Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Immunol ; 203(12): 3247-3255, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31722989

RESUMEN

Extracellular adenosine accumulates in tumors and causes suppression of immune cells. Suppressive adenosine signaling is achieved through adenosine A2A and A2B receptors, which are Gs coupled, and their activation elevates cAMP levels. Gs-coupled GPCR signaling causes cAMP accumulation, which plays an anti-inflammatory role in immune cells. Protein kinase A (PKA) and exchange protein directly activated by cAMP (Epac) are two intracellular receptors of cAMP. In this study we showed that adenosine receptor signaling polarizes activated murine dendritic cells (DCs) into a tumor-promoting suppressive phenotype. Adenosine receptor signaling activates cAMP pathway and upregulates the negative regulators of NF-κB but does not influence phosphorylation of immediate inflammatory signaling molecules downstream of TLR signaling. Pharmacologic activation of both PKA and Epac pathways by specific cAMP analogues phenocopied the effects of adenosine signaling on murine DCs, such as suppression of proinflammatory cytokines, elevation of anti-inflammatory IL-10, increased expression of regulators of NF-κB pathway, and finally suppression of T cell activation. Inhibition of effector cytokine, IL-12p40 production, and increased immunosuppressive IL-10 production by adenosine signaling is significantly reversed only when both PKA and Epac pathways were inhibited together. Adenosine signaling increased IL-10 secretion while decreasing IL-12p40 secretion in human monocyte-derived DCs. Stimulation of both PKA and Epac pathways also caused combinatorial effects in regulation of IL-12p40 secretion in human monocyte-derived DCs. Interestingly, PKA signaling alone caused similar increase in IL-10 secretion to that of adenosine signaling in human monocyte-derived DCs. Our data suggest adenosine/cAMP signaling targets both PKA/Epac pathways to fully differentiate DCs into a suppressive phenotype.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Fenotipo , Receptores Purinérgicos P1/metabolismo , Transducción de Señal , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Susceptibilidad a Enfermedades , Femenino , Inmunomodulación , Ratones , Fosforilación , Transducción de Señal/efectos de los fármacos
2.
J Immunol ; 188(1): 198-205, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22116822

RESUMEN

The accumulation of high levels of adenosine in tumors activates A(2A) and A(2B) receptors on immune cells and inhibits their ability to suppress tumor growth. Deletion of adenosine A(2A) receptors (A(2A)ARs) has been reported to activate antitumor T cells, stimulate dendritic cell (DC) function, and inhibit angiogenesis. In this study, we evaluated the effects of intermittent intratumor injection of a nonselective adenosine receptor antagonist, aminophylline (AMO; theophylline ethylenediamine) and, for the first time to our knowledge, a selective A(2B)AR antagonist, ATL801. AMO and ATL801 slowed the growth of MB49 bladder and 4T1 breast tumors in syngeneic mice and reduced by 85% metastasizes of breast cancer cells from mammary fat to lung. Based on experiments with A(2A)AR(-/-) or adenosine A(2B) receptor(-/-) mice, the effect of AMO injection was unexpectedly attributed to A(2B)AR and not to A(2A)AR blockade. AMO and ATL801 significantly increased tumor levels of IFN-γ and the IFN-inducible chemokine CXCL10, which is a ligand for CXCR3. This was associated with an increase in activated tumor-infiltrating CXCR3(+) T cells and a decrease in endothelial cell precursors within tumors. Tumor growth inhibition by AMO or ATL801 was eliminated in CXCR3(-/-) mice and RAG1(-/-) mice that lack mature T cells. In RAG1(-/-) mice, A(2B)AR deletion enhanced CD86 expression on CD11b(-) DCs. Bone marrow chimera experiments demonstrated that CXCR3 and A(2B)AR expression on bone marrow cells is required for the antitumor effects of AMO. The data suggest that blockade of A(2B)ARs enhances DC activation and CXCR3-dependent antitumor responses.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/farmacología , Células Dendríticas/inmunología , Neoplasias Mamarias Animales/tratamiento farmacológico , Receptor de Adenosina A2B/inmunología , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Antígeno B7-2/genética , Antígeno B7-2/inmunología , Antígeno B7-2/metabolismo , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Línea Celular Tumoral , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Quimiocina CXCL10/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/patología , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/inmunología , Proteínas de Homeodominio/metabolismo , Interferón gamma/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/inmunología , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Trasplante de Neoplasias , Receptor de Adenosina A2B/genética , Receptor de Adenosina A2B/metabolismo , Receptores CXCR3/genética , Receptores CXCR3/inmunología , Receptores CXCR3/metabolismo , Quimera por Trasplante/genética , Quimera por Trasplante/inmunología , Quimera por Trasplante/metabolismo , Trasplante Isogénico , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología
3.
Arterioscler Thromb Vasc Biol ; 32(9): 2097-103, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22772752

RESUMEN

Adenosine regulates the interaction between lymphocytes and the vasculature, and is important for controlling lymphocyte trafficking in response to tissue injury or infection. Adenosine can blunt the effects of T cell receptor activation primarily by activating adenosine A(2A) receptors and signaling via cyclic AMP and protein kinase A. Protein kinase A reduces proximal T cell receptor signaling by phosphorylation of C-terminal Src kinase, nuclear factor of activated T cells and cyclic AMP response element-binding protein. Protein kinase A activation can either enhance or inhibit the survival of T cells depending on the strength and duration of signaling. Inducible enzymes such as CD73 and CD39 regulate adenosine formation and degradation in vivo. The extravasation of lymphocytes through blood vessels is influenced by A(2A) receptors-mediated suppression of intercellular adhesion molecule 1 expression on lymphocytes and diminished production of interferon γ and interferon γ-inducible chemokines that are chemotactic to activated lymphocytes. Adenosine also decreases the barrier function of vascular endothelium by activating A(2B)Rs. In sum, adenosine signaling is influenced by tissue inflammation and injury through induction of receptors and enzymes and has generally inhibitory effects on lymphocyte migration into inflamed tissues due to protein kinase A-mediated effects on adhesion molecules, interferon γ production, and endothelial barrier function.


Asunto(s)
Adenosina/metabolismo , Endotelio Vascular/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Animales , Células Presentadoras de Antígenos/metabolismo , Moléculas de Adhesión Celular/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citocinas/metabolismo , Humanos , Rodamiento de Leucocito , Activación de Linfocitos , Receptor de Adenosina A2A/metabolismo , Receptor de Adenosina A2B/metabolismo , Linfocitos T Reguladores/metabolismo
4.
J Immunol ; 186(7): 3858-65, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21339365

RESUMEN

We previously showed that monophosphoryl lipid A (MLA) activates TLR4 in dendritic cells (DCs) in a Toll/IL-1R domain-containing adaptor inducing IFN-ß (TRIF)-biased manner: MLA produced from Salmonella minnesota Re595 induced signaling events and expression of gene products that were primarily TRIF dependent, whereas MyD88-dependent signaling was impaired. Moreover, when tested in TRIF-intact/MyD88-deficient DCs, synthetic MLA of the Escherichia coli chemotype (sMLA) showed the same activity as its diphosphoryl, inflammatory counterpart (synthetic diphosphoryl lipid A), indicating that TRIF-mediated signaling is fully induced by sMLA. Unexpectedly, we found that the transcript level of one proinflammatory cytokine was increased in sMLA-treated cells by MyD88 deficiency to the higher level induced by synthetic diphosphoryl lipid A, which suggested MyD88 may paradoxically help restrain proinflammatory signaling by TRIF-biased sMLA. In this article, we demonstrate that sMLA induces MyD88 recruitment to TLR4 and activates the anti-inflammatory lipid phosphatase SHIP1 in an MyD88-dependent manner. At the same time, MyD88-dependent signaling activity at the level of IL-1R-associated kinase 1 is markedly reduced. Increased SHIP1 activity is associated with reductions in sMLA-induced IκB kinase α/ß and IFN regulatory factor 3 activation and with restrained expression of their downstream targets, endothelin-1 and IFN-ß, respectively. Results of this study identify a pattern that is desirable in the context of vaccine adjuvant design: TRIF-biased sMLA can stimulate partial MyD88 activity, with MyD88-dependent SHIP1 helping to reduce proinflammatory signaling in DCs.


Asunto(s)
Adyuvantes Inmunológicos/fisiología , Células Dendríticas/inmunología , Mediadores de Inflamación/fisiología , Lípido A/análogos & derivados , Factor 88 de Diferenciación Mieloide/fisiología , Monoéster Fosfórico Hidrolasas/fisiología , Transducción de Señal/inmunología , Receptor Toll-Like 4/metabolismo , Adyuvantes Inmunológicos/antagonistas & inhibidores , Adyuvantes Inmunológicos/metabolismo , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/microbiología , Células de la Médula Ósea/patología , Células Dendríticas/microbiología , Células Dendríticas/patología , Escherichia coli/inmunología , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Inositol Polifosfato 5-Fosfatasas , Lípido A/fisiología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Monoéster Fosfórico Hidrolasas/deficiencia , Monoéster Fosfórico Hidrolasas/genética , Salmonella/inmunología , Transducción de Señal/genética , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/fisiología
5.
Sci Signal ; 14(674)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33727336

RESUMEN

The nucleoside adenosine accumulates extracellularly in solid tumors and inhibits CD8+ T cells by activating adenosine receptors. The cytokine interleukin-7 (IL-7), which is produced by various tissues and tumors, promotes the survival and maintenance of T cells. Adenosine and IL-7 signaling are being clinically targeted separately or in combination with other therapies for solid tumor indications. Here, we found that IL-7 signaling promoted the accumulation of tumor-associated CD8+ T cells, in part, by preventing adenosine-mediated immunosuppression. Inhibition of the transcription factor FoxO1 downstream of IL-7 receptor signaling was important for protecting CD8+ T cells from suppression by adenosine. These findings have implications for the development of new approaches for cancer immunotherapies that target the adenosine pathway.


Asunto(s)
Adenosina/farmacología , Linfocitos T CD8-positivos , Terapia de Inmunosupresión , Interleucina-7 , Animales , Tolerancia Inmunológica , Ratones
6.
Front Cell Dev Biol ; 9: 647058, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33928082

RESUMEN

Ecto-5'-nucleotidase (CD73) is an enzyme present on the surface of tumor cells whose primary described function is the production of extracellular adenosine. Due to the immunosuppressive properties of adenosine, CD73 is being investigated as a target for new antitumor therapies. We and others have described that CD73 is present at the surface of different CD8+ T cell subsets. Nonetheless, there is limited information as to whether CD73 affects CD8+ T cell proliferation and survival. In this study, we assessed the impact of CD73 deficiency on CD8+ T cells by analyzing their proliferation and survival in antigenic and homeostatic conditions. Results obtained from adoptive transfer experiments demonstrate a paradoxical role of CD73. On one side, it favors the expression of interleukin-7 receptor α chain on CD8+ T cells and their homeostatic survival; on the other side, it reduces the survival of activated CD8+ T cells under antigenic stimulation. Also, upon in vitro antigenic stimulation, CD73 decreases the expression of interleukin-2 receptor α chain and the anti-apoptotic molecule Bcl-2, findings that may explain the reduced CD8+ T cell survival observed in this condition. These results indicate that CD73 has a dual effect on CD8+ T cells depending on whether they are subject to an antigenic or homeostatic stimulus, and thus, special attention should be given to these aspects when considering CD73 blockade in the design of novel antitumor therapies.

7.
Cancer Res ; 81(12): 3319-3332, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33863778

RESUMEN

Extracellular adenosine in tumors can suppress immune responses and promote tumor growth. Adenosine deaminase 2 (ADA2) converts adenosine into inosine. The role of ADA2 in cancer and whether it can target adenosine for cancer therapy has not been investigated. Here we show that increased ADA2 expression is associated with increased patient survival and enrichment of adaptive immune response pathways in several solid tumor types. Several ADA2 variants were created to improve catalytic efficiency, and PEGylation was used to prolong systemic exposure. In mice, PEGylated ADA2 (PEGADA2) inhibited tumor growth by targeting adenosine in an enzyme activity-dependent manner and thereby modulating immune responses. These findings introduce endogenous ADA2 expression as a prognostic factor and PEGADA2 as a novel immunotherapy for cancer. SIGNIFICANCE: This study identifies ADA2 as a prognostic factor associated with prolonged cancer patient survival and introduces the potential of enzymatic removal of adenosine with engineered ADA2 for cancer immunotherapy.


Asunto(s)
Adenosina Desaminasa/metabolismo , Adenosina/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias/prevención & control , Adenosina Desaminasa/genética , Animales , Apoptosis , Proliferación Celular , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Neoplasias/enzimología , Neoplasias/patología , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Biol Chem ; 284(46): 31982-91, 2009 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-19759006

RESUMEN

TLR4 stimulation by lipopolysaccharide can cause both MAL/MyD88- and TRAM/TRIF (Toll IL-1 receptor domain-containing adaptor-inducing IFNbeta)-dependent signaling events. Monophosphoryl lipid A (MPLA), a low toxicity derivative of endotoxic lipopolysaccharide, enhances antibody responses, T cell expansion, and recall responses against antigens without causing excessive inflammatory side effects. Previously, we proposed that TRIF-biased activation of TLR4 by MPLA is responsible for its reduced toxicity while retaining potent adjuvant effects. However, some TRIF-associated genes, such as MCP-1, are only weakly expressed, and some MyD88-associated inflammatory and anti-inflammatory cytokines, such as tumor necrosis factor alpha and interleukin-10, are strongly activated after MPLA stimulation despite weak NF-kappaB but strong IRF3 activation. We now report that synthetic derivatives of MPLA retained TRIF bias as compared with synthetic diphosphoryl lipid A, indicating a change in a single phosphoryl group is sufficient for TRIF-biased TLR4 stimulation. We extend our previous observations by showing that sMLA induces strong p38 MAPK but weak JNK activation, resulting in high IP-10 (interferon-inducible protein 10), tumor necrosis factor alpha, and interleukin-10 but low MCP-1 transcript levels. Results of this study identify a novel biochemical mechanism for regulation of sMLA-induced gene expression.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/fisiología , Adyuvantes Inmunológicos/farmacología , Células Dendríticas/metabolismo , Lípido A/análogos & derivados , Factor 88 de Diferenciación Mieloide/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Western Blotting , Médula Ósea/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Lípido A/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
9.
J Immunol ; 181(10): 6797-802, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18981097

RESUMEN

TLR 4 stimulation of innate immune cells induces a MyD88-independent signaling pathway that leads to the production of IFN-beta. In this study, we demonstrate glycogen synthase kinase 3-beta (GSK3-beta) plays a fundamental role in this process. Suppression of GSK3-beta activity by either pharmacological inhibition, small interfering RNA-mediated gene silencing, or ectopic expression of a kinase-dead GSK3-beta mutant enhanced IFN-beta production by TLR4-stimulated macrophages. Conversely, ectopic expression of a constitutively active GSK3-beta mutant severely attenuated IFN-beta production. GSK3-beta was found to negatively control the cellular levels of the transcription factor c-Jun and its nuclear association with ATF-2. Small interfering RNA-mediated knockdown of c-Jun levels abrogated the ability of GSK3-beta inhibition to augment IFN-beta, demonstrating that the ability of GSK3 to control IFN-beta production was due to its ability to regulate c-Jun levels. The ability of GSK3 inhibition to control IFN-beta production was confirmed in vivo as mice treated with a GSK3 inhibitor exhibited enhanced systemic levels of IFN-beta upon LPS challenge. These findings identify a novel regulatory pathway controlling IFN-beta production by TLR4-stimulated innate immune cells.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Interferón beta/biosíntesis , Macrófagos/inmunología , Receptor Toll-Like 4/inmunología , Factor de Transcripción Activador 2/inmunología , Factor de Transcripción Activador 2/metabolismo , Animales , Western Blotting , Glucógeno Sintasa Quinasa 3/inmunología , Glucógeno Sintasa Quinasa 3 beta , Interferón beta/inmunología , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Fosforilación , Proteínas Proto-Oncogénicas c-jun/inmunología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Curr Opin Pharmacol ; 53: 134-145, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-33022543

RESUMEN

Hypoxia, metabolic activity, cell death and immune responses influence the adenosine concentrations in the extracellular space. Cellular responses to hypoxia and inflammation in myeloid cells promote activation of adenosine sensing circuit, which involves increased expression of ectoenzymes that converts phospho-nucleotides such as ATP to adenosine and increased expression of G protein-coupled adenosine receptors. Adenosine sensing circuitry also involves feedforward signaling, which leads to increased expression of hypoxia-inducible factor 1-alpha (HIF1 and feedback signaling, which leads to the suppression of inflammatory transcription factor, the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation. In this review we will discuss how different subsets of myeloid cells sense adenosine accumulation and how adenosine sensing by myeloid cells influence progression of different immune-related conditions including cancer.


Asunto(s)
Adenosina/inmunología , Células Mieloides/inmunología , Receptores Purinérgicos P1/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Células Dendríticas/inmunología , Humanos , Infecciones/inmunología , Neoplasias/inmunología , Sepsis/inmunología , Transducción de Señal
11.
Cancer Immunol Res ; 8(8): 1064-1074, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32381524

RESUMEN

Accumulating evidence suggests that inhibiting adenosine-generating ecto-enzymes (CD39 and CD73) and/or adenosine A2A or A2B receptors (R) stimulates antitumor immunity and limits tumor progression. Although activating A2ARs or A2BRs causes similar immunosuppressive and protumoral functions, few studies have investigated the distinct role of A2BR in cancer. Here, we showed that A2BR expression by hematopoietic cells was primarily responsible for promoting tumor growth. Deletion of A2BR profoundly enhanced anticancer T-cell immunity. Although T-cell A2BR plays an insignificant role for A2BR-mediated immunosuppression and tumor promotion, A2BR deficiency in tumor-bearing mice caused increased infiltration of myeloid and CD103+ dendritic cells, which was associated with more effective cross-priming of adoptively transferred tumor antigen-specific CD8+ T cells. A2BR deletion also intrinsically favored accumulation of myeloid and CD11bdim antigen-presenting cells (APC) in the tumor microenvironment. Both myeloid-specific or CD11c-specific conditional deletion of A2BR delayed primary tumor growth. Myeloid, but not CD11c-specific conditional, depletion delayed lung metastasis. Pharmacologic blockade of A2BR improved the antitumor effect of adoptive T-cell therapy. Overall, these results suggested that A2BR expression on myeloid cells and APCs indirectly suppressed CD8+ T-cell responses and promoted metastasis. These data provide a strong rationale to combine A2BR inhibition with T-cell-based immunotherapy for the treatment of tumor growth and metastasis.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Mieloides/inmunología , Neoplasias/inmunología , Receptor de Adenosina A2B/metabolismo , Animales , Células Presentadoras de Antígenos/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Tolerancia Inmunológica , Ratones , Ratones Noqueados , Células Mieloides/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Receptor de Adenosina A2B/genética , Receptor de Adenosina A2B/inmunología , Microambiente Tumoral
12.
Clin Cancer Res ; 24(8): 1987-2001, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29386221

RESUMEN

Purpose: Tamoxifen remains an important hormonal therapy for ER-positive breast cancer; however, development of resistance is a major obstacle in clinics. Here, we aimed to identify novel mechanisms of tamoxifen resistance and provide actionable drug targets overcoming resistance.Experimental Design: Whole-transcriptome sequencing, downstream pathway analysis, and drug repositioning approaches were used to identify novel modulators [here: phosphodiesterase 4D (PDE4D)] of tamoxifen resistance. Clinical data involving tamoxifen-treated patients with ER-positive breast cancer were used to assess the impact of PDE4D in tamoxifen resistance. Tamoxifen sensitization role of PDE4D was tested in vitro and in vivo Cytobiology, biochemistry, and functional genomics tools were used to elucidate the mechanisms of PDE4D-mediated tamoxifen resistance.Results: PDE4D, which hydrolyzes cyclic AMP (cAMP), was significantly overexpressed in both MCF-7 and T47D tamoxifen-resistant (TamR) cells. Higher PDE4D expression predicted worse survival in tamoxifen-treated patients with breast cancer (n = 469, P = 0.0036 for DMFS; n = 561, P = 0.0229 for RFS) and remained an independent prognostic factor for RFS in multivariate analysis (n = 132, P = 0.049). Inhibition of PDE4D by either siRNAs or pharmacologic inhibitors (dipyridamole and Gebr-7b) restored tamoxifen sensitivity. Sensitization to tamoxifen is achieved via cAMP-mediated induction of unfolded protein response/ER stress pathway leading to activation of p38/JNK signaling and apoptosis. Remarkably, acetylsalicylic acid (aspirin) was predicted to be a tamoxifen sensitizer using a drug repositioning approach and was shown to reverse resistance by targeting PDE4D/cAMP/ER stress axis. Finally, combining PDE4D inhibitors and tamoxifen suppressed tumor growth better than individual groups in vivoConclusions: PDE4D plays a pivotal role in acquired tamoxifen resistance via blocking cAMP/ER stress/p38-JNK signaling and apoptosis. Clin Cancer Res; 24(8); 1987-2001. ©2018 AACR.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Resistencia a Antineoplásicos , Receptores de Estrógenos/metabolismo , Sistemas de Mensajero Secundario/efectos de los fármacos , Tamoxifeno/farmacología , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Modelos Biológicos , Inhibidores de Fosfodiesterasa 4/farmacología , Estrés Fisiológico/genética , Tamoxifeno/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Nat Rev Immunol ; 16(3): 177-92, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26922909

RESUMEN

Cellular stress or apoptosis triggers the release of ATP, ADP and other nucleotides into the extracellular space. Extracellular nucleotides function as autocrine and paracrine signalling molecules by activating cell-surface P2 purinergic receptors that elicit pro-inflammatory immune responses. Over time, extracellular nucleotides are metabolized to adenosine, leading to reduced P2 signalling and increased signalling through anti-inflammatory adenosine (P1 purinergic) receptors. Here, we review how local purinergic signalling changes over time during tissue responses to injury or disease, and we discuss the potential of targeting purinergic signalling pathways for the immunotherapeutic treatment of ischaemia, organ transplantation, autoimmunity or cancer.


Asunto(s)
Inmunidad Adaptativa/inmunología , Inmunidad Innata/inmunología , Purinas/inmunología , Transducción de Señal/inmunología , Animales , Humanos
14.
Nat Commun ; 6: 6354, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25724068

RESUMEN

ATP-binding cassette transporter G1 (ABCG1) promotes cholesterol efflux from cells and regulates intracellular cholesterol homeostasis. Here we demonstrate a role of ABCG1 as a mediator of tumour immunity. Abcg1(-/-) mice have dramatically suppressed subcutaneous MB49-bladder carcinoma and B16-melanoma growth and prolonged survival. We show that reduced tumour growth in Abcg1(-/-) mice is myeloid cell intrinsic and is associated with a phenotypic shift of the macrophages from a tumour-promoting M2 to a tumour-fighting M1 within the tumour. Abcg1(-/-) macrophages exhibit an intrinsic bias towards M1 polarization with increased NF-κB activation and direct cytotoxicity for tumour cells in vitro. Overall, our study demonstrates that the absence of ABCG1 inhibits tumour growth through modulation of macrophage function within the tumour, and illustrates a link between cholesterol homeostasis and cancer.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Carcinoma/inmunología , Colesterol/metabolismo , Homeostasis/fisiología , Lipoproteínas/metabolismo , Macrófagos/inmunología , Melanoma/inmunología , Neoplasias de la Vejiga Urinaria/inmunología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Carcinoma/fisiopatología , Línea Celular Tumoral , Citometría de Flujo , Fluorescencia , Lipoproteínas/genética , Macrófagos/citología , Melanoma/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Neoplasias de la Vejiga Urinaria/fisiopatología
15.
Science ; 350(6263): 985-90, 2015 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-26494174

RESUMEN

The immune system plays an important role in regulating tumor growth and metastasis. Classical monocytes promote tumorigenesis and cancer metastasis, but how nonclassical "patrolling" monocytes (PMo) interact with tumors is unknown. Here we show that PMo are enriched in the microvasculature of the lung and reduce tumor metastasis to lung in multiple mouse metastatic tumor models. Nr4a1-deficient mice, which specifically lack PMo, showed increased lung metastasis in vivo. Transfer of Nr4a1-proficient PMo into Nr4a1-deficient mice prevented tumor invasion in the lung. PMo established early interactions with metastasizing tumor cells, scavenged tumor material from the lung vasculature, and promoted natural killer cell recruitment and activation. Thus, PMo contribute to cancer immunosurveillance and may be targets for cancer immunotherapy.


Asunto(s)
Vigilancia Inmunológica/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Monocitos/inmunología , Animales , Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/terapia , Ratones , Ratones Mutantes , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/secundario , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética
16.
Cancer Res ; 74(24): 7239-49, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25341542

RESUMEN

Adenosine A(2A) receptor (A(2A)R) blockade enhances innate and adaptive immune responses. However, mouse genetic studies have shown that A(2A)R deletion does not inhibit the growth of all tumor types. In the current study, we showed that growth rates for ectopic melanoma and bladder tumors are increased in Adora2a(-/-) mice within 2 weeks of tumor inoculation. A(2A)R deletion in the host reduced numbers of CD8(+) T cells and effector-memory differentiation of all T cells. To examine intrinsic functions in T cells, we generated mice harboring a T-cell-specific deletion of A(2A)R. In this host strain, tumor-bearing mice displayed increased growth of ectopic melanomas, decreased numbers of tumor-associated T cells, reduced effector-memory differentiation, and reduced antiapoptotic IL7Rα (CD127) expression on antigen-experienced cells. Intratumoral pharmacologic blockade similarly reduced CD8(+) T-cell density within tumors in wild-type hosts. We found that A(2A)R-proficient CD8(+) T cells specific for melanoma cells displayed a relative survival advantage in tumors. Thus, abrogating A(2A)R signaling appeared to reduce IL7R expression, survival, and differentiation of T cells in the tumor microenvironment. One implication of these results is that the antitumor effects of A(2A)R blockade that can be mediated by activation of cytotoxic T cells may be overcome in some tumor microenvironments as a result of impaired T-cell maintenance and effector-memory differentiation. Thus, our findings imply that the efficacious application of A(2A)R inhibitors for cancer immunotherapy may require careful dose optimization to prevent activation-induced T-cell death in tumors.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/administración & dosificación , Linfocitos T CD8-positivos/efectos de los fármacos , Inmunoterapia , Receptor de Adenosina A2A/metabolismo , Inmunidad Adaptativa/efectos de los fármacos , Animales , Linfocitos T CD8-positivos/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunidad Innata/efectos de los fármacos , Subunidad alfa del Receptor de Interleucina-7/biosíntesis , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Melanoma/patología , Ratones , Receptor de Adenosina A2A/genética , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/patología
17.
Cancer Res ; 74(24): 7250-9, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25377469

RESUMEN

High concentrations of adenosine in tumor microenvironments inhibit antitumor cytotoxic lymphocyte responses. Although T cells express inhibitory adenosine A2A receptors (A2AR) that suppress their activation and inhibit immune killing of tumors, a role for myeloid cell A2ARs in suppressing the immune response to tumors has yet to be investigated. In this study, we show that the growth of transplanted syngeneic B16F10 melanoma or Lewis lung carcinoma cells is slowed in Adora2a(f/f)-LysMCre(+/-) mice, which selectively lack myeloid A2ARs. Reduced melanoma growth is associated with significant increases in MHCII and IL12 expression in tumor-associated macrophages and with >90% reductions in IL10 expression in tumor-associated macrophages, dendritic cells (DC), and Ly6C(+) or Ly6G(+) myeloid-derived suppressor cells (MDSC). Myeloid deletion of A2ARs significantly increases CD44 expression on tumor-associated T cells and natural killer (NK) cells. Depletion of CD8(+) T cells or NK cells in tumor-bearing mice indicates that both cell types initially contribute to slowing melanoma growth in mice lacking myeloid A2A receptors, but tumor suppression mediated by CD8(+) T cells is more persistent. Myeloid-selective A2AR deletion significantly reduces lung metastasis of melanomas that express luciferase (for in vivo tracking) and ovalbumin (as a model antigen). Reduced metastasis is associated with increased numbers and activation of NK cells and antigen-specific CD8(+) T cells in lung infiltrates. Overall, the findings indicate that myeloid cell A2ARs have direct myelosuppressive effects that indirectly contribute to the suppression of T cells and NK cells in primary and metastatic tumor microenvironments. The results indicate that tumor-associated myeloid cells, including macrophages, DCs, and MDSCs all express immunosuppressive A2ARs that are potential targets of adenosine receptor blockers to enhance immune killing of tumors.


Asunto(s)
Inmunoterapia , Melanoma Experimental/terapia , Receptor de Adenosina A2A/metabolismo , Linfocitos T Citotóxicos/inmunología , Antagonistas del Receptor de Adenosina A2/administración & dosificación , Animales , Linfocitos T CD8-positivos/inmunología , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/metabolismo , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Células Mieloides/inmunología , Receptor de Adenosina A2A/biosíntesis , Receptor de Adenosina A2A/inmunología , Microambiente Tumoral/inmunología
18.
J Exp Med ; 210(12): 2693-706, 2013 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-24145516

RESUMEN

Adenosine produced as a byproduct of metabolic activity is present in all tissues and produces dose-dependent suppression of TCR signaling. Naive T cell maintenance depends on inhibition of TCR signals by environmental sensors, which are yet to be fully defined. We produced mice with a floxed adenosine A2A receptor (A2AR) gene, Adora2a, and show that either global A2AR deletion or cre-mediated T cell deletion elicits a decline in the number of naive but not memory T cells. A2AR signaling maintains naive T cells in a quiescent state by inhibiting TCR-induced activation of the phosphatidylinositide 3-kinase (PI3K)-AKT pathway, thereby reducing IL-7Rα down-regulation and naive T cell apoptosis. Patterns of IL-7Rα expression on T cells in chimeric mice reconstituted with Adora2a(+/+) and Adora2a(-/-) bone marrow cells suggest that decreased IL-7Rα in naive T cells is a cell-intrinsic consequence of Adora2a deletion. In addition, A2AR expression increases in early thymic T cell development and contributes to progression of double-negative thymic precursors to single-positive thymocytes with increased IL-7Rα expression. Therefore, A2AR signaling regulates T cell development and maintenance to sustain normal numbers of naive T cells in the periphery.


Asunto(s)
Adenosina/metabolismo , Linfocitos T/metabolismo , Animales , Apoptosis , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Espacio Extracelular/metabolismo , Femenino , Regulación de la Expresión Génica , Memoria Inmunológica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Adenosina A2A/deficiencia , Receptor de Adenosina A2A/genética , Receptor de Adenosina A2A/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Interleucina-7/genética , Receptores de Interleucina-7/metabolismo , Transducción de Señal , Linfocitos T/citología , Linfocitos T/inmunología
19.
Science ; 316(5831): 1628-32, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17569868

RESUMEN

The inflammatory toxicity of lipopolysaccharide (LPS), a component of bacterial cell walls, is driven by the adaptor proteins myeloid differentiation factor 88 (MyD88) and Toll-interleukin 1 receptor domain-containing adapter inducing interferon-beta (TRIF), which together mediate signaling by the endotoxin receptor Toll-like receptor 4 (TLR4). Monophosphoryl lipid A (MPLA) is a low-toxicity derivative of LPS with useful immunostimulatory properties, which is nearing regulatory approval for use as a human vaccine adjuvant. We report here that, in mice, the low toxicity of MPLA's adjuvant function is associated with a bias toward TRIF signaling, which we suggest is likely caused by the active suppression, rather than passive loss, of proinflammatory activity of this LPS derivative. This finding may have important implications for the development of future vaccine adjuvants.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Adyuvantes Inmunológicos , Lípido A/análogos & derivados , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/toxicidad , Traslado Adoptivo , Animales , Citocinas/biosíntesis , Inmunización , Lípido A/administración & dosificación , Lípido A/inmunología , Lípido A/toxicidad , Lipopolisacáridos/inmunología , Activación de Linfocitos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Ovalbúmina/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Receptor Toll-Like 4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA