Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Breast Cancer Res ; 25(1): 27, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36922898

RESUMO

Inflammatory alterations of the extracellular matrix shape the tumor microenvironment and promote all stages of carcinogenesis. This study aims to determine the impact of cellular fibronectin on inflammatory facets of tumor-associated macrophages (TAMs) in breast cancer. Cellular fibronectin (FN) harboring the alternatively spliced extra domain A (FN-EDA) was determined to be a matrix component produced by the triple-negative breast cancer (TNBC) cells. High levels of FN-EDA correlated with poor survival in breast cancer patients. The proinflammatory cytokine IL-1ß enhanced the expression of cellular fibronectin including FN-EDA. TAMs were frequently observed in the tumor areas rich in FN-EDA. Conditioned media from TNBC cells induced the differentiation of CD206+CD163+ macrophages and stimulated the STAT3 pathway, ex vivo. In the macrophages, the STAT3 pathway enhanced FN-EDA-induced IL-1ß secretion and NF-κB signaling. In conclusion, our data indicate a self-reinforcing mechanism sustained by FN-EDA and IL-1ß through NF-κB and STAT3 signaling in TAMs which fosters an inflammatory environment in TNBC.


Assuntos
NF-kappa B , Neoplasias de Mama Triplo Negativas , Humanos , NF-kappa B/genética , NF-kappa B/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Fibronectinas/farmacologia , Retroalimentação , Neoplasias de Mama Triplo Negativas/genética , Transdução de Sinais , Microambiente Tumoral/genética
2.
J Cell Sci ; 134(4)2021 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-33526715

RESUMO

Cellular fibronectin (FN; also known as FN1) variants harboring one or two alternatively spliced so-called extra domains (EDB and EDA) play a central bioregulatory role during development, repair processes and fibrosis. Yet, how the extra domains impact fibrillar assembly and function of the molecule remains unclear. Leveraging a unique biological toolset and image analysis pipeline for direct comparison of the variants, we demonstrate that the presence of one or both extra domains impacts FN assembly, function and physical properties of the matrix. When presented to FN-null fibroblasts, extra domain-containing variants differentially regulate pH homeostasis, survival and TGF-ß signaling by tuning the magnitude of cellular responses, rather than triggering independent molecular switches. Numerical analyses of fiber topologies highlight significant differences in variant-specific structural features and provide a first step for the development of a generative model of FN networks to unravel assembly mechanisms and investigate the physical and functional versatility of extracellular matrix landscapes.This article has an associated First Person interview with the first author of the paper.


Assuntos
Processamento Alternativo , Fibronectinas , Células Cultivadas , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Humanos
3.
Curr Opin Oncol ; 33(3): 168-174, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33720067

RESUMO

PURPOSE OF REVIEW: Head and neck squamous cell carcinoma (HNSCC) tissue is composed of multiple cell types embedded in an extracellular matrix (ECM) that actively participates in disease progression, spread and treatment response. In this review, we provide an update of our current knowledge about the ECM landscape of HNSCC, its functions, methods of analysis, and nonimmunological stromal targeting strategies that modify the tumor ECM to improve conventional and emerging therapies. RECENT FINDINGS: The tumor ECM differs significantly from that of normal tissue in abundance, composition, organization and mechanical properties. In HNSCC, signaling between malignant epithelial cells and stromal cells prompts the upregulation of a set of ECM components that serve as substrates for carcinoma cell migration, modulate the cytokine environment and promote immune evasion in these tumors. Advanced imaging techniques and molecular profiling at the single-cell level have provided valuable insights into our understanding of the tumor ECM and its role in malignancy, and opened new avenues for predictive and potentially actionable biomarker discovery for more effective management of the disease. SUMMARY: ECM components upregulated in HNSCC can impact several cancer hallmarks by sustaining proliferative signaling, promoting angiogenesis, facilitating invasion and metastasis, modulating growth suppressor activity, and suppressing antitumoral immunity. The tumor ECM is also involved in treatment resistance, making it a potential therapeutic target.


Assuntos
Matriz Extracelular/patologia , Neoplasias de Cabeça e Pescoço/patologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Progressão da Doença , Células Epiteliais/patologia , Humanos , Células Estromais/patologia , Microambiente Tumoral
4.
J Biol Chem ; 291(20): 10684-99, 2016 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-27002148

RESUMO

Glioblastomas are the most common primary brain tumors, highly vascularized, infiltrating, and resistant to current therapies. This cancer leads to a fatal outcome in less than 18 months. The aggressive behavior of glioblastomas, including resistance to current treatments and tumor recurrence, has been attributed to glioma stemlike/progenitor cells. The transcription factor EGR1 (early growth response 1), a member of a zinc finger transcription factor family, has been described as tumor suppressor in gliomas when ectopically overexpressed. Although EGR1 expression in human glioblastomas has been associated with patient survival, its precise location in tumor territories as well as its contribution to glioblastoma progression remain elusive. In the present study, we show that EGR1-expressing cells are more frequent in high grade gliomas where the nuclear expression of EGR1 is restricted to proliferating/progenitor cells. We show in primary cultures of glioma stemlike cells that EGR1 contributes to stemness marker expression and proliferation by orchestrating a PDGFA-dependent growth-stimulatory loop. In addition, we demonstrate that EGR1 acts as a positive regulator of several important genes, including SHH, GLI1, GLI2, and PDGFA, previously linked to the maintenance and proliferation of glioma stemlike cells.


Assuntos
Comunicação Autócrina , Neoplasias Encefálicas/metabolismo , Proliferação de Células , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fator de Crescimento Derivado de Plaquetas/biossíntese , Neoplasias Encefálicas/patologia , Feminino , Glioblastoma/patologia , Humanos , Masculino , Células-Tronco Neoplásicas/patologia , Células Tumorais Cultivadas
5.
Stem Cells ; 31(7): 1252-65, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23533157

RESUMO

Stem cell-like properties of glioma initiating cells (GiCs) fuel glioblastoma (GBM) development by providing the different cell types that comprise the tumor. It is therefore likely that the molecular circuitries that regulate their decision to self-renew or commit to a more differentiated state may offer targets for future innovative therapies. In previous micro-RNA profiling studies to search for regulators of stem cell plasticity, we identified miR-18a* as a potential candidate and its expression correlated with the stemness state. Here, using human GiCs we found that miR-18a* expression promotes clonal proliferation in vitro and tumorigenicity in vivo. Mechanistically, ERK-dependent induction of miR-18a* directly represses expression of DLL3, an autocrine inhibitor of NOTCH, thus enhancing the level of activated NOTCH-1. Activated NOTCH-1 in turn is required for sustained ERK activation. This feed-forward loop, driven by miR-18a*, is required to turn on the SHH-GLI-NANOG network, essential for GiC self-renewal. Hence, by tightly regulating expression of DLL3, miR-18a* constitutes an important signaling mediator for fine tuning the level of GiC self-renewal.


Assuntos
Glioma/genética , MicroRNAs/genética , Receptor Notch1/metabolismo , Idoso , Animais , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Regulação para Baixo , Glioma/metabolismo , Glioma/patologia , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos NOD , MicroRNAs/biossíntese , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Receptor Notch1/genética , Transfecção
6.
Elife ; 122023 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-37585334

RESUMO

Endothelial cell interactions with their extracellular matrix are essential for vascular homeostasis and expansion. Large-scale proteomic analyses aimed at identifying components of integrin adhesion complexes have revealed the presence of several RNA binding proteins (RBPs) of which the functions at these sites remain poorly understood. Here, we explored the role of the RBP SAM68 (Src associated in mitosis, of 68 kDa) in endothelial cells. We found that SAM68 is transiently localized at the edge of spreading cells where it participates in membrane protrusive activity and the conversion of nascent adhesions to mechanically loaded focal adhesions by modulation of integrin signaling and local delivery of ß-actin mRNA. Furthermore, SAM68 depletion impacts cell-matrix interactions and motility through induction of key matrix genes involved in vascular matrix assembly. In a 3D environment SAM68-dependent functions in both tip and stalk cells contribute to the process of sprouting angiogenesis. Altogether, our results identify the RBP SAM68 as a novel actor in the dynamic regulation of blood vessel networks.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , RNA , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células Endoteliais/metabolismo , Proteômica , Proteínas de Ciclo Celular/metabolismo , Integrinas/metabolismo , Membrana Basal/metabolismo
7.
Biol Imaging ; 3: e25, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38510171

RESUMO

Due to the complex architectural diversity of biological networks, there is an increasing need to complement statistical analyses with a qualitative and local description of their spatial properties. One such network is the extracellular matrix (ECM), a biological scaffold for which changes in its spatial organization significantly impact tissue functions in health and disease. Quantifying variations in the fibrillar architecture of major ECM proteins should considerably advance our understanding of the link between tissue structure and function. Inspired by the analysis of functional magnetic resonance imaging (fMRI) images, we propose a novel statistical analysis approach embedded into a machine learning paradigm, to measure and detect local variations of meaningful ECM parameters. We show that parametric maps representing fiber length and pore directionality can be analyzed within the proposed framework to differentiate among various tissue states. The parametric maps are derived from graph-based representations that reflect the network architecture of fibronectin (FN) fibers in a normal, or disease-mimicking in vitro setting. Such tools can potentially lead to a better characterization of dynamic matrix networks within fibrotic tumor microenvironments and contribute to the development of better imaging modalities for monitoring their remodeling and normalization following therapeutic intervention.

8.
J Cell Sci ; 123(Pt 22): 3989-99, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20980391

RESUMO

Cellular fibronectin (cFN) variants harboring extra FN type 3 repeats, namely extra domains B and A, are major constituents of the extracellular matrix around newly forming blood vessels during development and angiogenesis. Their expression is induced by angiogenic stimuli and their assembly into fibrillar arrays is driven by cell-generated tension at α5ß1 integrin-based adhesions. Here, we examined the role and functional redundancy of cFN variants in cultured endothelial cells by isoform-selective RNA interference. We show that FN fibrillogenesis is a cell-autonomous process whereby basally directed secretion and assembly of cellular FN are tightly coupled events that play an important role not only in signaling at cell-matrix adhesions but also at cell-cell contacts. Silencing of cFN variants differentially affects integrin usage, cell spreading, motility and capillary morphogenesis in vitro. cFN-deficient cells undergo a switch from α5ß1- to αvß3-based adhesion, accompanied by a Src-regulated disruption of adherens junctions. These studies identify a crucial role for autocrine FN in subendothelial matrix assembly and junctional integrity that provides spatially and temporally restricted control of endothelial plasticity during angiogenic blood vessel remodeling.


Assuntos
Comunicação Celular/fisiologia , Células Endoteliais/fisiologia , Fibronectinas/fisiologia , Adesão Celular/fisiologia , Ciclo Celular/fisiologia , Junções Célula-Matriz/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiologia , Fibronectinas/metabolismo , Humanos , Integrinas/metabolismo , Transdução de Sinais , Transfecção
9.
Matrix Biol ; 111: 26-52, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35537652

RESUMO

The extracellular matrix (ECM) is a fundamental component of the tissue of multicellular organisms that is comprised of an intricate network of multidomain proteins and associated factors, collectively known as the matrisome. The ECM creates a biophysical environment that regulates essential cellular processes such as adhesion, proliferation and migration and impacts cell fate decisions. The composition of the ECM varies across organs, developmental stages and diseases. Interestingly, most ECM genes generate transcripts that undergo extensive alternative splicing events, producing multiple protein variants from one gene thus enhancing ECM complexity and impacting matrix architecture. Extensive studies over the past several decades have linked ECM remodeling and expression of alternatively spliced ECM isoforms to cancer, and reprogramming of the alternative splicing patterns in cells has recently been proposed as a new hallmark of tumor progression. Indeed, tumor-associated alternative splicing occurs in both malignant and non-malignant cells of the tumor environment and growing evidence suggests that expression of specific ECM splicing variants could be a key step for stromal activation. In this review, we present a general overview of alternative splicing mechanisms, featuring examples of ECM components. The importance of ECM variant expression during essential physiological processes, such as tissue organization and embryonic development is discussed as well as the dysregulation of alternative splicing in cancer. The overall aim of this review is to address the complexity of the ECM by highlighting the importance of the yet-to-be-fully-characterized "alternative" matrisome in physiological and pathological states such as cancer.


Assuntos
Proteínas da Matriz Extracelular , Neoplasias , Processamento Alternativo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Homeostase/genética , Humanos , Neoplasias/genética , Neoplasias/metabolismo
10.
Front Immunol ; 12: 666233, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33936105

RESUMO

The integrative analysis of tumor immune microenvironment (TiME) components, their interactions and their microanatomical distribution is mandatory to better understand tumor progression. Imaging Mass Cytometry (IMC) is a high dimensional tissue imaging system which allows the comprehensive and multiparametric in situ exploration of tumor microenvironments at a single cell level. We describe here the design of a 39-antibody IMC panel for the staining of formalin-fixed paraffin-embedded human tumor sections. We also provide an optimized staining procedure and details of the experimental workflow. This panel deciphers the nature of immune cells, their functions and their interactions with tumor cells and cancer-associated fibroblasts as well as with other TiME structural components known to be associated with tumor progression like nerve fibers and tumor extracellular matrix proteins. This panel represents a valuable innovative and powerful tool for fundamental and clinical studies that could be used for the identification of prognostic biomarkers and mechanisms of resistance to current immunotherapies.


Assuntos
Citometria por Imagem/métodos , Microambiente Tumoral/imunologia , Biomarcadores Tumorais/imunologia , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Progressão da Doença , Humanos , Imuno-Histoquímica , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Fluxo de Trabalho
11.
Exp Cell Res ; 315(12): 2092-104, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19332054

RESUMO

Fibronectin (FN) fibrillogenesis is an essential biological process mediated by alpha5beta1 integrin and cellular contractile forces. Assembly of a FN matrix by activated endothelial cells occurs during angiogenic blood vessel remodeling and signaling components that control this event represent attractive therapeutic targets. Here we examined the role of individual Rho GTPases in FN matrix remodeling by selectively attenuating their expression in cultured endothelial cells. Whereas pharmacological ablation of myosin-regulated contractility abrogated matrix assembly, no significant decrease was detected in the amount of FN deposited by RhoA, RhoB-, RhoC-, Rac1-, or Cdc42-depleted cells. Rather, distinct differences in fiber arrangement were observed. Most strikingly, RhoA silenced cells assembled a fine FN meshwork beneath alpha5beta1 integrin-based fibrillar adhesions, in the absence of classical focal adhesions and actin stress fibers, indicating that alpha5beta1 integrin translocation and FN fibril elongation can occur in low tension states such as those encountered by newly-forming vessels in tissue. In contrast, highly contractile Cdc42-deficient cells deposited FN globules and Rac-deficient cells assembled long arrays, reflecting their increased motility. We propose that regulation of FN scaffolds by Rho GTPase signaling impacts bidirectional communications and mechanical interactions between endothelial cells and their extracellular matrix during vascular morphogenesis.


Assuntos
Capilares/fisiologia , Células Endoteliais/fisiologia , Endotélio Vascular/fisiologia , Fibronectinas/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Bovinos , Células Cultivadas , Matriz Extracelular/fisiologia , Morfogênese , Transdução de Sinais , Proteína cdc42 de Ligação ao GTP/fisiologia , Proteínas rac1 de Ligação ao GTP/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia
12.
Front Oncol ; 10: 641, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32426283

RESUMO

Normal tissue homeostasis and architecture restrain tumor growth. Thus, for a tumor to develop and spread, malignant cells must overcome growth-repressive inputs from surrounding tissue and escape immune surveillance mechanisms that curb cancer progression. This is achieved by promoting the conversion of a physiological microenvironment to a pro-tumoral state and it requires a constant dialog between malignant cells and ostensibly normal cells of adjacent tissue. Pro-tumoral reprogramming of the stroma is accompanied by an upregulation of certain extracellular matrix (ECM) proteins and their cognate receptors. Fibronectin (FN) is one such component of the tumor matrisome. This large multidomain glycoprotein dimer expressed over a wide range of human cancers is assembled by cell-driven forces into a fibrillar array that provides an obligate scaffold for the deposition of other matrix proteins and binding sites for functionalization by soluble factors in the tumor microenvironment. Encoded by a single gene, FN regulates the proliferation, motile behavior and fate of multiple cell types, largely through mechanisms that involve integrin-mediated signaling. These processes are coordinated by distinct isoforms of FN, collectively known as cellular FN (as opposed to circulating plasma FN) that arise through alternative splicing of the FN1 gene. Cellular FN isoforms differ in their solubility, receptor binding ability and spatiotemporal expression, and functions that have yet to be fully defined. FN induction at tumor sites constitutes an important step in the acquisition of biological capabilities required for several cancer hallmarks such as sustaining proliferative signaling, promoting angiogenesis, facilitating invasion and metastasis, modulating growth suppressor activity and regulating anti-tumoral immunity. In this review, we will first provide an overview of ECM reprogramming through tumor-stroma crosstalk, then focus on the role of cellular FN in tumor progression with respect to these hallmarks. Last, we will discuss the impact of dysregulated ECM on clinical efficacy of classical (radio-/chemo-) therapies and emerging treatments that target immune checkpoints and explore how our expanding knowledge of the tumor ECM and the central role of FN can be leveraged for therapeutic benefit.

13.
Cancer Immunol Res ; 8(9): 1122-1138, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32665262

RESUMO

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.


Assuntos
Neoplasias Bucais/imunologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/imunologia , Tenascina/imunologia , Animais , Quimiocina CCL21/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Bucais/patologia , Receptores CCR7/imunologia , Proteínas Recombinantes/farmacologia , Linfócitos T Reguladores/imunologia , Tenascina/farmacologia , Microambiente Tumoral/imunologia
14.
Mol Cell Biol ; 26(9): 3582-94, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16611998

RESUMO

Cytoskeletal rearrangements are central to endothelial cell physiology and are controlled by soluble factors, matrix proteins, cell-cell interactions, and mechanical forces. We previously reported that aortic endothelial cells can rearrange their cytoskeletons into complex actin-based structures called podosomes when a constitutively active mutant of Cdc42 is expressed. We now report that transforming growth factor beta (TGF-beta) promotes podosome formation in primary aortic endothelial cells. TGF-beta-induced podosomes assembled together into large ring- or crescent-shaped structures. Their formation was dependent on protein synthesis and required functional Src, phosphatidylinositide 3-kinase, Cdc42, RhoA, and Smad signaling. MT1-MMP and metalloprotease 9 (MMP9), both upregulated by TGF-beta, were detected at sites of podosome formation, and MT1-MMP was found to be involved in the local degradation of extracellular matrix proteins beneath the podosomes and required for the invasion of collagen gels by endothelial cells. We propose that TGF-beta plays an important role in endothelial cell physiology by inducing the formation of podosomal structures endowed with metalloprotease activity that may contribute to arterial remodeling.


Assuntos
Citoesqueleto/fisiologia , Células Endoteliais/citologia , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinases da Matriz/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Aorta/citologia , Aorta/efeitos dos fármacos , Bovinos , Citoesqueleto/enzimologia , Citoesqueleto/ultraestrutura , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Humanos , Metaloproteinase 9 da Matriz/análise , Metaloproteinases da Matriz/análise , Metaloproteinases da Matriz Associadas à Membrana , Fosfatidilinositol 3-Quinases/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Transdução de Sinais , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Quinases da Família src/metabolismo
15.
Sci Rep ; 9(1): 1926, 2019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30760745

RESUMO

High resolution imaging of molecules at the cell-substrate interface is required for understanding key biological processes. Here we propose a complete pipeline for multi-angle total internal reflection fluorescence microscopy (MA-TIRF) going from instrument design and calibration procedures to numerical reconstruction. Our custom setup is endowed with a homogeneous field illumination and precise excitation beam angle. Given a set of MA-TIRF acquisitions, we deploy an efficient joint deconvolution/reconstruction algorithm based on a variational formulation of the inverse problem. This algorithm offers the possibility of using various regularizations and can run on graphics processing unit (GPU) for rapid reconstruction. Moreover, it can be easily used with other MA-TIRF devices and we provide it as an open-source software. This ensemble has enabled us to visualize and measure with unprecedented nanometric resolution, the depth of molecular components of the fibronectin assembly machinery at the basal surface of endothelial cells.

16.
Cancers (Basel) ; 11(2)2019 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-30795607

RESUMO

It is generally accepted that carcinogenesis and aging are two biological processes, which are known to be associated. Notably, the frequency of certain cancers (including lung cancer), increases significantly with the age of patients and there is now a wealth of data showing that multiple mechanisms leading to malignant transformation and to aging are interconnected, defining the so-called common biology of aging and cancer. OncoAge, a consortium launched in 2015, brings together the multidisciplinary expertise of leading public hospital services and academic laboratories to foster the transfer of scientific knowledge rapidly acquired in the fields of cancer biology and aging into innovative medical practice and silver economy development. This is achieved through the development of shared technical platforms (for research on genome stability, (epi)genetics, biobanking, immunology, metabolism, and artificial intelligence), clinical research projects, clinical trials, and education. OncoAge focuses mainly on two pilot pathologies, which benefit from the expertise of several members, namely lung and head and neck cancers. This review outlines the broad strategic directions and key advances of OncoAge and summarizes some of the issues faced by this consortium, as well as the short- and long-term perspectives.

17.
Acta Oncol ; 47(8): 1502-12, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18607838

RESUMO

BACKGROUND: The gene encoding cortactin, CTTN (locus 11q13), an actin-binding substrate of Src kinases, is frequently amplified in breast and head and neck squamous cell carcinomas (HNSCC) and cortactin overexpression is thought to contribute in a significant way to the invasive phenotype of these tumors. Elevated Epidermal Growth Factor receptor (EGFR) expression is also commonly observed in HNSCC and has been associated with poor prognosis and resistance to cytotoxic agents, including ionizing radiation. It has been suggested that cortactin overexpression may increase EGFR levels in these tumors by affecting receptor downregulation, however we recently found by multivariate analysis, that cortactin expression status remained an independent prognostic factor for local recurrence, disease-free survival, and overall survival. MATERIAL AND METHODS: To examine the potential link between cortactin overexpression and EGFR status, we compared cortactin and EGFR levels in a series of tumor lines derived from HNSCC. RNAi-mediated silencing was performed in cortactin overexpressing cells and in vivo tumoral potential with respect to cortactin and EGFR status was analyzed. RESULTS AND DISCUSSION: Cortactin and EGFR levels were not strictly coupled in these lines and cortactin depletion did not decrease steady state receptor levels, although it did affect the epithelial to mesenchymal phenotypic conversion of cells. These results, together with clinical findings point to the existence of an EGFR-independent role of cortactin in HNSCC that may have important implications regarding the design of targeted therapies to combat tumor spread.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Cortactina/metabolismo , Receptores ErbB/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Animais , Southern Blotting , Western Blotting , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Células Cultivadas , Cortactina/genética , Receptores ErbB/genética , Feminino , Imunofluorescência , Amplificação de Genes , Neoplasias de Cabeça e Pescoço/genética , Humanos , Técnicas Imunoenzimáticas , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos Nus , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia
18.
FASEB J ; 20(9): 1489-91, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16723384

RESUMO

Extracellular matrix (ECM) receptors of the integrin family initiate changes in cell shape and motility by recruiting signaling components that coordinate these events. Integrin-linked kinase (ILK) is one such partner of beta1 integrins that participates in dynamic rearrangement of cell-matrix adhesions and cell spreading by mechanisms that are not well understood. To further elucidate the role of ILK in these events, we engineered a chimeric molecule comprising ILK fused to a membrane-targeted green fluorescent protein (ILK-GFP-F). ILK-GFP-F is highly enriched in cell-matrix adhesions, and its expression in fibroblasts leads to an accumulation of focal adhesions (2-5 microm) and elongated adhesions (>5 microm). ILK-GFP-F enhances cell spreading on fibronectin and induces a constitutive increase in the levels of GTP-bound Rac-1. Conversely, ILK knock-down by siRNA transfection decreases active Rac-1. Endogenous ILK was found to associate with PKL (paxillin kinase linker) and the Rac/Cdc42 guanine nucleotide exchange factor betaPIX. Further, expression of a dominant negative betaPIX mutant reversed the increase in active Rac-1 levels of ILK-GFP-F-expressing cells, thus placing betaPIX in the pathway leading from ILK to Rac-1 activation. However, expression of constitutively active Rac only partially restores the spreading defects of ILK-depleted cells, suggesting that an additional ILK-dependent signal is required for cell spreading.


Assuntos
Endotélio Vascular/fisiologia , Matriz Extracelular/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Aorta , Bovinos , Células Cultivadas , Endotélio Vascular/citologia , Vetores Genéticos , Cinética , Proteínas Serina-Treonina Quinases/genética , RNA Interferente Pequeno/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/fisiologia , Transfecção , Proteínas rac1 de Ligação ao GTP/genética
19.
Sci Rep ; 7(1): 12762, 2017 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-28986537

RESUMO

Cellular fibronectin (FN) and tenascin-C (TNC) are prominent development- and disease-associated matrix components with pro- and anti-adhesive activity, respectively. Whereas both are present in the tumour vasculature, their functional interplay on vascular endothelial cells remains unclear. We have previously shown that basally-oriented deposition of a FN matrix restricts motility and promotes junctional stability in cultured endothelial cells and that this effect is tightly coupled to expression of FN. Here we report that TNC induces FN expression in endothelial cells. This effect counteracts the potent anti-adhesive activity of TNC and leads to the assembly of a dense highly-branched subendothelial matrix that enhances tubulogenic activity. These findings suggest that pro-angiogenic remodelling of the perivascular matrix may involve TNC-induced upregulation of FN in endothelial cells.


Assuntos
Fibronectinas/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Tenascina/metabolismo , Capilares/metabolismo , Adesão Celular , Movimento Celular , Junções Célula-Matriz , Humanos , Modelos Biológicos , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/metabolismo , Neovascularização Fisiológica , Transdução de Sinais
20.
Nat Commun ; 8: 14105, 2017 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-28102238

RESUMO

Functional interplay between tumour cells and their neoplastic extracellular matrix plays a decisive role in malignant progression of carcinomas. Here we provide a comprehensive data set of the human HNSCC-associated fibroblast matrisome. Although much attention has been paid to the deposit of collagen, we identify oncofetal fibronectin (FN) as a major and obligate component of the matrix assembled by stromal fibroblasts from head and neck squamous cell carcinomas (HNSCC). FN overexpression in tumours from 435 patients corresponds to an independent unfavourable prognostic indicator. We show that migration of carcinoma collectives on fibrillar FN-rich matrices is achieved through αvß6 and α9ß1 engagement, rather than α5ß1. Moreover, αvß6-driven migration occurs independently of latent TGF-ß activation and Smad-dependent signalling in tumour epithelial cells. These results provide insights into the adhesion-dependent events at the tumour-stroma interface that govern the collective mode of migration adopted by carcinoma cells to invade surrounding stroma in HNSCC.


Assuntos
Carcinoma de Células Escamosas , Movimento Celular/efeitos dos fármacos , Fibronectinas/metabolismo , Neoplasias de Cabeça e Pescoço , Adulto , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Matriz Extracelular , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Integrinas/genética , Integrinas/metabolismo , Masculino , Pessoa de Meia-Idade , Carcinoma de Células Escamosas de Cabeça e Pescoço
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa