Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Sci ; 114(10): 3957-3971, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37496288

RESUMO

Lysyl oxidase-like 2 (LOXL2) is a matrix-remodeling enzyme that has recently been identified as an important regulator of tumor progression and metastasis. This study discovered that LOXL2 expression in oral squamous cell carcinoma (OSCC) tissues was significantly associated with tumor clinical stage, lymph node metastasis and patients' overall survival time. LOXL2-overexpressing human buccal SCC TW2.6 (TW2.6/LOXL2) and hypopharyngeal SCC FaDu (FaDu/LOXL2) cells exhibited enhanced migration, invasion, epithelial-mesenchymal transition (EMT), and cancer stem cell (CSC) phenotypes, independently of its enzymatic activity. Moreover, TW2.6/LOXL2 significantly increased tumor-initiating frequency in SCID mice. We further demonstrated that LOXL2 increased the levels of interferon-induced protein with tetratricopeptide repeats 1 (IFIT1) and IFIT3 in TW2.6/LOXL2 and FaDu/LOXL2 cells. We also identified IFIT1 and IFIT3 as key downstream components of LOXL2 action in migration, invasion, EMT, and CSC phenotypes in TW2.6 and FaDu cells. Furthermore, a significant positive correlation between LOXL2 expression and IFIT1 and IFIT3 overexpression in human OSCC tissues was observed. In addition, TW2.6/LOXL2 and FaDu/LOXL2 cells were 3.3- to 3.6-fold more susceptible to the epidermal growth factor receptor (EGFR) inhibitor gefitinib than were their respective control cells. The antitumor effect of gefitinib on orthotopic TW2.6/LOXL2 xenograft tumor was fourfold higher than that on controls. Our results indicate that LOXL2 expression is a strong prognostic factor for OSCC and may be used as a marker to identify patients most likely to respond to EGFR-targeted therapy.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Animais , Camundongos , Humanos , Gefitinibe/farmacologia , Carcinoma de Células Escamosas/patologia , Proteína-Lisina 6-Oxidase , Camundongos SCID , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Proteínas de Ligação a RNA/genética , Receptores ErbB , Regulação Neoplásica da Expressão Gênica , Transição Epitelial-Mesenquimal , Linhagem Celular Tumoral , Peptídeos e Proteínas de Sinalização Intracelular
2.
Artigo em Inglês | MEDLINE | ID: mdl-37471157

RESUMO

Autotransplantation has been proven as a viable method of reconstructing missing teeth. During preparation of the recipient site, the location of bone reduction depends largely on the experience of the surgeon. Inappropriate overpreparation can cause biologic and esthetic complications such as buccal or lingual bone resorption. The aim of this paper is to provide an innovative method to aid clinicians in precisely preparing a recipient site with the assistance of medical image processing software and a real-time navigation system. This case report presents the autotransplantation of a mandibular molar using this technique with good short-term (6 months) clinical outcomes, including radiographic bone fill, normal probing pocket depth, physiologic tooth mobility, acceptable gingival level, and satisfactory restoration.

3.
J Periodontal Res ; 57(6): 1219-1226, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36205057

RESUMO

OBJECTIVE AND BACKGROUND: Gingival overgrowth (GO) is a common side effect of some drugs such as anticonvulsants, immunosuppressant, and calcium channel blockers. Among them, the antiepileptic agent phenytoin is the most common agent related to this condition due to its high incidence. Transforming growth factor ß (TGFß) importantly contributes to the pathogenesis of GO. Connective tissue growth factor (CTGF or CCN2) is a key mediator of tissue fibrosis and is positively associated with the degree of fibrosis in GO. We previously showed that Src, c-jun N-terminal kinase, and Smad3 mediate TGFß1-induced CCN2 protein expression in human gingival fibroblasts (HGFs). This study investigates whether phenytoin can induce CCN2 synthesis through activated latent TGFß in HGFs and its mechanisms. METHODS: CCN2 synthesis, latent TGFß1 activation, and cellular reactive oxygen species (ROS) generation in HGFs were studied using western blot analysis, a TGFß1 Emax® ImmunoAssay System, and 2',7'-dichlorodihydrofluorescein diacetate (an oxidation-sensitive fluorescent probe), respectively. RESULTS: Phenytoin significantly stimulated CCN2 synthesis, latent TGFß1 activation, and ROS generation in HGFs. Addition of an TGFß-neutralizing antibody, TGFß receptor kinase inhibitor SB431542, and Smad3 inhibitor SIS3 completely inhibited phenytoin-induced CCN2 synthesis. General antioxidant N-acetylcysteine, NADPH oxidase (NOX) inhibitor diphenylene iodonium, and specific NOX4 inhibitor plumbagin almost completely suppressed phenytoin-induced total cellular ROS and latent TGFß1 activation. Curcumin dose-dependently decreased phenytoin-induced TGFß1 activation and CCN2 synthesis in HGFs. CONCLUSIONS: Our findings indicated that NOX4-derived ROS play pivotal roles in phenytoin-induced latent TGFß1 activation. Molecular targeting the phenytoin/NOX4/ROS/TGFß1 pathway may provide promising strategies for the prevention and treatment of GO. Curcumin-inhibited phenytoin-induced CCN2 synthesis is caused by the suppression of latent TGFß1 activation.


Assuntos
Curcumina , Crescimento Excessivo da Gengiva , Humanos , Gengiva/metabolismo , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fator de Crescimento do Tecido Conjuntivo/farmacologia , Curcumina/farmacologia , NADPH Oxidase 4/metabolismo , NADPH Oxidase 4/farmacologia , Fenitoína/efeitos adversos , Espécies Reativas de Oxigênio/metabolismo , Células Cultivadas , Fibroblastos , Fator de Crescimento Transformador beta1/metabolismo , Crescimento Excessivo da Gengiva/induzido quimicamente , Fibrose
4.
PLoS One ; 17(3): e0258979, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35235557

RESUMO

Circular shell rings along the South Atlantic Coast of North America are the remnants of some of the earliest villages that emerged during the Late Archaic (5000-3000 BP). Many of these villages, however, were abandoned during the Terminal Late Archaic (ca 3800-3000 BP). We combine Bayesian chronological modeling with mollusk shell geochemistry and oyster paleobiology to understand the nature and timing of environmental change associated with the emergence and abandonment of circular shell ring villages on Sapelo Island, Georgia. Our Bayesian models indicate that Native Americans occupied the three Sapelo shell rings at varying times with some generational overlap. By the end of the complex's occupation, only Ring III was occupied before abandonment ca. 3845 BP. Ring III also consists of statistically smaller oysters harvested from less saline estuaries compared to earlier occupations. Integrating shell biochemical and paleobiological data with recent tree ring analyses shows a clear pattern of environmental fluctuations throughout the period in which the rings were occupied. We argue that as the environment became unstable around 4300 BP, aggregation at villages provided a way to effectively manage fisheries that are highly sensitive to environmental change. However, with the eventual collapse of oyster fisheries and subsequent rebound in environmental conditions ca. post-3800 BP, people dispersed from shell rings, and shifted to non-marine subsistence economies and other types of settlements. This study provides the most comprehensive evidence for correlations between large-scale environmental change and societal transformations on the Georgia coast during the Late Archaic period.


Assuntos
Teorema de Bayes
5.
Life Sci ; 231: 116563, 2019 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-31200003

RESUMO

AIMS: In the present study, we investigated the roles of renin-angiotensin system (RAS) activation and imbalance of matrix metalloproteinase-9 (MMP-9)/tissue inhibitor of matrix metalloproteinase-1 (TIMP-1) in cold-induced stroke during chronic hypertension, as well as the protective effects of captopril and recombinant human TIMP-1 (rhTIMP-1). MAIN METHODS: Rats were randomly assigned to sham; 2-kidney, 2-clip (2K-2C); 2K-2C + captopril, and 2K-2C + rhTIMP-1 groups. After blood pressure values had stabilized, each group was randomly divided into an acute cold exposure (ACE) group (12-h light at 22 °C/12-h dark at 4 °C) and a non-acute cold exposure (NACE) group (12-h light/12-h dark at 22 °C), each of which underwent three cycles of exposure. Captopril treatment was administered via gavage (50 mg/kg/d), while rhTIMP-1 treatment was administered via the tail vein (60 µg/kg/36 h). KEY FINDINGS: In the 2K-2C group, angiotensin II (AngII) and MMP-9 levels increased in both the plasma and cortex, while no such changes in TIMP-1 expression were observed. Cold exposure further upregulated AngII and MMP-9 levels and increased stroke incidence. Captopril and rhTIMP-1 treatment inhibited MMP-9 expression and activation and decreased stroke incidence in response to cold exposure. SIGNIFICANCE: The present study is the first to demonstrate that cold exposure exacerbates imbalance between MMP-9 and TIMP-1 by activating the RAS, which may be critical in the initiation of stroke during chronic hypertension. In addition, our results suggest that captopril and rhTIMP-1 exert protective effects against cold-induced stroke by ameliorating MMP-9/TIMP-1 imbalance.


Assuntos
Metaloproteinase 9 da Matriz/metabolismo , Sistema Renina-Angiotensina/fisiologia , Acidente Vascular Cerebral/metabolismo , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Angiotensina II/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Captopril/metabolismo , Captopril/farmacologia , Proteínas de Ciclo Celular/metabolismo , Temperatura Baixa/efeitos adversos , Humanos , Rim/metabolismo , Masculino , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Sistema Renina-Angiotensina/genética , Acidente Vascular Cerebral/fisiopatologia , Inibidor Tecidual de Metaloproteinase-1/farmacologia , Inibidor Tecidual de Metaloproteinase-2
6.
J Formos Med Assoc ; 117(6): 527-534, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28720506

RESUMO

BACKGROUND/PURPOSE: Oral submucous fibrosis (OSF) is a premalignant condition caused by the chewing of areca nut (AN). Transforming growth factor ß (TGFß) plays a central role in the pathogenesis of OSF. Connective tissue growth factor (CTGF or CCN2) and early growth response-1 (Egr-1) are important mediators in the fibrotic response to TGFß in several fibrotic disorders including OSF. Arecoline, a major AN alkaloid, induced the synthesis of CCN2 and Egr-1 in human buccal mucosal fibroblast (BMFs). The aims of this study were to investigate whether arecoline-induced CCN2 and Egr-1 syntheses are mediated through TGFß1 signaling and to inspect the detailed mechanisms involved. METHODS: Western blot and TGFß1 Emax® ImmunoAssay were used to measure the effect of arecoline on the TGFß signaling pathways. 2',7'-dichlorodihydrofluorescein diacetate and MitoSOX™ Red were used to measure the effect of arecoline on the cellular and mitochondrial reactive oxygen species (ROS). RESULTS: Arecoline induced latent TGFß1 activation, Smad2 phosphorylation, and mitochondrial and total cellular ROS in BMFs. TGFß-neutralizing antibody completely inhibited the arecoline-induced synthesis of CCN2 and Egr-1. Mito-TEMPO, a mitochondria-targeted antioxidant, completely suppressed arecoline-induced latent TGFß1 activation and mitochondrial and total cellular ROS. Epigallocatechin-3-gallate (EGCG) dose-dependently inhibited arecoline-induced TGFß1 activation and mitochondrial ROS in BMFs. CONCLUSION: Our results indicated that arecoline-induced mitochondrial ROS plays pivotal roles in the activation of latent TGFß1 leading to the initiation of TGFß1 signaling and subsequent increase in the synthesis of CCN2 and Egr-1. EGCG can be a useful agent in the chemoprevention and treatment of OSF.


Assuntos
Areca/efeitos adversos , Arecolina/farmacologia , Catequina/análogos & derivados , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Western Blotting , Catequina/farmacologia , Células Cultivadas , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Imunoensaio , Mitocôndrias/metabolismo , Mucosa Bucal/patologia , Fibrose Oral Submucosa/induzido quimicamente , Fosforilação/efeitos dos fármacos , Extratos Vegetais/farmacologia , Plantas Tóxicas/efeitos adversos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/genética , Fator de Crescimento Transformador beta1/genética
7.
J Formos Med Assoc ; 117(3): 212-219, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28438434

RESUMO

BACKGROUND/PURPOSE: Deregulation of metabolic pathways is one of the hallmarks of cancer progression. Connective tissue growth factor (CTGF/CCN2) acts as a tumor suppressor in oral squamous cell carcinoma (OSCC). However, the role of CTGF in modulating cancer metabolism is still unclear. METHODS: OSCC cells stably overexpressing CTGF (SAS/CTGF) and shRNA against CTGF (TW2.6/shCTGF) were established. Oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) were examined by the Seahorse XF24 analyzer. The expression of CTGF and mitochondrial biogenesis related genes was measured by real-time polymerase chain reaction or Western blot analysis. RESULTS: CTGF decreased OCR, ECAR, adenosine triphosphate (ATP) generation, mitochondrial DNA (mtDNA), and mitochondrial transcription factor A (mtTFA) protein expression in OSCC cells. Overexpression of mtTFA restored CTGF-decreased OCR, ECAR, mtDNA copy number, migration and invasion of SAS/CTGF cells. Immunoprecipitation assay showed a higher level of ubiquitinated mtTFA protein after CTGF treatment. MG132, an inhibitor of proteasomal degradation, reversed the effect of CTGF on mtTFA protein expression in SAS cells. CONCLUSION: CTGF can decrease glycolysis, mitochondrial oxidative phosphorylation, ATP generation, and mtDNA copy number by increasing mtTFA protein degradation through ubiquitin proteasome pathway and in turn reduces migration and invasion of OSCC cells. Therefore, CTGF may be developed as a potential additive therapeutic drug for oral cancer in the near future.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Fator de Crescimento do Tecido Conjuntivo/fisiologia , Proteínas de Ligação a DNA/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Neoplasias Bucais/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitina/fisiologia , Linhagem Celular Tumoral , Humanos
8.
Oral Oncol ; 66: 100-107, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28012797

RESUMO

OBJECTIVES: Relapse is the most serious problem affecting the morbidity and mortality rates of patients with head and neck squamous cell carcinoma (HNSCC). Although HNSCC has been studied for several decades, the exact mechanism of cancer recurrence remains unclear. MATERIALS AND METHODS: ataxia-telangiectasia mutated interactor (ATMIN) messenger RNA(mRNA) expression was detected in HNSCC samples by quantitative RT-PCR, and was analyzed with patients' clinical outcomes by Kaplan-Meier analyses. The ectopic ATMIN expression or ATMIN silencing on invasion ability was evaluated in HNSCC cell lines. Lymph node metastasis ability was investigated by buccal orthotopic implantation in vivo. All statistical tests were two-sided. RESULTS: ATMIN mRNA expression was positively correlated with patients' clinical outcomes. ATMIN blockage reduced invasion, migration, and metastasis abilities both in vitro and in vivo. Evidence from a buccal orthotopic implantation mice model showed that silenced ATMIN expression prolongs mice survival and reduced lymph node metastasis. In high-throughput microarray and bioinformative analyses, KRas was identified as a crucial downstream effector in ATMIN-mediated HNSCC metastasis and was positively associated with patients' clinical stages and ATMIN mRNA expression. CONCLUSIONS: The role of ATMIN and its regulatory mechanisms in HNSCC progression are reported for the first time. The study results improve our understanding of the ATMIN-KRas axis leading to HNSCC migration or invasion and metastasis and facilitates the identification of possible therapy targets of downstream genes for designing effective therapeutic strategies in personalized medicine.


Assuntos
Carcinoma de Células Escamosas/patologia , Genes ras , Neoplasias de Cabeça e Pescoço/patologia , Metástase Linfática/genética , Fatores de Transcrição/fisiologia , Progressão da Doença , Feminino , Técnicas de Silenciamento de Genes , Humanos , Masculino , Pessoa de Meia-Idade , Carcinoma de Células Escamosas de Cabeça e Pescoço , Fatores de Transcrição/genética
9.
Clin Oral Investig ; 20(9): 2343-2352, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26822174

RESUMO

OBJECTIVES: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been regarded as a promising candidate for cancer therapy. However, most of oral cancer cell lines are resistant to the TRAIL-induced cytotoxicity. The aim of this study was to investigate the ability of phenethyl isothiocyanate (PEITC) to sensitize TRAIL-induced apoptosis in TRAIL-resistant oral cancer cells and xenografts. MATERIALS AND METHODS: Terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay, Western blotting, and a mouse xenograft model were used to study the effects of PEITC and TRAIL on two TRAIL-resistant human oral cancer cells, SAS and Ca9-22. RESULTS: PEITC upregulated death receptor 4 (DR4) and DR5 protein expression and increased reactive oxygen species (ROS) production in both SAS and Ca9-22 cells. Antioxidant N-acetyl-L-cysteine (NAC) and c-Jun NH2-terminal kinase (JNK) inhibitor SP600125 inhibited PEITC-induced DR4 and DR5 expression. Inhibitor experiments showed that PEITC induced apoptosis through ROS-mediated JNK activation and upregulation of DR4 and DR5. Furthermore, treatment with PEITC significantly increased TRAIL-induced apoptosis in both cells. Combined treatment with PEITC and TRAIL had greater effect on the inhibition of tumor growth than either agent alone. CONCLUSIONS: We showed for the first time that PEITC overcomes TRAIL resistance in oral cancer cells and enhance the therapeutic potential of TRAIL in vivo. CLINICAL RELEVANCE: PEITC, either alone or in combination with TRAIL, can be used as a new therapeutic approach for the treatment of oral cancers.


Assuntos
Apoptose/efeitos dos fármacos , Isotiocianatos/farmacologia , Neoplasias Bucais/tratamento farmacológico , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Acetilcisteína/farmacologia , Animais , Antracenos/farmacologia , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular , Resistencia a Medicamentos Antineoplásicos , Xenoenxertos , Humanos , Marcação In Situ das Extremidades Cortadas , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
10.
Head Neck ; 37(4): 493-7, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24436257

RESUMO

BACKGROUND: Early growth response-1 (Egr-1) protein plays an important role in many human fibrotic diseases. Areca nut chewing is the most important risk factor of oral submucous fibrosis (OSF). METHODS: Egr-1 protein expression in OSF was examined using antibody to Egr-1. Arecoline-induced Egr-1 expression and its signaling pathways were assessed by Western blot analyses in human buccal mucosal fibroblasts (BMFs). RESULTS: Elevated Egr-1 staining was observed in epithelial cells, fibroblast, and inflammatory cells in 7 of 10 OSF cases. Arecoline, a main alkaloid found in the areca nut, stimulated Egr-1 synthesis in BMFs. Pretreatment with antioxidant N-acetyl-L-cysteine, c-Jun NH2-terminal kinase inhibitor SP600125, and extracellular signal-regulated kinase inhibitor PD98059 significantly reduced arecoline-induced Egr-1 synthesis. Epigallocatechin-3-gallate (EGCG) inhibited arecoline-induced Egr-1 synthesis and collagen gel contraction in a dose-responsive manner. CONCLUSION: Constitutive Egr-1 expression during areca nut chewing may play a role in the pathogenesis of OSF. EGCG could be a good candidate for prevention or treatment of OSF.


Assuntos
Arecolina/farmacologia , Catequina/análogos & derivados , Agonistas Colinérgicos/farmacologia , Proteína 1 de Resposta de Crescimento Precoce/efeitos dos fármacos , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Mucosa Bucal/citologia , Fibrose Oral Submucosa/metabolismo , Western Blotting , Catequina/farmacologia , Bochecha , Humanos , Imuno-Histoquímica , Mucosa Bucal/metabolismo , Transdução de Sinais/fisiologia
11.
J Formos Med Assoc ; 114(9): 860-4, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24035571

RESUMO

BACKGROUND/PURPOSE: Connective tissue growth factor (CCN2) has been associated with the pathogenesis of various fibrotic diseases, including oral submucous fibrosis (OSF). The chemical constituents of areca nut along with the mechanical trauma cause OSF. The coarse fibers of areca nut injure the mucosa and hence sphingosine-1-phosphate (S1P) is released at the wounded sites. Recent studies have shown that S1P is involved in wound healing and the development of fibrosis. The aims of this study were to investigate the effects of S1P on CCN2 expression in human buccal fibroblasts (HBFs) and identify the potential targets for drug intervention or chemoprevention of OSF. METHODS: Western blot analyses were used to study the effects of S1P on CCN2 expression and its signaling pathways in HBFs and whether epigallocatechin-3-gallate (EGCG), the main and most significant polyphenol in green tea, could inhibit this pathway. RESULTS: S1P significantly enhanced CCN2 synthesis in HBFs. This effect can be inhibited by c-Jun NH2-terminal kinase (JNK) inhibitor and extracellular signal-regulated kinase inhibitor but not by P38 mitogen-activated protein kinase inhibitor. Interestingly, EGCG completely blocked S1P-induced CCN2 expression via suppressing S1P-induced JNK phosphorylation. CONCLUSION: S1P released by repetitive mechanical trauma during AN chewing may contribute to the pathogenesis of OSF through upregulating CCN2 expression in HBFs. EGCG could be an adjuvant to the current offered therapy options or the prevention of OSF through suppression of JNK activation.


Assuntos
Catequina/análogos & derivados , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fibroblastos/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Fibrose Oral Submucosa/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Areca , Catequina/farmacologia , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Humanos , Esfingosina/farmacologia , Regulação para Cima/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
12.
Oral Oncol ; 51(2): 170-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25435433

RESUMO

OBJECTIVES: MicroRNA (miRNA) machinery regulates cancer cell behavior, and has been implicated in patients' clinical status and prognosis. We found that microRNA-29b (miR-29b) increased significantly in advanced migratory cells. However, miR-29b controls the migration ability, and its regulatory mechanism in oral squamous cell carcinoma (OSCC) remains unknown. MATERIALS AND METHODS: We triggered miR-29b expression in OSCC patients and cell lines by conducting real-time quantitative PCR. We determined the functions of miR-29b in the migration of OSCC cells by using gain- and loss-of-function approaches. We elevated the target genes of miR29b through software predictions and a luciferase report assay. We used an orthotopic OSCC animal model to investigate the effects of miR29b on OSCC cell metastasis in vivo. RESULTS: The clinical data revealed that miR-29b expression was correlated with lymph node metastasis and an advanced tumor stage in 98 OSCC patients. Furthermore, multivariate analysis revealed that miR-29b expression was significantly correlated with recurrence, and indicated poor survival. MiR-29b promoted OSCC cell migration and downregulated CX3CL1, a cell-cell adhesion regulator, which plays an essential role in miR-29b-regulated OSCC cell migration machinery. Furthermore, we found that CX3CL1 expression was correlated with lymph node metastasis and an early tumor stage in OSCC patients, and negatively correlated with miR-29b expression. CONCLUSION: MiR-29b acts as an oncomir, promoting cell migration through CX3CL1 suppression, and could be a potential therapeutic target for preventing OSCC progression.


Assuntos
Carcinoma de Células Escamosas/patologia , MicroRNAs/fisiologia , Neoplasias Bucais/patologia , Metástase Neoplásica , Animais , Quimiocina CX3CL1/genética , Inativação Gênica , Humanos , Metástase Linfática , Camundongos , MicroRNAs/genética , Análise de Sobrevida
13.
J Formos Med Assoc ; 113(11): 828-32, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24560449

RESUMO

BACKGROUND/PURPOSE: Transforming growth factor-ß (TGF-ß) plays an important role in the pathogenesis of cyclosporine A (CsA)-induced gingival overgrowth (GO). Connective tissue growth factor (CTGF/CCN2) acts as a cofactor with TGF-ß to induce the maximal profibrotic effects of TGF-ß. We investigated the effects of CsA on CCN2 expression in human gingival fibroblasts (HGFs) and the potential chemopreventive agent for CsA-induced GO. METHODS: Western blot analyses were used to examine the signaling pathways of CsA-induced CCN2 expression in HGFs and whether epigallocatechin-3-gallate (EGCG), curcumin, or lovastatin can inhibit CsA-induced CCN2 expression. RESULTS: CsA significantly stimulated CCN2 synthesis in HGFs. This effect can be inhibited by c-Jun NH(2)-terminal kinase (JNK) and Smad3 inhibitors but not by TGF-ß neutralizing antibody and TGF-ß type I receptor inhibitor. Furthermore, EGCG completely blocked CsA-induced CCN2 expression. CONCLUSION: CsA-induced CCN2 protein expression is mediated through JNK and Smad signaling. CsA may contribute to the pathogenesis of GO through upregulation of CCN2 expression in HGFs. EGCG could be an adjuvant for the prevention of CsA-induced GO.


Assuntos
Catequina/análogos & derivados , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Ciclosporina/efeitos adversos , Fibroblastos/efeitos dos fármacos , Crescimento Excessivo da Gengiva/induzido quimicamente , Fator de Crescimento Transformador beta1/metabolismo , Catequina/farmacologia , Gengiva/citologia , Humanos , Cultura Primária de Células
14.
J Formos Med Assoc ; 113(1): 50-5, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24445011

RESUMO

BACKGROUND/PURPOSE: Connective tissue growth factor (CTGF/CCN2) is involved in the development and progression of fibrotic diseases, including gingival overgrowth (GO). Recent studies indicate that lysophosphatidic acid (LPA) is also significantly involved in wound healing and the development of fibrosis. This study investigated whether epigallocatechin-3-gallate (EGCG) can inhibit LPA-induced CCN2 expression in human gingival fibroblast (GF) and its mechanism. METHODS: Western blot analyses were used to study the signaling pathways of LPA-induced CCN2 expression in human GFs and the effects of EGCG on this pathway. RESULTS: LPA stimulated CCN2 synthesis in human GFs. This effect can be significantly inhibited bytransforming growth factor-ß type I receptor/ALK5, Smad3, and JNK inhibitors but not ERK, P38, and MAPK inhibitors. EGCG completely inhibited LPA-induced CCN2 expression through attenuating the LPA-induced JNK and Smad3 phosphorylation in human GFs. CONCLUSION: LPA produced at the surgical wound may contribute to the recurrence of GO by upregulating CCN2 expression in human GFs. This effect was mediated by Smad3 and JNK activation and ALK5 transactivation. EGCG could be a useful agent for reducing the recurrence of GO after surgery through suppression of JNK and Smad3 activations.


Assuntos
Catequina/análogos & derivados , Fator de Crescimento do Tecido Conjuntivo/fisiologia , Fibroblastos/metabolismo , Gengiva/citologia , Lisofosfolipídeos/farmacologia , Proteína Smad3/antagonistas & inibidores , Catequina/farmacologia , Células Cultivadas , Fator de Crescimento do Tecido Conjuntivo/análise , Curcumina/farmacologia , Humanos , MAP Quinase Quinase 4/antagonistas & inibidores , Cicatrização/efeitos dos fármacos
15.
J Endod ; 39(11): 1407-12, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24139263

RESUMO

INTRODUCTION: Methacrylate resin-based materials could release components into adjacent environment even after polymerization. The major components leached include triethylene glycol dimethacrylate (TEGDMA). TEGDMA has been shown to induce the expression of cyclooxygenase-2 (COX-2). However, the mechanisms are not completely understood. The aims of this study were to investigate the molecular mechanism underlying TEGDMA-induced COX-2 in 2 oral cell types, the primary culture of human dental pulp (HDP) cells and the human embryonic palatal mesenchymal (HEPM) pre-osteoblasts, and to propose potential strategy to prevent or ameliorate the TEGDMA-induced inflammation in oral tissues. METHODS: TEGDMA-induced COX-2 expression and its signaling pathways were assessed by Western blot analyses in HDP and HEPM cells. The inhibition of TEGDMA-induced COX-2 protein expression using various dietary phytochemicals was investigated. RESULTS: COX-2 protein expression was increased after exposure to TEGDMA at concentrations as low as 5 µmol/L. TEGDMA-induced COX-2 expression was associated with reaction oxygen species, the extracellular signal-regulated kinase 1/2, and the p38 mitogen-activated protein kinase signaling pathways in HDP and HEPM cells. The activation of p38 mitogen-activated protein kinase was directly associated with reactive oxygen species. Epigallocatechin-3-gallate suppressed TEGDMA-induced COX-2 expression by inhibiting phosphorylation of extracellular signal-regulated kinase 1/2. CONCLUSIONS: Cells exposed to low concentrations of TEGDMA may induce inflammatory responses of the adjacent tissues, and this should be taken into consideration during common dental practice. Green tea, which has a long history of safe beverage consumption, may be a useful agent for the prevention or treatment of TEGDMA-induced inflammation in oral tissues.


Assuntos
Catequina/análogos & derivados , Resinas Compostas/farmacologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , Ciclo-Oxigenase 2/efeitos dos fármacos , Polpa Dentária/enzimologia , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , Células-Tronco Mesenquimais/enzimologia , Palato/citologia , Polietilenoglicóis/farmacologia , Ácidos Polimetacrílicos/farmacologia , Catequina/farmacologia , Técnicas de Cultura de Células , Linhagem Celular , Células Cultivadas , Polpa Dentária/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteoblastos/enzimologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/efeitos dos fármacos
16.
Clin Oral Investig ; 17(2): 455-61, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22415218

RESUMO

OBJECTIVES: Transforming growth factor ß (TGFß) has been suggested as the main trigger for the increased collagen production and decreased matrix degradation pathways in oral submucous fibrosis (OSF). Connective tissue growth factor (CTGF/CCN2) and cyclooxygenase-2 (COX-2) were found to overexpress in OSF. The aim of this study was to investigate the molecular mechanism underlying the TGFß-induced CCN2 expressions in human buccal mucosal fibroblasts (BMFs) to identify the potential targets for drug intervention or chemoprevention of OSF. MATERIALS AND METHODS: TGFß-induced CCN2 expression and its signaling pathways were assessed by Western blot analyses in BMFs. RESULTS: TGFß1 stimulated CCN2 synthesis in BMFs. Pretreatment with c-Jun NH(2)-terminal kinase (JNK) inhibitor SP600125, p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580, and activin receptor-like kinase 5 (ALK5) inhibitor SB431542 significantly reduced TGFß1-induced CCN2 synthesis. Epigallocatechin-3-gallate (EGCG) completely blocked TGFß1-induced CCN2 synthesis by inhibiting the phosphorylation of JNK and p38 MAPK. Prostaglandin E(2) (PGE(2)) inhibited the TGFß1-induced CCN2 synthesis in human fetal lung fibroblasts IMR90 but not in BMFs. CONCLUSIONS: The TGFß1-induced CCN2 synthesis in BMFs could be mediated by the ALK5, JNK, and p38 MAPK pathways. EGCG blocks TGFß1-induced CCN2 by suppressing JNK and p38 in BMFs. CLINICAL RELEVANCE: The exceptional signal transduction pathways of TGFß1-induced CCN2 production in BMFs contribute to the resistance of PGE(2) downregulation of CCN2 expression; therefore, the CTGF/CCN2 levels are maintained in the OSF tissues in the presence of COX-2. EGCG may serve as a useful agent in controlling OSF.


Assuntos
Catequina/análogos & derivados , Fator de Crescimento do Tecido Conjuntivo/antagonistas & inibidores , Fibroblastos/efeitos dos fármacos , MAP Quinase Quinase 4/antagonistas & inibidores , Mucosa Bucal/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Fator de Crescimento Transformador beta1/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Antracenos/farmacologia , Benzamidas/farmacologia , Catequina/farmacologia , Linhagem Celular , Células Cultivadas , Fator de Crescimento do Tecido Conjuntivo/efeitos dos fármacos , Dinoprostona/farmacologia , Dioxóis/farmacologia , Relação Dose-Resposta a Droga , Fibroblastos/enzimologia , Flavonoides/farmacologia , Humanos , Imidazóis/farmacologia , Pulmão/citologia , Pulmão/efeitos dos fármacos , Mucosa Bucal/citologia , Fibrose Oral Submucosa/enzimologia , Fibrose Oral Submucosa/patologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Piridinas/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores
17.
Head Neck ; 34(8): 1089-94, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21953860

RESUMO

BACKGROUND: Connective tissue growth factor (CTGF/CCN2) is associated with many human fibrotic disorders and was found to overexpress in oral submucous fibrosis (OSF). OSF is the result of persistent chemical irritation and microtrauma to oral mucosa from areca nut. Microtrauma could lead to the release of thrombin. METHODS: Thrombin-induced CCN2 expression and its signaling pathways were assessed by Western blot analyses in human buccal mucosal fibroblasts. RESULTS: Thrombin stimulated CCN2 synthesis in buccal mucosal fibroblasts via activation of protease-activated receptor-1. Pretreatment with antioxidant N-acetyl-L-cysteine, apoptosis signal-regulating kinase 1 inhibitor thioredoxin, and c-Jun NH(2) -terminal kinase inhibitor SP600125 significantly reduced thrombin-induced CCN2 synthesis. Epigallocatechin-3-gallate completely inhibited thrombin-induced CCN2 synthesis. CONCLUSION: Thrombin produced by microtrauma may contribute to the pathogenesis of OSF by up-regulating CCN2 expression. This effect could be mediated by protease-activated receptor-1, reactive oxygen species, apoptosis signal-regulating kinase 1, and c-Jun NH(2) -terminal kinase pathways and prevented by epigallocatechin-3-gallate.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fibroblastos/metabolismo , Mucosa Bucal/citologia , Trombina/farmacologia , Clorometilcetonas de Aminoácidos/farmacologia , Anticarcinógenos/farmacologia , Areca , Western Blotting , Catequina/análogos & derivados , Catequina/farmacologia , Células Cultivadas , Hemostáticos/farmacologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase Quinase 5/metabolismo , Mastigação , Fibrose Oral Submucosa/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptor PAR-1/metabolismo , Inibidores de Serino Proteinase/farmacologia , Regulação para Cima
18.
Oral Oncol ; 47(4): 256-61, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21317023

RESUMO

Cyr61 is associated with growth and progression of many types of tumors and is an independent poor prognostic indicator for oral cancer patients. Areca nut (AN) chewing is the most important etiological factor in the pathogenesis of oral cancer in India and many Southeast Asian countries. Yet, the molecular mechanisms involved in the AN-induced oral cancer remain largely unknown. In this study, we show that arecoline, a main alkaloid found in AN, stimulated Cyr61 synthesis in human gingival epithelial S-G cells. Constitutive overexpression of Cyr61 protein in oral epithelial cells during AN chewing may play a role in the pathogenesis of oral cancer. ERK inhibitor PD98059, N-acetyl-L-cysteine, Rho-associated protein kinase (ROCK) selective inhibitor Y-27632 and a geranylgeranyltransferase inhibitor reduced the arecoline-stimulated levels of Cyr61 protein by ∼31%, 47%, 65% and 100%, respectively. Lovastatin also completely inhibited arecoline-induced Cyr61 synthesis and the inhibition is dose-dependent. Decreased of geranylgeranylated proteins could be the mechanism that lovastatin regulates Cyr61 synthesis and lovastatin could serve as a useful agent in controlling AN-induced oral cancer.


Assuntos
Arecolina/farmacologia , Carcinoma de Células Escamosas/induzido quimicamente , Proteína Rica em Cisteína 61/metabolismo , Gengiva/efeitos dos fármacos , Lovastatina/uso terapêutico , Neoplasias Bucais/induzido quimicamente , Extratos Vegetais/farmacologia , Areca/efeitos adversos , Areca/química , Arecolina/antagonistas & inibidores , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Regulação Neoplásica da Expressão Gênica , Gengiva/metabolismo , Gengiva/patologia , Humanos , Masculino , Neoplasias Bucais/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
19.
J Oral Pathol Med ; 40(9): 699-705, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23647076

RESUMO

BACKGROUND: Insulin-like growth factor II mRNA-binding protein 3 (IGF2BP3), an oncofetal RNA-binding protein, has been implicated in the enhancement of proliferation and invasion in various cancers. This study aimed to investigate the clinical significance and functional role of IGF2BP3 expression in oral squamous cell carcinoma (OSCC). METHODS: IGF2BP3 expression in 93 OSCC patients was investigated using immunohistochemical staining and correlated with clinical parameters and patients' survival. The effect of IGF2BP3 on cell invasion ability was evaluated by RNA interference in OSCC cell line. RESULTS: High expression of IGF2BP3 in OSCC was significantly correlated with large tumor size and lymph node metastasis. Kaplan-Meier analysis revealed that oral cancer patients with high IGF2BP3 expression had a significantly lower 5-year survival (P = 0.0017). Multivariate analysis of clinical samples demonstrated IGF2BP3 to be an independent prognosis factor (P = 0.003). Moreover, the IGF2BP3 shRNA significantly suppressed the invasion ability of OSCC in vitro, and the knockdown of endogenous IGF2BP3 expression also inhibited tumor formation in vivo. CONCLUSIONS: IGF2BP3 enhances cell invasion ability and tumorigenicity in human OSCC in vitro and in vivo. IGF2BP3 is an independent prognostic factor in patients with OSCC. Targeting of IGF2BP3 could potentially suppress the tumor growth and metastasis to improve the outcome of patients with OSCC.


Assuntos
Carcinoma de Células Escamosas/patologia , Transformação Celular Neoplásica/patologia , Neoplasias Bucais/patologia , RNA Mensageiro/análise , Proteínas de Ligação a RNA/análise , Idoso , Biomarcadores Tumorais/análise , Western Blotting , Carcinoma de Células Escamosas/secundário , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Inativação Gênica , Humanos , Imuno-Histoquímica , Metástase Linfática/patologia , Masculino , Gradação de Tumores , Invasividade Neoplásica , Estadiamento de Neoplasias , Prognóstico , Interferência de RNA/fisiologia , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética , Taxa de Sobrevida
20.
Mol Cancer Ther ; 8(9): 2718-25, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19737941

RESUMO

Suberoylanilide hydroxamic acid has been shown to selectively induce tumor apoptosis in cell cultures and animal models in several types of cancers and is about as a promising new class of chemotherapeutic agents. In addition, suberoylanilide hydroxamic acid showed synergistic anticancer activity with radiation, cisplatin, and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in some cancers. Here, we report suberoylanilide hydroxamic acid also induced apoptosis in human oral cancer cells. Western blotting showed suberoylanilide hydroxamic acid increased Fas, Fas ligand, DR4, and DR5 protein expression and activated caspase-8 and caspase-9. The apoptosis was almost completely inhibited by caspase-8 inhibitor Z-IETD-FMK and attenuated by caspase-9 inhibitor Z-LEHD-FMK. Human recombinant DR5/Fc chimera protein but not Fas/Fc or DR4/Fc significantly inhibited apoptosis induced by suberoylanilide hydroxamic acid. These results suggest that suberoylanilide hydroxamic acid induces apoptosis mainly through activation of DR5/TRAIL death pathway. Furthermore, subtoxic concentrations of suberoylanilide hydroxamic acid sensitize two TRAIL resistant human oral cancer cells, SAS and Ca9-22, to exogenous recombinant TRAIL-induced apoptosis in a p53-independent manner. Combined treatment of suberoylanilide hydroxamic acid and TRAIL may be used as a new promising therapy for oral cancer.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/patologia , Ácidos Hidroxâmicos/farmacologia , Neoplasias Bucais/patologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Apoptose/fisiologia , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Sinergismo Farmacológico , Humanos , Marcação In Situ das Extremidades Cortadas , Neoplasias Bucais/metabolismo , Vorinostat
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...