Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Am J Physiol Cell Physiol ; 318(3): C542-C554, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31913695

RESUMO

Chemokines are a family of soluble cytokines that act as chemoattractants to guide the migration of cells, in particular of immune cells. However, chemokines are also involved in cell proliferation, differentiation, and survival. Chemokines are associated with a variety of human diseases including chronic inflammation, immune dysfunction, cancer, and metastasis. This review discusses the expression of CC and CXC chemokines in the tumor microenvironment and their supportive and inhibitory roles in tumor progression, angiogenesis, metastasis, and tumor immunity. We also specially focus on the diverse roles of CXC chemokines (CXCL9-11, CXCL4 and its variant CXCL4L1) and their two chemokine receptor CXCR3 isoforms, CXCR3-A and CXCR3-B. These two distinct isoforms have divergent roles in tumors, either promoting (CXCR3-A) or inhibiting (CXCR3-B) tumor progression. Their effects are mediated not only directly in tumor cells but also indirectly via the regulation of angiogenesis and tumor immunity. A full comprehension of their mechanisms of action is critical to further validate these chemokines and their receptors as biomarkers or therapeutic targets in cancer.


Assuntos
Biomarcadores Tumorais/fisiologia , Quimiocina CXCL9/fisiologia , Fator Plaquetário 4/fisiologia , Receptores CXCR3/fisiologia , Microambiente Tumoral/fisiologia , Animais , Progressão da Doença , Humanos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia
2.
Methods ; 94: 65-74, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26342258

RESUMO

The study of cell behavior in constricted environment is particularly relevant to our understanding of the mechanisms of cell invasion. In this regard, microfluidic systems offer promising platforms as microfabricated fluidic chips provide well-controlled physical, chemical and confined environments to study cell phenotype and behavior. Here, we report a fast and effective manufacturing process of user-friendly microfluidic chips ideally suited for quantitative live cell analysis in combination with immunofluorescence microscopy. The chip body, made of polydimethylsiloxane, is composed of two incubation chambers connected by one rectangular intermediate entry channel which provides access to a series of transversal slits where the observation can be made. The height of the slit is designed to be slightly smaller than that of the cells under study. To validate the chip performance, we analyzed the reorganization of the cytoskeleton of endothelial cells under various degree of spatial confinement. We illustrate how the constricted environment affects endothelial cell behavior in inducing the formation of podosomes. Moreover, the process was stimulated further when the surface of the slit was coated with a thin layer of fibronectin. The study demonstrates the suitability of this technological process for cost-effective fabrication of custom-made single-use chips for biological applications.


Assuntos
Citoesqueleto de Actina/fisiologia , Células Endoteliais/fisiologia , Dispositivos Lab-On-A-Chip , Podossomos/fisiologia , Análise de Célula Única/instrumentação , Citoesqueleto de Actina/ultraestrutura , Animais , Bovinos , Células Cultivadas , Meios de Cultura , Células Endoteliais/ultraestrutura , Microscopia de Fluorescência
3.
Biochim Biophys Acta ; 1836(2): 287-95, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23994549

RESUMO

CXC chemokines are involved in chemotaxis, regulation of cell growth, induction of apoptosis and modulation of angiostatic effects. CXCL9, CXCL10, CXCL11, CXCL4 and its variant CXCL4L1 are members of the CXC chemokine family, which bind to the CXCR3 receptor to exert their biological effects. These chemokines are associated with a variety of human diseases including chronic inflammation, immune dysfunction, cancer and metastasis. In this review, we focus on accumulating evidence demonstrating the pivotal role of CXCR3 in tumor progression. Its effects are mediated directly in tumor cells or indirectly through the regulation of angiogenesis and tumor immunity. Understanding the emerging role of CXCR3 and its signaling mechanisms further validates this receptor as a biomarker and therapeutic target for tumor progression and tumor angiogenesis.


Assuntos
Transformação Celular Neoplásica/patologia , Quimiocinas CXC/metabolismo , Neoplasias/patologia , Neovascularização Patológica , Receptores CXCR3/metabolismo , Animais , Progressão da Doença , Humanos , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo
4.
Sci Transl Med ; 14(661): eaax8933, 2022 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-36070364

RESUMO

Brain metastasis is a complication of increasing incidence in patients with breast cancer at advanced disease stage. It is a severe condition characterized by a rapid decline in quality of life and poor prognosis. There is a critical clinical need to develop effective therapies to prevent and treat brain metastases. Here, we describe a unique and robust spontaneous preclinical model of breast cancer metastasis to the brain (4T1-BM2) in mice that has been instrumental in uncovering molecular mechanisms guiding metastatic dissemination and colonization of the brain. Key experimental findings were validated in the additional murine D2A1-BM2 model and in human MDA231-BrM2 model. Gene expression analyses and functional studies, coupled with clinical transcriptomic and histopathological investigations, identified connexins (Cxs) and focal adhesion kinase (FAK) as master molecules orchestrating breast cancer colonization of the brain. Cx31 promoted homotypic tumor cell adhesion, heterotypic tumor-astrocyte interaction, and FAK phosphorylation. FAK signaling prompted NF-κB activation inducing Lamc2 expression and laminin 332 (laminin 5) deposition, α6 integrin-mediated adhesion, and sustained survival and growth within brain parenchyma. In the MDA231-BrM2 model, the human homologous molecules CX43, LAMA4, and α3 integrin were involved. Systemic treatment with FAK inhibitors reduced brain metastasis progression. In conclusion, we report a spontaneous model of breast cancer metastasis to the brain and identified Cx-mediated FAK-NF-κB signaling as a mechanism promoting cell-autonomous and microenvironmentally controlled cell survival for brain colonization. Considering the limited therapeutic options for brain metastatic disease in cancer patients, we propose FAK as a therapeutic candidate to further pursue in the clinic.


Assuntos
Neoplasias Encefálicas , Neoplasias da Mama , Animais , Encéfalo/metabolismo , Neoplasias da Mama/genética , Conexinas/metabolismo , Feminino , Proteína-Tirosina Quinases de Adesão Focal/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Melanoma , Camundongos , NF-kappa B/metabolismo , Qualidade de Vida , Neoplasias Cutâneas , Melanoma Maligno Cutâneo
5.
J Cell Sci ; 122(Pt 23): 4311-8, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19887587

RESUMO

Podosomes are specialized plasma-membrane actin-based microdomains that combine adhesive and proteolytic activities to spatially restrict sites of matrix degradation in in vitro assays, but the physiological relevance of these observations remain unknown. Inducible rings of podosomes (podosome rosettes) form in cultured aortic cells exposed to the inflammatory cytokine TGFbeta. In an attempt to prove the existence of podosomes in living tissues, we developed an ex vivo endothelium observation model. This system enabled us to visualize podosome rosettes in the endothelium of native arterial vessel exposed to biologically active TGFbeta. Podosomes induced in the vessel appear similar to those formed in cultured cells in terms of molecular composition, but in contrast to the latter, arrange in a protruding structure that is similar to invadopodia. Local degradation of the basement membrane scaffold protein collagen-IV, is observed underneath the structures. Our results reveal for the first time the presence of podosome rosettes in the native endothelium and provide evidence for their capacity to degrade the basement membrane, opening up new avenues to study their role in vascular pathophysiology. We propose that podosome rosettes are involved in arterial vessel remodeling.


Assuntos
Membrana Basal/metabolismo , Colágeno/metabolismo , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Membrana Basal/efeitos dos fármacos , Células Cultivadas , Endotélio/efeitos dos fármacos , Endotélio/metabolismo , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência
6.
Biomedicines ; 9(10)2021 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-34680548

RESUMO

BACKGROUND: LRP-1 is a multifunctional scavenger receptor belonging to the LDLR family. Due to its capacity to control pericellular levels of various growth factors and proteases, LRP-1 plays a crucial role in membrane proteome dynamics, which appears decisive for tumor progression. METHODS: LRP-1 involvement in a TNBC model was assessed using an RNA interference strategy in MDA-MB-231 cells. In vivo, tumorigenic and angiogenic effects of LRP-1-repressed cells were evaluated using an orthotopic xenograft model and two angiogenic assays (Matrigel® plugs, CAM). DCE-MRI, FMT, and IHC were used to complete a tumor longitudinal follow-up and obtain morphological and functional vascular information. In vitro, HUVECs' angiogenic potential was evaluated using a tumor secretome, subjected to a proteomic analysis to highlight LRP-1-dependant signaling pathways. RESULTS: LRP-1 repression in MDA-MB-231 tumors led to a 60% growth delay because of, inter alia, morphological and functional vascular differences, confirmed by angiogenic models. In vitro, the LRP-1-repressed cells secretome restrained HUVECs' angiogenic capabilities. A proteomics analysis revealed that LRP-1 supports tumor growth and angiogenesis by regulating TGF-ß signaling and plasminogen/plasmin system. CONCLUSIONS: LRP-1, by its wide spectrum of interactions, emerges as an important matricellular player in the control of cancer-signaling events such as angiogenesis, by supporting tumor vascular morphology and functionality.

7.
Eur J Cell Biol ; 87(8-9): 543-54, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18397815

RESUMO

Podosomes are punctate actin-rich adhesion structures which spontaneously form in cells of the myelomonocytic lineage. Their formation is dependent on Src and RhoGTPases. Recently, podosomes have also been described in vascular cells. These podosomes differ from the former by the fact that they are inducible. In endothelial cells, such a signal can be provided by either constitutively active Cdc42, the PKC activator PMA or TGFbeta, depending on the model. Consequently, other regulatory pathways have been reported to contribute to podosome formation. To get more insight into the mechanisms by which podosomes form in endothelial cells, we have explored the respective contribution of signal transducers such as Cdc42-related GTPases, Smads and PKCs in three endothelial cell models. Results presented demonstrate that, in addition to Cdc42, TC10 and TCL GTPases can also promote podosome formation in endothelial cells. We also show that PKCalpha can be either necessary or entirely dispensable, depending on the cell model. In contrast, PKCdelta is essential for podosome formation in endothelial cells but not smooth muscle cells. Finally, although podosomes vary very little in their molecular composition, the signalling pathways involved in their assembly appear very diverse.


Assuntos
Citoesqueleto de Actina/enzimologia , Células Endoteliais/enzimologia , Transdução de Sinais , Citoesqueleto de Actina/ultraestrutura , Actinas/metabolismo , Animais , Bovinos , Adesão Celular/fisiologia , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Células Endoteliais/metabolismo , Humanos , Microscopia de Fluorescência , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C-delta/metabolismo , Proteínas Smad/metabolismo , Suínos , Proteína cdc42 de Ligação ao GTP/metabolismo
8.
J Cell Biochem ; 104(3): 826-39, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18189245

RESUMO

During epithelial-mesenchymal transition (EMT), epithelial cells are converted into isolated motile and invasive mesenchymal cells. In model systems, EMT is induced most often by the activation of tyrosine kinase receptors through signaling pathways involving translational and post-translational regulation. In this study, we have used the NBT-II bladder carcinoma cell system to investigate in vitro Fibroblast Growth Factor-1 (FGF-1)-induced EMT. Transcriptome analyses were performed on NBT-II cells stimulated for 2, 6, 24, and 48 h with FGF-1. As some phenotypic changes occurred around 6 h post-stimulation, a supervised analysis was designed to identify transcript variations across defined time-periods. Our results clearly indicate that immediately after FGF-1 stimulation a set of genes assigned to transcriptional regulation (e.g., jun-B and v-ets) and to EMT induction (e.g., Notch 1) is transiently up-regulated. A set of genes involved in proteolytic systems (e.g., MMP-13 and uPAR) is immediately up-regulated but subsequently maintained throughout FGF-1 stimulation. Then follows a second wave of gene expression that includes a strong but transient up-regulation of ephrin B1 and arginase I. Finally, a third group of genes is stably modulated over 48 h which consists primarily of down-regulated genes specifically associated with the EMT-based loss of the epithelial phenotype and maintenance of the mesenchymal and invasive phenotype of carcinoma cells. Using genome-wide oligoarray technology, we have identified novel expressions of immediate, immediate-early and later EMT biomarkers that are specifically activated downstream of the FGF/FGFR pathway and which might be significant prognostic factors for tumor progression of carcinoma.


Assuntos
Carcinoma/metabolismo , Transformação Celular Neoplásica , Epitélio/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Mesoderma/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Animais , Biomarcadores Tumorais , Linhagem Celular Tumoral , Modelos Estatísticos , Fenótipo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
9.
Mol Biol Cell ; 15(10): 4725-34, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15282342

RESUMO

Fibroblast growth factor (FGF)-1 and -2 have potent biological activities implicated in malignant tumor development. Their autocrine and nonautocrine activity in tumor progression of carcinoma was investigated in the NBT-II cell system. Cells were manipulated to either produce and be autocrine for FGF-1 or -2 or to only produce but not respond to these factors. The autocrine cells are highly invasive and tumorigenic and the determination of specific targets of FGF/fibroblast growth factor receptor (FGFR) signaling was assessed. In vitro studies showed that nonautocrine cells behave like epithelial parental cells, whereas autocrine cells have a mesenchymal phenotype correlated with the overexpression of urokinase plasminogen activator receptor (uPAR), the internalization of E-cadherin, and the redistribution of beta-catenin from the cell surface to the cytoplasm and nucleus. uPAR was defined as an early target, whereas E-cadherin and the leukocyte common antigen-related protein-tyrosine phosphatase (LAR-PTP) were later targets of FGF signaling, with FGFR1 activation more efficient than FGFR2 at modulating these targets. Behavior of autocrine cells was consistent with a decrease of tumor-suppressive activities of both E-cadherin and LAR-PTP. These molecular analyses show that the potential of these two growth factors in tumor progression is highly dependent on specific FGFR signaling and highlights its importance as a target for antitumor therapy.


Assuntos
Carcinoma/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/fisiologia , Animais , Comunicação Autócrina , Caderinas/metabolismo , Linhagem Celular Tumoral , Forma Celular , Proteínas do Citoesqueleto/metabolismo , Desmoplaquinas , Invasividade Neoplásica , Ratos , Receptores de Superfície Celular/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Transativadores/metabolismo , beta Catenina
10.
ACS Chem Biol ; 12(11): 2767-2778, 2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-28945356

RESUMO

CXCL4 chemokines have antiangiogenic properties, mediated by different mechanisms, including CXCR3 receptor activation. Chemokines have distinct oligomerization states that are correlated with their biological functions. CXCL4 exists as a stable tetramer under physiological conditions. It is unclear whether the oligomerization state impacts CXCL4-receptor interaction. We found that the CXCL4 tetramer is sensitive to pH and salt concentration. Residues Glu28 and Lys50 were important for tetramer formation, and the first ß-strand and the C-terminal helix are critical for dimerization. By mutating the critical residues responsible for oligomerization, we generated CXCL4 mutants that behave as dimers or monomers under neutral/physiological conditions. The CXCL4 monomer acts as the minimal active unit for interacting CXCR3A, and sulfation of N-terminal tyrosine residues on the receptor is important for binding. Noticeably, CXCL4L1, a CXCL4 variant that differs by three residues in the C-terminal helix, could activate CXCR3A. CXCL4L1 showed a higher tendency to dissociate into monomers, but native CXCL4 did not. This result indicates that monomeric CXCL4 behaves like CXCL4L1. Thus, in this chemokine family, being in the monomeric state seems critical for interaction with CXCR3A.


Assuntos
Fator Plaquetário 4/metabolismo , Receptores CXCR3/metabolismo , Linhagem Celular , Humanos , Concentração de Íons de Hidrogênio , Cinética , Modelos Moleculares , Fator Plaquetário 4/química , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Receptores CXCR3/química
11.
Nat Commun ; 8(1): 1571, 2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-29146996

RESUMO

CXCR3 plays important roles in angiogenesis, inflammation, and cancer. However, the precise mechanism of regulation and activity in tumors is not well known. We focused on CXCR3-A conformation and on the mechanisms controlling its activity and trafficking and investigated the role of CXCR3/LRP1 cross talk in tumor cell invasion. Here we report that agonist stimulation induces an anisotropic response with conformational changes of CXCR3-A along its longitudinal axis. CXCR3-A is internalized via clathrin-coated vesicles and recycled by retrograde trafficking. We demonstrate that CXCR3-A interacts with LRP1. Silencing of LRP1 leads to an increase in the magnitude of ligand-induced conformational change with CXCR3-A focalized at the cell membrane, leading to a sustained receptor activity and an increase in tumor cell migration. This was validated in patient-derived glioma cells and patient samples. Our study defines LRP1 as a regulator of CXCR3, which may have important consequences for tumor biology.


Assuntos
Neoplasias Encefálicas/patologia , Movimento Celular/fisiologia , Glioblastoma/patologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Receptores CXCR3/metabolismo , Animais , Membrana Celular/metabolismo , Embrião de Galinha , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Masculino , Camundongos , Camundongos Knockout , Invasividade Neoplásica/patologia , Ligação Proteica , Transporte Proteico/fisiologia , Esferoides Celulares , Células Tumorais Cultivadas
12.
Cancer Res ; 76(22): 6507-6519, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27634764

RESUMO

The CXCL4 paralog CXCL4L1 is a less studied chemokine that has been suggested to exert an antiangiogenic function. However, CXCL4L1 is also expressed in patient tumors, tumor cell lines, and murine xenografts, prompting a more detailed analysis of its role in cancer pathogenesis. We used genetic and antibody-based approaches to attenuate CXCL4L1 in models of pancreatic ductal adenocarcinoma (PDAC). Mechanisms of expression were assessed in cell coculture experiments, murine, and avian xenotransplants, including through an evaluation of CpG methylation and mutation of critical CpG residues. CXCL4L1 gene expression was increased greatly in primary and metastatic PDAC. We found that myofibroblasts triggered cues in the tumor microenvironment, which led to induction of CXCL4L1 in tumor cells. CXCL4L1 expression was also controlled by epigenetic modifications at critical CpG islands, which were mapped. CXCL4L1 inhibited angiogenesis but also affected tumor development more directly, depending on the tumor cell type. In vivo administration of an mAb against CXCL4L1 demonstrated a blockade in the growth of tumors positive for CXCR3, a critical receptor for CXCL4 ligands. Our findings define a protumorigenic role in PDAC development for endogenous CXCL4L1, which is independent of its antiangiogenic function. Cancer Res; 76(22); 6507-19. ©2016 AACR.


Assuntos
Inibidores da Angiogênese/genética , Neoplasias Pancreáticas/genética , Receptores CXCR3/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células , Quimiocinas , Humanos , Camundongos , Neovascularização Patológica , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Fator Plaquetário 4 , Análise de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Oncogene ; 21(53): 8128-39, 2002 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-12444548

RESUMO

FGF-1 and FGF-2 are pleiotropic growth factors for many cell types, operating through the activation of specific transmembrane FGF receptors (FGFRs). The role of these factors in tumor progression was investigated, with specific discrimination between their autocrine and non autocrine cellular activity. The rat bladder carcinoma NBT-II cells were engineered to produce FGF-1 or 18 kDa FGF-2 in the presence or absence of their specific receptor. Non-autocrine cells that produced FGF-1 or FGF-2 but lacked FGFRs were epithelial and reminiscent of the parental NBT-II cells. Whilst autocrine cells, which both constitutively produced and secreted the growth factor and expressed FGFRs, had a highly invasive mesenchymal phenotype. Correspondingly, the autocrine cells were highly tumorigenic in vivo compared to the parental and non-autocrine cells, which correlated with the increased production of uPAR and active uPA and increased in vitro invasive potential. Although all cells produced VEGF, only tumors derived from cells that produced FGF-1 or FGF-2 were highly vascularized, suggesting that these two growth factors could be involved in the angiogenic process by activating host endothelial cells. As a result of activation of the FGFR in autocrine cells, changes in cell morphology and an increase in the invasive and tumorigenic properties were observed, however no in vitro or in vivo differential functions between FGF-1 and FGF-2 could be identified in this system. In conclusion, our data demonstrates that rapid tumor development is not dependent upon increased tumor vascularization, suggesting that 'basal' angiogenesis, probably mediated by VEGF, is sufficient to support tumor growth.


Assuntos
Carcinoma/patologia , Fator 1 de Crescimento de Fibroblastos/fisiologia , Fator 2 de Crescimento de Fibroblastos/fisiologia , Proteínas de Neoplasias/fisiologia , Neovascularização Patológica/fisiopatologia , Neoplasias da Bexiga Urinária/patologia , Animais , Comunicação Autócrina , Carcinoma/irrigação sanguínea , Carcinoma/genética , Carcinoma/metabolismo , Progressão da Doença , Fatores de Crescimento Endotelial/metabolismo , Células Epiteliais/patologia , Feminino , Fator 1 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/genética , Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Linfocinas/metabolismo , Metaloproteinases da Matriz/biossíntese , Metaloproteinases da Matriz/genética , Mesoderma , Camundongos , Camundongos Nus , Invasividade Neoplásica , Transplante de Neoplasias , Neovascularização Patológica/genética , Fenótipo , Ratos , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/fisiologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Proteínas Recombinantes de Fusão/fisiologia , Deleção de Sequência , Inibidor Tecidual de Metaloproteinase-2/biossíntese , Inibidor Tecidual de Metaloproteinase-2/genética , Transfecção , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/patologia , Células Tumorais Cultivadas/transplante , Neoplasias da Bexiga Urinária/irrigação sanguínea , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Ativador de Plasminogênio Tipo Uroquinase/genética , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
14.
Oncogene ; 23(40): 6769-78, 2004 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-15273729

RESUMO

The hypothesis that tumor growth is angiogenesis-dependent has been documented by a considerable body of direct and indirect experimental data. Since the discovery of the vascular endothelial growth factor (VEGF), most attention has been focused on the VEGF system. Although fibroblast growth factors 1 and 2 (FGF-1 and FGF-2) can exert a strong angiogenic activity when they are supplied as a single pharmacological agent, their role in pathological angiogenesis in preclinical models remains controversial. To decipher the contribution of FGF receptors in various models of angiogenesis, we took advantage of the anti-idiotypic strategy to obtain circulating agonists specific for FGFR-1 and FGFR-2 (AIdF-1 and AIdF-2). They mimicked FGF-1 and FGF-2 for receptor binding, signal transduction, proliferation of endothelial cells and differentiation of the bladder carcinoma cell NBT-II which expresses FGFR-2b but not FGFR-1. The constitutive expression of FGFR-1 allowed binding of FGF-2 and AIdF-2 and inhibition of the proliferation of NBT-II cells. AIdF-1 and AIdF-2 induced angiogenesis in the corneal pocket assay. Although FGFR-1 dimerization achieved by AIdF-2 injection led to highly differentiated and smaller NBT-II tumors, no sign of reduction of tumor angiogenesis was observed, thus suggesting that endothelial cells are resistant to FGF.


Assuntos
Fator 2 de Crescimento de Fibroblastos/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Neoplasias da Bexiga Urinária/patologia , Córtex Suprarrenal/irrigação sanguínea , Animais , Anticorpos Anti-Idiotípicos/imunologia , Capilares , Divisão Celular , Linhagem Celular Tumoral , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Humanos , Camundongos , Camundongos Nus , Fosforilação , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Transdução de Sinais/fisiologia , Transplante Heterólogo , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/imunologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
15.
Mol Biol Cell ; 23(2): 297-309, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22114353

RESUMO

Invadosomes are F-actin structures capable of degrading the matrix through the activation of matrix metalloproteases. As fibrillar type I collagen promotes pro-matrix metalloproteinase 2 activation by membrane type 1 matrix metalloproteinase, we aimed at investigating the functional relationships between collagen I organization and invadosome induction. We found that fibrillar collagen I induced linear F-actin structures, distributed along the fibrils, on endothelial cells, macrophages, fibroblasts, and tumor cells. These structures share features with conventional invadosomes, as they express cortactin and N-WASP and accumulate the scaffold protein Tks5, which proved essential for their formation. On the basis of their ability to degrade extracellular matrix elements and their original architecture, we named these structures "linear invadosomes." Interestingly, podosomes or invadopodia were replaced by linear invadosomes upon contact of the cells with fibrillar collagen I. However, linear invadosomes clearly differ from classical invadosomes, as they do not contain paxillin, vinculin, and ß1/ß3 integrins. Using knockout mouse embryonic fibroblasts and RGD peptide, we demonstrate that linear invadosome formation and activity are independent of ß1 and ß3 integrins. Finally, linear invadosomes also formed in a three-dimensional collagen matrix. This study demonstrates that fibrillar collagen I is the physiological inducer of a novel class of invadosomes.


Assuntos
Colágeno Tipo I/metabolismo , Colágeno Tipo I/ultraestrutura , Matriz Extracelular/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Bovinos , Linhagem Celular , Cricetinae , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Humanos , Integrina beta1/metabolismo , Integrina beta3/metabolismo , Camundongos , Camundongos Knockout , Oligopeptídeos/farmacologia , Suínos , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo
16.
Cancer Res ; 69(3): 1027-36, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19176369

RESUMO

We have investigated the role of phosphoinositide 3-kinases (PI3Ks) in the in vitro pathophysiology of acute promyelocytic leukemia (APL) and in the response to treatment with all-trans-retinoic-acid (ATRA), utilizing a range of novel inhibitors that target individual or all catalytic class I isoforms of PI3K (p110alpha, p110beta, p110delta, and p110gamma). ATRA-induced phosphorylation of the Akt kinase and ribosomal S6 protein in APL cells was sensitive to class I PI3K, and p110beta or p110delta inhibitors, and to the mammalian target of rapamycin (mTOR) inhibitor rapamycin. In primary APL, inhibition of p110beta or p110delta triggered apoptosis in the absence or presence of ATRA. Class I PI3K inhibition could also reverse ATRA-induced protection of these cells against doxorubicin and arsenic trioxide, correlating with impaired induction of the antiapoptotic MCL-1 protein. The differentiation-inducing effects of ATRA were not dependent on class I PI3K/mTOR. In summary, class I PI3K signaling, mediated by p110beta and p110delta, plays an important role in basal and ATRA-induced cell survival mechanisms in APL. Addition of PI3K inhibitors to induction treatment regimens may provide therapeutic benefit.


Assuntos
Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/enzimologia , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Tretinoína/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Processos de Crescimento Celular , Humanos , Isoenzimas , Leucemia Promielocítica Aguda/patologia , Proteína de Sequência 1 de Leucemia de Células Mieloides , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/administração & dosagem , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Especificidade por Substrato , Serina-Treonina Quinases TOR , Tretinoína/administração & dosagem , Regulação para Cima/efeitos dos fármacos
17.
Bull Cancer ; 95(1): 51-6, 2008 Jan.
Artigo em Francês | MEDLINE | ID: mdl-18230570

RESUMO

It is widely accepted that the development of carcinoma is not only due to somatic mutations in epithelial cells but also is influenced by the tumor microenvironment ie the stroma. The different stroma components produced growth factors, cytokines, the extra cellular matrix and also participated to the recruitment of the endothelial cells necessary for the tumor neovascularisation. The stroma favored the oncogenesis through synergistic reciprocal paracrine signals with the tumor cells. The stroma is determinant for the tumor progression and therefore is an important therapeutic target.


Assuntos
Carcinoma/patologia , Comunicação Celular , Neoplasias/patologia , Células Estromais/patologia , Carcinoma/fisiopatologia , Membrana Celular/fisiologia , Progressão da Doença , Fatores de Crescimento Endotelial/metabolismo , Inibidores Enzimáticos/uso terapêutico , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Inibidores de Metaloproteinases de Matriz , Metaloproteinases da Matriz/metabolismo , Invasividade Neoplásica , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Neovascularização Patológica/etiologia , Células Estromais/fisiologia , Fatores de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA