Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Biochem Biophys Res Commun ; 530(2): 471-478, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32593416

RESUMEN

Tenascin-C (TNC) and tenascin-W (TNW), large hexameric glycoproteins overexpressed in the tumor microenvironment, are useful tumor biomarkers for theranostic applications. For now, polyclonal and monoclonal antibodies, as well as aptamers targeting TNC and TNW have been developed. However, the immunostaining sensitivity of antibodies is very heterogenous. The main aim of this study was to generate antibodies in dromedary that detect TNC and TNW, respectively. We show that immune sera from immunized dromedaries are able to specifically bind native TNC and TNW by ELISA and also to detect TNC and TNW in matrix tracks of mammary tumors by immunostaining. Furthermore, we demonstrate that purified IgG subtypes are able to interact specifically with TNC or TNW by ELISA and immunostaining. These camelid antibodies are a good basis to develop tools for the detection of TNC and TNW in the tumor microenvironment and could potentially have a broader application for early diagnosis of solid cancers.


Asunto(s)
Anticuerpos/inmunología , Camelus/inmunología , Tenascina/inmunología , Animales , Anticuerpos/análisis , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/inmunología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Células HEK293 , Humanos , Inmunización , Ratones , Microscopía Fluorescente , Tenascina/análisis , Microambiente Tumoral
2.
Matrix Biol ; 116: 1-27, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36669744

RESUMEN

Tracks rich in matrix and cells, as described in several cancer types, have immunosuppressive functions and separate tumor nests and stroma, yet their origin is unknown. Immunostainings of cryosections from mouse breast tumors show that these tracks are bordered by an endothelial-like basement membrane, filled with fibers of collagen adjacent to tenascin-C (TNC) and low-tension fibronectin (Fn) fibers. While present in early-stage tumors and maturing with time, tracks still form under TNC KO conditions, however, host (not tumor cell)-derived TNC is important for track maturation. Tumor infiltrating leukocytes (mostly M2 macrophages and CD8+ T cells) are retained in tracks of early-stage tumors. Following track maturation, retained tumor infiltrating leukocyte (TIL) numbers get reduced and more CD8+ TIL enter the tumor nests in the absence of TNC. As these tracks are enriched with platelets and fibrinogen and have a demarcating endothelial-like basement membrane often adjacent to endothelial cells, this suggests a role of blood vessels in the formation of these tracks. The Fn fiber tension probe FnBPA5 colocalizes with TNC and immune cells in the tracks and shows decreased binding in tracks lacking TNC. Consequently, FnBPA5 can serve as probe for tumor matrix tracks that have immune suppressive properties.


Asunto(s)
Fibronectinas , Neoplasias , Ratones , Animales , Fibronectinas/metabolismo , Células Endoteliales/metabolismo , Neoplasias/patología , Macrófagos/metabolismo , Tenascina/metabolismo , Linfocitos T CD8-positivos/metabolismo , Linfocitos T/metabolismo
3.
Front Immunol ; 12: 635166, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33790905

RESUMEN

The extracellular matrix (ECM) molecule Tenascin-C (TNC) is well-known to promote tumor progression by multiple mechanisms. However, reliable TNC detection in tissues of tumor banks remains limited. Therefore, we generated dromedary single-domain nanobodies Nb3 and Nb4 highly specific for human TNC (hTNC) and characterized the interaction with TNC by several approaches including ELISA, western blot, isothermal fluorescence titration and negative electron microscopic imaging. Our results revealed binding of both nanobodies to distinct sequences within fibronectin type III repeats of hTNC. By immunofluroescence and immunohistochemical imaging we observed that both nanobodies detected TNC expression in PFA and paraffin embedded human tissue from ulcerative colitis, solid tumors and liver metastasis. As TNC impairs cell adhesion to fibronectin we determined whether the nanobodies abolished this TNC function. Indeed, Nb3 and Nb4 restored adhesion of tumor and mesangial cells on a fibronectin/TNC substratum. We recently showed that TNC orchestrates the immune-suppressive tumor microenvironment involving chemoretention, causing tethering of CD11c+ myeloid/dendritic cells in the stroma. Here, we document that immobilization of DC2.4 dendritic cells by a CCL21 adsorbed TNC substratum was blocked by both nanobodies. Altogether, our novel TNC specific nanobodies could offer valuable tools for detection of TNC in the clinical practice and may be useful to inhibit the immune-suppressive and other functions of TNC in cancer and other diseases.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Camelus/inmunología , Anticuerpos de Dominio Único/inmunología , Tenascina/antagonistas & inhibidores , Animales , Anticuerpos Neutralizantes/farmacología , Especificidad de Anticuerpos , Sitios de Unión de Anticuerpos , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Colitis Ulcerosa/inmunología , Colon/inmunología , Ensayo de Inmunoadsorción Enzimática , Células HEK293 , Humanos , Inmunohistoquímica , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/secundario , Unión Proteica , Anticuerpos de Dominio Único/farmacología , Tenascina/administración & dosificación , Tenascina/inmunología
4.
EMBO Mol Med ; 13(6): e13270, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33988305

RESUMEN

Immune checkpoint therapy, where CD8 tumor infiltrating T lymphocytes (TIL) are reactivated, is a promising anti-cancer treatment approach, yet with low response rates. The extracellular matrix, in particular tenascin-C, may generate barriers for TIL. To investigate this possibility, we used a MMTV-NeuNT and syngeneic mammary gland grafting model derived thereof with engineered tenascin-C levels and observed accumulation of CD8 TIL in tenascin-C-rich stroma. Inhibition studies revealed that tenascin-C induced CXCL12 through TLR4. By binding CXCL12, tenascin-C retained CD8 TIL in the stroma. Blockade of CXCR4, the receptor of CXCL12, enhanced macrophage and CD8 TIL infiltration and reduced tumor growth and subsequent metastasis. Retention of CD8 TIL by tenascin-C/CXCL12 was also observed in human breast cancer by tissue staining. Moreover, whereas high CD8 TIL numbers correlated with longer metastasis-free survival, this was not the case when also tenascin-C and CXCL12 levels were high. Altogether, these results may be useful for improving tumor immunity as diagnostic tool and to formulate a future "TIL-matrix-release-and-reactivate" strategy.


Asunto(s)
Linfocitos Infiltrantes de Tumor , Neoplasias , Linfocitos T CD8-positivos , Quimiocina CXCL12 , Matriz Extracelular , Humanos , Tenascina
5.
Cancer Immunol Res ; 8(3): 368-382, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31941671

RESUMEN

The interplay between cancer cells and immune cells is a key determinant of tumor survival. Here, we uncovered how tumors exploit the immunomodulatory properties of the extracellular matrix to create a microenvironment that enables their escape from immune surveillance. Using orthotopic grafting of mammary tumor cells in immunocompetent mice and autochthonous models of breast cancer, we discovered how tenascin-C, a matrix molecule absent from most healthy adult tissues but expressed at high levels and associated with poor patient prognosis in many solid cancers, controls the immune status of the tumor microenvironment. We found that, although host-derived tenascin-C promoted immunity via recruitment of proinflammatory, antitumoral macrophages, tumor-derived tenascin-C subverted host defense by polarizing tumor-associated macrophages toward a pathogenic, immune-suppressive phenotype. Therapeutic monoclonal antibodies that blocked tenascin-C activation of Toll-like receptor 4 reversed this phenotypic switch in vitro and reduced tumor growth and lung metastasis in vivo, providing enhanced benefit in combination with anti-PD-L1 over either treatment alone. Combined tenascin-C:macrophage gene-expression signatures delineated a significant survival benefit in people with breast cancer. These data revealed a new approach to targeting tumor-specific macrophage polarization that may be effective in controlling the growth and spread of breast tumors.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/inmunología , Macrófagos/inmunología , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/inmunología , Femenino , Humanos , Vigilancia Inmunológica , Inmunoterapia/métodos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Ratones , Fenotipo , Tenascina/inmunología , Células Tumorales Cultivadas , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
6.
Cancer Immunol Res ; 8(9): 1122-1138, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32665262

RESUMEN

Inherent immune suppression represents a major challenge in the treatment of human cancer. The extracellular matrix molecule tenascin-C promotes cancer by multiple mechanisms, yet the roles of tenascin-C in tumor immunity are incompletely understood. Using a 4NQO-induced oral squamous cell carcinoma (OSCC) model with abundant and absent tenascin-C, we demonstrated that tenascin-C enforced an immune-suppressive lymphoid stroma via CCL21/CCR7 signaling, leading to increased metastatic tumors. Through TLR4, tenascin-C increased expression of CCR7 in CD11c+ myeloid cells. By inducing CCL21 in lymphatic endothelial cells via integrin α9ß1 and binding to CCL21, tenascin-C immobilized CD11c+ cells in the stroma. Inversion of the lymph node-to-tumor CCL21 gradient, recruitment of T regulatory cells, high expression of anti-inflammatory cytokines, and matrisomal components were hallmarks of the tenascin-C-instructed lymphoid stroma. Ablation of tenascin-C or CCR7 blockade inhibited the lymphoid immune-suppressive stromal properties, reducing tumor growth, progression, and metastasis. Thus, targeting CCR7 could be relevant in human head and neck tumors, as high tenascin-C expression and an immune-suppressive stroma correlate to poor patient survival.


Asunto(s)
Neoplasias de la Boca/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Tenascina/inmunología , Animales , Quimiocina CCL21/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias de la Boca/patología , Receptores CCR7/inmunología , Proteínas Recombinantes/farmacología , Linfocitos T Reguladores/inmunología , Tenascina/farmacología , Microambiente Tumoral/inmunología
7.
Matrix Biol ; 83: 26-47, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31288084

RESUMEN

Metastasis is a major cause of death in cancer patients. The extracellular matrix molecule tenascin-C is a known promoter of metastasis, however the underlying mechanisms are not well understood. To further analyze the impact of tenascin-C on cancer progression we generated MMTV-NeuNT mice that develop spontaneous mammary tumors, on a tenascin-C knockout background. We also developed a syngeneic orthotopic model in which tumor cells derived from a MMTV-NeuNT tumor. Tumor cells were transfected with control shRNA or with shRNA to knockdown tenascin-C expression and, were grafted into the mammary gland of immune competent, wildtype or tenascin-C knockout mice. We show that stromal-derived tenascin-C increases metastasis by reducing apoptosis and inducing the cellular plasticity of cancer cells located in pulmonary blood vessels invasions (BVI), before extravasation. We characterized BVI as organized structures of tightly packed aggregates of proliferating tumor cells with epithelial characteristics, surrounded by Fsp1+ cells, internally located platelets and, a luminal monolayer of endothelial cells. We found extracellular matrix, in particular, tenascin-C, between the stromal cells and the tumor cell cluster. In mice lacking stromal-derived tenascin-C, the organization of pulmonary BVI was significantly affected, revealing novel functions of host-derived tenascin-C in supporting the integrity of the endothelial cell coat, increasing platelet abundance, tumor cell survival, epithelial plasticity, thereby promoting overall lung metastasis. Many effects of tenascin-C observed in BVI including enhancement of cellular plasticity, survival and migration, could be explained by activation of TGF-ß signaling. Finally, in several human cancers, we also observed BVI to be surrounded by an endothelial monolayer and to express tenascin-C. Expression of tenascin-C is specific to BVI and is not observed in lymphatic vascular invasions frequent in breast cancer, which lack an endothelial lining. Given that BVI have prognostic significance for many tumor types, such as shorter cancer patient survival, increased metastasis, vessel occlusion, and organ failure, our data revealing a novel mechanism by which stromal tenascin-C promotes metastasis in human cancer, may have potential for diagnosis and therapy.


Asunto(s)
Vasos Sanguíneos/patología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/patología , Receptor ErbB-2/genética , Tenascina/genética , Animales , Vasos Sanguíneos/metabolismo , Línea Celular Tumoral , Femenino , Técnicas de Inactivación de Genes , Humanos , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/genética , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Transgénicos , Ratas , Transducción de Señal , Células del Estroma , Tenascina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
8.
Methods Cell Biol ; 143: 371-400, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29310788

RESUMEN

The extracellular matrix molecule tenascin-C (TNC) was discovered over 30 years ago, and its tightly regulated pattern of expression since sparked keen interest in the scientific community. In adult tissues, TNC expression is restricted to specific niches and areas of active remodeling or high mechanical strain. However, while most healthy tissues contain little TNC, its transient expression upon cellular stress or tissue injury helps to mediate repair and restore homeostasis. Persistent expression of TNC is associated with chronic inflammation, fibrosis, and cancer, where methods for its detection are emerging as a reliable means to predict disease onset, prognosis, and response to treatment. Because studying the expression of this large matrix molecule is not always straightforward, here we describe basic techniques to examine tissue levels of TNC mRNA and protein. We also describe methods for purifying recombinant TNC, knocking down its expression, and creating cell-derived matrices with or without TNC within.


Asunto(s)
Bioensayo/métodos , Matriz Extracelular/metabolismo , Imagen Molecular/métodos , Tenascina/análisis , Animales , Bioensayo/instrumentación , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Técnicas de Silenciamiento del Gen , Humanos , Ratones , ARN Mensajero/análisis , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/análisis , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Tenascina/genética , Tenascina/aislamiento & purificación , Tenascina/metabolismo
9.
Methods Cell Biol ; 143: 401-428, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29310789

RESUMEN

The extracellular matrix molecule tenascin-C (TNC) has received a lot of attention since its discovery 30 years ago because of its multiple roles in tissue repair, and in pathologies such as chronic inflammation, fibrosis, and cancer. Mouse models with high or no TNC expression have enabled the validation of key roles for TNC in immunity and angiogenesis. In parallel, many approaches including primary cell or organ cultures have shed light on the cellular and molecular mechanisms by which TNC exerts its multiple actions in vivo. Here, we will describe assays that investigate its antiadhesive properties and that measure the effect of TNC on the actin cytoskeleton, cell survival, proliferation, and migration. We will also describe assays to assess the impact of TNC on endothelial and immune cells in cell and organ culture, and to compare the responses of fibroblasts from normal and diseased tissues.


Asunto(s)
Bioensayo/métodos , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Imagen Molecular/métodos , Tenascina/fisiología , Citoesqueleto de Actina/metabolismo , Animales , Bioensayo/instrumentación , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Pollos , Membrana Corioalantoides , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Modelos Animales , Neovascularización Fisiológica/fisiología , Tenascina/análisis
10.
Cancer Res ; 78(4): 950-961, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29259017

RESUMEN

Tenascin-C is an extracellular matrix molecule that drives progression of many types of human cancer, but the basis for its actions remains obscure. In this study, we describe a cell-autonomous signaling mechanism explaining how tenascin-C promotes cancer cell migration in the tumor microenvironment. In a murine xenograft model of advanced human osteosarcoma, tenascin-C and its receptor integrin α9ß1 were determined to be essential for lung metastasis of tumor cells. We determined that activation of this pathway also reduced tumor cell-autonomous expression of target genes for the transcription factor YAP. In clinical specimens, a genetic signature comprising four YAP target genes represents prognostic impact. Taken together, our results illuminate how tumor cell deposition of tenascin-C in the tumor microenvironment promotes invasive migration and metastatic progression.Significance: These results illuminate how the extracellular matrix glycoprotein tenascin-C in the tumor microenvironment promotes invasive migration and metastatic progression by employing integrin α9ß1, abolishing actin stress fiber formation, inhibiting YAP and its target gene expression, with potential implications for cancer prognosis and therapy. Cancer Res; 78(4); 950-61. ©2017 AACR.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Integrinas/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Tenascina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Xenoinjertos , Humanos , Integrinas/genética , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Osteosarcoma/genética , Osteosarcoma/metabolismo , Osteosarcoma/patología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transducción de Señal , Tenascina/genética , Factores de Transcripción , Transfección , Microambiente Tumoral , Proteínas Señalizadoras YAP
11.
Cancer Lett ; 386: 57-64, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27816490

RESUMEN

The vast majority of cancer deaths are caused by the formation of metastases rather than the primary tumor itself. Despite this clinical importance, the molecular and cellular events that support the dissemination of cancer cells are not yet fully unraveled. We have previously shown that CDX2, a homeotic transcription factor essential for gut development, acts as a colon-specific tumor suppressor and opposes metastasis. Here, using a combination of biochemical, biophysical, and immunofluorescence techniques, we further investigated the mechanisms promoted by CDX2 that might antagonize tumor cell dissemination. We found that CDX2 expression regulates the transcription of RHO GEFs, thereby activating RHO signaling cascades that lead to reorganization of the actin cytoskeleton and enhanced adherent junctions. Accordingly, we observed by atomic force microscopy (AFM) that colon cancer cells expressing CDX2 are less deformable, a feature that has been shown to correlate with poor metastatic potential. Thus, this study illustrates how the loss of expression of a transcription factor during colon cancer progression modifies the biomechanical characteristics of tumor cells and hence facilitates invasion and metastasis.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Factor de Transcripción CDX2/metabolismo , Movimiento Celular , Neoplasias del Colon/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Citoesqueleto de Actina/patología , Uniones Adherentes/metabolismo , Uniones Adherentes/patología , Animales , Fenómenos Biomecánicos , Factor de Transcripción CDX2/genética , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Técnica del Anticuerpo Fluorescente , Genes APC , Predisposición Genética a la Enfermedad , Células HT29 , Humanos , Ratones Transgénicos , Microscopía de Fuerza Atómica , Metástasis de la Neoplasia , Fenotipo , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas c-vav/metabolismo , Interferencia de ARN , Transducción de Señal , Transfección , Proteínas Supresoras de Tumor/genética , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
12.
Cell Rep ; 17(10): 2607-2619, 2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27926865

RESUMEN

High expression of the extracellular matrix component tenascin-C in the tumor microenvironment correlates with decreased patient survival. Tenascin-C promotes cancer progression and a disrupted tumor vasculature through an unclear mechanism. Here, we examine the angiomodulatory role of tenascin-C. We find that direct contact of endothelial cells with tenascin-C disrupts actin polymerization, resulting in cytoplasmic retention of the transcriptional coactivator YAP. Tenascin-C also downregulates YAP pro-angiogenic target genes, thus reducing endothelial cell survival, proliferation, and tubulogenesis. Glioblastoma cells exposed to tenascin-C secrete pro-angiogenic factors that promote endothelial cell survival and tubulogenesis. Proteomic analysis of their secretome reveals a signature, including ephrin-B2, that predicts decreased survival of glioma patients. We find that ephrin-B2 is an important pro-angiogenic tenascin-C effector. Thus, we demonstrate dual activities for tenascin-C in glioblastoma angiogenesis and uncover potential targeting and prediction opportunities.


Asunto(s)
Efrina-B2/genética , Glioblastoma/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Proteínas Nucleares/genética , Tenascina/administración & dosificación , Factores de Transcripción/genética , Animales , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Glioblastoma/patología , Humanos , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Proteómica , Transducción de Señal , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA