Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomed Res ; 35(6): 381-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25743344

RESUMO

CXCL14/BRAK (BRAK) is a secreted chemokine with anti-tumor activity, and its expression is suppressed in tumor cells. We previously reported the anti-tumor activity of BRAK in cell lines of head and neck squamous cell carcinoma (HNSCC) and the suppression of BRAK secretion in these cells. BRAK secretion in fibrosarcoma cells is restored by Fasudil, which is a Rho-kinase (ROCK) inhibitor. In this study, we examined the anti-tumor effect of BRAK by evaluating its gene expression and protein secretion in HNSCC cell lines. We found that BRAK mediated the suppressive effect of Fasudil against HNSCC cells. Tumor development in female BALB/cAJclnu/nu mice was suppressed by Fasudil. Also secretion of BRAK protein by tumor cell lines in vitro was significantly stimulated by Fasudil treatment. Similarly, the production of BRAK protein was significantly increased by the addition of Fasudil to cultured tumor cells. Furthermore Fasudil significantly increased BRAK gene expression at the mRNA level in HNSCC cell line. Inhibition of the RhoA/ROCK pathway by siRNAs significantly stimulated BRAK gene expression. These results show that the tumor-suppressive effect of Fasudil was mediated by BRAK, suggesting that Fasudil may therefore be useful for the treatment of HNSCC.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , Carcinoma de Células Escamosas/patologia , Quimiocinas CXC/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias de Cabeça e Pescoço/patologia , Inibidores de Proteínas Quinases/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Feminino , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Carcinoma de Células Escamosas de Cabeça e Pescoço
2.
J Pharmacol Sci ; 120(3): 241-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23099322

RESUMO

We previously reported that chemokine CXCL14/BRAK (BRAK) has antitumor activity in several carcinoma cells indicating that BRAK secretion suppresses carcinoma cells. Ras-homologous small GTPase (RhoA) and Rho-associated coiled-coil-containing protein kinase (ROCK) are important regulators of secretory processes, and activation of the RhoA/ROCK signaling pathway stimulates tumor invasion and metastasis. We investigated the effects of fasudil, a specific ROCK inhibitor, on BRAK secretion and tumor progression in mesenchymal fibrosarcoma cells (MC57). We demonstrated the antitumor activity of secreted BRAK using MC57 transplantation of BRAK in overexpressed transgenic mice. Further, to eliminate the influence of change in the mRNA expression of endogenous BRAK, we produced stable MC57 cell lines expressing BRAK (MC57-BRAK) or mock vector (MC57-MOCK). Fasudil significantly increased BRAK secretion by MC57-BRAK cells in a dose-dependent manner. To determine the effect of fasudil on tumor growth, MC57-BRAK and MC57-MOCK cells were transplanted into wild-type mice. Fasudil treatment suppressed tumor growth only in mice that had received MC57-BRAK cell transplants. These results indicate that fasudil inhibits fibrosarcoma growth by stimulating BRAK secretion and suggests that fasudil therapy might have clinical efficacy.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , Antineoplásicos/uso terapêutico , Quimiocinas CXC/metabolismo , Fibrossarcoma/tratamento farmacológico , Proteínas de Neoplasias/metabolismo , Inibidores de Proteínas Quinases/uso terapêutico , Quinases Associadas a rho/antagonistas & inibidores , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/uso terapêutico , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quimiocinas CXC/genética , Fibrossarcoma/metabolismo , Fibrossarcoma/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Inibidores de Proteínas Quinases/farmacologia , Proteínas Recombinantes/metabolismo , Transdução de Sinais/efeitos dos fármacos , Carga Tumoral/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/metabolismo
3.
Biochem Biophys Res Commun ; 420(2): 217-22, 2012 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-22382027

RESUMO

The chemokine BRAK/CXCL14 (BRAK) is expressed in normal squamous epithelium, but is not expressed or is expressed at negligible levels in head and neck squamous cell carcinoma. Malignant cells are known to be dedifferentiated compared with normal epithelial cells, suggesting a role for differentiation cues in the expression of BRAK. Thus, we examined the relationship between BRAK expression and stages of differentiation level in epithelial cells. Immunohistochemical analysis showed that BRAK protein was expressed in cells above the spinous cell layer in normal epithelia. In HSC-3 cells in culture, expression of BRAK mRNA was significantly upregulated by cell contact in a cell density-dependent manner, and mRNA expression of cell differentiation markers such as involucrin, cystatin-A, TGM1, TGM3, and TGM5 was concomitantly augmented. Furthermore, the upregulation of BRAK induced by cell contact was suppressed by chlorpromazine, a specific inhibitor of calmodulin. We previously reported that GC boxes and a TATA-like sequence in the BRAK promoter region are associated with the expression of BRAK. Using a promoter assay and ChIP, we demonstrated that binding of the stimulating protein-1 (SP1) transcription factor to a GC box upstream of the BRAK transcription start site was necessary for cell density-dependent upregulation of BRAK. These results indicated that upregulation of BRAK was accompanied by differentiation of epithelial cells induced by calcium/calmodulin signaling, and that SP1 binding to the BRAK promoter region played an important role in this signaling.


Assuntos
Cálcio/metabolismo , Calmodulina/metabolismo , Diferenciação Celular/genética , Quimiocinas CXC/genética , Células Epiteliais/citologia , Regulação da Expressão Gênica , Fator de Transcrição Sp1/metabolismo , Calmodulina/antagonistas & inibidores , Comunicação Celular/genética , Contagem de Células , Clorpromazina/farmacologia , Cistatina A/metabolismo , Células Epiteliais/metabolismo , Humanos , Regiões Promotoras Genéticas , Transdução de Sinais , Transglutaminases/metabolismo , Células Tumorais Cultivadas , Regulação para Cima
4.
Anticancer Res ; 31(1): 105-11, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21273587

RESUMO

BACKGROUND: NK4 inhibits vascularisation in tumour tissues, thereby arresting tumour growth. However, the antitumour efficacy of individual antiangiogenic molecules expressed in vivo is not sufficiently potent to induce regression in animal models. One of the strategies to overcome this disadvantage is to use chemotherapy. MATERIALS AND METHODS: This study evaluated the efficacy of combining NK4 gene therapy with cisplatin to treat experimental squamous cell carcinomas. For gene therapy, biodegradable cationised gelatin microspheres were used for the controlled release of NK4 plasmid DNA. RESULTS: A combined regimen of antiangiogenic gene therapy and low-dose cisplatin led to a marked decrease in tumour volume and vascularity, and caused increased apoptosis compared to NK4 gene therapy alone. Moreover, combination treatment of NK4 gene therapy and low-dose cisplatin dramatically inhibited the formation of lung metastases. CONCLUSION: NK4 gene therapy combined with low-dose cisplatin may be an effective regimen for treating oral squamous cell carcinoma.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma de Células Escamosas/secundário , Cisplatino/uso terapêutico , Terapia Genética , Fator de Crescimento de Hepatócito/genética , Neoplasias Pulmonares/secundário , Neoplasias Cutâneas/patologia , Animais , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/terapia , Terapia Combinada , Ensaio de Imunoadsorção Enzimática , Humanos , Técnicas Imunoenzimáticas , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/terapia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Nus , Neovascularização Patológica/prevenção & controle , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/terapia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Free Radic Res ; 44(8): 913-24, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20815772

RESUMO

The present study investigated the effects of oxidative stress induced by reactive oxygen species (ROS), such as hydrogen peroxide (H(2)O(2)) and hydroxyl radical (HO(*)), on the expression of both BRAK , which is also known as non-ELR motif angiostatic CXC chemokine ligand 14 (CXCL14), in head and neck squamous cell carcinoma (HNSCC) cells. When HNSCC cells were cultured in the presence of ROS, the expression of BRAK was significantly decreased whereas that of IL-8 was increased. Interestingly, the effects on the expression of both genes in HNSCC cells were much greater with HO(blacksquare, square, filled) than with H(2)O(2). The effects of ROS on both BRAK and IL-8 expression were attenuated by pre-treatment with N-acetyl-L-cysteine (NAC), epidermal growth factor receptor (EGFR), and mitogen-activated protein kinase (MAPK) inhibitors. These results indicate that oxidative stress induced by H(2)O(2) or HO(*) stimulates angiogenesis and tumuor progression by altering the gene expression of BRAK and IL-8 via the EGFR/MEK/ERK pathway in human HNSCC cells.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Quimiocinas CXC/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Sobrevivência Celular/efeitos dos fármacos , Quimiocinas CXC/metabolismo , Compostos Ferrosos/farmacologia , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Radical Hidroxila/análise , Radical Hidroxila/metabolismo , Interleucina-8/biossíntese , Estresse Oxidativo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Células Tumorais Cultivadas
6.
Biochem Biophys Res Commun ; 396(4): 1060-4, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20478268

RESUMO

The mitogen-activated protein kinase (MAPK) family comprises ERK, JNK, p38 and ERK5 (big-MAPK, BMK1). UV irradiation of squamous cell carcinoma cells induced up-regulation of gene expression of chemokine BRAK/CXCL14, stimulated p38 phosphorylation, and down-regulated the phosphorylation of ERK. Human p38 MAPKs exist in 4 isoforms: p38 alpha, beta, gamma and delta. The UV stimulation of p38 phosphorylation was not inhibited by the presence of SB203580 or PD169316, inhibitors of p38 alpha and beta, suggesting p38 phosphorylation was not dependent on these 2 isoforms and that p38 gamma and/or delta was responsible for the phosphorylation. In fact, inhibition of each of these 4 p38 isoforms by the introduction of short hairpin (sh) RNAs for respective isoforms revealed that only shRNA for p38 delta attenuated the UV-induced up-regulation of BRAK/CXCL14 gene expression. In addition, over-expression of p38 isoforms in the cells showed the association of p38 delta with ERK1 and 2, concomitant with down-regulation of ERK phosphorylation. The usage of p38 delta isoform by UV irradiation is not merely due to the abundance of this p38 isoform in the cells. Because serum deprivation of the cells also induced an increase in BRAK/CXCL14 gene expression, and in this case p38 alpha and/or beta isoform is responsible for up-regulation of BRAK/CXCL14 gene expression. Taken together, the data indicate that the respective stress-dependent action of p38 isoforms is responsible for the up-regulation of the gene expression of the chemokine BRAK/CXCL14.


Assuntos
Quimiocinas CXC/genética , Regulação da Expressão Gênica , Expressão Gênica/efeitos da radiação , Proteína Quinase 13 Ativada por Mitógeno/metabolismo , Raios Ultravioleta , Linhagem Celular Tumoral , Humanos , Proteína Quinase 13 Ativada por Mitógeno/genética , Regulação para Cima
7.
Transgenic Res ; 19(6): 1109-17, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20333465

RESUMO

We reported previously that the forced expression of the chemokine BRAK, also called CXCL14 in head and neck squamous cell carcinoma (HNSCC) cells decreased the rate of tumor formation and size of tumor xenografts compared with mock-vector treated cells in athymic nude mice or in severe combined immunodeficiency mice. This suppression occurred even though the growth rates of these cells were the same under in vitro culture conditions, suggesting that a high expression level of the gene in tumor cells is important for the suppression of tumor establishment in vivo. The aim of this study was to determine whether CXCL14/BRAK transgenic mice show resistance to tumor cell xenografts or not. CXCL14/BRAK cDNA was introduced into male C57BL/6 J pronuclei, and 10 founder transgenic mice (Tg) were obtained. Two lines of mice expressed over 10 times higher CXCL14/BRAK protein levels (14 and 11 ng/ml plasma, respectively) than normal blood level (0.9 ng/ml plasma), without apparent abnormality. The sizes of Lewis lung carcinoma and B16 melanoma cell xenografts in Tg mice were significantly smaller than those in control wild-type mice, indicating that CXCL14/BRAK, first found as a suppressor of tumor progression of HNSCC, also suppresses the progression of a carcinoma of other tissue origin. Immunohistochemical studies showed that invasion of blood vessels into tumors was suppressed in tumor xenografts of CXCL14/BRAK Tg mice. These results indicate that CXCL14/BRAK suppressed tumor cell xenografts by functioning paracrine or endocrine fashion and that CXCL14/BRAK is a very promising molecular target for tumor suppression without side effects.


Assuntos
Quimiocinas CXC/genética , Quimiocinas CXC/imunologia , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Animais , Sequência de Bases , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/terapia , Linhagem Celular Tumoral , Primers do DNA/genética , Feminino , Expressão Gênica , Humanos , Rim/imunologia , Masculino , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transplante de Neoplasias , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/genética , RNA Mensageiro/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Transplante Heterólogo
8.
Biomed Res ; 30(5): 271-7, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19887723

RESUMO

Oral mucosal tissue can serve as a long-term fluoride reservoir following topical application and retain a small amount of fluoride in oral environment for prevention of dental caries. The aim of this study was to determine the effect of low level sodium fluoride (NaF) on the proliferation and migration of epithelial cells in vitro. Human primary gingival epithelial cells and human epidermal HaCaT keratinocytes were used. Cultured epithelial cells, treated with various concentrations of NaF ranging from 5 microM to 500 microM, were investigated by 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt (MTS) assay, wound healing assay, invasion assay and quantitative real-time PCR. MTS assay revealed that fluoride added to human gingival epithelial cells elevated cell proliferation at a concentration of 5 microM or more. The wound healing assay and invasion assay confirmed this observation. Quantitative real-time PCR revealed that low concentration of NaF up-regulated fibronectin mRNA expression in fluoride-treated cells compared with controls. These results suggest that a low concentration of NaF is able to induce cell proliferation, migration, and matrix production in epithelial cells. Our results provide new information on epithelial cell adhesion and may thus aid in the understanding of periodontal physiology.


Assuntos
Movimento Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Fluoreto de Sódio/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/citologia , Gengiva/citologia , Humanos
9.
Biomed Res ; 30(5): 315-8, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19887729

RESUMO

SCID mice are a model of human severe combined immunodeficiency disease and are deficient in B cell function in addition to T cell function. Tumors from other species are easily transplanted into SCID mice and will grow without being rejected. We previously reported that the chemokine BRAK/CXCL14 is expressed in normal cells but its expression is down regulated in an in vitro cancer progression model, suggesting that it has the potential for antitumor activity. Here we report that the growth of BRAK/CXCL14 expression vector-transfected oral cancer cells was completely (100%) suppressed in SCID mouse xenografts even though mock-vector introduced control tumor cells grew well with 100% of animals developing tumors. In addition, suppression of xenografts was much faster and the rate was much higher in SCID mice than in T cell function-deficient nude mice. These data indicate the possibility that BRAK expression inhibits tumor cell establishment by regulating interactions between tumor stem cells and NK cells and/or suppressing formation of tumor microvessels.


Assuntos
Quimiocinas CXC/imunologia , Camundongos SCID , Neoplasias Bucais/imunologia , Transplante Heterólogo/imunologia , Animais , Quimiocinas CXC/genética , Feminino , Humanos , Camundongos , Camundongos Nus/imunologia , Camundongos SCID/imunologia , Neoplasias Bucais/patologia , Transplante de Neoplasias , Transplante Heterólogo/patologia
10.
Cancer Sci ; 100(11): 2202-9, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19673887

RESUMO

Clinical efficacy of gefitinib (ZD1839, Iressa), which is an inhibitor specific for epidermal growth factor (EGF) receptor tyrosine kinase, has been shown in non-small-cell lung carcinoma patients with EGF receptor mutations, so these mutations are useful marker(s) to find a responder for the drug. Recent studies have shown that the EGF receptor gene mutation is rare in squamous cell carcinoma in the esophageal and head and neck regions. We previously reported that the expression of the chemokine BRAK/CXCL14 in head and neck squamous cell carcinoma (HNSCC) cells was down-regulated by EGF treatment, and that forced expression of BRAK in tumor cells decreased the tumorigenicity of the cells in xenografts. Thus, we investigated the relationship between restoration of BRAK expression by gefitinib and the efficacy of the drug for tumor suppression. We found that EGF down-regulated BRAK expression through the MEK-extracellular signal regulated kinase pathway and that this down-regulated expression was restored by gefitinib in vitro. Oral administration of gefitinib significantly (P < 0.001) reduced tumor growth of xenografts of three HNSCC cell lines (HSC-2, HSC-3, and HSC-4), in female athymic nude mice, accompanied by an increase in BRAK expression specifically in tumor tissue. This tumor-suppressing effect of the drug was not observed in the case of BRAK non-expressing cells. Furthermore introduction of BRAK shRNA vector reduced both the expression levels of BRAK in HSC-3 cells and the antitumor efficacy of gefitinib in vivo. Our data showing an inverse relationship between BRAK expression levels in tumor cells and the tumor growth rate indicate that the gefitinib-induced increase in BRAK expression is beneficial for tumor suppression in vivo.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Quimiocinas CXC/genética , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Quinazolinas/farmacologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/genética , Gefitinibe , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Mutação , RNA Mensageiro/análise
11.
J Biomater Appl ; 24(4): 327-42, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18987021

RESUMO

We examine the osteogenicity of a sponge biomaterial consisting of a biodegradable mixture of gelatin and beta-tricalcium phosphate (betaTCP) that bound bone morphogenetic protein 2 (BMP-2) in critical-sized bone defects in rats. Gelatin-betaTCP sponges containing either phosphate buffered saline or incorporating BMP-2 are implanted into 5 mm diameter bone defects created in rat mandibles. We assess the defects biweekly for 8 weeks following implantation. There is significantly higher osteoinductive activity and significantly more Gla-osteocalcin content at bone-defect healing sites treated with gelatin-betaTCP sponges incorporating BMP-2 than there is in those treated with sponges that did not contain BMP-2. Histologically, new bone that contains bone marrow and that is connected to the original bone almost entirely replaces the regenerated bone. These results show that biodegradable gelatin-betaTCP incorporating BMP-2 is osteogenic enough to promote healing in large bone defects.


Assuntos
Materiais Biocompatíveis/química , Proteína Morfogenética Óssea 2/metabolismo , Regeneração Óssea , Fosfatos de Cálcio/uso terapêutico , Gelatina/uso terapêutico , Implantes Absorvíveis , Animais , Substitutos Ósseos , Masculino , Microscopia Eletrônica de Varredura/métodos , Neuroglia/metabolismo , Osteocalcina/química , Osteogênese , Ratos , Ratos Wistar , Suínos
12.
FEBS J ; 274(12): 3171-83, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17540003

RESUMO

Acidic extracellular pH is a common feature of tumor tissues. We have reported that culturing cells at acidic pH (5.4-6.5) induced matrix metalloproteinase-9 expression through phospholipase D, extracellular signal regulated kinase 1/2 and p38 mitogen-activated protein kinases and nuclear factor-kappaB. Here, we show that acidic extracellular pH signaling involves both pathways of phospholipase D triggered by Ca2+ influx and acidic sphingomyelinase in mouse B16 melanoma cells. We found that BAPTA-AM [1,2-bis(2-aminophenoxy)-ethane-N,N,N',N'-tetraacetic acid tetrakis (acetoxymethyl) ester], a chelator of intracellular free calcium, and the voltage dependent Ca2+ channel blockers, mibefradil (for T-type) and nimodipine (for L-type), dose-dependently inhibited acidic extracellular pH-induced matrix metalloproteinase-9 expression. Intracellular free calcium concentration ([Ca2+]i) was transiently elevated by acidic extracellular pH, and this [Ca2+]i elevation was repressed by EGTA and the voltage dependent Ca2+ channel blockers but not by phospholipase C inhibitor, suggesting that acidic extracellular pH increased [Ca2+]i through voltage dependent Ca2+ channel. In contrast, SR33557, an L-type voltage dependent Ca2+ channel blocker and acidic sphingomyelinase inhibitor, attenuated matrix metalloproteinase-9 induction but did not affect calcium influx. We found that acidic sphingomyelinase activity was induced by acidic extracellular pH and that the specific acidic sphingomyelinase inhibitors (perhexiline and desipramine) and siRNA targeting aSMase/smpd1 could inhibit acidic extracellular pH-induced matrix metalloproteinase-9 expression. BAPTA-AM reduced acidic extracellular pH-induced phospholipase D but not acidic sphingomyelinase acitivity. The acidic sphingomyelinase inhibitors did not affect the phosphorylation of extracellular signal regulated kinase 1/2 and p38, but they suppressed nuclear factor-kappaB activity. These data suggest that the calcium influx-triggered phospholipase D and acidic sphingomyelinase pathways of acidic extracellular pH induced matrix metalloproteinase-9 expression, at least in part, through nuclear factor-kappaB activation.


Assuntos
Cálcio/metabolismo , Líquido Extracelular/química , Metaloproteinase 9 da Matriz/biossíntese , Melanoma/enzimologia , Fosfolipase D/metabolismo , Esfingomielina Fosfodiesterase/metabolismo , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/fisiologia , Linhagem Celular Tumoral , Quelantes/farmacologia , Desipramina/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Ativação Enzimática , Concentração de Íons de Hidrogênio , Indolizinas/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Metaloproteinase 9 da Matriz/genética , Melanoma/secundário , Mibefradil/farmacologia , Camundongos , NF-kappa B/metabolismo , Nimodipina/farmacologia , Perexilina/farmacologia , Fenetilaminas/farmacologia , Fosfolipase D/antagonistas & inibidores , Fosforilação , Regiões Promotoras Genéticas , Esfingomielina Fosfodiesterase/antagonistas & inibidores
13.
Biochem Biophys Res Commun ; 348(2): 406-12, 2006 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-16884687

RESUMO

In order to find a suppressor(s) of tumor progression in vivo for oral carcinoma (OC), we searched for molecules down-regulated in OC cells when the cells were treated with epidermal growth factor (EGF), whose receptor is frequently over-activated in OC. The expression of BRAK, which is also known as CXC chemokine ligand14 (CXCL14), was down-regulated significantly by the treatment of OC cells with EGF as observed by cDNA microarray analysis followed by reverse-transcriptase polymerase chain reaction analysis. The EGF effect was attenuated by the co-presence of a MEK inhibitor. The rate of tumor formation in vivo of BRAK-expressing vector-transfected tumor cells in athymic nude mice was significantly lower than that of mock vector-transfected ones. In addition tumors formed in vivo by the BRAK-expressing cells were significantly smaller than those of the mock-transfected ones. These results indicate that BRAK/CXCL14 is a chemokine, having suppressive activity toward tumor progression of OC in vivo.


Assuntos
Carcinoma de Células Escamosas/fisiopatologia , Quimiocinas CXC/fisiologia , Neoplasias da Língua/fisiopatologia , Animais , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Quimiocinas CXC/uso terapêutico , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Camundongos , Transplante de Neoplasias , Transdução de Sinais , Neoplasias da Língua/tratamento farmacológico , Neoplasias da Língua/patologia , Transfecção , Proteínas Supressoras de Tumor/fisiologia
14.
Pathol Int ; 56(8): 457-61, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16872441

RESUMO

Inverted ductal papilloma (IDP) is a type of ductal papilloma arising in ducts of minor salivary glands. Very few cases, and no cases in Japan, have been reported. Reported herein is a case of IDP with a review of the literature. The patient was a 49-year-old man presenting with a lump in the right buccal mucosa of the premolar area of the mandible. The tumor was excised en bloc after a biopsy diagnosis of IDP. On the surface of the covering epithelium, an opening was seen to be filled with mucinous material. On cut surface the opening led to the tumor cavity. The major portion of the tumor parenchyma was made up of papillary proliferation of basaloid squamous cells. Some crypts, microcysts, and mucous cells were seen. There were no findings suggestive of a malignant tumor. The patient's postoperative course was uneventful and there has been no recurrence after 1 year's follow up. Immunohistochemical analysis of the present case supports the hypothesis that IDP originates from squamous metaplasia and proliferation of minor salivary gland duct cells.


Assuntos
Papiloma Invertido/patologia , Ductos Salivares/patologia , Neoplasias das Glândulas Salivares/patologia , Glândulas Salivares Menores/patologia , Biomarcadores Tumorais/análise , Humanos , Queratinas/análise , Masculino , Pessoa de Meia-Idade , Mucosa Bucal/química , Mucosa Bucal/patologia , Mucina-1/análise , Papiloma Invertido/química , Papiloma Invertido/cirurgia , Ductos Salivares/química , Ductos Salivares/cirurgia , Neoplasias das Glândulas Salivares/química , Neoplasias das Glândulas Salivares/cirurgia , Glândulas Salivares Menores/química , Glândulas Salivares Menores/cirurgia
15.
Redox Rep ; 11(2): 71-7, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16686997

RESUMO

Oral manifestations of Down syndrome include high susceptibility to gingival inflammation with early onset and rapidly progressive periodontitis. The influence of reactive oxygen species (ROS) on periodontitis of Down syndrome is unclear. The aim of this study was to characterize ROS formation in Down syndrome-gingival fibroblasts (DS-GF) using electron spin resonance (ESR) spin trapping with 5,5-dimetyl-1-pyrolline-N-oxide (DMPO), and to determine whether ROS generation plays a role in the pathogenesis of periodontitis in Down syndrome patients. We observed formation of the DMPO-OH spin adduct, indicating HO* generation from cultured DS-GF and non-DS-GF. The increased HO* generation in cultured DS-GF was strongly decreased in the presence of the H2O2 scavenger, catalase, or the iron chelator, desferal. This may due to the enzymatic ability of over-expressed CuZn-superoxide dismutase in Down syndrome to catalyze the formation of H2O2 from O2*-, thereby increasing the availability of substrate H2O2 for the iron-dependent generation of HO* via the Fenton reaction, suggesting that HO* generated from DS-GF may be involved in progressive periodontitis of Down syndrome.


Assuntos
Síndrome de Down/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Fibroblastos/metabolismo , Gengiva/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Adulto , Catalase/metabolismo , Catalase/farmacologia , Técnicas de Cultura de Células , Desferroxamina/farmacologia , Síndrome de Down/patologia , Feminino , Fibroblastos/citologia , Gengiva/citologia , Humanos , Peróxido de Hidrogênio/metabolismo , Radical Hidroxila/metabolismo , Masculino , Modelos Químicos , Oxirredução/efeitos dos fármacos , Estresse Oxidativo , Periodontite/metabolismo , Periodontite/patologia , Superóxido Dismutase/metabolismo , Superóxidos/metabolismo
16.
Cancer Sci ; 97(4): 313-21, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16630124

RESUMO

Double-stranded RNA (dsRNA) plays a major role in RNA interference (RNAi), a process in which segments of dsRNA are initially cleaved by the Dicer into shorter segments (21-23 nt) called small interfering RNA (siRNA). These siRNA then specifically target homologous mRNA molecules causing them to be degraded by cellular ribonucleases. RNAi down regulates endogenous gene expression in mammalian cells. Vascular endothelial growth factor (VEGF) is a key molecule in vasculogenesis as well as in angiogenesis. Tumor growth is an angiogenesis-dependent process, and therapeutic strategies aimed at inhibiting angiogenesis are theoretically attractive. To investigate the feasibility of using siRNA for VEGF in the specific knockdown of VEGF mRNA, thereby inhibiting angiogenesis, we have performed experiments with a DNA vector based on a siRNA system that targets VEGF (siVEGF). It almost completely inhibited the expression of three different isoforms (VEGF120, VEGF164 and VEGF188) of VEGF mRNA and the secretion of VEGF protein in mouse squamous cell carcinoma NRS-1 cells. The siVEGF released from cationized gelatin microspheres suppressed tumor growth in vivo. A marked reduction in vascularity accompanied the inhibition of a siVEGF-transfected tumor. Fluorescent microscopic study showed that the complex of siVEGF with cationized gelatin microspheres was still present around the tumor 10 days after injection, while free siVEGF had vanished by that time. siVEGF gene therapy increased the fraction of vessels covered by pericytes and induced expression of angiopoietin-1 by pericytes. These data suggest that cationized-gelatin microspheres containing siVEGF can be used to normalize tumor vasculature and inhibit tumor growth in a NRS-1 squamous cell carcinoma xenograft model.


Assuntos
Carcinoma de Células Escamosas/terapia , Cátions , DNA/administração & dosagem , Gelatina/administração & dosagem , Terapia Genética , RNA Interferente Pequeno/farmacologia , Neoplasias Cutâneas/terapia , Fator A de Crescimento do Endotélio Vascular/genética , Angiopoietina-1/metabolismo , Animais , Carcinoma de Células Escamosas/genética , Sistemas de Liberação de Medicamentos , Camundongos , Camundongos Endogâmicos C3H , Microesferas , Neovascularização Patológica , Plasmídeos , Reação em Cadeia da Polimerase , Neoplasias Cutâneas/genética , Transfecção , Células Tumorais Cultivadas/transplante , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Matrix Biol ; 25(1): 47-58, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16266799

RESUMO

In order to investigate the mechanisms by which 1alpha,25(OH)2 vitamin D3 (VD3) stimulates the differentiation of human osteoblasts, we cultured MG-63, which is a human osteoblastic cell line, in the presence or absence of VD3 and/or L-ascorbic acid 2-phosphate (Asc 2-P), a long-acting vitamin C derivative. The cell growth rate was decreased by the presence of VD3 in the culture medium. Type I collagen synthesis and alkaline phosphatase (ALP) activity, which are markers of early stage osteoblast differentiation, were stimulated by the presence of VD3 as well as by that of Asc 2-P. The co-presence of Asc 2-P and VD3 had a synergistic effect on the collagen synthesis and ALP activity of the cells. Inhibition of collagen synthesis by the addition of inhibitors of collagen synthesis to the medium attenuated the stimulative effect of VD3 and Asc 2-P on the ALP activity. Transfection of the cells with siRNA-expressing vectors for COL1A1 decreased the expression level of ALP mRNA in addition to that of COL1A1. On the other hand, ALP activity was significantly increased, and the growth rate was decreased, when the cells were cultured on type I collagen-coated dishes. These effects were not seen when the cells were cultured on dishes coated with heat-denatured collagen. VD3 also increased the mRNA levels for Runx2 and osterix, which are transcription factors critical for osteoblast differentiation, as well as those of differentiation markers such as bone/liver/kidney type ALP, COL1A1, (the gene for the alpha1 chain of type I collagen), and osteocalcin, in the cells. Normal human osteoblasts and human bone marrow-derived mesenchymal stem cells (hBMSC) showed quite similar responses to VD3. These results indicate that VD3-stimulated gene expression of type I collagen and that mature type I collagen produced in the presence of Asc 2-P mediates at least a part of the stimulative effects of Asc 2-P and VD3 on the differentiation of these human osteoblastic cells. Levels of mRNAs for ALP and COL1A1 were increased, but the level of Runx2 was decreased, by the expression of osterix in MG-63 cells. These results also suggest that VD3 controls the growth and differentiation of human osteoblastic cells by regulating the gene expression of osteoblast-related transcription factors as well as that of type I collagen, and that the co-presence of both signals is essential for VD3 to express full activity toward the differentiation of human osteoblasts.


Assuntos
Diferenciação Celular/fisiologia , Colecalciferol/metabolismo , Colágeno Tipo I/metabolismo , Osteoblastos/fisiologia , Fatores de Transcrição/metabolismo , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Animais , Biomarcadores/metabolismo , Linhagem Celular Tumoral , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Regulação da Expressão Gênica , Humanos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Fator de Transcrição Sp7 , Fatores de Transcrição/genética
18.
J Immunol ; 173(8): 4976-84, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15470040

RESUMO

NKT cells produce large amounts of cytokines associated with both the Th1 (IFN-gamma) and Th2 (IL-4) responses following stimulation of their invariant Valpha14 Ag receptor. The role of adhesion molecules in the activation of NKT cells by the Valpha14 ligand alpha-galactosylceramide (alpha-GalCer) remains unclear. To address this issue, LFA-1-/- (CD11a-/-) mice were used to investigate IL-4 and IFN-gamma production by NKT cells following alpha-GalCer stimulation. Intriguingly, LFA-1-/- mice showed increased IL-4, IL-5, and IL-13 production and polarized Th2-type responses in response to alpha-GalCer in vitro and in vivo. Furthermore, the Th2-specific transcription factor GATA-3 was up-regulated in alpha-GalCer-activated NKT cells from LFA-1-/- mice. These results provide the first genetic evidence that the adhesion receptor LFA-1 has a crucial role in Th2-polarizing functions of NKT cells.


Assuntos
Galactosilceramidas/farmacologia , Células Matadoras Naturais/fisiologia , Ativação Linfocitária/efeitos dos fármacos , Antígeno-1 Associado à Função Linfocitária/fisiologia , Células Th2/imunologia , Animais , Apresentação de Antígeno , Adesão Celular , Proteínas de Ligação a DNA/genética , Fator de Transcrição GATA3 , Regulação da Expressão Gênica , Interferon gama/biossíntese , Interleucina-4/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Transativadores/genética
19.
Redox Rep ; 9(6): 331-6, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15720828

RESUMO

Reactive oxygen species (ROS) have been implicated in the pathogenesis of temporomandibular disorders. In the present study, we provide the first evidence of ROS generation in the synovial fluid from human temporomandibular disorder patients, as shown by electron spin resonance (ESR) and spin trapping. Three distinct ESR spectra of DMPO spin adducts were observed in the synovial fluid. They corresponded to three free radical species: hydroxyl radical (HO(*)), hydrogen radical (H(*)), and carbon-center radical (R(*)). Among them, the 5,5-dimethyl-1-pyrroline-N-oxide (DMPO)-OH spectrum was the most prominent, suggesting that HO(*) was dominantly generated in the synovial fluid from temporomandibular disorder patients. Desferrioxamine (DFO), an iron chelator, strongly depressed the DMPO-OH signal intensity in the synovial fluid from patients with temporomandibular disorders. We successfully demonstrated ROS-induced oxidative stress in the synovial fluid from temporomandibular disorder patients. ROS generation in the temporomandibular joint could lead to exacerbation of inflammation and activation of cartilage matrix degrading enzymes that proceed to degenerative change of the temporomandibular joint. Thus, iron-dependent generation of HO( *) might have a crucial role in the pathogenesis of temporomandibular disorders.


Assuntos
Espécies Reativas de Oxigênio/metabolismo , Líquido Sinovial/metabolismo , Transtornos da Articulação Temporomandibular/metabolismo , Adolescente , Adulto , Desferroxamina/farmacologia , Espectroscopia de Ressonância de Spin Eletrônica , Feminino , Humanos , Radical Hidroxila/metabolismo , Masculino , Pessoa de Meia-Idade , Marcadores de Spin , Superóxido Dismutase/metabolismo , Transtornos da Articulação Temporomandibular/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...