Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Gut ; 73(1): 131-155, 2023 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-36977556

RESUMO

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.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Camundongos , Humanos , Animais , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Apresentação de Antígeno , Evasão da Resposta Imune , Proteômica , Neoplasias Pancreáticas/patologia , Carcinoma Ductal Pancreático/patologia , Microambiente Tumoral , Linhagem Celular Tumoral
2.
Br J Cancer ; 127(10): 1893-1905, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36138073

RESUMO

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.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animais , Humanos , Camundongos , Carcinoma Ductal Pancreático/patologia , Células Endoteliais/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas
3.
Elife ; 92020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31959281

RESUMO

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.


Assuntos
Quinase 1 de Adesão Focal , Inibidores de Checkpoint Imunológico , Linfócitos T , Animais , Antígeno B7-1/imunologia , Antígeno B7-1/metabolismo , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Quinase 1 de Adesão Focal/antagonistas & inibidores , Quinase 1 de Adesão Focal/imunologia , Humanos , Inibidores de Checkpoint Imunológico/imunologia , Inibidores de Checkpoint Imunológico/farmacologia , Camundongos , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/metabolismo , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
4.
Oncoimmunology ; 8(8): 1608106, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31413918

RESUMO

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.

5.
J Immunol ; 203(6): 1579-1588, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31427445

RESUMO

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.


Assuntos
Inflamação/metabolismo , Integrinas/metabolismo , Neutrófilos/metabolismo , Animais , Células CHO , Adesão Celular/fisiologia , Linhagem Celular , Cricetulus , Proteínas Ativadoras de GTPase/metabolismo , Camundongos , Infiltração de Neutrófilos/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/fisiologia
6.
Sci Signal ; 10(508)2017 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-29208683

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas/imunologia , Quinase 1 de Adesão Focal/metabolismo , Redes Reguladoras de Genes , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Neoplasias Cutâneas/metabolismo , Evasão Tumoral/genética , Animais , Carcinoma de Células Escamosas/genética , Núcleo Celular/imunologia , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Quinase 1 de Adesão Focal/genética , Humanos , Proteína 1 Semelhante a Receptor de Interleucina-1/genética , Interleucina-33/genética , Isoenxertos , Queratinócitos/imunologia , Camundongos , Camundongos Transgênicos , Proteômica , Células Tumorais Cultivadas
7.
Cancer Res ; 77(19): 5301-5312, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28807942

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas/patologia , Núcleo Celular/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Neoplasias Cutâneas/patologia , Animais , Apoptose , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Ciclo Celular , Núcleo Celular/genética , Proliferação de Células , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Camundongos , Camundongos Knockout , Camundongos Nus , Fosforilação , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Células Tumorais Cultivadas
8.
Oncotarget ; 7(10): 11539-52, 2016 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-26883193

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Receptor ErbB-2/metabolismo , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal , Feminino , Humanos , Lapatinib , Terapia de Alvo Molecular , Prognóstico , Proteômica , Quinazolinas/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Transdução de Sinais
9.
Cell ; 163(1): 160-73, 2015 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-26406376

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas/imunologia , Quimiocina CCL5/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Neoplasias Cutâneas/imunologia , Linfócitos T Reguladores/imunologia , Evasão Tumoral , Aminopiridinas/administração & dosagem , Animais , Carcinoma de Células Escamosas/metabolismo , Quimiocina CCL5/imunologia , Modelos Animais de Doenças , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Proteína-Tirosina Quinases de Adesão Focal/genética , Humanos , Queratinócitos/metabolismo , Camundongos , Camundongos Nus , Neoplasias Cutâneas/metabolismo , Transcrição Gênica
10.
J Cell Sci ; 126(Pt 2): 393-401, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23525005

RESUMO

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.


Assuntos
Caderinas/metabolismo , Integrinas/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Animais , Adesão Celular/fisiologia , Comunicação Celular/fisiologia , Linhagem Celular Tumoral , Humanos , Invasividade Neoplásica , Metástase Neoplásica , Transdução de Sinais , Microambiente Tumoral
11.
Int J Cancer ; 131(2): 287-97, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21823119

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/patologia , Movimento Celular , Proliferação de Células/efeitos dos fármacos , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Animais , Carcinoma de Células Escamosas/genética , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Polaridade Celular , Dasatinibe , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Proteína-Tirosina Quinases de Adesão Focal/deficiência , Proteína-Tirosina Quinases de Adesão Focal/genética , Indóis/farmacologia , Camundongos , Camundongos Transgênicos , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Transdução de Sinais , Sulfonamidas/farmacologia , Tiazóis/farmacologia , Quinases da Família src/metabolismo
12.
Cell Adh Migr ; 5(4): 360-5, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21836391

RESUMO

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.


Assuntos
Caderinas/metabolismo , Movimento Celular , Quinase 1 de Adesão Focal/metabolismo , Quinases da Família src/metabolismo , Junções Aderentes/metabolismo , Animais , Adesão Celular , Transição Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Neoplasias/genética , Neoplasias/patologia , Transdução de Sinais
13.
Pharmaceutics ; 3(2): 141-70, 2011 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-24310493

RESUMO

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.

14.
Cancer Res ; 70(22): 9413-22, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21045155

RESUMO

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.


Assuntos
Caderinas/metabolismo , Movimento Celular , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Integrinas/metabolismo , Transdução de Sinais , Quinases da Família src/metabolismo , Animais , Caderinas/genética , Adesão Celular , Comunicação Celular , Linhagem Celular Tumoral , Proliferação de Células , Endocitose , Recuperação de Fluorescência Após Fotodegradação/métodos , Proteína-Tirosina Quinases de Adesão Focal/genética , Humanos , Immunoblotting , Integrina beta1/genética , Integrina beta1/metabolismo , Integrinas/genética , Camundongos , Simulação de Dinâmica Molecular , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Interferência de RNA , Quinases da Família src/genética
15.
Cell Adh Migr ; 4(4): 491-501, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20595808

RESUMO

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.


Assuntos
Caderinas/metabolismo , Membrana Celular/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Junções Aderentes/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Clonagem Molecular , Dinaminas/antagonistas & inibidores , Endocitose , Recuperação de Fluorescência Após Fotodegradação , Humanos , Hidrazonas/farmacologia , Estimulação Luminosa , Transporte Proteico
16.
Curr Biol ; 20(12): 1086-92, 2010 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-20493699

RESUMO

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.


Assuntos
Polaridade Celular , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/patologia , Receptores de Superfície Celular/metabolismo , Quimiotaxia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Proteínas de Ligação ao GTP/genética , Humanos , Mutação , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Receptores de Quinase C Ativada , Receptores de Superfície Celular/genética
17.
Cell Adh Migr ; 3(4): 351-4, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19690469

RESUMO

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.


Assuntos
Imagem Molecular/tendências , Invasividade Neoplásica/patologia , Metástase Neoplásica/patologia , Animais , Humanos , Microscopia de Fluorescência/métodos , Microscopia de Fluorescência/tendências , Imagem Molecular/métodos
18.
Cancer Res ; 69(7): 2714-9, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19318551

RESUMO

The ability of tumor cells to invade and metastasize requires deregulation of interactions with adjacent cells and the extracellular matrix. A major challenge of cancer biology is to observe the dynamics of the proteins involved in this process in their functional and physiologic context. Here, for the first time, we have used photobleaching and photoactivation to compare the mobility of cell adhesion and plasma membrane probes in vitro and in tumors grown in mice (in vivo). We find differences between in vitro and in vivo recovery dynamics of two key molecules, the tumor suppressor E-cadherin and the membrane-targeting sequence of H-Ras. Our data show that E-cadherin dynamics are significantly faster in vivo compared with cultured cells, that the ratio of E-cadherin stabilized in cell-cell junctions is significantly higher in vivo, and that E-cadherin mobility correlates with cell migration. Moreover, quantitative imaging has allowed us to assess the effects of therapeutic intervention on E-cadherin dynamics using dasatinib, a clinically approved Src inhibitor, and show clear differences in the efficacy of drug treatment in vivo. Our results show for the first time the utility of photobleaching and photoactivation in the analysis of dynamic biomarkers in living animals. Furthermore, this work highlights critical differences in molecular dynamics in vitro and in vivo, which have important implications for the use of cultured disease models as surrogates for living tissue.


Assuntos
Caderinas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Comunicação Celular/fisiologia , Membrana Celular/fisiologia , Animais , Caderinas/química , Carcinoma de Células Escamosas/tratamento farmacológico , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Movimento Celular/fisiologia , Modelos Animais de Doenças , Desenho de Fármacos , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Camundongos Nus , Fotodegradação , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Transplante Heterólogo , Proteínas ras/metabolismo
19.
Acta Otorrinolaringol Esp ; 58(8): 347-51, 2007 Oct.
Artigo em Espanhol | MEDLINE | ID: mdl-17949661

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Quinase 1 de Adesão Focal/genética , Genes p53 , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Escamosas/cirurgia , Feminino , Humanos , Neoplasias Laríngeas/cirurgia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Resultado do Tratamento
20.
Acta otorrinolaringol. esp ; 58(8): 347-351, oct. 2007. ilus, tab
Artigo em Es | IBECS | ID: ibc-056394

RESUMO

Introducción y objetivos: La expresión de la proteína FAK frecuentemente se encuentra elevada en múltiples tipos de cánceres, como los carcinomas epidermoides de cabeza y cuello. Sin embargo, los mecanismos que regulan la expresión de dicha proteína son poco conocidos. Nuestro objetivo es analizar si el supresor tumoral p53 está implicado en la regulación de la expresión de FAK en los carcinomas epidermoides de laringe. Material y método: Se estudia mediante inmunohistoquímica la expresión de las proteínas FAK y p53 en una muestra de 102 carcinomas epidermoides de laringe supraglótica tratados quirúrgicamente (con radioterapia postoperatoria en 49 casos). Resultados: Todos los tumores presentaban algún grado de expresión de FAK, que era moderada o intensa en 60 (59 %) de los casos. También en 60 casos hubo positividad de p53 (> 10 % de las células tumorales con tinción nuclear). No se halló relación entre la positividad de p53 y la expresión de FAK (p = 0,54). La expresión de cualquiera de las dos proteínas no mostró relación con el pronóstico de los pacientes. Conclusiones: Nuestros resultados indican que la actividad de p53 no se relaciona con la expresión de FAK en los carcinomas epidermoides de laringe


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


Assuntos
Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Idoso , Idoso de 80 Anos ou mais , Humanos , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/metabolismo , Biomarcadores Tumorais/análise , Neoplasias Laríngeas/patologia , Neoplasias Laríngeas/metabolismo , Proteínas Tirosina Quinases/análise , Proteína Supressora de Tumor p53/análise , Prognóstico , Resultado do Tratamento , Estadiamento de Neoplasias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...