Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biosensors (Basel) ; 14(4)2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38667185

RESUMO

Separase is a key cysteine protease in the separation of sister chromatids through the digestion of the cohesin ring that inhibits chromosome segregation as a trigger of the metaphase-anaphase transition in eukaryotes. Its activity is highly regulated by binding with securin and cyclinB-CDK1 complex. These bindings prevent the proteolytic activity of separase until the onset of anaphase. Chromosome missegregation and aneuploidy are frequently observed in malignancies. However, there are some difficulties in biochemical examinations due to the instability of separase in vitro and the fact that few spatiotemporal resolution approaches exist for monitoring live separase activity throughout mitotic processes. Here, we have developed FRET-based molecular sensors, including GFP variants, with separase-cleavable sequences as donors and covalently attached fluorescent dyes as acceptor molecules. These are applicable to conventional live cell imaging and flow cytometric analysis because of efficient live cell uptake. We investigated the performance of equivalent molecular sensors, either localized or not localized inside the nucleus under cell cycle control, using flow cytometry. Synchronized cell cycle progression rendered significant separase activity detections in both molecular sensors. We obtained consistent outcomes with localized molecular sensor introduction and cell cycle control by fluorescent microscopic observations. We thus established live cell separase activity monitoring systems that can be used specifically or statistically, which could lead to the elucidation of separase properties in detail.


Assuntos
Ciclo Celular , Segregação de Cromossomos , Transferência Ressonante de Energia de Fluorescência , Separase , Separase/metabolismo , Humanos , Técnicas Biossensoriais , Células HeLa
2.
Nature ; 608(7924): 784-794, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35798028

RESUMO

Cancer relapse after chemotherapy remains a main cause of cancer-related death. Although the relapse is thought to result from the propagation of resident cancer stem cells1, a lack of experimental platforms that enable the prospective analysis of cancer stem cell dynamics with sufficient spatiotemporal resolution has hindered the testing of this hypothesis. Here we develop a live genetic lineage-tracing system that allows the longitudinal tracking of individual cells in xenotransplanted human colorectal cancer organoids, and identify LGR5+ cancer stem cells that exhibit a dormant behaviour in a chemo-naive state. Dormant LGR5+ cells are marked by the expression of p27, and intravital imaging provides direct evidence of the persistence of LGR5+p27+ cells during chemotherapy, followed by clonal expansion. Transcriptome analysis reveals that COL17A1-a cell-adhesion molecule that strengthens hemidesmosomes-is upregulated in dormant LGR5+p27+ cells. Organoids in which COL17A1 is knocked out lose the dormant LGR5+p27+ subpopulation and become sensitive to chemotherapy, which suggests that the cell-matrix interface has a role in the maintenance of dormancy. Chemotherapy disrupts COL17A1 and breaks the dormancy in LGR5+p27+ cells through FAK-YAP activation. Abrogation of YAP signalling prevents chemoresistant cells from exiting dormancy and delays the regrowth of tumours, highlighting the therapeutic potential of YAP inhibition in preventing cancer relapse. These results offer a viable therapeutic approach to overcome the refractoriness of human colorectal cancer to conventional chemotherapy.


Assuntos
Neoplasias do Colo , Células-Tronco Neoplásicas , Autoantígenos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem da Célula , Proliferação de Células , Rastreamento de Células , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Quinase 1 de Adesão Focal/metabolismo , Perfilação da Expressão Gênica , Xenoenxertos , Humanos , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/patologia , Colágenos não Fibrilares/metabolismo , Organoides/metabolismo , Organoides/patologia , Receptores Acoplados a Proteínas G/metabolismo , Fatores de Transcrição/metabolismo , Colágeno Tipo XVII
3.
Arthritis Rheumatol ; 71(11): 1923-1934, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31173491

RESUMO

OBJECTIVE: To assess the preclinical efficacy and mechanism of action of an anti-CX3 CL1 monoclonal antibody (mAb) in systemic sclerosis (SSc). METHODS: Cultured human dermal fibroblasts were used to evaluate the direct effect of anti-CX3 CL1 mAb on fibroblasts. In addition, bleomycin-induced and growth factor-induced models of SSc were used to investigate the effect of anti-CX3 CL1 mAb on leukocyte infiltration, collagen deposition, and vascular damage in the skin. RESULTS: Anti-CX3 CL1 mAb treatment significantly inhibited Smad3 phosphorylation (P < 0.05) and expression of type I collagen and fibronectin 1 (P < 0.01) in dermal fibroblasts stimulated with transforming growth factor ß1 (TGFß1). In the bleomycin model, daily subcutaneous bleomycin injection increased serum CX3 CL1 levels (P < 0.05) and augmented lesional CX3 CL1 expression. Simultaneous administration of anti-CX3 CL1 mAb or CX3 CR1 deficiency significantly suppressed the dermal thickness, collagen content, and capillary loss caused by bleomycin (P < 0.05). Injection of bleomycin induced expression of pSmad3 and TGFß1 in the skin, which was inhibited by anti-CX3 CL1 mAb. Further, the dermal infiltration of CX3 CR1+ cells, macrophages (inflammatory and alternatively activated [M2-like] subsets), and CD3+ cells significantly decreased following anti-CX3 CL1 mAb therapy (P < 0.05), as did the enhanced skin expression of fibrogenic molecules, such as thymic stromal lymphopoietin and secreted phosphoprotein 1 (P < 0.05). However, the treatment did not significantly reduce established skin fibrosis. In the second model, simultaneous anti-mCX3 CL1 mAb therapy significantly diminished the skin fibrosis induced by serial subcutaneous injection of TGFß and connective tissue growth factor (P < 0.01). CONCLUSION: Anti-CX3 CL1 mAb therapy may be a novel approach for treating early skin fibrosis in inflammation-driven fibrotic skin disorders such as SSc.


Assuntos
Anticorpos Monoclonais/farmacologia , Receptor 1 de Quimiocina CX3C/imunologia , Capilares/efeitos dos fármacos , Quimiocina CX3CL1/antagonistas & inibidores , Colágeno/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Escleroderma Sistêmico/imunologia , Pele/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/toxicidade , Bleomicina/toxicidade , Capilares/patologia , Quimiocina CX3CL1/imunologia , Colágeno/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Fibroblastos/patologia , Fibrose/induzido quimicamente , Humanos , Técnicas In Vitro , Inflamação , Camundongos , Escleroderma Sistêmico/patologia , Transdução de Sinais , Pele/imunologia , Pele/patologia , Proteína Smad3/efeitos dos fármacos , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta3/toxicidade
4.
Cell Rep ; 23(4): 974-982, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29694905

RESUMO

Recent studies have revealed that newly emerging transformed cells are often eliminated from epithelial tissues via cell competition with the surrounding normal epithelial cells. This cancer preventive phenomenon is termed epithelial defense against cancer (EDAC). However, it remains largely unknown whether and how EDAC is diminished during carcinogenesis. In this study, using a cell competition mouse model, we show that high-fat diet (HFD) feeding substantially attenuates the frequency of apical elimination of RasV12-transformed cells from intestinal and pancreatic epithelia. This process involves both lipid metabolism and chronic inflammation. Furthermore, aspirin treatment significantly facilitates eradication of transformed cells from the epithelial tissues in HFD-fed mice. Thus, our work demonstrates that obesity can profoundly influence competitive interaction between normal and transformed cells, providing insights into cell competition and cancer preventive medicine.


Assuntos
Transformação Celular Neoplásica/imunologia , Gorduras na Dieta/efeitos adversos , Células Epiteliais/imunologia , Imunidade Inata/efeitos dos fármacos , Mucosa Intestinal/imunologia , Obesidade/imunologia , Pâncreas/imunologia , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Gorduras na Dieta/farmacologia , Cães , Células Epiteliais/patologia , Imunidade Inata/genética , Mucosa Intestinal/patologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Metabolismo dos Lipídeos/imunologia , Células Madin Darby de Rim Canino , Camundongos , Obesidade/induzido quimicamente , Obesidade/genética , Obesidade/patologia , Pâncreas/patologia
5.
Methods Mol Biol ; 1763: 153-163, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29476497

RESUMO

Most of preclinical cancer studies use xenograft models established from human cell lines or patient-derived cancer cells subcutaneously implanted into the flank of immunocompromised mice. These models are often assumed to represent the original diseases and are valuable tools, at least to some extent, for understanding both the basic biology of cancer and for proof-of-concept studies of molecularly targeted therapies. However, analyzing the cellular behavior of individual components within xenografts, including tumor cells, stromal cells, immune cells, and blood vessels, is challenging. In particular, it has been difficult and urgently required to trace the whole process of heterogeneous tumor microenvironment formation mediated by various components described above. Here we demonstrate a method for monitoring this process using a window device system that we have recently developed and a subcutaneous xenograft model that accurately recapitulates the histology of human lung adenocarcinoma. Use of our imaging window device and a multiphoton laser scanning microscope provides a powerful tool for investigating tumor heterogeneity and responses to drug treatments in an in vivo live imaging system.


Assuntos
Adenocarcinoma/patologia , Neoplasias Pulmonares/patologia , Imagem Molecular/instrumentação , Animais , Feminino , Humanos , Injeções Subcutâneas , Camundongos , Camundongos Nus , Camundongos SCID , Células Tumorais Cultivadas , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
6.
JCI Insight ; 2(3): e90905, 2017 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-28194443

RESUMO

In the central nervous system, endothelial cells (ECs) and pericytes (PCs) of blood vessel walls cooperatively form a physical and chemical barrier to maintain neural homeostasis. However, in diabetic retinopathy (DR), the loss of PCs from vessel walls is assumed to cause breakdown of the blood-retina barrier (BRB) and subsequent vision-threatening vascular dysfunctions. Nonetheless, the lack of adequate DR animal models has precluded disease understanding and drug discovery. Here, by using an anti-PDGFRß antibody, we show that transient inhibition of the PC recruitment to developing retinal vessels sustained EC-PC dissociations and BRB breakdown in adult mouse retinas, reproducing characteristic features of DR such as hyperpermeability, hypoperfusion, and neoangiogenesis. Notably, PC depletion directly induced inflammatory responses in ECs and perivascular infiltration of macrophages, whereby macrophage-derived VEGF and placental growth factor (PlGF) activated VEGFR1 in macrophages and VEGFR2 in ECs. Moreover, angiopoietin-2 (Angpt2) upregulation and Tie1 downregulation activated FOXO1 in PC-free ECs locally at the leaky aneurysms. This cycle of vessel damage was shut down by simultaneously blocking VEGF, PlGF, and Angpt2, thus restoring the BRB integrity. Together, our model provides new opportunities for identifying the sequential events triggered by PC deficiency, not only in DR, but also in various neurological disorders.


Assuntos
Anticorpos/farmacologia , Retinopatia Diabética/imunologia , Pericitos/citologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Angiopoietina-2/metabolismo , Animais , Barreira Hematorretiniana , Retinopatia Diabética/tratamento farmacológico , Modelos Animais de Doenças , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Proteínas de Membrana , Camundongos , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Proteínas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Genes Cells ; 18(11): 985-98, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24112238

RESUMO

Tight junctions (TJs) and adherens junctions (AJs) form an apical junctional complex at the apical side of the lateral membranes of epithelial cells, in which TJs are aligned at the apical side of AJs. Many cell adhesion molecules (CAMs) and cell polarity molecules (CPMs) cooperatively regulate the formation of the apical junctional complex, but the mechanism for the alignment of TJs at the apical side of AJs is not fully understood. We developed a cellular system with which epithelial-like TJs and AJs were reconstituted in fibroblasts and analyzed the cooperative roles of CAMs and CPMs. We exogenously expressed various combinations of CAMs and CPMs in fibroblasts that express negligible amounts of these molecules endogenously. In these cells, the nectin-based cell-cell adhesion was formed at the apical side of the junctional adhesion molecule (JAM)-based cell-cell adhesion, and cadherin and claudin were recruited to the nectin-3- and JAM-based cell-cell adhesion sites to form AJ-like and TJ-like domains, respectively. This inversed alignment of the AJ-like and TJ-like domains was reversed by complementary expression of CPMs Par-3, atypical protein kinase C, Par-6, Crb3, Pals1 and Patj. We describe the cooperative roles of these CAMs and CPMs in the apico-basal alignment of TJs and AJs in epithelial cells.


Assuntos
Junções Aderentes/metabolismo , Moléculas de Adesão Celular/metabolismo , Células Epiteliais/metabolismo , Moléculas de Adesão Juncional/metabolismo , Junções Íntimas/metabolismo , Animais , Caderinas/metabolismo , Adesão Celular , Moléculas de Adesão Celular/genética , Polaridade Celular , Claudina-1/metabolismo , Humanos , Moléculas de Adesão Juncional/genética , Células L , Camundongos , Células NIH 3T3 , Nectinas , Junções Íntimas/genética
8.
J Biol Chem ; 288(41): 29356-68, 2013 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-23990464

RESUMO

Adherens junction (AJ) is a specialized cell-cell junction structure that plays a role in mechanically connecting adjacent cells to resist strong contractile forces and to maintain tissue structure, particularly in the epithelium. AJ is mainly comprised of cell adhesion molecules cadherin and nectin and their associating cytoplasmic proteins including ß-catenin, α-catenin, p120(ctn), and afadin. Our series of studies have revealed that nectin first forms cell-cell adhesion and then recruits cadherin to form AJ. The recruitment of cadherin by nectin is mediated by the binding of α-catenin and p120(ctn) to afadin. Recent studies showed that PLEKHA7 binds to p120(ctn), which is associated with E-cadherin, and maintains the integrity of AJ in epithelial cells. In this study, we showed that PLEKHA7 bound to afadin in addition to p120(ctn) and was recruited to the nectin-3α-based cell-cell adhesion site in a manner dependent on afadin, but not on p120(ctn). The binding of PLEKHA7 to afadin was required for the proper formation of AJ, but not for the formation of tight junction, in EpH4 mouse mammary gland epithelial cells. These results indicate that PLEKHA7 plays a cooperative role with nectin and afadin in the proper formation of AJ in epithelial cells.


Assuntos
Junções Aderentes/metabolismo , Proteínas de Transporte/metabolismo , Moléculas de Adesão Celular/metabolismo , Células Epiteliais/metabolismo , Proteínas dos Microfilamentos/metabolismo , Animais , Western Blotting , Caderinas/genética , Caderinas/metabolismo , Proteínas de Transporte/genética , Cateninas/genética , Cateninas/metabolismo , Adesão Celular/genética , Moléculas de Adesão Celular/genética , Células Cultivadas , Células HEK293 , Humanos , Células L , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/genética , Microscopia de Fluorescência , Nectinas , Interferência de RNA , Junções Íntimas/metabolismo , delta Catenina
9.
J Biol Chem ; 287(51): 42455-68, 2012 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-23100251

RESUMO

During epithelial junctional development, both vesicle transport and reorganization of the actin cytoskeleton must be spatiotemporally regulated. Coordination of these cellular functions is especially important, but the precise mechanism remains elusive. Previously, we identified junctional Rab13-binding protein (JRAB)/molecules interacting with CasL-like 2 (MICAL-L2) as an effector of the Rab13 small G protein, and we found that the Rab13-JRAB system may be involved in the formation of cell-cell adhesions via transport of adhesion molecules. Here, we showed that JRAB interacts with two actin-binding proteins, actinin-1 and -4, and filamentous actin via different domains and regulates actin cross-linking and stabilization through these interactions. During epithelial junctional development, JRAB is prominently enriched in the actin bundle at the free border; subsequently, JRAB undergoes a Rab13-dependent conformational change that is required for maturation of cell-cell adhesion sites. These results suggest that Rab13 and JRAB regulate reorganization of the actin cytoskeleton throughout epithelial junctional development from establishment to maturation of cell-cell adhesion.


Assuntos
Citoesqueleto de Actina/metabolismo , Proteínas do Citoesqueleto/metabolismo , Epitélio/crescimento & desenvolvimento , Epitélio/metabolismo , Junções Íntimas/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Actinina/metabolismo , Actinas/metabolismo , Animais , Linhagem Celular , Membrana Celular/metabolismo , Proteínas do Citoesqueleto/química , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Camundongos , Proteínas dos Microfilamentos , Modelos Biológicos , Ligação Proteica , Multimerização Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas rab de Ligação ao GTP/química
10.
J Biol Chem ; 286(42): 36667-76, 2011 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-21880726

RESUMO

In normal epithelial cells, integrin α(6)ß(4) is abundantly expressed and forms hemidesmosomes, which is a cellular structure that mediates cell-extracellular matrix binding. In many types of cancer cells, integrin α(6)ß(4) is up-regulated, laminin is cleaved, and hemidesmosomes are disrupted, eventually causing an enhancement of cancer cell movement and facilitation of their invasion. We previously showed that the immunoglobulin-like cell adhesion molecule Necl-2 (Nectin-like molecule 2), known as a tumor suppressor, inhibits cancer cell movement by suppressing the ErbB3/ErbB2 signaling. We show here that Necl-2 interacts in cis with integrin α(6)ß(4). The binding of Necl-2 with integrin ß(4) was mediated by its extracellular region. In human colorectal adenocarcinoma Caco-2 cells, integrin α(6)ß(4) was localized at hemidesmosomes. Small interfering RNA-mediated suppression of Necl-2 expression enhanced the phorbol ester-induced disruption of the integrin α(6)ß(4) complex at hemidesmosomes, whereas expression of Necl-2 suppressed the disruption of this structure. These results indicate that tumor-suppressive functions of Necl-2 are mediated by the stabilization of the hemidesmosome structure in addition to the inhibition of the ErbB3/ErbB2 signaling.


Assuntos
Moléculas de Adesão Celular/metabolismo , Hemidesmossomos/metabolismo , Imunoglobulinas/metabolismo , Integrina alfa6beta4/metabolismo , Células CACO-2 , Molécula 1 de Adesão Celular , Moléculas de Adesão Celular/genética , Células HEK293 , Hemidesmossomos/genética , Humanos , Imunoglobulinas/genética , Integrina alfa6beta4/genética , Integrina beta4/genética , Integrina beta4/metabolismo , Laminina/biossíntese , Laminina/genética , Ligação Proteica , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia
11.
Genes Cells ; 15(11): 1123-35, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20964795

RESUMO

Microtubules (MTs) search for and grow toward the leading edge of moving cells, followed by their stabilization at a specific structure at the rear site of the leading edge. This dynamic re-orientation of MTs is critical to directional cell movement. We previously showed that Necl-5/poliovirus receptor (PVR) interacts with platelet-derived growth factor (PDGF) receptor and integrin α(v) ß(3) at the leading edge of moving NIH3T3 cells, resulting in an enhancement of their directional movement. We studied here the role of Necl-5 in the PDGF-induced attraction of growing MTs to the leading edge of NIH3T3 cells. Necl-5 enhanced the PDGF-induced growth of MTs and attracted them near to the plasma membrane of the leading edge of NIH3T3 cells in an integrin α(v) ß(3) -dependent manner. Furthermore, Necl-5 enhanced the PDGF-induced attraction of the plus-end-tracking proteins (+TIPs), including EB1, CLIP170, an intermediate chain subunit of cytoplasmic dynein, and p150(Glued) , a subunit of dynactin, near to the plasma membrane of the leading edge. Thus, Necl-5 plays a role in the attraction of growing MTs to the plasma membrane of the leading edge of moving cells.


Assuntos
Membrana Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Microtúbulos/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Receptores Virais/metabolismo , Animais , Integrina alfaVbeta3/metabolismo , Camundongos , Células NIH 3T3 , Proteínas de Neoplasias/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Ligação Proteica
12.
Genes Cells ; 15(9): 995-1001, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20695903

RESUMO

Integrin alpha(6)beta(4) is abundantly expressed in normal epithelial cells and forms hemidesmosomes, one of cell-extracellular matrix junctions. In many types of cancer cells, integrin alpha(6)beta(4) is up-regulated, laminin, an integrin alpha(6)beta(4)-binding extracellular matrix protein, is cleaved, and hemidesmosomes are disrupted, eventually causing an enhancement of cancer cell movement and a facilitation of their invasion. It was previously shown that integrin alpha(6)beta(4) interacts with ErbB1 and ErbB2 and enhances cell proliferation and motility. Here we show that integrin alpha(6)beta(4) interacts with ErbB3 but not with ErbB1, ErbB2 or ErbB4, and enhances the heregulin-induced, ErbB3/ErbB2 heterodimer-mediated DNA synthesis, but not cell motility, in A549 cells.


Assuntos
DNA/biossíntese , Integrina alfa6beta4/metabolismo , Neuregulina-1/farmacologia , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Western Blotting , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , DNA/genética , Células HEK293 , Humanos , Integrina alfa6beta4/genética , Ligação Proteica , Multimerização Proteica/efeitos dos fármacos , Interferência de RNA , Receptor ErbB-2/química , Receptor ErbB-2/genética , Receptor ErbB-3/química , Receptor ErbB-3/genética , Transdução de Sinais/efeitos dos fármacos , Transfecção
13.
J Biol Chem ; 284(35): 23793-805, 2009 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-19561085

RESUMO

ErbB2 and ErbB3, members of the EGF receptor/ErbB family, form a heterodimer upon binding of a ligand, inducing the activation of Rac small G protein and Akt protein kinase for cell movement and survival, respectively. The enhanced ErbB3/ErbB2 signaling causes tumorigenesis, invasion, and metastasis. We found here that the ErbB3/ErbB2 signaling is regulated by immunoglobulin-like Necl-2, which is down-regulated in various cancer cells and serves as a tumor suppressor. The extracellular region of ErbB3, but not ErbB2, interacted in cis with that of Necl-2. This interaction reduced the ligand-induced, ErbB2-catalyzed tyrosine phosphorylation of ErbB3 and inhibited the consequent ErbB3-mediated activation of Rac and Akt, resulting in the inhibition of cancer cell movement and survival. These inhibitory effects of Necl-2 were mediated by the protein-tyrosine phosphatase PTPN13 which interacted with the cytoplasmic tail of Necl-2. We describe here this novel mechanism for silencing of the ErbB3/ErbB2 signaling by Necl-2.


Assuntos
Imunoglobulinas/metabolismo , Proteínas de Membrana/metabolismo , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Transdução de Sinais , Animais , Molécula 1 de Adesão Celular , Moléculas de Adesão Celular , Linhagem Celular , Dimerização , Humanos , Imunoglobulinas/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Ligação Proteica , Receptor ErbB-2/química , Receptor ErbB-2/genética , Receptor ErbB-3/química , Receptor ErbB-3/genética
14.
Nat Rev Mol Cell Biol ; 9(8): 603-15, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18648374

RESUMO

Nectins and nectin-like molecules (Necls) are immunoglobulin-like transmembrane cell adhesion molecules that are expressed in various cell types. Homophilic and heterophilic engagements between family members provide cells with molecular tools for intercellular communications. Nectins primarily regulate cell-cell adhesions, whereas Necls are involved in a greater variety of cellular functions. Recent studies have revealed that nectins and NECL-5, in cooperation with integrin alphavbeta3 and platelet-derived growth factor receptor, are crucial for the mechanisms that underlie contact inhibition of cell movement and proliferation; this has important implications for the development and tissue regeneration of multicellular organisms and the phenotypes of cancer cells.


Assuntos
Moléculas de Adesão Celular/fisiologia , Movimento Celular/fisiologia , Proliferação de Células , Inibição de Contato/fisiologia , Animais , Adesão Celular , Humanos , Modelos Biológicos , Nectinas , Receptores Virais/fisiologia
15.
Genes Cells ; 13(6): 549-69, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18422604

RESUMO

Moving cells form protrusions, such as filopodia and lamellipodia, and focal complexes at leading edges, which eventually enhance cell movement. The Rho family small G proteins, Rac1, Cdc42 and RhoA, are involved in the formation of these leading edge structures. We investigated the role of another small G protein Rap1 in the platelet-derived growth factor (PDGF)-induced formation of leading edge structures and cell movement. Upon stimulation of NIH3T3 cells by PDGF, leading edge structures were formed and Necl-5, integrin alpha(V)beta(3), and PDGF receptor were accumulated at leading edges. Rap1, upstream regulators of Rap1 such as Crk and C3G, and a downstream effector RalGDS, were accumulated at peripheral ruffles over lamellipodia. Over-expression of Rap1GAP, which inactivates Rap1, and knockdown of Rap1 inhibited the PDGF-induced formation of leading edge structures, accumulation of these molecules, and cell movement. In addition, Rap1 activation subsequently induced accumulation of Rac1, Vav2 and PAK at peripheral ruffles, which was inhibited by Rap1GAP and knockdown of Rap1. These results indicate that Rap1, activated by PDGF, is recruited to leading edges and that Rac1 is thereby activated locally at peripheral ruffles. This process is pivotal for the PDGF-induced formation of leading edge structures and cell movement.


Assuntos
Movimento Celular , Neuropeptídeos/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/metabolismo , Integrinas/metabolismo , Camundongos , Células NIH 3T3 , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-vav/metabolismo , Pseudópodes/metabolismo , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP
16.
Genes Cells ; 13(3): 269-84, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18298801

RESUMO

It was previously shown that platelet-derived growth factor (PDGF) receptor physically and functionally interacts with integrin alpha(v)beta(3), effectively inducing cell movement. We previously showed that Necl-5, originally identified as a poliovirus receptor, interacts with integrin alpha(v)beta(3) and enhances its clustering and the formation of focal complexes at the leading edges of moving cells, resulting in an enhancement of cell movement. We showed here that Necl-5 additionally interacts with PDGF receptor in NIH3T3 cells and regulates the interaction between PDGF receptor and integrin alpha(v)beta(3), effectively inducing directional cell movement. PDGF receptor co-localized with Necl-5 and integrin alpha(v)beta(3) at peripheral ruffles over lamellipodia, which were formed at the leading edges of moving cells in response to PDGF, but not at the focal complexes under these ruffles, whereas Necl-5 and integrin alpha(v)beta(3) co-localized at these focal complexes. The clustering of these three molecules at peripheral ruffles required the activation of integrin alpha(v)beta(3) by vitronectin and the PDGF-induced activation of the small G protein Rac and subsequent re-organization of the actin cytoskeleton. These results indicate a key role of Necl-5 in directional cell movement by physically and functionally interacting with both integrin alpha(v)beta(3) and PDGF receptor.


Assuntos
Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/metabolismo , Movimento Celular/fisiologia , Proteínas de Neoplasias/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Antígenos de Neoplasias/genética , Moléculas de Adesão Celular/genética , Células Cultivadas , Imunoprecipitação da Cromatina/métodos , Humanos , Integrina alfaVbeta3/metabolismo , Camundongos , Células NIH 3T3 , Proteínas de Neoplasias/genética , Ligação Proteica/fisiologia , Pseudópodes/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Proteínas rac de Ligação ao GTP/metabolismo
17.
J Biol Chem ; 282(25): 18481-18496, 2007 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-17446174

RESUMO

Integrin alphavbeta3, which forms focal complexes at leading edges in moving cells, is up-regulated in cancer cells and so is implicated in their invasiveness. Necl-5, originally identified as a poliovirus receptor and also up-regulated in cancer cells, colocalizes with integrin alphavbeta3 at leading edges in moving cells and enhances growth factor-induced cell movement. Here, we show that Necl-5 interacts directly, in cis, with integrin alphavbeta3, and enhances integrin alphavbeta3 clustering and focal complex formation at leading edges in NIH3T3 cells. The extracellular region of Necl-5, but not the cytoplasmic region, is necessary for its interaction with integrin alphavbeta3; however, both regions are necessary for its action. An interaction between integrin alphavbeta3 and vitronectin and PDGF-induced activation of Rac are also necessary for integrin alphavbeta3 clustering. The interaction between Necl-5 and integrin alphavbeta3 enhances PDGF-induced Rac activation, facilitating integrin alphavbeta3 clustering presumably in a feedback amplification manner. Thus, Necl-5 has a critical role in integrin alphavbeta3 clustering and focal complex formation.


Assuntos
Regulação da Expressão Gênica , Integrina alfaVbeta3/metabolismo , Proteínas de Membrana/fisiologia , Receptores Virais/fisiologia , Animais , Adesão Celular , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Microscopia de Fluorescência , Células NIH 3T3 , Fator de Crescimento Derivado de Plaquetas/metabolismo , Ligação Proteica , RNA Interferente Pequeno/metabolismo , Receptores Virais/metabolismo
18.
Genes Cells ; 12(3): 345-57, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17352739

RESUMO

Necl-5, known as a poliovirus receptor and up-regulated in many cancer cells, enhances platelet-derived growth factor (PDGF)-induced activation of Ras-Raf-MEK-ERK signaling, but not PDGF-induced tyrosine phosphorylation of PDGF receptor, resulting in facilitation of cell proliferation. Here, we showed that Necl-5 interacted with Sprouty2, known to be a negative regulator of growth factor-induced signaling, and reduced the inhibitory effect of Sprouty2 on PDGF-induced Ras signaling. Necl-5 was reported to be down-regulated by its trans-interaction with nectin-3 upon cell-cell contact, initiating cooperative cell-cell adhesion with cadherin. This down-regulation of Necl-5 caused tyrosine phosphorylation of Sprouty2 by c-Src, which was activated by PDGF receptor in response to PDGF, and inhibited PDGF-induced Ras signaling. Thus, Necl-5 and Sprouty2 cooperatively regulate PDGF-induced Ras signaling. The roles of Necl-5 and Sprouty2 in contact inhibition for cell proliferation are also discussed.


Assuntos
Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Receptores Virais/metabolismo , Proteínas ras/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos de Neoplasias/genética , Becaplermina , Proteína Tirosina Quinase CSK , Moléculas de Adesão Celular/genética , Regulação para Baixo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana/genética , Camundongos , Modelos Biológicos , Células NIH 3T3 , Proteínas de Neoplasias/genética , Fosforilação , Proteínas Serina-Treonina Quinases , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-sis , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores Virais/genética , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Quinases da Família src
19.
Biochem Biophys Res Commun ; 352(4): 856-60, 2007 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-17156749

RESUMO

Normal cells show contact inhibition of cell movement and proliferation, but this is lost following transformation. We found that Necl-5, originally identified as a poliovirus receptor and up-regulated in many cancer cells, enhances growth factor-induced cell movement and proliferation. We showed that when cells contact other cells, Necl-5 interacts in trans with nectin-3 and is removed by endocytosis from the cell surface, resulting in a reduction of cell movement and proliferation. We show here that up-regulation of the gene encoding Necl-5 by the oncogene V12-Ki-Ras causes enhanced cell movement and proliferation. Upon cell-cell contact, de novo synthesis of Necl-5 exceeds the rate of Necl-5 endocytosis, eventually resulting in a net increase in the amount of Necl-5 at the cell surface. In addition, expression of the gene encoding nectin-3 is markedly reduced in transformed cells. Thus, up-regulation of Necl-5 following transformation contributes to the loss of contact inhibition in transformed cells.


Assuntos
Proteínas de Membrana/metabolismo , Receptores Virais/metabolismo , Transformação Genética/genética , Regulação para Cima , Animais , Moléculas de Adesão Celular/metabolismo , Endocitose , Proteínas de Membrana/genética , Camundongos , Células NIH 3T3 , Nectinas , Ligação Proteica , Receptores Virais/genética , Proteínas ras/genética , Proteínas ras/metabolismo
20.
J Cell Biol ; 171(1): 165-73, 2005 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-16216929

RESUMO

Immunoglobulin-like Necl-5/Tage4/poliovirus receptor (PVR)/CD155, originally identified as the PVR, has been shown to be up-regulated in cancer cells and to enhance growth factor-induced cell movement and proliferation. In addition, Necl-5 heterophilically trans-interacts with nectin-3, a cell-cell adhesion molecule known to form adherens junctions in cooperation with cadherin. We show here that Necl-5 was down-regulated from cell surface upon cell-cell contacts in NIH3T3 cells. This down-regulation of Necl-5 was initiated by its interaction with nectin-3 and was mainly mediated by clathrin-dependent endocytosis. Then, the down-regulation of Necl-5 induced in this way reduced movement and proliferation of NIH3T3 cells. These results indicate that the down-regulation of Necl-5 induced by its interaction with nectin-3 upon cell-cell contacts may be at least one mechanism underlying contact inhibition of cell movement and proliferation.


Assuntos
Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Animais , Antígenos de Neoplasias/genética , Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/genética , Clatrina/metabolismo , Regulação para Baixo , Endocitose , Camundongos , Células NIH 3T3 , Nectinas , Proteínas de Neoplasias/genética , RNA Interferente Pequeno/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA