed following adoptive transfer of low numbers of CTL to mice with subcutaneously implanted MC38 tumors. Tumor infiltrating lymphocytes were isolated for analysis of effector functions. An in vitro assay was established to compare CTL function in response to acute and chronic restimulation with antigen-pulsed tumor cells. The expression of effector and exhaustion-associated proteins by Ptpn22WT and Ptpn22KO T cells was followed over time in vitro and in vivo using the ID8 tumor model. Finally, the effect of PD-1 and TIM-3 blockade on Ptpn22KO CTL tumor control was assessed using monoclonal antibodies and CRISPR/Cas9-mediated knockout. RESULTS: Despite having improved effector function at the time of transfer, Ptpn22KO CTL became more exhausted than Ptpn22WT CTL, characterized by more rapid loss of effector functions, and earlier and higher expression of inhibitory receptors (IRs), particularly the terminal exhaustion marker TIM-3. TIM-3 expression, under the control of the transcription factor NFIL3, was induced by IL-2 signaling which was enhanced in Ptpn22KO cells. Antitumor responses of Ptpn22KO CTL were improved following PD-1 blockade in vivo, yet knockout or antibody-mediated blockade of TIM-3 did not improve but further impaired tumor control, indicating TIM-3 signaling itself did not drive the diminished function seen in Ptpn22KO CTL. CONCLUSIONS: This study questions whether TIM-3 plays a role as an IR and highlights that genetic manipulation of T cells for ACT needs to balance short-term augmented effector function against the risk of T cell exhaustion in order to achieve longer-term protection.


Assuntos
Receptor Celular 2 do Vírus da Hepatite A , Neoplasias , Camundongos , Animais , Receptor de Morte Celular Programada 1 , Exaustão das Células T , Proteínas Tirosina Fosfatases , Terapia Baseada em Transplante de Células e Tecidos , Microambiente Tumoral
18.
Eur J Immunol ; 41(8): 2137-41, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21725971

RESUMO

The mammalian target of rapamycin (mTOR) integrates signalling responses to growth factors and nutrients. The macrolide rapamycin inhibits mTOR function and has been used extensively to demonstrate a critical role for mTOR in immune responses. This mini-review summarizes recent evidence demonstrating an integral role for mTOR in the differentiation of T helper cell subsets and the development, maturation and antigen-presenting capacity of DCs in both mice and humans.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Serina-Treonina Quinases TOR/imunologia , Animais , Apresentação de Antígeno/imunologia , Células Dendríticas/metabolismo , Humanos , Camundongos , Modelos Imunológicos , Transdução de Sinais/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Serina-Treonina Quinases TOR/metabolismo
19.
Front Immunol ; 13: 913184, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35958566

RESUMO

T cell activation is dependent upon the integration of antigenic, co-stimulatory and cytokine-derived signals and the availability and acquisition of nutrients from the environment. Furthermore, T cell activation is accompanied by reprogramming of cellular metabolism to provide the energy and building blocks for proliferation, differentiation and effector function. Transforming growth factor ß (TGFß) has pleiotropic effects on T cell populations, having both an essential role in the maintenance of immune tolerance but also context-dependent pro-inflammatory functions. We set out to define the mechanisms underpinning the suppressive effects of TGFß on mouse CD8+ T cell activation. RNA-sequencing analysis of TCR-stimulated T cells determined that Myc-regulated genes were highly enriched within gene sets downregulated by TGFß. Functional analysis demonstrated that TGFß impeded TCR-induced upregulation of amino acid transporter expression, amino acid uptake and protein synthesis. Furthermore, TCR-induced upregulation of Myc-dependent glycolytic metabolism was substantially inhibited by TGFß treatment with minimal effects on mitochondrial respiration. Thus, our data suggest that inhibition of Myc-dependent metabolic reprogramming represents a major mechanism underpinning the suppressive effects of TGFß on CD8+ T cell activation.


Assuntos
Linfócitos T CD8-Positivos , Fator de Crescimento Transformador beta , Animais , Citocinas/metabolismo , Ativação Linfocitária , Camundongos , Receptores de Antígenos de Linfócitos T/metabolismo , Fator de Crescimento Transformador beta/metabolismo
20.
J Immunol ; 183(11): 7388-97, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19917692

RESUMO

Ribosomal protein S6 (rpS6) is a key component of the translational machinery in eukaryotic cells and is essential for ribosome biogenesis. rpS6 is phosphorylated on evolutionarily conserved serine residues, and data indicate that rpS6 phosphorylation might regulate cell growth and protein synthesis. Studies in cell lines have shown an important role for the serine kinase mammalian target of rapamycin (mTOR) in rpS6 phosphorylation, further linking rpS6 to control of cellular metabolism. rpS6 is essential in T cells because its deletion in mouse double-positive thymocyte cells results in a complete block in T cell development; however, the signaling pathway leading to rpS6 phosphorylation downstream of TCR stimulation has yet to be fully characterized. We show that maximal TCR-induced rpS6 phosphorylation in CD8 T cells requires both Lck and Fyn activity and downstream activation of PI3K, mTOR, and MEK/ERK MAPK pathways. We demonstrate that there is cross-talk between the PI3K and MAPK pathways as well as PI3K-independent mTOR activity, which result in differential phosphorylation of specific rpS6 serine residues. These results place rpS6 phosphorylation as a point of convergence for multiple crucial signaling pathways downstream of TCR triggering.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proteínas de Transporte/imunologia , Proteínas Quinases Ativadas por Mitógeno/imunologia , Fosfatidilinositol 3-Quinases/imunologia , Fosfotransferases (Aceptor do Grupo Álcool)/imunologia , Proteínas Quinases S6 Ribossômicas/imunologia , Transdução de Sinais/imunologia , Animais , Western Blotting , Linfócitos T CD8-Positivos/metabolismo , Proteínas de Transporte/metabolismo , Citometria de Fluxo , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/imunologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Proto-Oncogênicas c-fyn/imunologia , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Receptor Cross-Talk , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Quinases S6 Ribossômicas/metabolismo , Serina-Treonina Quinases TOR
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA