Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cell ; 163(1): 160-73, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26406376

RESUMEN

Focal adhesion kinase (FAK) promotes anti-tumor immune evasion. Specifically, the kinase activity of nuclear-targeted FAK in squamous cell carcinoma (SCC) cells drives exhaustion of CD8(+) T cells and recruitment of regulatory T cells (Tregs) in the tumor microenvironment by regulating chemokine/cytokine and ligand-receptor networks, including via transcription of Ccl5, which is crucial. These changes inhibit antigen-primed cytotoxic CD8(+) T cell activity, permitting growth of FAK-expressing tumors. Mechanistically, nuclear FAK is associated with chromatin and exists in complex with transcription factors and their upstream regulators that control Ccl5 expression. Furthermore, FAK's immuno-modulatory nuclear activities may be specific to cancerous squamous epithelial cells, as normal keratinocytes do not have nuclear FAK. Finally, we show that a small-molecule FAK kinase inhibitor, VS-4718, which is currently in clinical development, also drives depletion of Tregs and promotes a CD8(+) T cell-mediated anti-tumor response. Therefore, FAK inhibitors may trigger immune-mediated tumor regression, providing previously unrecognized therapeutic opportunities.


Asunto(s)
Carcinoma de Células Escamosas/inmunología , Quimiocina CCL5/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Neoplasias Cutáneas/inmunología , Linfocitos T Reguladores/inmunología , Escape del Tumor , Aminopiridinas/administración & dosificación , Animales , Carcinoma de Células Escamosas/metabolismo , Quimiocina CCL5/inmunología , Modelos Animales de Enfermedad , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Humanos , Queratinocitos/metabolismo , Ratones , Ratones Desnudos , Neoplasias Cutáneas/metabolismo , Transcripción Genética
2.
Gut ; 73(1): 131-155, 2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-36977556

RESUMEN

OBJECTIVE: Immunotherapy for the treatment of pancreatic ductal adenocarcinoma (PDAC) has shown limited efficacy. Poor CD8 T-cell infiltration, low neoantigen load and a highly immunosuppressive tumour microenvironment contribute to this lack of response. Here, we aimed to further investigate the immunoregulatory function of focal adhesion kinase (FAK) in PDAC, with specific emphasis on regulation of the type-II interferon response that is critical in promoting T-cell tumour recognition and effective immunosurveillance. DESIGN: We combined CRISPR, proteogenomics and transcriptomics with mechanistic experiments using a KrasG12Dp53R172H mouse model of pancreatic cancer and validated findings using proteomic analysis of human patient-derived PDAC cell lines and analysis of publicly available human PDAC transcriptomics datasets. RESULTS: Loss of PDAC cell-intrinsic FAK signalling promotes expression of the immunoproteasome and Major Histocompatibility Complex class-I (MHC-I), resulting in increased antigen diversity and antigen presentation by FAK-/- PDAC cells. Regulation of the immunoproteasome by FAK is a critical determinant of this response, optimising the physicochemical properties of the peptide repertoire for high affinity binding to MHC-I. Expression of these pathways can be further amplified in a STAT1-dependent manner via co-depletion of FAK and STAT3, resulting in extensive infiltration of tumour-reactive CD8 T-cells and further restraint of tumour growth. FAK-dependent regulation of antigen processing and presentation is conserved between mouse and human PDAC, but is lost in cells/tumours with an extreme squamous phenotype. CONCLUSION: Therapies aimed at FAK degradation may unlock additional therapeutic benefit for the treatment of PDAC through increasing antigen diversity and promoting antigen presentation.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Ratones , Humanos , Animales , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Presentación de Antígeno , Evasión Inmune , Proteómica , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/patología , Microambiente Tumoral , Línea Celular Tumoral
3.
Br J Cancer ; 127(10): 1893-1905, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36138073

RESUMEN

BACKGROUND: Pancreatic Cancer is one of the most lethal cancers, with less than 8% of patients surviving 5 years following diagnosis. The last 40 years have seen only small incremental improvements in treatment options, highlighting the continued need to better define the cellular and molecular pathways contributing to therapy response and patient prognosis. METHODS: We combined CRISPR, shRNA and flow cytometry with mechanistic experiments using a KrasG12Dp53R172H mouse model of pancreatic cancer and analysis of publicly available human PDAC transcriptomic datasets. RESULTS: Here, we identify that expression of the immune checkpoint, Programmed Death Ligand 2 (PD-L2), is associated with poor prognosis, tumour grade, clinical stage and molecular subtype in patients with Pancreatic Ductal Adenocarcinoma (PDAC). We further show that PD-L2 is predominantly expressed in the stroma and, using an orthotopic murine model of PDAC, identify cancer cell-intrinsic Focal Adhesion Kinase (FAK) signalling as a regulator of PD-L2 stromal expression. Mechanistically, we find that FAK regulates interleukin-6, which can act in concert with interleukin-4 secreted by CD4 T-cells to drive elevated expression of PD-L2 on tumour-associated macrophages, dendritic cells and endothelial cells. CONCLUSIONS: These findings identify further complex heterocellular signalling networks contributing to FAK-mediated immune suppression in pancreatic cancer.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animales , Humanos , Ratones , Carcinoma Ductal Pancreático/patología , Células Endoteliales/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas
4.
J Immunol ; 203(6): 1579-1588, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31427445

RESUMEN

Neutrophils are abundant circulating leukocytes that are rapidly recruited to sites of inflammation in an integrin-dependent fashion. Contrasting with the well-characterized regulation of integrin activation, mechanisms regulating integrin inactivation remain largely obscure. Using mouse neutrophils, we demonstrate in this study that the GTPase activating protein ARAP3 is a critical regulator of integrin inactivation; experiments with Chinese hamster ovary cells indicate that this is not restricted to neutrophils. Specifically, ARAP3 acts in a negative feedback loop downstream of PI3K to regulate integrin inactivation. Integrin ligand binding drives the activation of PI3K and of its effectors, including ARAP3, by outside-in signaling. ARAP3, in turn, promotes localized integrin inactivation by negative inside-out signaling. This negative feedback loop reduces integrin-mediated PI3K activity, with ARAP3 effectively switching off its own activator, while promoting turnover of substrate adhesions. In vitro, ARAP3-deficient neutrophils display defective PIP3 polarization, adhesion turnover, and transendothelial migration. In vivo, ARAP3-deficient neutrophils are characterized by a neutrophil-autonomous recruitment defect to sites of inflammation.


Asunto(s)
Inflamación/metabolismo , Integrinas/metabolismo , Neutrófilos/metabolismo , Animales , Células CHO , Adhesión Celular/fisiología , Línea Celular , Cricetulus , Proteínas Activadoras de GTPasa/metabolismo , Ratones , Infiltración Neutrófila/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/fisiología
5.
J Cell Sci ; 126(Pt 2): 393-401, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23525005

RESUMEN

E-cadherin is a single-pass transmembrane protein that mediates homophilic cell-cell interactions. Tumour progression is often associated with the loss of E-cadherin function and the transition to a more motile and invasive phenotype. This requires the coordinated regulation of both E-cadherin-mediated cell-cell adhesions and integrin-mediated adhesions that contact the surrounding extracellular matrix (ECM). Regulation of both types of adhesion is dynamic as cells respond to external cues from the tumour microenvironment that regulate polarity, directional migration and invasion. Here, we review the mechanisms by which tumour cells control the cross-regulation between dynamic E-cadherin-mediated cell-cell adhesions and integrin-mediated cell-matrix contacts, which govern the invasive and metastatic potential of tumours. In particular, we will discuss the role of the adhesion-linked kinases Src, focal adhesion kinase (FAK) and integrin-linked kinase (ILK), and the Rho family of GTPases.


Asunto(s)
Cadherinas/metabolismo , Integrinas/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Animales , Adhesión Celular/fisiología , Comunicación Celular/fisiología , Línea Celular Tumoral , Humanos , Invasividad Neoplásica , Metástasis de la Neoplasia , Transducción de Señal , Microambiente Tumoral
6.
Int J Cancer ; 131(2): 287-97, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21823119

RESUMEN

Focal adhesion kinase (FAK) is upregulated in several epithelial tumours and there has been considerable interest in developing small molecule kinase inhibitors of FAK. However, FAK also has important adaptor functions within the cell, integrating signals from both integrins and growth factors. To investigate the role of FAKs kinase domain, we generated fak-deficient squamous cell carcinoma (SCC) cell lines. Re-expression of a wild type or kinase dead FAK allowed us to delineate its kinase dependent functions. In addition, we used the novel FAK kinase inhibitor PF-562,271. The kinase activity of FAK was important for tumour cell migration and polarity but more striking was its requirement for the anchorage independent 3 dimensional (3D) proliferation of SCC cells and their growth as xenografts in mice. Inhibition of FAK activity and prevention of growth in 3D correlated with Src inhibition. We further identified a mechanism whereby FAK regulates proliferation in 3D via regulation of the kinase activity of Src. This was dependent on the kinase activity of FAK and its resulting phosphorylation on Y397 that provides a high affinity binding site for Src. These data support the further development of FAK kinase inhibitors as agents that have the potential to inhibit both tumour cell migration and proliferation.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/patología , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Animales , Carcinoma de Células Escamosas/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Polaridad Celular , Dasatinib , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Proteína-Tirosina Quinasas de Adhesión Focal/deficiencia , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Indoles/farmacología , Ratones , Ratones Transgénicos , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Transducción de Señal , Sulfonamidas/farmacología , Tiazoles/farmacología , Familia-src Quinasas/metabolismo
7.
Elife ; 92020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31959281

RESUMEN

Focal Adhesion Kinase (FAK) inhibitors are currently undergoing clinical testing in combination with anti-PD-1 immune checkpoint inhibitors. However, which patients are most likely to benefit from FAK inhibitors, and what the optimal FAK/immunotherapy combinations are, is currently unknown. We identify that cancer cell expression of the T-cell co-stimulatory ligand CD80 sensitizes murine tumors to a FAK inhibitor and show that CD80 is expressed by human cancer cells originating from both solid epithelial cancers and some hematological malignancies in which FAK inhibitors have not been tested clinically. In the absence of CD80, we identify that targeting alternative T-cell co-stimulatory receptors, in particular OX-40 and 4-1BB in combination with FAK, can drive enhanced anti-tumor immunity and even complete regression of murine tumors. Our findings provide rationale supporting the clinical development of FAK inhibitors in combination with patient selection based on cancer cell CD80 expression, and alternatively with therapies targeting T-cell co-stimulatory pathways.


Asunto(s)
Quinasa 1 de Adhesión Focal , Inhibidores de Puntos de Control Inmunológico , Linfocitos T , Animales , Antígeno B7-1/inmunología , Antígeno B7-1/metabolismo , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
8.
Oncoimmunology ; 8(8): 1608106, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31413918

RESUMEN

Immunization of patients with autologous, ex vivo matured dendritic cell (DC) preparations, in order to prime antitumor T-cell responses, is the focus of intense research. Despite progress and approval of clinical approaches, significant enhancement of these personalized immunotherapies is urgently needed to improve efficacy. We show that immunotherapeutic murine and human DC, generated in the presence of the antimicrobial host defense peptide LL-37, have dramatically enhanced expansion and differentiation of cells with key features of the critical CD103+/CD141+ DC subsets, including enhanced cross-presentation and co-stimulatory capacity, and upregulation of CCR7 with improved migratory capacity. These LL-37-DC enhanced proliferation, activation and cytokine production by CD8+ (but not CD4+) T cells in vitro and in vivo. Critically, tumor antigen-presenting LL-37-DC increased migration of primed, activated CD8+ T cells into established squamous cell carcinomas in mice, and resulted in tumor regression. This advance therefore has the potential to dramatically enhance DC immunotherapy protocols.

9.
Clin Cancer Res ; 12(11 Pt 1): 3272-9, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16740747

RESUMEN

UNLABELLED: The development of human malignancies can involve the aberrant regulation of intracellular signal transduction pathways that regulate cell-extracellular matrix interactions. PURPOSE: In the current study, we aimed to evaluate focal adhesion kinase (FAK) at both genetic and protein expression levels in head and neck squamous cell carcinomas (HNSCC) and to explore the prognostic significance of FAK. EXPERIMENTAL DESIGN: A total of 211 tissue specimens, including 147 primary tumors, 56 lymph node metastases, 3 benign hyperplasias, and 5 dysplasias, were analyzed using immunohistochemistry. The fak gene dosage was determined in 33 tumors. Correlations among DNA, protein, and clinicopathologic variables were analyzed. RESULTS: FAK protein was overexpressed in HNSCCs compared with corresponding normal mucosa. High expression levels were found in 62% of the samples. Positive immunostaining was also detected in benign hyperplasias and preinvasive dysplastic lesions. All lymph node metastases examined showed FAK overexpression, with significant correlation with the expression in matched primary tumor. DNA copy number ratios for fak were higher in 39% of the tumors compared with normal mucosa. However, elevated FAK expression did not correlate with gains on DNA level, and not all cases with an amplification of the fak gene displayed protein overexpression. Similar data were obtained in five HNSCC-derived cell lines, in which FAK mRNA levels were precisely correlated with FAK protein levels. FAK protein overexpression in tumors correlated with nodal metastases. CONCLUSIONS: These findings suggest an involvement of FAK in the onset and progression of HNSCC and provide an insight into a mechanism of FAK activation alternative to gene amplification.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Dosificación de Gen , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/enzimología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Inmunohistoquímica , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Familia-src Quinasas/metabolismo
10.
Arch Otolaryngol Head Neck Surg ; 133(2): 145-50, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17309983

RESUMEN

OBJECTIVE: To explore the value of E-cadherin and focal adhesion kinase (FAK) expression in the prediction of cervical lymph node metastases in squamous cell carcinoma of the supraglottic larynx. DESIGN: Immunohistochemical analysis of retrospectively selected cases. Patients The study population was composed of 95 previously untreated men with squamous cell carcinoma of the supraglottic larynx. Intervention All the patients underwent surgical resection of the tumor and bilateral neck dissection. MAIN OUTCOME MEASURES: E-cadherin and FAK expression in relation to nodal metastases. RESULTS: Decreased E-cadherin expression was correlated with the presence of nodal metastases (P = .006). The combination of E-cadherin and FAK expression resulted in a superior accuracy in assessing nodal metastasis (P = .001). Histological grade was also associated with nodal metastases (P = .005). Multivariate analysis confirmed that these parameters were independent predictors of nodal metastases. In addition, the cases with decreased E-cadherin and increased FAK expression presented a significantly reduced disease-specific survival (P = .005). CONCLUSION: The combination of the expression of E-cadherin and FAK could increase our ability to identify patients with clinically negative lymph nodes who are at considerable risk for occult metastases.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Cadherinas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Neoplasias Laríngeas/metabolismo , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/secundario , Humanos , Inmunohistoquímica , Neoplasias Laríngeas/mortalidad , Neoplasias Laríngeas/patología , Modelos Logísticos , Metástasis Linfática/diagnóstico , Masculino , Análisis Multivariante , Recurrencia Local de Neoplasia , Estudios Retrospectivos , Tasa de Supervivencia
11.
Acta Otorrinolaringol Esp ; 58(8): 347-51, 2007 Oct.
Artículo en Español | MEDLINE | ID: mdl-17949661

RESUMEN

INTRODUCTION AND OBJECTIVES: FAK expression is frequently elevated in human cancers, including head and neck squamous cell carcinomas. However, the regulation of FAK expression is poorly understood. The aim of this study is to establish the possible role of the p53 tumour suppressor gene in the regulation of FAK expression in squamous cell carcinomas of the larynx. MATERIAL AND METHOD: The expression of FAK and p53 proteins were studied using immunohistochemistry in 102 samples from surgically-treated squamous cell carcinomas of the supraglottic larynx (with postoperative radiotherapy in 49 cases). RESULTS: All the cases showed some degree of FAK expression, which was moderate to intense in 60 cases (59 %). Also 60 cases showed positive p53 expression (>10 % of tumour cells with nuclear staining). No relationship was observed between p53 positivity and FAK expression (P= .54). In addition, none of these proteins presented association with the prognosis of the patients. CONCLUSIONS: Our results suggests that p53 activity does not correlate with FAK expression in squamous cell carcinomas of the larynx.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Quinasa 1 de Adhesión Focal/genética , Genes p53 , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/patología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/cirugía , Femenino , Humanos , Neoplasias Laríngeas/cirugía , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Resultado del Tratamiento
12.
Cancer Res ; 77(19): 5301-5312, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28807942

RESUMEN

Nuclear focal adhesion kinase (FAK) is a potentially important regulator of gene expression in cancer, impacting both cellular function and the composition of the surrounding tumor microenvironment. Here, we report in a murine model of skin squamous cell carcinoma (SCC) that nuclear FAK regulates Runx1-dependent transcription of insulin-like growth factor binding protein 3 (IGFBP3), and that this regulates SCC cell-cycle progression and tumor growth in vivo Furthermore, we identified a novel molecular complex between FAK and Runx1 in the nucleus of SCC cells and showed that FAK interacted with a number of Runx1-regulatory proteins, including Sin3a and other epigenetic modifiers known to alter Runx1 transcriptional function through posttranslational modification. These findings provide important new insights into the role of FAK as a scaffolding protein in molecular complexes that regulate gene transcription. Cancer Res; 77(19); 5301-12. ©2017 AACR.


Asunto(s)
Carcinoma de Células Escamosas/patología , Núcleo Celular/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/fisiología , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Neoplasias Cutáneas/patología , Animales , Apoptosis , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Ciclo Celular , Núcleo Celular/genética , Proliferación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Ratones , Ratones Noqueados , Ratones Desnudos , Fosforilación , Transducción de Señal , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Células Tumorales Cultivadas
13.
Sci Signal ; 10(508)2017 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-29208683

RESUMEN

Focal adhesion kinase (FAK) mediates tumor cell-intrinsic behaviors that promote tumor growth and metastasis. We previously showed that FAK also induces the expression of inflammatory genes that inhibit antitumor immunity in the microenvironment. We identified a crucial, previously unknown role for the dual-function cytokine interleukin-33 (IL-33) in FAK-dependent immune evasion. In murine squamous cell carcinoma (SCC) cells, specifically nuclear FAK enhanced the expression of the genes encoding IL-33, the chemokine CCL5, and the soluble, secreted form of the IL-33 receptor, called soluble ST2 (sST2). The abundance of IL-33 and CCL5 was increased in FAK-positive SCC cells but not in normal keratinocytes. IL-33 associated with FAK in the nucleus, and the FAK-IL-33 complex interacted with a network of chromatin modifiers and transcriptional regulators, including TAF9, WDR82, and BRD4, which promote the activity of nuclear factor κB (NF-κB) and its induction of genes encoding chemokines, including CCL5. We did not detect secretion of IL-33 from FAK-positive SCC cells; thus, we propose that the increased production and secretion of sST2 likely sequesters IL-33 secreted by other cell types within the tumor environment, thus blocking its stimulatory effects on infiltrating host immune cells. Depleting FAK, IL-33, or sST2 from SCC cells before implantation induced tumor regression in syngeneic mice, except when CD8+ T cells were co-depleted. Our data provide mechanistic insight into how FAK controls the tumor immune environment, namely, through a transcriptional regulatory network mediated by nuclear IL-33. Targeting this axis may boost antitumor immunity in patients.


Asunto(s)
Carcinoma de Células Escamosas/inmunología , Quinasa 1 de Adhesión Focal/metabolismo , Redes Reguladoras de Genes , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Neoplasias Cutáneas/metabolismo , Escape del Tumor/genética , Animales , Carcinoma de Células Escamosas/genética , Núcleo Celular/inmunología , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Quinasa 1 de Adhesión Focal/genética , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/genética , Interleucina-33/genética , Isoinjertos , Queratinocitos/inmunología , Ratones , Ratones Transgénicos , Proteómica , Células Tumorales Cultivadas
14.
Oncotarget ; 7(10): 11539-52, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26883193

RESUMEN

Resistance to human epidermal growth factor receptor 2 (HER2)-targeted therapies in the treatment of HER2-positive breast cancer is a major clinical problem. To identify pathways linked to resistance, we generated HER2-positive breast cancer cell lines which are resistant to either lapatinib or AZD8931, two pan-HER family kinase inhibitors. Resistance was HER2 independent and was associated with epithelial-to-mesenchymal transition (EMT), resulting in increased proliferation and migration of the resistant cells. Using a global proteomics approach, we identified a novel set of EMT-associated proteins linked to HER2-independent resistance. We demonstrate that a subset of these EMT-associated genes is predictive of prognosis within the ERBB2 subtype of human breast cancers. Furthermore, targeting the EMT-associated kinases Src and Axl potently inhibited proliferation of the resistant cells, and inhibitors to these kinases may provide additional options for the treatment of HER2-independent resistance in tumors.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Receptor ErbB-2/metabolismo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Femenino , Humanos , Lapatinib , Terapia Molecular Dirigida , Pronóstico , Proteómica , Quinazolinas/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Transducción de Señal
15.
Cell Adh Migr ; 5(4): 360-5, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21836391

RESUMEN

Recent advances in confocal and multi-photon microscopy, together with fluorescent probe development, have enabled cancer biology studies to go beyond the culture dish and interrogate cancer-associated processes in the complex in vivo environment. Regulation of the tumor suppressor protein E-cadherin plays an important role in cancer development and progression and may contribute to the decision between 'single cell' and 'collective invasion' in vivo. Mounting evidence from in vitro and in vivo experiments places the two non-receptor protein tyrosine kinases Src and Focal Adhesion Kinase, at the heart of E-cadherin regulation, and the crosstalk between integrins and cadherins. Here we discuss recent insights, attained using high resolution fluorescent in vivo imaging, into the regulation of E-cadherin and collective invasion. We focus on the regulatory crosstalk between the Src/FAK signaling axis and E-cadherin in vivo.


Asunto(s)
Cadherinas/metabolismo , Movimiento Celular , Quinasa 1 de Adhesión Focal/metabolismo , Familia-src Quinasas/metabolismo , Uniones Adherentes/metabolismo , Animales , Adhesión Celular , Transición Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Neoplasias/genética , Neoplasias/patología , Transducción de Señal
16.
Pharmaceutics ; 3(2): 141-70, 2011 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24310493

RESUMEN

Dynamic regulation of specific molecular processes and cellular phenotypes in live cell systems reveal unique insights into cell fate and drug pharmacology that are not gained from traditional fixed endpoint assays. Recent advances in microscopic imaging platform technology combined with the development of novel optical biosensors and sophisticated image analysis solutions have increased the scope of live cell imaging applications in drug discovery. We highlight recent literature examples where live cell imaging has uncovered novel insight into biological mechanism or drug mode-of-action. We survey distinct types of optical biosensors and associated analytical methods for monitoring molecular dynamics, in vitro and in vivo. We describe the recent expansion of live cell imaging into automated target validation and drug screening activities through the development of dedicated brightfield and fluorescence kinetic imaging platforms. We provide specific examples of how temporal profiling of phenotypic response signatures using such kinetic imaging platforms can increase the value of in vitro high-content screening. Finally, we offer a prospective view of how further application and development of live cell imaging technology and reagents can accelerate preclinical lead optimization cycles and enhance the in vitro to in vivo translation of drug candidates.

17.
Cancer Res ; 70(22): 9413-22, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21045155

RESUMEN

Most cancer-related deaths are due to the development of metastatic disease, and several new molecularly targeted agents in clinical development have the potential to prevent disease progression. However, it remains difficult to assess the efficacy of antimetastatic agents in the clinical setting, and an increased understanding of how such agents work at different stages of the metastatic cascade is important in guiding their clinical use. We used optical window chambers combined with photobleaching, photoactivation, and photoswitching to quantitatively measure (a) tumor cell movement and proliferation by tracking small groups of cells in the context of the whole tumor, and (b) E-cadherin molecular dynamics in vivo following perturbation of integrin signaling by inhibiting focal adhesion kinase (FAK) and Src. We show that inhibition of Src and FAK suppresses E-cadherin-dependent collective cell movement in a complex three-dimensional tumor environment, and modulates cell-cell adhesion strength and endocytosis in vitro. This shows a novel role for integrin signaling in the regulation of E-cadherin internalization, which is linked to regulation of collective cancer cell movement. This work highlights the power of fluorescent, direct, in vivo imaging approaches in the preclinical evaluation of chemotherapeutic agents, and shows that inhibition of the Src/FAK signaling axis may provide a strategy to prevent tumor cell spread by deregulating E-cadherin-mediated cell-cell adhesions.


Asunto(s)
Cadherinas/metabolismo , Movimiento Celular , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Integrinas/metabolismo , Transducción de Señal , Familia-src Quinasas/metabolismo , Animales , Cadherinas/genética , Adhesión Celular , Comunicación Celular , Línea Celular Tumoral , Proliferación Celular , Endocitosis , Recuperación de Fluorescencia tras Fotoblanqueo/métodos , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Humanos , Immunoblotting , Integrina beta1/genética , Integrina beta1/metabolismo , Integrinas/genética , Ratones , Simulación de Dinámica Molecular , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Interferencia de ARN , Familia-src Quinasas/genética
18.
Cell Adh Migr ; 4(4): 491-501, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20595808

RESUMEN

The dynamic control of E-cadherin is critical for establishing and maintaining cell-cell junctions in epithelial cells. The concentration of E-cadherin molecules at adherens junctions (AJs) is regulated by lateral movement of E-cadherin within the plasma membrane and endocytosis. Here we set out to study the interplay between these processes and their contribution to E-cadherin dynamics. Using photoactivation (PA) and fluorescence recovery after photobleaching (FRAP) we were able to monitor the fate of E-cadherin molecules within the plasma membrane. Our results suggest that the motility of E-cadherin within, and away from, the cell surface are not exclusive or independent mechanisms and there is a fine balance between the two which when perturbed can have dramatic effects on the regulation of AJs.


Asunto(s)
Cadherinas/metabolismo , Membrana Celular/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Uniones Adherentes/metabolismo , Adhesión Celular , Línea Celular Tumoral , Clonación Molecular , Dinaminas/antagonistas & inhibidores , Endocitosis , Recuperación de Fluorescencia tras Fotoblanqueo , Humanos , Hidrazonas/farmacología , Estimulación Luminosa , Transporte de Proteínas
19.
Curr Biol ; 20(12): 1086-92, 2010 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-20493699

RESUMEN

A fundamental question in cell biology concerns how cells respond to their environment by polarizing after sensing directional cues. This requires the differential localization of protein complexes in cells, and it is important to identify and understand how these complexes function. Here we describe a novel "direction-sensing" pathway that links the integrin effector focal adhesion kinase (FAK), the molecular scaffold protein RACK1, and activity of one of its client proteins, PDE4D5, a cAMP-degrading phosphodiesterase. The complex is recruited to nascent adhesions and promotes cell polarity. We identify FAK FERM domain residues whose mutation impairs RACK1 binding. When re-expressed in cancer cells in which endogenous fak is deleted by Cre-lox-mediated recombination, the RACK1-binding-impaired FAK mutant protein does not support formation of nascent actin adhesion structures as cells spread. These cancer cells, like FAK-deficient cells, cannot undergo directional responses, including wound-induced polarization or chemotactic invasion into three-dimensional matrix gels. We show that RACK1 serves as the molecular bridge linking FAK to the recruitment of PDE4D5. FAK/RACK1/PDE4D5 is a novel 'direction-sensing' complex that acts to recruit specific components of the cAMP second-messenger system to nascent integrin adhesions and to the leading edge of polarizing cells.


Asunto(s)
Polaridad Celular , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/patología , Receptores de Superficie Celular/metabolismo , Quimiotaxis , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4 , Proteínas de Unión al GTP/genética , Humanos , Mutación , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Receptores de Cinasa C Activada , Receptores de Superficie Celular/genética
20.
Cell Adh Migr ; 3(4): 351-4, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19690469

RESUMEN

Despite our advanced understanding of primary cancer development and progression, metastasis and the systemic spread of the disease to secondary sites remains the leading cause of cancer-associated death. The metastatic process is therefore a major potential therapeutic target area for cancer researchers and elucidating the key steps that are susceptible to therapeutic intervention will be critical to improve our treatment strategies. Recent advances in intravital imaging are rapidly improving our insight into this process and are helping in the design of stage-specific drug regimes for the treatment of metastatic cancer. Here we discuss current developments in intravital imaging and our recent use of photobleaching and photoactivation in the analysis of dynamic biomarkers in living animals to assess the efficacy of therapeutic intervention on early stages of tumor cell metastasis.


Asunto(s)
Imagen Molecular/tendencias , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/patología , Animales , Humanos , Microscopía Fluorescente/métodos , Microscopía Fluorescente/tendencias , Imagen Molecular/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA