Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Biol Chem ; 287(46): 38716-28, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-22992737

RESUMO

Matrix metalloproteinases (MMPs) are extracellular zinc-dependent endopeptidases involved in the degradation and remodeling of extracellular matrix in physiological and pathological processes. MMPs also have a role in cell proliferation, migration, differentiation, angiogenesis, and apoptosis. We previously identified cancer invasion-related factors by comparing the gene expression profiles between parent and the highly invasive clone of cancer cells. Matrix metalloproteinase-13 (MMP-13) was identified as a common up-regulated gene by cancer invasion-related factors. Although MMP-13 slightly promoted tumor invasion, we found that MMP-13 was involved in tumor angiogenesis. Conditioned medium from MMP-13-overexpressing cells promoted capillary formation of immortalized human umbilical vein endothelial cells. Furthermore, treatment with recombinant MMP-13 protein enhanced capillary tube formation both in vitro and in vivo. MMP-13-promoted capillary tube formation was mediated by activation of focal adhesion kinase and ERK. Interestingly, MMP-13 promoted the secretion of VEGF-A from fibroblasts and endothelial cells. By immunohistochemical analysis, we found a possible correlation between MMP-13 expression and the number of blood vessels in human cancer cases. In summary, these findings suggest that MMP-13 may directly and indirectly promote tumor angiogenesis.


Assuntos
Metaloproteinase 13 da Matriz/metabolismo , Neovascularização Patológica , Animais , Linhagem Celular Tumoral , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Imuno-Histoquímica/métodos , Masculino , Metástase Neoplásica , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/química , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
J Biol Chem ; 287(30): 25419-33, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22654110

RESUMO

The contribution of hyaluronan (HA) to the regulatory network of the hematopoietic microenvironment was studied using knock-out mice of three hyaluronan synthase genes (Has1, Has2, and Has3). The number of hematopoietic progenitors was decreased in bone marrow and increased in extramedullary sites of Prx1-Cre;Has2(flox/flox);Has1(-/-);Has3(-/-) triple knock-out (tKO) mice as compared with wild type (WT) and Has1(-/-);Has3(-/-) double knock-out (dKO) mice. In line with this observation, decreased hematopoietic activity was observed in long term bone marrow cultures (LTBMC) from tKO mice, whereas the formation of the adherent layer and generation of hematopoietic cells in WT and dKO cultures was not different. 4-Methylumbelliferone (4MU) was used to pharmacologically inhibit the production of HA in LTBMC. Treatment with 4MU inhibited HA synthesis, decreased expression of HAS2 and HAS3, and eliminated hematopoiesis in LTBMC, and this effect was alleviated by the addition of exogenous HA. Exogenous HA also augmented the cell motility in LTBMC, which correlated with the HA-stimulated production of chemokines and growth factors. Conditioned media from HA-induced LTBMC enhanced the chemotaxis of hematopoietic stem/progenitor cells (HSPC) in response to SDF-1. Exposure of endothelial cells to 4MU decreased their ability to support HSPC rolling and adhesion. In addition, migration of transplanted HSPC into the marrow of 4MU-pretreated mice was lower than in untreated mice. Collectively, the results suggest that HA depletion reduces the ability of the microenvironment to support HSPC, and confirm a role for HA as a necessary regulatory element in the structure of the hematopoietic microenvironment.


Assuntos
Medula Óssea/metabolismo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Ácido Hialurônico/metabolismo , Nicho de Células-Tronco/fisiologia , Animais , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Quimiotaxia/efeitos dos fármacos , Quimiotaxia/fisiologia , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Humanos , Hialuronan Sintases , Ácido Hialurônico/genética , Himecromona/análogos & derivados , Himecromona/farmacologia , Camundongos , Camundongos Knockout , Nicho de Células-Tronco/efeitos dos fármacos
3.
J Oral Pathol Med ; 40(1): 77-82, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21070371

RESUMO

Spindle cell carcinoma (SpCC) is a biphasic tumor composed of conventional squamous cell carcinoma and a malignant spindle cell component. SpCC expresses both epithelial and mesenchymal markers by immunohistochemical analysis. There is mounting evidence for sarcomatoid transformation from the epithelial component, supporting the theory that SpCC is a monoclonal neoplasm originating from a stem cell giving rise to both components. The loss of E-cadherin and the gain of N-cadherin expression are known as the "cadherin switching". Cadherin switching is a major hallmark of epithelial-mesenchymal transition (EMT). EMT is a crucial process in cancer progression providing cancer cells with the ability to escape from the primary focus, to invade stromal tissues, and to migrate to distant regions. Although E-cadherin down-regulation is well known in various cancers, there are a few studies on N-cadherin expression in cancer. Here, therefore, we investigated N-cadherin expression in the progression of head and neck SpCC. First, we examined cadherin switching in our established SpCC cell lines, SpCC-1 and SpCC-2. SpCC-1 and SpCC-2 cells were spindle in shape and showed cadherin switching. Moreover, we examined N-cadherin expression in 15 SpCC cases by immunohistochemistry. Although N-cadherin expression was not observed in non-neoplastic squamous epithelium, high expression of N-cadherin was observed in 10 of 15 SpCC cases. Interestingly, 6 of 7 SpCC cases with metastasis showed high expression of N-cadherin. In conclusion, our findings suggest that N-cadherin may play an important role in metastasis of SpCC in addition to the pathogenesis of SpCC of the head and neck.


Assuntos
Caderinas/metabolismo , Carcinoma/metabolismo , Transformação Celular Neoplásica/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Caderinas/genética , Carcinoma/patologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Células Clonais/metabolismo , Células Clonais/patologia , Feminino , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade
4.
Artigo em Japonês | MEDLINE | ID: mdl-22259844

RESUMO

To establish a detection method for enterohemorrhagic Escherichia coli (EHEC) O111 in meat, a single-laboratory evaluation and a collaborative study were conducted focusing on comparisons of the efficiencies in combination with enrichment, a direct plating method and a plating method with immunomagnetic separation (IMS-plating method) using various agar media for EHEC O111, loop-mediated isothermal amplification (LAMP) assay targeting the Verocytotoxin (VT) gene as a molecular detection method. On a single-laboratory evaluation, enrichment in modified EC at 36 degrees C was inferior to that in modified EC supplemented with novobiocin (NmEC) and mEC at 42 degrees C to isolate EHEC O111 by plating methods. On a collaborative study, there were no significant differences between combinations of enrichment in NmEC at 42 degrees C-LAMP assay and enrichment in mEC at 42 degrees C-LAMP assay. The combinations of enrichment in NmEC at 42 degrees C-direct plating and enrichment in NmEC at 42 degrees C-IMS-plating were superior to combinations of enrichment in mEC at 42 degrees C-direct plating and enrichment in mEC at 42 degrees C-IMS-plating (p<0.05). There were no significant differences among the six different agar media by the direct plating and IMS-plating methods. As a result, it was suggested that the following methods are adequate for detection of EHEC O111 in beef: combinations of enrichment in NmEC at 42 degrees C, and direct plating and IMS-plating methods, or LAMP assay as a screening assay to detect VT gene followed by direct plating and IMS-plating methods.


Assuntos
Técnicas Bacteriológicas/métodos , Escherichia coli Êntero-Hemorrágica/isolamento & purificação , Microbiologia de Alimentos/métodos , Carne/microbiologia , Animais , Bovinos , Separação Imunomagnética , Toxina Shiga I/genética
5.
Cancer Res ; 81(6): 1472-1485, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33414172

RESUMO

Invadopodia are actin-based proteolytic membrane protrusions required for invasive behavior and tumor growth. In this study, we used our high-content screening assay to identify kinases whose activity affects invadopodia formation. Among the top hits selected for further analysis was TAO3, an STE20-like kinase of the GCK subfamily. TAO3 was overexpressed in many human cancers and regulated invadopodia formation in melanoma, breast, and bladder cancers. Furthermore, TAO3 catalytic activity facilitated melanoma growth in three-dimensional matrices and in vivo. A novel, potent catalytic inhibitor of TAO3 was developed that inhibited invadopodia formation and function as well as tumor cell extravasation and growth. Treatment with this inhibitor demonstrated that TAO3 activity is required for endosomal trafficking of TKS5α, an obligate invadopodia scaffold protein. A phosphoproteomics screen for TAO3 substrates revealed the dynein subunit protein LIC2 as a relevant substrate. Knockdown of LIC2 or expression of a phosphomimetic form promoted invadopodia formation. Thus, TAO3 is a new therapeutic target with a distinct mechanism of action. SIGNIFICANCE: An unbiased screening approach identifies TAO3 as a regulator of invadopodia formation and function, supporting clinical development of this class of target.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Endossomos/metabolismo , Invasividade Neoplásica/patologia , Podossomos/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Dineínas do Citoplasma/genética , Dineínas do Citoplasma/metabolismo , Conjuntos de Dados como Assunto , Matriz Extracelular , Feminino , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Ensaios de Triagem em Larga Escala , Humanos , Masculino , Melanoma/tratamento farmacológico , Melanoma/patologia , Camundongos , Invasividade Neoplásica/prevenção & controle , Podossomos/patologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia , Imagem com Lapso de Tempo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Eur J Cell Biol ; 99(7): 151122, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33070041

RESUMO

The scaffold protein Tks5α is required for invadopodia-mediated cancer invasion both in vitro and in vivo. We have previously also revealed a role for Tks5 in tumor cell growth using three-dimensional (3D) culture model systems and mouse transplantation experiments. Here we use both 3D and high-density fibrillar collagen (HDFC) culture to demonstrate that native collagen-I, but not a form lacking the telopeptides, stimulated Tks5-dependent growth, which was dependent on the DDR collagen receptors. We used microenvironmental microarray (MEMA) technology to determine that laminin, fibronectin and tropoelastin also stimulated invadopodia formation. A Tks5α-specific monoclonal antibody revealed its expression both on microtubules and at invadopodia. High- and super-resolution microscopy of cells in and on collagen was then used to place Tks5α at the base of invadopodia, separated from much of the actin and cortactin, but coincident with both matrix metalloprotease and cathepsin proteolytic activity. Inhibition of the Src family kinases, cathepsins or metalloproteases all reduced invadopodia length but each had distinct effects on Tks5α localization. These studies highlight the crosstalk between invadopodia and extracellular matrix components, and reveal the invadopodium to be a spatially complex structure.


Assuntos
Matriz Extracelular/metabolismo , Podossomos/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Camundongos , Isoformas de Proteínas
7.
J Periodontol ; 79(3): 535-40, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18315437

RESUMO

BACKGROUND: Enamel matrix derivative (EMD) is used clinically to promote periodontal tissue regeneration with variable efficacy. EMD application results in significantly higher frequencies of sites without clinical signs of inflammation; additionally, patients receiving EMD therapy report significantly less post-treatment discomfort. However, there are few reports that focus on defining the biologic mechanisms for the observed anti-inflammatory effects of EMD. The aim of this study was to evaluate the influence of EMD on inflammatory-associated markers using an in vitro monocyte assay. METHODS: Rat monocytes were exposed to lipopolysaccharide (LPS; 100 ng/ml from Escherichia coli or Actinobacillus actinomycetemcomitans) along with EMD (0, 50, 100, or 200 microg/ml). Levels of tumor necrosis factor-alpha (TNF-alpha) and prostaglandin E(2) (PGE(2)) in conditioned media were analyzed by enzyme-linked immunosorbent assay. In addition, the effects of exogenous PGE(2) on TNF-alpha production from LPS-stimulated monocytes were determined. RESULTS: LPS-stimulated monocytes exposed to EMD exhibited a decrease in TNF-alpha production (0.10- to 0.52-fold) and an increase in PGE(2) production (1.31- to 2.71-fold) compared to controls not treated with EMD. Exogenously applied PGE(2) decreased TNF-alpha production by LPS-stimulated monocytes in a dose-dependent manner, and EMD treatment enhanced this PGE(2)-mediated inhibition of TNF-alpha production. CONCLUSION: In addition to EMD's published role in inducing proliferation, migration, adhesion, mineralization, and differentiation of periodontal ligament cells, our results indicated that EMD modulates two inflammation-associated factors, TNF-alpha and PGE(2), in monocytes.


Assuntos
Anti-Inflamatórios/farmacologia , Proteínas do Esmalte Dentário/farmacologia , Dinoprostona/agonistas , Monócitos/efeitos dos fármacos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Aggregatibacter actinomycetemcomitans/química , Animais , Células Cultivadas , Dinoprostona/biossíntese , Dinoprostona/farmacologia , Escherichia coli/química , Lipopolissacarídeos , Monócitos/metabolismo , Ratos , Fator de Necrose Tumoral alfa/biossíntese
8.
Foodborne Pathog Dis ; 5(1): 97-103, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18260820

RESUMO

To detect various serotypes of Shiga toxin-producing Escherichia coli (STEC) in food, methods independent of serotyping are needed. We established procedures to isolate STEC using a rapid and sensitive loop-mediated isothermal amplification (LAMP) assay targeting the Shiga toxin (ST) gene and a method of plating LAMP assay positive dilutions onto media for the selection of E. coli. After incubation, suspensions of a colony or some colonies were tested in the LAMP assay. Positive suspensions were diluted and plated onto selective media. The procedure was repeated. Finally, LAMP positive colonies were confirmed as STEC and serotype. As a result of surveillance in beef in 2005-2007, 11 of 720 samples (1.5%) tested positive for the ST gene by LAMP assay. Serotype O8, O128, and O-untypeable STEC were isolated from the samples by the newly established procedure. It was demonstrated that the procedure was effective for detecting STEC independent of serotype.


Assuntos
Contaminação de Alimentos/análise , Carne/microbiologia , Toxinas Shiga/genética , Escherichia coli Shiga Toxigênica/isolamento & purificação , Animais , Austrália/epidemiologia , Bovinos , Contagem de Colônia Microbiana , Meios de Cultura , Microbiologia de Alimentos , Humanos , Separação Imunomagnética , Prevalência , Sensibilidade e Especificidade , Sorotipagem , Toxinas Shiga/biossíntese , Escherichia coli Shiga Toxigênica/classificação , Escherichia coli Shiga Toxigênica/genética , Estados Unidos/epidemiologia
9.
Mol Cell Biol ; 38(6)2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29263157

RESUMO

The transcription factor gene MYC is important in breast cancer, and its mRNA is maintained at a high level even in the absence of gene amplification. The mechanism(s) underlying increased MYC mRNA expression is unknown. Here, we demonstrate that MYC mRNA was stabilized upon estrogen stimulation of estrogen receptor-positive breast cancer cells via SRC-dependent effects on a recently described RNA-binding protein, IMP1 with an N-terminal deletion (ΔN-IMP1). We also show that loss of the tumor suppressor p53 increased MYC mRNA levels even in the absence of estrogen stimulation. However, in cells with wild-type p53, SRC acted to overcome p53-mediated inhibition of estrogen-stimulated cell cycle entry and progression. SRC thus promotes cell proliferation in two ways: by stabilizing MYC mRNA and by inhibiting p53 function. Since estrogen receptor-positive breast cancers typically express wild-type p53, these studies establish a rationale for p53 status to be predictive for effective SRC inhibitor treatment in this subtype of breast cancer.


Assuntos
Neoplasias da Mama/genética , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Mensageiro/metabolismo , Proteína Supressora de Tumor p53/antagonistas & inibidores , Quinases da Família src/metabolismo , Animais , Neoplasias da Mama/metabolismo , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Estradiol/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Xenoenxertos , Humanos , Células MCF-7 , Camundongos Nus , Proteínas Proto-Oncogênicas c-myc/biossíntese , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Receptores de Estrogênio/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Quinases da Família src/genética
10.
Sci Adv ; 4(9): eaat7828, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30214939

RESUMO

High lethality rates associated with metastatic cancer highlight an urgent medical need for improved understanding of biologic mechanisms driving metastatic spread and identification of biomarkers predicting late-stage progression. Numerous neoplastic cell intrinsic and extrinsic mechanisms fuel tumor progression; however, mechanisms driving heterogeneity of neoplastic cells in solid tumors remain obscure. Increased mutational rates of neoplastic cells in stressed environments are implicated but cannot explain all aspects of tumor heterogeneity. We present evidence that fusion of neoplastic cells with leukocytes (for example, macrophages) contributes to tumor heterogeneity, resulting in cells exhibiting increased metastatic behavior. Fusion hybrids (cells harboring hematopoietic and epithelial properties) are readily detectible in cell culture and tumor-bearing mice. Further, hybrids enumerated in peripheral blood of human cancer patients correlate with disease stage and predict overall survival. This unique population of neoplastic cells provides a novel biomarker for tumor staging, as well as a potential therapeutic target for intervention.


Assuntos
Carcinoma Ductal Pancreático/patologia , Células Neoplásicas Circulantes/patologia , Neoplasias Pancreáticas/patologia , Animais , Biomarcadores Tumorais/sangue , Carcinoma Ductal Pancreático/mortalidade , Fusão Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Células Epiteliais/patologia , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Células Híbridas , Cariotipagem , Macrófagos/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Pancreáticas/mortalidade , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Sci Rep ; 7: 40187, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-28054649

RESUMO

Ameloblastin (AMBN), the most abundant non-amelogenin enamel matrix protein, plays a role in ameloblast differentiation. Previously, we found that AMBN promoted osteogenic differentiation via the interaction between CD63 and integrin ß1, leading to the inactivation of Src; however, how AMBN affects the malignant behavior of osteosarcoma is still unclear. Osteosarcoma affects the bone and is associated with poor prognosis because of the high rate of pulmonary metastases and drug resistance. Here we demonstrated that stable overexpression of AMBN induced apoptosis and suppressed colony formation and cell migration via the inactivation of Src-Stat3 pathway in human osteosarcoma cells. Moreover, AMBN induced chemosensitivity to doxorubicin. Thus, AMBN induced a tumor suppressive phenotype and chemosensitivity to doxorubicin via the AMBN-Src-Stat3 axis in osteosarcoma. Indeed, immunohistochemical expression of AMBN was significantly correlated with better outcome of osteosarcoma patients. Our findings suggest that AMBN can be a new prognostic marker and therapeutic target for osteosarcoma combined with conventional doxorubicin treatment.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Proteínas do Esmalte Dentário/metabolismo , Doxorrubicina/farmacologia , Proteína Oncogênica pp60(v-src)/antagonistas & inibidores , Osteossarcoma/tratamento farmacológico , Fator de Transcrição STAT3/antagonistas & inibidores , Animais , Apoptose , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Camundongos Nus , Modelos Biológicos , Transplante de Neoplasias , Osteossarcoma/patologia , Resultado do Tratamento
12.
Oncotarget ; 7(48): 78473-78486, 2016 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-27802184

RESUMO

Metastatic cancer cells are characterized by their ability to degrade and invade through extracellular matrix. We previously showed that the Tks adaptor proteins, Tks4 and Tks5, are required for invadopodia formation and/or function in Src-transformed fibroblasts and a number of human cancer cell types. In this study, we investigated the role of Tks adaptor proteins in melanoma cell invasion and metastasis. Knockdown of either Tks4 or Tks5 in both mouse and human melanoma cell lines resulted in a decreased ability to form invadopodia and degrade extracellular matrix. In addition, Tks-knockdown melanoma cells had decreased proliferation in a 3-dimensional type l collagen matrix, but not in 2-dimensional culture conditions. We also investigated the role of Tks proteins in melanoma progression in vivo using xenografts and experimental metastasis assays. Consistent with our in vitro results, reduction of Tks proteins markedly reduced subcutaneous melanoma growth as well as metastatic growth in the lung. We explored the clinical relevance of Tks protein expression in human melanoma specimens using a tissue microarray. Compared to non-malignant nevi, both Tks proteins were highly expressed in melanoma tissues. Moreover, metastatic melanoma cases showed higher expression of Tks5 than primary melanoma cases. Taken together, these findings suggest the importance of Tks adaptor proteins in melanoma growth and metastasis in vivo, likely via functional invadopodia formation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Movimento Celular , Proliferação de Células , Neoplasias Pulmonares/metabolismo , Melanoma/metabolismo , Fosfoproteínas/metabolismo , Podossomos/metabolismo , Neoplasias Cutâneas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundário , Melanoma/genética , Melanoma/secundário , Camundongos , Camundongos Nus , Invasividade Neoplásica , Proteínas de Ligação a Fosfato , Fosfoproteínas/genética , Podossomos/patologia , Interferência de RNA , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Fatores de Tempo , Transfecção , Carga Tumoral
13.
PLoS One ; 10(3): e0121003, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25826475

RESUMO

The ability of cancer cells to invade underlies metastatic progression. One mechanism by which cancer cells can become invasive is through the formation of structures called invadopodia, which are dynamic, actin-rich membrane protrusions that are sites of focal extracellular matrix degradation. While there is a growing consensus that invadopodia are instrumental in tumor metastasis, less is known about whether they are involved in tumor growth, particularly in vivo. The adaptor protein Tks5 is an obligate component of invadopodia, and is linked molecularly to both actin-remodeling proteins and pericellular proteases. Tks5 appears to localize exclusively to invadopodia in cancer cells, and in vitro studies have demonstrated its critical requirement for the invasive nature of these cells, making it an ideal surrogate to investigate the role of invadopodia in vivo. In this study, we examined how Tks5 contributes to human breast cancer progression. We used immunohistochemistry and RNA sequencing data to evaluate Tks5 expression in clinical samples, and we characterized the role of Tks5 in breast cancer progression using RNA interference and orthotopic implantation in SCID-Beige mice. We found that Tks5 is expressed to high levels in approximately 50% of primary invasive breast cancers. Furthermore, high expression was correlated with poor outcome, particularly in those patients with late relapse of stage I/II disease. Knockdown of Tks5 expression in breast cancer cells resulted in decreased growth, both in 3D in vitro cultures and in vivo. Moreover, our data also suggest that Tks5 is important for the integrity and permeability of the tumor vasculature. Together, this work establishes an important role for Tks5 in tumor growth in vivo, and suggests that invadopodia may play broad roles in tumor progression.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Neoplasias da Mama/patologia , Divisão Celular/fisiologia , Animais , Xenoenxertos , Humanos , Técnicas In Vitro , Camundongos , Camundongos SCID
14.
Cancers (Basel) ; 6(1): 396-415, 2014 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-24531055

RESUMO

Extracellular matrix degradation by matrix metalloproteinases (MMPs) plays a pivotal role in cancer progression by promoting motility, invasion and angiogenesis. Studies have shown that MMP expression is increased in head and neck squamous cell carcinomas (HNSCCs), one of the most common cancers in the world, and contributes to poor outcome. In this review, we examine the expression pattern of MMPs in HNSCC by microarray datasets and summarize the current knowledge of MMPs, specifically MMP-1, -3, -7 -10, -12, -13, 14 and -19, that are highly expressed in HNSCCs and involved cancer invasion and angiogenesis.

15.
J Cell Biol ; 201(2): 279-92, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23589494

RESUMO

Notch regulates cell-cell contact-dependent signaling and is activated by hypoxia, a microenvironmental condition that promotes cellular invasion during both normal physiology and disease. The mechanisms by which hypoxia and Notch regulate cellular invasion are not fully elucidated. In this paper, we show that, in cancer cells, hypoxia increased the levels and activity of the ADAM12 metalloprotease in a Notch signaling-dependent manner, leading to increased ectodomain shedding of the epidermal growth factor (EGF) receptor (EGFR) ligand heparin-binding EGF-like growth factor. Released HB-EGF induced the formation of invadopodia, cellular structures that aid cancer cell invasion. Thus, we describe a signaling pathway that couples cell contact-dependent signaling with the paracrine activation of the EGFR, indicating cross talk between the Notch and EGFR pathways in promoting cancer cell invasion. This signaling pathway might regulate the coordinated acquisition of invasiveness by neighboring cells and mediate the communication between normoxic and hypoxic areas of tumors to facilitate cancer cell invasion.


Assuntos
Proteínas ADAM/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Pseudópodes/metabolismo , Receptores Notch/metabolismo , Proteínas ADAM/genética , Proteína ADAM12 , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/genética , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Epitélio/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/química , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Proteínas de Membrana/genética , Modelos Biológicos , Estrutura Terciária de Proteína , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
16.
PLoS One ; 7(8): e44488, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22952986

RESUMO

BACKGROUND: Metastasis to regional lymph nodes via lymphatic vessels plays a key role in cancer progression. Tumor lymphangiogenesis is known to promote lymphatic metastasis, and vascular endothelial growth factor C (VEGF-C) is a critical activator of tumor lymphangiogenesis during the process of metastasis. We previously identified periostin as an invasion- and angiogenesis-promoting factor in head and neck squamous cell carcinoma (HNSCC). In this study, we discovered a novel role for periostin in tumor lymphangiogenesis. METHODS AND FINDINGS: Periostin overexpression upregulated VEGF-C mRNA expression in HNSCC cells. By using conditioned media from periostin-overexpressing HNSCC cells, we examined tube formation of lymphatic endothelial cells. Conditioned media from periostin-overexpressing cells promoted tube formation. To know the correlation between periostin and VEGF-C, we compared Periostin expression with VEGF-C expression in 54 HNSCC cases by immunohistochemistry. Periostin expression was correlated well with VEGF-C expression in HNSCC cases. Moreover, correlation between periostin and VEGF-C secretion was observed in serum from HNSCC patients. Interestingly, periostin itself promoted tube formation of lymphatic endothelial cells independently of VEGF-C. Periostin-promoted lymphangiogenesis was mediated by Src and Akt activity. Indeed possible correlation between periostin and lymphatic status in periostin-overexpressing xenograft tumors and HNSCC cases was observed. CONCLUSIONS: Our findings suggest that periostin itself as well as periostin-induced upregulation of VEGF-C may promote lymphangiogenesis. We suggest that periostin may be a marker for prediction of malignant behaviors in HNSCC and a potential target for future therapeutic intervention to obstruct tumoral lymphatic invasion and lymphangiogenesis in HNSCC patients.


Assuntos
Moléculas de Adesão Celular/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Linfangiogênese , Moléculas de Adesão Celular/genética , Movimento Celular , Proliferação de Células , Células Endoteliais/patologia , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/irrigação sanguínea , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Linfangiogênese/genética , Modelos Biológicos , Neovascularização Patológica
17.
Mol Cell Biol ; 31(4): 783-92, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21149578

RESUMO

Ameloblastin, the most abundant nonamelogenin enamel matrix protein, plays a role in ameloblast differentiation. Here, we found that ameloblastin was expressed in osteosarcoma cells; to explore the potential functions of ameloblastin in osteoblasts, we investigated whether this protein is involved in osteogenic differentiation and bone formation on the premise that CD63, a member of the transmembrane-4 glycoprotein superfamily, interacts with integrins in the presence of ameloblastin. Ameloblastin bound to CD63 and promoted CD63 binding to integrin ß1. The interaction between CD63 and integrin ß1 induced Src kinase inactivation via the binding of CD63 to Src. The reduction of Src activity and osteogenic differentiation mediated by ameloblastin were abrogated by treatment with anti-CD63 antibody and overexpression of constitutively active Src, respectively. Therefore, our results suggest that ameloblastin is expressed in osteoblasts and functions as a promoting factor for osteogenic differentiation via a novel pathway through the interaction between CD63 and integrin ß1.


Assuntos
Antígenos CD/metabolismo , Proteínas do Esmalte Dentário/metabolismo , Integrina beta1/metabolismo , Osteogênese/fisiologia , Glicoproteínas da Membrana de Plaquetas/metabolismo , Quinases da Família src/antagonistas & inibidores , Animais , Antígenos CD/genética , Sequência de Bases , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Primers do DNA/genética , Proteínas do Esmalte Dentário/genética , Humanos , Minerais/metabolismo , Modelos Biológicos , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteossarcoma/genética , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Glicoproteínas da Membrana de Plaquetas/genética , RNA Interferente Pequeno/genética , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Tetraspanina 30 , Quinases da Família src/metabolismo
18.
PLoS One ; 6(10): e25438, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21998657

RESUMO

BACKGROUND: Periostin, IFN-induced transmembrane protein 1 (IFITM1) and Wingless-type MMTV integration site family, member 5B (Wnt-5b) were previously identified as the invasion promoted genes of head and neck squamous cell carcinoma (HNSCC) by comparing the gene expression profiles between parent and a highly invasive clone. We have previously reported that Periostin and IFITM1 promoted the invasion of HNSCC cells. Here we demonstrated that Wnt-5b overexpression promoted the invasion of HNSCC cells. Moreover, stromelysin-2 (matrix metalloproteinase-10; MMP-10) was identified as a common up-regulated gene among Periostin, IFITM1 and Wnt-5b overexpressing HNSCC cells by using microarray data sets. In this study, we investigated the roles of MMP-10 in the invasion of HNSCC. METHODS AND FINDINGS: We examined the expression of MMP-10 in HNSCC cases by immunohistochemistry. High expression of MMP-10 was frequently observed and was significantly correlated with the invasiveness and metastasis in HNSCC cases. Next, we examined the roles of MMP-10 in the invasion of HNSCC cells in vitro. Ectopic overexpression of MMP-10 promoted the invasion of HNSCC cells, and knockdown of MMP-10 suppressed the invasion of HNSCC cells. Moreover, MMP-10 knockdown suppressed Periostin and Wnt-5b-promoted invasion. Interestingly, MMP-10 overexpression induced the decreased p38 activity and MMP-10 knockdown induced the increased p38 activity. In addition, treatment with a p38 inhibitor SB203580 in HNSCC cells inhibited the invasion. CONCLUSIONS: These results suggest that MMP-10 plays an important role in the invasion and metastasis of HNSCC, and that invasion driven by MMP-10 is partially associated with p38 MAPK inhibition. We suggest that MMP-10 can be used as a marker for prediction of metastasis in HNSCC.


Assuntos
Carcinoma de Células Escamosas/patologia , Neoplasias de Cabeça e Pescoço/patologia , Metaloproteinase 10 da Matriz/metabolismo , Antígenos de Diferenciação/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Moléculas de Adesão Celular/genética , Linhagem Celular Tumoral , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Metaloproteinase 10 da Matriz/deficiência , Metaloproteinase 10 da Matriz/genética , Invasividade Neoplásica , Metástase Neoplásica , Inibidores de Proteínas Quinases/farmacologia , Regulação para Cima/efeitos dos fármacos , Proteínas Wnt/genética , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
19.
Plast Reconstr Surg ; 124(2): 540-550, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19644272

RESUMO

BACKGROUND: The concept of deep tissue injury under intact skin helps us understand the pathogenesis of pressure ulcers, but the best method for detecting and evaluating deep tissue injury remains to be established. METHODS: Intermediate-frequency (10-MHz) ultrasonography was performed to evaluate deep tissue injury. The authors analyzed 12 patients (nine male patients and three female patients aged 16 to 92 years) who showed deep tissue injury-related abnormal findings on ultrasonography at the first examination and were followed up until the pressure ulcer reached a final stage. RESULTS: The stage of ulcer worsened in six of 12 cases compared with baseline, and healed in the remaining six patients. The authors recognized four types of abnormal signs unique to deep tissue damage in ultrasonography: unclear layered structure, hypoechoic lesion, discontinuous fascia, and heterogeneous hypoechoic area. Unclear layered structure, hypoechoic lesion, discontinuous fascia, and heterogeneous hypoechoic area were detected at the first examination in 12, 10, seven, and five patients, respectively. Unclear layered structure and hypoechoic lesion were more commonly seen in pressure ulcers in deep tissue injury than the other features, but the follow-up study suggested that discontinuous fascia and heterogeneous hypoechoic area are more reliable predictors of future progression of pressure ulcers. CONCLUSIONS: The use of intermediate-frequency ultrasound reliably identified deep tissue injury and was believed to contribute to prevention and treatment of pressure-related ulcers. The results suggest that specific ultrasonographic characteristics may predict which pressure ulcers will progress.


Assuntos
Úlcera por Pressão/diagnóstico por imagem , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Progressão da Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Úlcera por Pressão/patologia , Prognóstico , Sensibilidade e Especificidade , Ultrassonografia , Adulto Jovem
20.
PLoS One ; 4(6): e5892, 2009 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-19521519

RESUMO

BACKGROUND: Runt-related transcription factor 3 (RUNX3) is a tumor suppressor of cancer and appears to be an important component of the transforming growth factor-beta (TGF-ss)-induced tumor suppression pathway. Surprisingly, we found that RUNX3 expression level in head and neck squamous cell carcinoma (HNSCC) tissues, which is one of the most common types of human cancer, was higher than that in normal tissues by a previously published microarray dataset in our preliminary study. Therefore, here we examined the oncogenic role of RUNX3 in HNSCC. PRINCIPAL FINDINGS: Frequent RUNX3 expression and its correlation with malignant behavior were observed in HNSCC. Ectopic RUNX3 overexpression promoted cell growth and inhibited serum starvation-induced apoptosis and chemotherapeutic drug induced apoptosis in HNSCC cells. These findings were confirmed by RUNX3 knockdown. Moreover, RUNX3 overexpression enhanced tumorsphere formation. RUNX3 expression level was well correlated with the methylation status in HNSCC cells. Moreover, RUNX3 expression was low due to the methylation of its promoter in normal oral epithelial cells. CONCLUSIONS/SIGNIFICANCE: Our findings suggest that i) RUNX3 has an oncogenic role in HNSCC, ii) RUNX3 expression observed in HNSCC may be caused in part by demethylation during cancer development, and iii) RUNX3 expression can be a useful marker for predicting malignant behavior and the effect of chemotherapeutic drugs in HNSCC.


Assuntos
Subunidade alfa 3 de Fator de Ligação ao Core/biossíntese , Subunidade alfa 3 de Fator de Ligação ao Core/fisiologia , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/metabolismo , Apoptose , Biomarcadores Tumorais , Carcinoma de Células Escamosas , Linhagem Celular Tumoral , Metilação de DNA , Células Epiteliais/metabolismo , Genes Supressores de Tumor , Humanos , Neoplasias Bucais/metabolismo , Neoplasias/metabolismo , RNA Interferente Pequeno/metabolismo , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA