Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
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.
Cell Immunol ; 278(1-2): 21-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23121972

RESUMEN

The immune surveillance hypothesis proposed over 50 years ago that many precancerous lesions are eliminated without a histological trace due to immunological pressure. Since then, it has become apparent that both the tumor and the anti-cancer immune response evolve over a long period to allow the eventual escape of nascent precancerous lesions into full-blown tumors. Although primarily focusing on loss of antigenicity, the immunoediting hypothesis has gradually evolved to appreciate the role of active immunosuppression in tumor progression, where myeloid leukocytes are increasingly recognized as the major driving force. This review highlights recent studies implicating how myeloid cells with antigen-presenting capabilities are co-opted by tumors to promote malignant progression. Because at least some advanced tumors remain significantly immunogenic, these new studies add a tweak to the immunoediting hypothesis as well as a rationale to block immunosuppressive mechanisms as a first-line intervention in cancer patients.


Asunto(s)
Células Dendríticas/inmunología , Vigilancia Inmunológica , Células Mieloides/inmunología , Neoplasias/inmunología , Linfocitos T/inmunología , Inmunidad Adaptativa , Antígenos de Neoplasias/inmunología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Citocinas/biosíntesis , Citocinas/inmunología , Células Dendríticas/patología , Progresión de la Enfermedad , Humanos , Tolerancia Inmunológica , Inmunidad Innata , Células Mieloides/patología , Linfocitos T/patología , Escape del Tumor
4.
J Autoimmun ; 36(1): 56-64, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21095100

RESUMEN

Experimental autoimmune encephalomyelitis (EAE), a model for the human disease multiple sclerosis (MS), is dependent upon the activation and effector functions of autoreactive CD4 T cells. Multiple interactions between CD4 T cells and major histocompatibility class II (MHCII)+ antigen presenting cells (APCs) must occur in both the periphery and central nervous system (CNS) to elicit autoimmunity. The identity of the MHCII+ APCs involved throughout this process remains in question. We investigated which APC in the periphery and CNS mediates disease using transgenic mice with MHCII expression restricted to dendritic cells (DCs). MHCII expression restricted to DCs results in normal susceptibility to peptide-mediated EAE. Indeed, radiation-sensitive bone marrow-derived DCs were sufficient for all APC functions during peptide-induced disease. However, DCs alone were inefficient at promoting disease after immunization with the myelin protein myelin oligodendrocyte glycoprotein (MOG), even in the presence of MHCII-deficient B cells. Consistent with a defect in disease induction following protein immunization, antigen presentation by DCs alone was incapable of mediating spontaneous optic neuritis. These results indicate that DCs are capable of perpetuating CNS-targeted autoimmunity when antigens are readily available, but other APCs are required to efficiently initiate pathogenic cognate CD4 T cell responses.


Asunto(s)
Presentación de Antígeno/inmunología , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Separación Celular , Encefalomielitis Autoinmune Experimental/patología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Antígenos de Histocompatibilidad Clase II/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Nervio Óptico/patología , Médula Espinal/patología
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
9.
J Vis Exp ; (85)2014 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-24748051

RESUMEN

Breast cancer is a heterogeneous disease involving complex cellular interactions between the developing tumor and immune system, eventually resulting in exponential tumor growth and metastasis to distal tissues and the collapse of anti-tumor immunity. Many useful animal models exist to study breast cancer, but none completely recapitulate the disease progression that occurs in humans. In order to gain a better understanding of the cellular interactions that result in the formation of latent metastasis and decreased survival, we have generated an inducible transgenic mouse model of YFP-expressing ductal carcinoma that develops after sexual maturity in immune-competent mice and is driven by consistent, endocrine-independent oncogene expression. Activation of YFP, ablation of p53, and expression of an oncogenic form of K-ras was achieved by the delivery of an adenovirus expressing Cre-recombinase into the mammary duct of sexually mature, virgin female mice. Tumors begin to appear 6 weeks after the initiation of oncogenic events. After tumors become apparent, they progress slowly for approximately two weeks before they begin to grow exponentially. After 7-8 weeks post-adenovirus injection, vasculature is observed connecting the tumor mass to distal lymph nodes, with eventual lymphovascular invasion of YFP+ tumor cells to the distal axillary lymph nodes. Infiltrating leukocyte populations are similar to those found in human breast carcinomas, including the presence of αß and γδ T cells, macrophages and MDSCs. This unique model will facilitate the study of cellular and immunological mechanisms involved in latent metastasis and dormancy in addition to being useful for designing novel immunotherapeutic interventions to treat invasive breast cancer.


Asunto(s)
Modelos Animales de Enfermedad , Neoplasias Mamarias Experimentales/patología , Adenoviridae/genética , Alelos , Animales , Femenino , Genes p53 , Genes ras , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/virología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
10.
Hum Pathol ; 44(11): 2549-57, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24074532

RESUMEN

Fibroblast activation protein (FAP) has long been known to be expressed in the stroma of breast cancer. However, very little is known if the magnitude of FAP expression within the stroma may have a prognostic value and reflect the heterogeneous biology of the tumor cell. An earlier study had suggested that stromal FAP expression in breast cancer was inversely proportional to prognosis. We, therefore, hypothesized that stromal FAP expression may correlate with clinicopathologic variables and may serve as an adjunct prognostic factor in breast cancer. We evaluated the expression of FAP in a panel of breast cancer tissues (n = 52) using a combination of immunostain analyses at the tissue and single-cell level using freshly frozen or freshly digested human breast tumor samples, respectively. Our results showed that FAP expression was abundantly expressed in the stroma across all breast cancer subtypes without significant correlation with clinicopathologic factors. We further identified a subset of FAP-positive (or FAP(+)) stromal cells that also expressed CD45, a pan-leukocyte marker. Using freshly dissociated human breast tumor specimens (n = 5), we demonstrated that some of these FAP(+)CD45(+) cells were CD11b(+)CD14(+)MHC-II(+), indicating that they were likely tumor-associated macrophages (TAMs). Although FAP(+)CD45(+) cells have been demonstrated in the mouse tumor stroma, our results demonstrating that human breast TAMs expressed FAP were novel and suggested that existing and future FAP-directed therapy may have dual-therapeutic benefits targeting both stromal mesenchymal cells and immune cells such as TAMs. More work is needed to explore the role of FAP as a potential targetable molecule in breast cancer treatment.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Gelatinasas/metabolismo , Proteínas de la Membrana/metabolismo , Serina Endopeptidasas/metabolismo , Actinas/metabolismo , Adulto , Anciano , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Estudios de Cohortes , Endopeptidasas , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Antígenos Comunes de Leucocito/metabolismo , Macrófagos/metabolismo , Persona de Mediana Edad , Pronóstico , Células del Estroma/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA