Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Immunol Res ; 11(11): 1508-1523, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37649096

RESUMO

Chimeric antigen receptor (CAR) T cells have shown promising results in the treatment of B-cell malignancies. Despite the successes, challenges remain. One of them directly involves the CAR T-cell manufacturing process and especially the ex vivo activation phase. While this is required to allow infection and expansion, ex vivo activation dampens the antitumor potential of CAR T cells. Optimizing the nature of the T cells harboring the CAR is a strategy to address this obstacle and has the potential to improve CAR T-cell therapy, including for solid tumors. Here, we describe a protocol to create CAR T cells without ex vivo preactivation by inhibiting the transcription factor FOXO1 (CAR TAS cells). This approach made T cells directly permissive to lentiviral infection, allowing CAR expression, with enhanced antitumor functions. FOXO1 inhibition in primary T cells (TAS cells) correlated with acquisition of a stem cell memory phenotype, high levels of granzyme B, and increased production of TNFα. TAS cells displayed enhanced proliferative and cytotoxic capacities as well as improved migratory properties. In vivo experiments showed that CAR TAS cells were more efficient at controlling solid tumor growth than classical CAR T cells. The production of CAR TAS from patients' cells confirmed the feasibility of the protocol in clinic.


Assuntos
Imunoterapia Adotiva , Linfócitos T , Humanos , Linhagem Celular Tumoral , Imunoterapia Adotiva/métodos , Fenótipo , Proteína Forkhead Box O1/metabolismo
2.
J Immunol ; 205(9): 2499-2510, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32978282

RESUMO

Glycosylation with O-linked ß-N-acetylglucosamine (O-GlcNAcylation) is a reversible posttranslational modification that regulates the activity of intracellular proteins according to glucose availability and its metabolism through the hexosamine biosynthesis pathway. This modification has been involved in the regulation of various immune cell types, including macrophages. However, little is known concerning the mechanisms that regulate the protein O-GlcNAcylation level in these cells. In the present work, we demonstrate that LPS treatment induces a marked increase in protein O-GlcNAcylation in RAW264.7 cells, bone marrow-derived and peritoneal mouse macrophages, as well as human monocyte-derived macrophages. Targeted deletion of OGT in macrophages resulted in an increased effect of LPS on NOS2 expression and cytokine production, suggesting that O-GlcNAcylation may restrain inflammatory processes induced by LPS. The effect of LPS on protein O-GlcNAcylation in macrophages was associated with an increased expression and activity of glutamine fructose 6-phosphate amidotransferase (GFAT), the enzyme that catalyzes the rate-limiting step of the hexosamine biosynthesis pathway. More specifically, we observed that LPS potently stimulated GFAT2 isoform mRNA and protein expression. Genetic or pharmacological inhibition of FoxO1 impaired the LPS effect on GFAT2 expression, suggesting a FoxO1-dependent mechanism. We conclude that GFAT2 should be considered a new LPS-inducible gene involved in regulation of protein O-GlcNAcylation, which permits limited exacerbation of inflammation upon macrophage activation.


Assuntos
Acetilglucosamina/metabolismo , Glutamina-Frutose-6-Fosfato Transaminase (Isomerizante)/metabolismo , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Animais , Vias Biossintéticas/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Glicosilação/efeitos dos fármacos , Humanos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Células RAW 264.7
4.
Cancer Immunol Res ; 7(11): 1849-1863, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31527069

RESUMO

ß-Adrenergic receptor (ß-AR) signaling exerts protumoral effects by acting directly on tumor cells and angiogenesis. In addition, ß-AR expression on immune cells affects their ability to mount antitumor immune responses. However, how ß-AR signaling impinges antitumor immune responses is still unclear. Using a mouse model of vaccine-based immunotherapy, we showed that propranolol, a nonselective ß-blocker, strongly improved the efficacy of an antitumor STxBE7 vaccine by enhancing the frequency of CD8+ T lymphocytes infiltrating the tumor (TIL). However, propranolol had no effect on the reactivity of CD8+ TILs, a result further strengthened by ex vivo experiments showing that these cells were insensitive to adrenaline- or noradrenaline-induced AR signaling. In contrast, naïve CD8+ T-cell activation was strongly inhibited by ß-AR signaling, and the beneficial effect of propranolol mainly occurred during CD8+ T-cell priming in the tumor-draining lymph node. We also demonstrated that the differential sensitivity of naïve CD8+ T cells and CD8+ TILs to ß-AR signaling was linked to a strong downregulation of ß2-AR expression related to their activation status, since in vitro-activated CD8+ T cells behaved similarly to CD8+ TILs. These results revealed that ß-AR signaling suppresses the initial priming phase of antitumor CD8+ T-cell responses, providing a rationale to use clinically available ß-blockers in patients to improve cancer immunotherapies.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Vacinas Anticâncer/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Antagonistas Adrenérgicos beta/uso terapêutico , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer/uso terapêutico , Células Cultivadas , Imunoterapia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Propranolol/farmacologia , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais/efeitos dos fármacos
5.
Nat Commun ; 10(1): 4131, 2019 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-31511510

RESUMO

Type I interferons (IFN) are being rediscovered as potent anti-tumoral agents. Activation of the STimulator of INterferon Genes (STING) by DMXAA (5,6-dimethylxanthenone-4-acetic acid) can induce strong production of IFNα/ß and rejection of transplanted primary tumors. In the present study, we address whether targeting STING with DMXAA also leads to the regression of spontaneous MMTV-PyMT mammary tumors. We show that these tumors are refractory to DMXAA-induced regression. This is due to a blockade in the phosphorylation of IRF3 and the ensuing IFNα/ß production. Mechanistically, we identify TGFß, which is abundant in spontaneous tumors, as a key molecule limiting this IFN-induced tumor regression by DMXAA. Finally, blocking TGFß restores the production of IFNα by activated MHCII+ tumor-associated macrophages, and enables tumor regression induced by STING activation. On the basis of these findings, we propose that type I IFN-dependent cancer therapies could be greatly improved by combinations including the blockade of TGFß.


Assuntos
Interferon-alfa/metabolismo , Interferon beta/metabolismo , Neoplasias Mamárias Animais/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Feminino , Fator Regulador 3 de Interferon/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Vírus do Tumor Mamário do Camundongo/metabolismo , Camundongos , Fosforilação/efeitos dos fármacos , Xantonas/farmacologia
6.
Nat Commun ; 10(1): 3345, 2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-31350404

RESUMO

Neuropilin-1 (Nrp-1) is a marker for murine CD4+FoxP3+ regulatory T (Treg) cells, a subset of human CD4+ Treg cells, and a population of CD8+ T cells infiltrating certain solid tumours. However, whether Nrp-1 regulates tumour-specific CD8 T-cell responses is still unclear. Here we show that Nrp-1 defines a subset of CD8+ T cells displaying PD-1hi status and infiltrating human lung cancer. Interaction of Nrp-1 with its ligand semaphorin-3A inhibits migration and tumour-specific lytic function of cytotoxic T lymphocytes. In vivo, Nrp-1+PD-1hi CD8+ tumour-infiltrating lymphocytes (TIL) in B16F10 melanoma are enriched for tumour-reactive T cells exhibiting an exhausted state, expressing Tim-3, LAG-3 and CTLA-4 inhibitory receptors. Anti-Nrp-1 neutralising antibodies enhance the migration and cytotoxicity of Nrp-1+PD-1hi CD8+ TIL ex vivo, while in vivo immunotherapeutic blockade of Nrp-1 synergises with anti-PD-1 to enhance CD8+ T-cell proliferation, cytotoxicity and tumour control. Thus, Nrp-1 could be a target for developing combined immunotherapies.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunoterapia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/terapia , Neuropilina-1/imunologia , Animais , Movimento Celular , Feminino , Humanos , Imunidade Celular , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/fisiopatologia , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Neuropilina-1/genética , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/imunologia , Semaforina-3A/imunologia , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/imunologia
7.
PLoS Pathog ; 15(5): e1007669, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31042779

RESUMO

HIV-1 is dependent on the host cell for providing the metabolic resources for completion of its viral replication cycle. Thus, HIV-1 replicates efficiently only in activated CD4+ T cells. Barriers preventing HIV-1 replication in resting CD4+ T cells include a block that limits reverse transcription and also the lack of activity of several inducible transcription factors, such as NF-κB and NFAT. Because FOXO1 is a master regulator of T cell functions, we studied the effect of its inhibition on T cell/HIV-1 interactions. By using AS1842856, a FOXO1 pharmacologic inhibitor, we observe that FOXO1 inhibition induces a metabolic activation of T cells with a G0/G1 transition in the absence of any stimulatory signal. One parallel outcome of this change is the inhibition of the activity of the HIV restriction factor SAMHD1 and the activation of the NFAT pathway. FOXO1 inhibition by AS1842856 makes resting T cells permissive to HIV-1 infection. In addition, we found that FOXO1 inhibition by either AS1842856 treatment or upon FOXO1 knockdown induces the reactivation of HIV-1 latent proviruses in T cells. We conclude that FOXO1 has a central role in the HIV-1/T cell interaction and that inhibiting FOXO1 with drugs such as AS1842856 may be a new therapeutic shock-and-kill strategy to eliminate the HIV-1 reservoir in human T cells.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Proteína Forkhead Box O1/antagonistas & inibidores , Regulação da Expressão Gênica , Infecções por HIV/virologia , HIV-1/imunologia , Ativação Viral/imunologia , Replicação Viral , Animais , Linfócitos T CD4-Positivos/virologia , Ciclo Celular , Proteína Forkhead Box O1/genética , Infecções por HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/metabolismo , Humanos , Células Jurkat , Ativação Linfocitária/imunologia , Macaca fascicularis , Masculino , Latência Viral
8.
Front Immunol ; 9: 852, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29780382

RESUMO

If misregulated, macrophage (Mϕ)-T cell interactions can drive chronic inflammation thereby causing diseases, such as rheumatoid arthritis (RA). We report that in a proinflammatory environment, granulocyte-Mϕ (GM-CSF)- and Mϕ colony-stimulating factor (M-CSF)-dependent Mϕs have dichotomous effects on T cell activity. While GM-CSF-dependent Mϕs show a highly stimulatory activity typical for M1 Mϕs, M-CSF-dependent Mϕs, marked by folate receptor ß (FRß), adopt an immunosuppressive M2 phenotype. We find the latter to be caused by the purinergic pathway that directs release of extracellular ATP and its conversion to immunosuppressive adenosine by co-expressed CD39 and CD73. Since we observed a misbalance between immunosuppressive and immunostimulatory Mϕs in human and murine arthritic joints, we devised a new strategy for RA treatment based on targeted delivery of a novel methotrexate (MTX) formulation to the immunosuppressive FRß+CD39+CD73+ Mϕs, which boosts adenosine production and curtails the dominance of proinflammatory Mϕs. In contrast to untargeted MTX, this approach leads to potent alleviation of inflammation in the murine arthritis model. In conclusion, we define the Mϕ extracellular purine metabolism as a novel checkpoint in Mϕ cell fate decision-making and an attractive target to control pathological Mϕs in immune-mediated diseases.


Assuntos
Artrite Reumatoide/imunologia , Diferenciação Celular , Macrófagos/imunologia , Macrófagos/metabolismo , Purinas/metabolismo , Adenosina/imunologia , Animais , Artrite Reumatoide/tratamento farmacológico , Proliferação de Células , Modelos Animais de Doenças , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Imunossupressores/administração & dosagem , Imunossupressores/uso terapêutico , Inflamação/tratamento farmacológico , Fator Estimulador de Colônias de Macrófagos/farmacologia , Masculino , Metotrexato/administração & dosagem , Metotrexato/uso terapêutico , Camundongos , Monócitos/efeitos dos fármacos , Líquido Sinovial/citologia , Líquido Sinovial/imunologia
9.
Cancer Res ; 77(24): 7072-7082, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29021139

RESUMO

CD8+/CD103+ tissue-resident memory T cells (TRM cells) accumulate in several human solid tumors, where they have been associated with a favorable prognosis. However, the role of CD103, the α subunit of the integrin αEß7 (also known as CD103), in the retention and functions of these TRM is undefined. In this report, we investigated the role of CD103 cytoplasmic domain and the focal adhesion-associated protein paxillin (Pxn) in downstream signaling and functional activities triggered through αE/CD103 chain. Binding to immobilized recombinant (r)E-cadherin-Fc of CD103 integrin expressed on tumor-specific CTL clones promotes phosphorylation of Pxn and Pyk2 and binding of Pxn to the αE/CD103 subunit tail. Inhibition of Pxn phosphorylation by the Src inhibitor saracatinib or its knockdown via shRNA dramatically altered adhesion and spreading of freshly isolated CD8+/CD103+ lung tumor-infiltrating lymphocytes and CD103+ tumor-specific CTL clones. Inhibition of Pxn phosphorylation with saracatinib in these CTL clones also severely compromised their functional activities toward autologous tumor cells. Using Jurkat T cells as a model to study CD103 integrin activation, we demonstrated a key role of serine residue S1163 of the αE chain intracellular domain in polarization of CD103 and recruitment of lysosomes and Pxn at the contact zone of T lymphocytes with rE-cadherin-Fc-coated beads. Overall, our results show how Pxn binding to the CD103 cytoplasmic tail triggers αEß7 integrin outside-in signaling that promotes CD8+ T-cell migratory behavior and effector functions. These results also explain the more favorable prognosis associated with retention of TRM cells in the tumor microenvironment. Cancer Res; 77(24); 7072-82. ©2017 AACR.


Assuntos
Antígenos CD/metabolismo , Linfócitos T CD8-Positivos , Adesão Celular , Citotoxicidade Imunológica/fisiologia , Cadeias alfa de Integrinas/metabolismo , Linfócitos do Interstício Tumoral , Paxilina/metabolismo , Antígenos CD/química , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Citoplasma/metabolismo , Células HEK293 , Humanos , Memória Imunológica/fisiologia , Cadeias alfa de Integrinas/química , Células Jurkat , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Microambiente Tumoral/imunologia
10.
Oncotarget ; 7(39): 63215-63225, 2016 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-27556504

RESUMO

Cell quiescence is controlled by regulated genome-encoded programs that actively express genes which are often down-regulated or inactivated in transformed cells. Among them is FoxO1, a transcription factor that imposes quiescence in several cell types, including T lymphocytes. In these cells, the FAM65B encoding gene is a major target of FOXO1. Here, we show that forced expression of FAM65B in transformed cells blocks their mitosis because of a defect of the mitotic spindle, leading to G2 cell cycle arrest and apoptosis. Upon cell proliferation arrest, FAM65B is engaged in a complex containing two proteins well known to be involved in cell proliferation i.e. the HDAC6 deacetylase and the 14.3.3 scaffolding protein. In primary T cells, FAM65B is down-regulated upon T cell receptor engagement, and maintaining its expression blocks their proliferation, establishing that the decrease of FAM65B expression is required for proliferation. Conversely, inhibiting FAM65B expression in naive T lymphocytes decreases their activation threshold. These results identify FAM65B as a potential new target for controlling proliferation of both transformed and normal cells.


Assuntos
Proliferação de Células , Proteína Forkhead Box O1/metabolismo , Proteínas/metabolismo , Linfócitos T/citologia , Moléculas de Adesão Celular , Ciclo Celular , Linhagem Celular Transformada , Linhagem Celular Tumoral , Células Cultivadas , Regulação para Baixo , Fase G2 , Regulação da Expressão Gênica , Humanos , Leucemia/metabolismo , Mitose , Fosforilação , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Fuso Acromático/metabolismo , Fatores de Transcrição/metabolismo
11.
J Biomed Nanotechnol ; 11(12): 2243-52, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26510317

RESUMO

Methotrexate is the first line of treatment of rheumatoid arthritis. Since many patients become unresponsive to methotrexate treatment, only very expensive biological therapies are effective and increased methotrexate tolerance strategies need to be identified. Here we propose the encapsulation of methotrexate in a new liposomal formulation using a hydrophobic fragment of surfactant protein conjugated to a linker and folate to enhance their tolerance and efficacy. In this study we aim to evaluate the efficiency of this system to treat rheumatoid arthritis, by targeting folate receptor ß present at the surface of activated macrophages, key effector cells in this pathology. The specificity of our liposomal formulation to target folate receptor ß was investigated both in vitro as in vivo using a mouse model of arthritis (collagen-induced arthritis in DBA/1J mice strain). In both systems, the liposomal constructs were shown to be highly specific and efficient in targeting folate receptor ß. These liposomal formulations also significantly increase the clinical benefit of the encapsulated methotrexate in vivo in arthritic mice, together with reduced expression of CD39 and CD73 ectonucleotidases by joint-infiltrating macrophages. Thus, our formulation might be a promising cost effective way to treat rheumatoid arthritis and delay or reduce methotrexate intolerance.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Ácido Fólico/química , Metotrexato/efeitos adversos , Metotrexato/farmacologia , Animais , Linhagem Celular , Receptores de Folato com Âncoras de GPI/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Lipossomos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Metotrexato/administração & dosagem , Metotrexato/uso terapêutico , Camundongos
12.
J Immunol ; 195(4): 1791-803, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26170390

RESUMO

An uncontrolled exaggerated Th17 response can drive the onset of autoimmune and inflammatory diseases. In this study, we show that, in T cells, Foxo1 is a negative regulator of the Th17 program. Using mixed bone marrow chimeras and Foxo1-deficient mice, we demonstrate that this control is effective in vivo, as well as in vitro during differentiation assays of naive T cells with specific inhibitor of Foxo1 or inhibitors of the PI3K/Akt pathway acting upstream of Foxo1. Consistently, expressing this transcription factor in T cells strongly decreases Th17 generation in vitro as well as transcription of both IL-17A and IL-23R RORγt-target genes. Finally, at the molecular level, we demonstrate that Foxo1 forms a complex with RORγt via its DNA binding domain to inhibit RORγt activity. We conclude that Foxo1 is a direct antagonist of the RORγt-Th17 program acting in a T cell-intrinsic manner.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Subpopulações de Linfócitos T/metabolismo , Células Th17/metabolismo , Animais , Diferenciação Celular/genética , Linhagem Celular , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Humanos , Imunofenotipagem , Interleucina-17/genética , Interleucina-17/metabolismo , Contagem de Linfócitos , Camundongos , Camundongos Knockout , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/antagonistas & inibidores , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/química , Fenótipo , Fosfatidilinositol 3-Quinases/metabolismo , Regiões Promotoras Genéticas , Domínios e Motivos de Interação entre Proteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Células Th17/citologia , Células Th17/imunologia , Transcrição Gênica
13.
Innate Immun ; 21(7): 685-97, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26055819

RESUMO

TLR2 has a prominent role in host defense against a wide variety of pathogens. Stimulation of TLR2 triggers MyD88-dependent signaling to induce NF-κB translocation, and activates a Rac1-PI 3-kinase dependent pathway that leads to transactivation of NF-κB through phosphorylation of the P65 NF-κB subunit. This transactivation pathway involves tyrosine phosphorylations. The role of the tyrosine kinases in TLR signaling is controversial, with discrepancies between studies using only chemical inhibitors and knockout mice. Here, we show the involvement of the tyrosine-kinase Lyn in TLR2-dependent activation of NF-κB in human cellular models, by using complementary inhibition strategies. Stimulation of TLR2 induces the formation of an activation cluster involving TLR2, CD14, PI 3-kinase and Lyn, and leads to the activation of AKT. Lyn-dependent phosphorylation of the p110 catalytic subunit of PI 3-kinase is essential to the control of PI 3-kinase biological activity upstream of AKT and thereby to the transactivation of NF-κB. Thus, Lyn kinase activity is crucial in TLR2-mediated activation of the innate immune response in human mononuclear cells.


Assuntos
NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptor 2 Toll-Like/metabolismo , Quinases da Família src/metabolismo , Células HEK293 , Humanos , Imunidade Inata , Receptores de Lipopolissacarídeos/metabolismo , Complexos Multiproteicos/metabolismo , Fosforilação , Transdução de Sinais , Ativação Transcricional
14.
Sci Transl Med ; 7(289): 289ra85, 2015 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-26019221

RESUMO

Inflammation adversely affects the health of millions of people worldwide, and there is an unmet medical need for better anti-inflammatory drugs. We evaluated the therapeutic interest of mycolactone, a polyketide-derived macrolide produced by Mycobacterium ulcerans. Bacterial production of mycolactone in human skin causes a combination of ulcerative, analgesic, and anti-inflammatory effects. Whereas ulcer formation is mediated by the proapoptotic activity of mycolactone on skin cells via hyperactivation of Wiskott-Aldrich syndrome proteins, analgesia results from neuronal hyperpolarization via signaling through angiotensin II type 2 receptors. Mycolactone also blunts the capacity of immune cells to produce inflammatory mediators by an independent mechanism of protein synthesis blockade. In an attempt to isolate the structural determinants of mycolactone's immunosuppressive activity, we screened a library of synthetic subunits of mycolactone for inhibition of cytokine production by activated T cells. The minimal structure retaining immunosuppressive activity was a truncated version of mycolactone, missing one of the two core-branched polyketide chains. This compound inhibited the inflammatory cytokine responses of human primary cells at noncytotoxic doses and bound to angiotensin II type 2 receptors comparably to mycolactone in vitro. Notably, it was considerably less toxic than mycolactone in human primary dermal fibroblasts modeling ulcerative activity. In mouse models of human diseases, it conferred systemic protection against chronic skin inflammation and inflammatory pain, with no apparent side effects. In addition to establishing the anti-inflammatory potency of mycolactone in vivo, our study therefore highlights the translational potential of mycolactone core-derived structures as prospective immunosuppressants.


Assuntos
Inflamação/tratamento farmacológico , Macrolídeos/uso terapêutico , Animais , Doença Crônica , Células HeLa , Humanos , Imunomodulação , Inflamação/patologia , Células Jurkat , Macrolídeos/química , Camundongos , Mycobacterium ulcerans/fisiologia , Dor/complicações , Dor/tratamento farmacológico , Substâncias Protetoras/uso terapêutico , Acetato de Tetradecanoilforbol/farmacologia
15.
J Autoimmun ; 56: 23-33, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25277651

RESUMO

CD31, a trans-homophilic inhibitory receptor expressed on both T- and B-lymphocytes, drives the mutual detachment of interacting leukocytes. Intriguingly, T cell CD31 molecules relocate to the immunological synapse (IS), where the T and B cells establish a stable interaction. Here, we show that intact CD31 molecules, which are able to drive an inhibitory signal, are concentrated at the periphery of the IS but are excluded from the center of the IS. At this site, were the cells establish the closest contact, the CD31 molecules are cleaved, and most of the extracellular portion of the protein, including the trans-homophilic binding sites, is shed from the cell surface. T cells lacking CD31 trans-homophilic binding sites easily establish stable interactions with B cells; at the opposite, CD31 signaling agonists inhibit T/B IS formation as well as the ensuing helper T cell activation and function. Confocal microscopy and flow cytometry analysis of experimental T/B IS shows that the T cell inhibitory effects of CD31 agonists depend on SHP-2 signaling, which reduces the phosphorylation of ZAP70. The analysis of synovial tissue biopsies from patients affected by rheumatoid arthritis showed that T cell CD31 molecules are excluded from the center of the T/B cell synapses in vivo. Interestingly, the administration of CD31 agonists in vivo significantly attenuated the development of the clinical signs of collagen-induced arthritis in DBA1/J mice. Altogether, our data indicate that the T cell co-inhibitory receptor CD31 prevents the formation of functional T/B immunological synapses and that therapeutic strategies aimed at sustaining CD31 signaling will attenuate the development of autoimmune responses in vivo.


Assuntos
Artrite Experimental/imunologia , Doenças Autoimunes/imunologia , Linfócitos B/imunologia , Sinapses Imunológicas/imunologia , Sinapses Imunológicas/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Idoso , Animais , Artrite Experimental/metabolismo , Doenças Autoimunes/diagnóstico , Doenças Autoimunes/metabolismo , Biópsia , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/imunologia , Linhagem Celular , Feminino , Humanos , Ativação Linfocitária/imunologia , Camundongos , Pessoa de Meia-Idade , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Transdução de Sinais , Membrana Sinovial/imunologia , Membrana Sinovial/patologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Proteína-Tirosina Quinase ZAP-70/metabolismo
16.
J Immunol ; 192(5): 2471-9, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24477908

RESUMO

The interaction of integrin αE(CD103)ß7, often expressed on tumor-infiltrating T lymphocytes, with its cognate ligand, the epithelial cell marker E-cadherin on tumor cells, plays a major role in antitumor CTL responses. CD103 is induced on CD8 T cells upon TCR engagement and exposure to TGF-ß1, abundant within the tumor microenvironment. However, the transcriptional mechanisms underlying the cooperative role of these two signaling pathways in inducing CD103 expression in CD8 T lymphocytes remain unknown. Using a human CTL system model based on a CD8(+)/CD103(-) T cell clone specific of a lung tumor-associated Ag, we demonstrated that the transcription factors Smad2/3 and NFAT-1 are two critical regulators of this process. We also identified promoter and enhancer elements of the human ITGAE gene, encoding CD103, involved in its induction by these transcriptional regulators. Overall, our results explain how TGF-ß1 can participate in CD103 expression on locally TCR-engaged Ag-specific CD8 T cells, thus contributing to antitumor CTL responses and cancer cell destruction.


Assuntos
Antígenos CD/imunologia , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Cadeias alfa de Integrinas/imunologia , Neoplasias Pulmonares/imunologia , Fatores de Transcrição NFATC/imunologia , Proteína Smad2/imunologia , Proteína Smad3/imunologia , Antígenos CD/biossíntese , Antígenos CD/genética , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Células HEK293 , Humanos , Cadeias alfa de Integrinas/biossíntese , Cadeias alfa de Integrinas/genética , Células Jurkat , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Elementos de Resposta/genética , Elementos de Resposta/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/imunologia , Fator de Crescimento Transformador beta1/metabolismo
17.
Oncoimmunology ; 2(1): e22841, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23483035

RESUMO

The destruction of tumor cells by the immune system is under the control of positive and negative receptors that tightly regulate T-cell effector functions. The T-cell receptor (TCR) inhibitory molecule CD5 critically contributes to the regulation of antitumor immune responses. Indeed, the modulation of CD5 within the tumor microenvironment corresponds to a strategy adopted by tumor-specific cytotoxic T lymphocytes (CTLs) to optimize their cytotoxic and cytokine secretion functions. In this review, we provide insights into the immunobiology of CD5 and its role in regulating antitumor CD8 T-cell responses, and suggest the possibility of targeting CD5 to improve the efficacy of current immunotherapeutic approaches against cancer.

18.
J Immunol ; 190(2): 748-55, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23241886

RESUMO

Forkhead box O (FOXO) transcription factors favor both T cell quiescence and trafficking through their control of the expression of genes involved in cell cycle progression, adhesion, and homing. In this article, we report that the product of the fam65b gene is a new transcriptional target of FOXO1 that regulates RhoA activity. We show that family with sequence similarity 65 member b (Fam65b) binds the small GTPase RhoA via a noncanonical domain and represses its activity by decreasing its GTP loading. As a consequence, Fam65b negatively regulates chemokine-induced responses, such as adhesion, morphological polarization, and migration. These results show the existence of a new functional link between FOXO1 and RhoA pathways, through which the FOXO1 target Fam65b tonically dampens chemokine-induced migration by repressing RhoA activity.


Assuntos
Movimento Celular/genética , Fatores de Transcrição Forkhead/metabolismo , Proteínas/genética , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Moléculas de Adesão Celular , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Quimiocinas/farmacologia , Proteína Forkhead Box O1 , Regulação da Expressão Gênica , Humanos , Ligação Proteica , Proteínas/metabolismo , Ativação Transcricional
19.
Eur J Immunol ; 42(1): 195-205, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21956609

RESUMO

The T lineage glycoprotein CD6 is generally considered to be a costimulator of T-cell activation. Here, we demonstrate that CD6 significantly reduces early and late T-cell responses upon superantigen stimulation or TCR triggering by Abs. Measuring calcium mobilization in single cells responding to superantigen, we found that human T cells expressing rat CD6 react significantly less well compared with T cells not expressing the exogenous receptor. When the cytoplasmic domain of rat CD6 was removed, calcium responses were recovered, indicating that the inhibitory properties of CD6 are attributable to its cytoplasmic domain. Calcium responses, and also late indicators of T-cell activation such as IL-2 release, were also diminished in TCR-activated Jurkat cells expressing human CD6, compared with CD6-deficient cells or cells expressing a cytoplasmic deletion mutant of human CD6. Similarly, calcium signals triggered by anti-CD3 were enhanced in human T lymphocytes following morpholino-mediated suppression of CD6 expression. Finally, the proliferation of T lymphocytes was increased when the CD6-CD166 interaction was blocked with anti-CD166 Abs, but inhibited when anti-CD6 Abs were used. Our data suggest that CD6 is a signaling attenuator whose expression alone, i.e. in the absence of ligand engagement, is sufficient to restrain signaling in T cells.


Assuntos
Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos T/imunologia , Cálcio/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Molécula de Adesão de Leucócito Ativado/imunologia , Animais , Complexo CD3/imunologia , Cálcio/análise , Citometria de Fluxo , Humanos , Células Jurkat , Ativação Linfocitária , Ratos , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/citologia , Transfecção
20.
Blood ; 119(1): 95-105, 2012 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-22049514

RESUMO

B lymphocytes can be triggered in lymph nodes by nonopsonized antigens (Ag), potentially in their native form. However, the mechanisms that promote encounter of B lymphocytes with unprocessed antigens in lymph nodes are still elusive. We show here that antigens are detected in B cells in the draining lymph nodes of mice injected with live, but not fixed, dendritic cells (DCs) loaded with antigens. This highlights active processes in DCs to promote Ag transfer to B lymphocytes. In addition, antigen-loaded DCs found in the draining lymph node were CD103+. Using 3 different model Ag, we then show that immature DCs efficiently take up Ag by macropinocytosis and store the internalized material in late endocytic compartments. We find that DCs have a unique ability to release antigens from these compartments in the extracellular medium, which is controlled by Rab27. B cells take up the regurgitated Ag and the chemokine CXCL13, essential to attract B cells in lymph nodes, enhances this transfer. Our results reveal a unique property of DCs to regurgitate unprocessed Ag that could play an important role in B-cell activation.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos/imunologia , Linfócitos B/imunologia , Células Dendríticas/imunologia , Linfonodos/imunologia , Pinocitose/imunologia , Animais , Antígenos CD/metabolismo , Linfócitos B/metabolismo , Western Blotting , Células Cultivadas , Quimiocina CXCL13/metabolismo , Células Dendríticas/metabolismo , Citometria de Fluxo , Humanos , Cadeias alfa de Integrinas/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...