Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Immunity ; 46(1): 51-64, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28099864

RESUMEN

Despite the importance of programmed cell death-1 (PD-1) in inhibiting T cell effector activity, the mechanisms regulating its expression remain poorly defined. We found that the chromatin organizer special AT-rich sequence-binding protein-1 (Satb1) restrains PD-1 expression induced upon T cell activation by recruiting a nucleosome remodeling deacetylase (NuRD) complex to Pdcd1 regulatory regions. Satb1 deficienct T cells exhibited a 40-fold increase in PD-1 expression. Tumor-derived transforming growth factor ß (Tgf-ß) decreased Satb1 expression through binding of Smad proteins to the Satb1 promoter. Smad proteins also competed with the Satb1-NuRD complex for binding to Pdcd1 enhancers, releasing Pdcd1 expression from Satb1-mediated repression, Satb1-deficient tumor-reactive T cells lost effector activity more rapidly than wild-type lymphocytes at tumor beds expressing PD-1 ligand (CD274), and these differences were abrogated by sustained CD274 blockade. Our findings suggest that Satb1 functions to prevent premature T cell exhaustion by regulating Pdcd1 expression upon T cell activation. Dysregulation of this pathway in tumor-infiltrating T cells results in diminished anti-tumor immunity.


Asunto(s)
Represión Epigenética/inmunología , Regulación de la Expresión Génica/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Proteínas de Unión a la Región de Fijación a la Matriz/biosíntesis , Receptor de Muerte Celular Programada 1/biosíntesis , Animales , Ensayo de Immunospot Ligado a Enzimas , Humanos , Inmunoprecipitación , Activación de Linfocitos/inmunología , Proteínas de Unión a la Región de Fijación a la Matriz/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/inmunología , Neoplasias/metabolismo
2.
Immunity ; 41(3): 427-439, 2014 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-25238097

RESUMEN

Tumor-reactive T cells become unresponsive in advanced tumors. Here we have characterized a common mechanism of T cell unresponsiveness in cancer driven by the upregulation of the transcription factor Forkhead box protein P1 (Foxp1), which prevents CD8⁺ T cells from proliferating and upregulating Granzyme-B and interferon-γ in response to tumor antigens. Accordingly, Foxp1-deficient lymphocytes induced rejection of incurable tumors and promoted protection against tumor rechallenge. Mechanistically, Foxp1 interacted with the transcription factors Smad2 and Smad3 in preactivated CD8⁺ T cells in response to microenvironmental transforming growth factor-ß (TGF-ß), and was essential for its suppressive activity. Therefore, Smad2 and Smad3-mediated c-Myc repression requires Foxp1 expression in T cells. Furthermore, Foxp1 directly mediated TGF-ß-induced c-Jun transcriptional repression, which abrogated T cell activity. Our results unveil a fundamental mechanism of T cell unresponsiveness different from anergy or exhaustion, driven by TGF-ß signaling on tumor-associated lymphocytes undergoing Foxp1-dependent transcriptional regulation.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Neoplasias/inmunología , Proteínas Represoras/inmunología , Linfocitos T Citotóxicos/inmunología , Factor de Crecimiento Transformador beta/inmunología , Escape del Tumor/inmunología , Traslado Adoptivo , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Femenino , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Granzimas/biosíntesis , Interferón gamma/biosíntesis , Proteínas Quinasas JNK Activadas por Mitógenos/biosíntesis , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Transducción de Señal/inmunología , Proteína Smad2/inmunología , Proteína smad3/inmunología , Linfocitos T Citotóxicos/trasplante , Transcripción Genética , Activación Transcripcional , Microambiente Tumoral/inmunología
3.
Clin Cancer Res ; 23(2): 441-453, 2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27435394

RESUMEN

PURPOSE: To define the safety and effectiveness of T cells redirected against follicle-stimulating hormone receptor (FSHR)-expressing ovarian cancer cells. EXPERIMENTAL DESIGN: FSHR expression was determined by Western blotting, immunohistochemistry, and qPCR in 77 human ovarian cancer specimens from 6 different histologic subtypes and 20 human healthy tissues. The effectiveness of human T cells targeted with full-length FSH in vivo was determined against a panel of patient-derived xenografts. Safety and effectiveness were confirmed in immunocompetent tumor-bearing mice, using constructs targeting murine FSHR and syngeneic T cells. RESULTS: FSHR is expressed in gynecologic malignancies of different histologic types but not in nonovarian healthy tissues. Accordingly, T cells expressing full-length FSHR-redirected chimeric receptors mediate significant therapeutic effects (including tumor rejection) against a panel of patient-derived tumors in vivo In immunocompetent mice growing syngeneic, orthotopic, and aggressive ovarian tumors, fully murine FSHR-targeted T cells also increased survival without any measurable toxicity. Notably, chimeric receptors enhanced the ability of endogenous tumor-reactive T cells to abrogate malignant progression upon adoptive transfer into naïve recipients subsequently challenged with the same tumor. Interestingly, FSHR-targeted T cells persisted as memory lymphocytes without noticeable PD-1-dependent exhaustion during end-stage disease, in the absence of tumor cell immunoediting. However, exosomes in advanced tumor ascites diverted the effector activity of this and other chimeric receptor-transduced T cells away from targeted tumor cells. CONCLUSIONS: T cells redirected against FSHR+ tumor cells with full-length FSH represent a promising therapeutic alternative against a broad range of ovarian malignancies, with negligible toxicity even in the presence of cognate targets in tumor-free ovaries. Clin Cancer Res; 23(2); 441-53. ©2016 AACR.


Asunto(s)
Inmunoterapia , Neoplasias Ováricas/terapia , Receptores de HFE/inmunología , Linfocitos T/inmunología , Animales , Ascitis/inmunología , Ascitis/patología , Exosomas/inmunología , Exosomas/patología , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inmunohistoquímica , Ratones , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de HFE/genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cancer Discov ; 7(1): 72-85, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27694385

RESUMEN

The role of estrogens in antitumor immunity remains poorly understood. Here, we show that estrogen signaling accelerates the progression of different estrogen-insensitive tumor models by contributing to deregulated myelopoiesis by both driving the mobilization of myeloid-derived suppressor cells (MDSC) and enhancing their intrinsic immunosuppressive activity in vivo Differences in tumor growth are dependent on blunted antitumor immunity and, correspondingly, disappear in immunodeficient hosts and upon MDSC depletion. Mechanistically, estrogen receptor alpha activates the STAT3 pathway in human and mouse bone marrow myeloid precursors by enhancing JAK2 and SRC activity. Therefore, estrogen signaling is a crucial mechanism underlying pathologic myelopoiesis in cancer. Our work suggests that new antiestrogen drugs that have no agonistic effects may have benefits in a wide range of cancers, independently of the expression of estrogen receptors in tumor cells, and may synergize with immunotherapies to significantly extend survival. SIGNIFICANCE: Ablating estrogenic activity delays malignant progression independently of the tumor cell responsiveness, owing to a decrease in the mobilization and immunosuppressive activity of MDSCs, which boosts T-cell-dependent antitumor immunity. Our results provide a mechanistic rationale to block estrogen signaling with newer antagonists to boost the effectiveness of anticancer immunotherapies. Cancer Discov; 7(1); 72-85. ©2016 AACR.See related commentary by Welte et al., p. 17This article is highlighted in the In This Issue feature, p. 1.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/inmunología , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Humanos , Janus Quinasa 2/metabolismo , Células MCF-7 , Ratones , Ratones Endogámicos C57BL , Células Supresoras de Origen Mieloide/metabolismo , Trasplante de Neoplasias , Neoplasias/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Familia-src Quinasas/metabolismo
5.
Cancer Res ; 76(21): 6253-6265, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27803104

RESUMEN

Targeted therapies elicit seemingly paradoxical and poorly understood effects on tumor immunity. Here, we show that the MEK inhibitor trametinib abrogates cytokine-driven expansion of monocytic myeloid-derived suppressor cells (mMDSC) from human or mouse myeloid progenitors. MEK inhibition also reduced the production of the mMDSC chemotactic factor osteopontin by tumor cells. Together, these effects reduced mMDSC accumulation in tumor-bearing hosts, limiting the outgrowth of KRas-driven breast tumors, even though trametinib largely failed to directly inhibit tumor cell proliferation. Accordingly, trametinib impeded tumor progression in vivo through a mechanism requiring CD8+ T cells, which was paradoxical given the drug's reported ability to inhibit effector lymphocytes. Confirming our observations, adoptive transfer of tumor-derived mMDSC reversed the ability of trametinib to control tumor growth. Overall, our work showed how the effects of trametinib on immune cells could partly explain its effectiveness, distinct from its activity on tumor cells themselves. More broadly, by providing a more incisive view into how MEK inhibitors may act against tumors, our findings expand their potential uses to generally block mMDSC expansion, which occurs widely in cancers to drive their growth and progression. Cancer Res; 76(21); 6253-65. ©2016 AACR.


Asunto(s)
Antineoplásicos/farmacología , Mutación , Mielopoyesis/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Proteínas Proto-Oncogénicas p21(ras)/genética , Piridonas/farmacología , Pirimidinonas/farmacología , Linfocitos T/fisiología , Animales , Línea Celular Tumoral , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Células Mieloides/efectos de los fármacos , Células Mieloides/fisiología , Neoplasias/genética , Neoplasias/fisiopatología , Osteopontina/biosíntesis
6.
Cancer Res ; 76(9): 2561-72, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26980764

RESUMEN

Many signal transduction inhibitors are being developed for cancer therapy target pathways that are also important for the proper function of antitumor lymphocytes, possibly weakening their therapeutic effects. Here we show that most inhibitors targeting multiple signaling pathways have especially strong negative effects on T-cell activation at their active doses on cancer cells. In particular, we found that recently approved MEK inhibitors displayed potent suppressive effects on T cells in vitro However, these effects could be attenuated by certain cytokines that can be administered to cancer patients. Among them, clinically available IL15 superagonists, which can activate PI3K selectively in T lymphocytes, synergized with MEK inhibitors in vivo to elicit potent and durable antitumor responses, including by a vaccine-like effect that generated resistance to tumor rechallenge. Our work identifies a clinically actionable approach to overcome the T-cell-suppressive effects of MEK inhibitors and illustrates how to reconcile the deficiencies of signal transduction inhibitors, which impede desired immunologic effects in vivo Cancer Res; 76(9); 2561-72. ©2016 AACR.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Neoplasias Experimentales/patología , Proteínas/farmacología , Animales , Western Blotting , Línea Celular Tumoral , Citometría de Flujo , Ensayos Analíticos de Alto Rendimiento , Humanos , Interleucina-15 , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibidores de Proteínas Quinasas/farmacología , Piridonas/farmacología , Pirimidinonas/farmacología , Proteínas Recombinantes de Fusión , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cell Rep ; 14(7): 1774-1786, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26876172

RESUMEN

Special AT-rich sequence-binding protein 1 (Satb1) governs genome-wide transcriptional programs. Using a conditional knockout mouse, we find that Satb1 is required for normal differentiation of conventional dendritic cells (DCs). Furthermore, Satb1 governs the differentiation of inflammatory DCs by regulating major histocompatibility complex class II (MHC II) expression through Notch1 signaling. Mechanistically, Satb1 binds to the Notch1 promoter, activating Notch expression and driving RBPJ occupancy of the H2-Ab1 promoter, which activates MHC II transcription. However, tumor-driven, unremitting expression of Satb1 in activated Zbtb46(+) inflammatory DCs that infiltrate ovarian tumors results in an immunosuppressive phenotype characterized by increased secretion of tumor-promoting Galectin-1 and IL-6. In vivo silencing of Satb1 in tumor-associated DCs reverses their tumorigenic activity and boosts protective immunity. Therefore, dynamic fluctuations in Satb1 expression govern the generation and immunostimulatory activity of steady-state and inflammatory DCs, but continuous Satb1 overexpression in differentiated DCs converts them into tolerogenic/pro-inflammatory cells that contribute to malignant progression.


Asunto(s)
Células Dendríticas/inmunología , Regulación Neoplásica de la Expresión Génica , Antígenos de Histocompatibilidad Clase II/inmunología , Proteínas de Unión a la Región de Fijación a la Matriz/inmunología , Neoplasias Ováricas/inmunología , Animales , Diferenciación Celular , Proliferación Celular , Transformación Celular Neoplásica , Células Dendríticas/patología , Femenino , Galectina 1/genética , Galectina 1/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Histonas/genética , Histonas/inmunología , Humanos , Tolerancia Inmunológica , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Proteínas de Unión a la Región de Fijación a la Matriz/antagonistas & inhibidores , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Ratones , Ratones Noqueados , Trasplante de Neoplasias , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Regiones Promotoras Genéticas , Unión Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/inmunología , Receptor Notch1/genética , Receptor Notch1/inmunología , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/inmunología
8.
Cancer Cell ; 27(1): 27-40, 2015 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-25533336

RESUMEN

The dominant TLR5(R392X) polymorphism abrogates flagellin responses in >7% of humans. We report that TLR5-dependent commensal bacteria drive malignant progression at extramucosal locations by increasing systemic IL-6, which drives mobilization of myeloid-derived suppressor cells (MDSCs). Mechanistically, expanded granulocytic MDSCs cause γδ lymphocytes in TLR5-responsive tumors to secrete galectin-1, dampening antitumor immunity and accelerating malignant progression. In contrast, IL-17 is consistently upregulated in TLR5-unresponsive tumor-bearing mice but only accelerates malignant progression in IL-6-unresponsive tumors. Importantly, depletion of commensal bacteria abrogates TLR5-dependent differences in tumor growth. Contrasting differences in inflammatory cytokines and malignant evolution are recapitulated in TLR5-responsive/unresponsive ovarian and breast cancer patients. Therefore, inflammation, antitumor immunity, and the clinical outcome of cancer patients are influenced by a common TLR5 polymorphism.


Asunto(s)
Interleucina-17/metabolismo , Interleucina-6/metabolismo , Microbiota , Neoplasias/inmunología , Neoplasias/patología , Receptor Toll-Like 5/genética , Receptor Toll-Like 5/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Galectina 1/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Trasplante de Neoplasias , Polimorfismo de Nucleótido Simple , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA