Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biomed Sci ; 30(1): 3, 2023 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-36627707

RESUMO

The tumor immune microenvironment represents a sophisticated ecosystem where various immune cell subtypes communicate with cancer cells and stromal cells. The dynamic cellular composition and functional characteristics of the immune landscape along the trajectory of cancer development greatly impact the therapeutic efficacy and clinical outcome in patients receiving systemic antitumor therapy. Mounting evidence has suggested that epigenetic mechanisms are the underpinning of many aspects of antitumor immunity and facilitate immune state transitions during differentiation, activation, inhibition, or dysfunction. Thus, targeting epigenetic modifiers to remodel the immune microenvironment holds great potential as an integral part of anticancer regimens. In this review, we summarize the epigenetic profiles and key epigenetic modifiers in individual immune cell types that define the functional coordinates of tumor permissive and non-permissive immune landscapes. We discuss the immunomodulatory roles of current and prospective epigenetic therapeutic agents, which may open new opportunities in enhancing cancer immunotherapy or overcoming existing therapeutic challenges in the management of cancer.


Assuntos
Ecossistema , Neoplasias , Humanos , Estudos Prospectivos , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Epigênese Genética , Imunoterapia , Microambiente Tumoral/genética
2.
NPJ Genom Med ; 7(1): 64, 2022 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-36309505

RESUMO

Hoyeraal-Hreidarsson syndrome (HHS) is the most severe form of dyskeratosis congenita (DC) and is caused by mutations in genes involved in telomere maintenance. Here, we identified male siblings from a family with HHS carrying a hemizygous mutation (c.1345C > G, p.R449G), located in the C-terminal nuclear localization signal (NLS) of the DKC1 gene. These patients exhibit progressive cerebellar hypoplasia, recurrent infections, pancytopenia due to bone marrow failure, and short leukocyte telomere lengths. Single-cell RNA sequencing analysis suggested defects in the NLRP3 inflammasome in monocytes and the activation and maturation of NK cells and B cells. In experiments using induced pluripotent stem cells (iPSCs) from patients, DKC1_R449G iPSCs had short telomere lengths due to reduced levels of human telomerase RNA (hTR) and increased cytosolic proportions of DKC1. Treatment with dihydroquinolizinone RG7834 and 3'deoxyanosine cordycepin rescued telomere length in patient-derived iPSCs. Together, our findings not only provide new insights into immunodeficiency in DC patients but also provide treatment options for telomerase insufficiency disorders.

3.
Int J Med Sci ; 18(15): 3452-3462, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34522171

RESUMO

Hepatocellular carcinoma (HCC) is a worldwide health problem. Currently, there is no effective therapeutic strategy for HCC patients. Chewing areca nut is closely associated with oral cancer and liver cirrhosis. The therapeutic effect of areca nut extract (ANE) on HCC is unknown. Our results revealed that ANE treatment caused a reduction in cell viability and an increase in cell apoptosis and suppressed tumor progression in xenograft models. ANE-treated didn't induce liver tumor in nude mice. For mechanism dissection, ANE treatment caused ROS-mediated autophagy and lysosome formation. Pretreatment with an ROS inhibitor, aminoguanidine hemisulfate (AGH), abolished ANE-induced ROS production. ANE treated cells caused an increase in light chain 3 (LC3)-I to -II conversion, anti-thymocyte globulin 5+12 (ATG5+12), and beclin levels, and apoptosis related-protein changes (an increases in BAX, cleaved poly(ADP-ribose) polymerase (c-PARP), and a decrease in the Bcl-2 level). In conclusion, our study demonstrated that the ANE may be a new potential compound for HCC therapy.


Assuntos
Areca/química , Autofagia/efeitos dos fármacos , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Extratos Vegetais/farmacologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Nus , Nozes/química , Espécies Reativas de Oxigênio/metabolismo
4.
Nat Commun ; 12(1): 2163, 2021 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-33846331

RESUMO

γδ T cells are a distinct subgroup of T cells that bridge the innate and adaptive immune system and can attack cancer cells in an MHC-unrestricted manner. Trials of adoptive γδ T cell transfer in solid tumors have had limited success. Here, we show that DNA methyltransferase inhibitors (DNMTis) upregulate surface molecules on cancer cells related to γδ T cell activation using quantitative surface proteomics. DNMTi treatment of human lung cancer potentiates tumor lysis by ex vivo-expanded Vδ1-enriched γδ T cells. Mechanistically, DNMTi enhances immune synapse formation and mediates cytoskeletal reorganization via coordinated alterations of DNA methylation and chromatin accessibility. Genetic depletion of adhesion molecules or pharmacological inhibition of actin polymerization abolishes the potentiating effect of DNMTi. Clinically, the DNMTi-associated cytoskeleton signature stratifies lung cancer patients prognostically. These results support a combinatorial strategy of DNMTis and γδ T cell-based immunotherapy in lung cancer management.


Assuntos
Citoesqueleto/metabolismo , Citotoxicidade Imunológica/genética , Epigênese Genética , Sinapses Imunológicas/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Animais , Linhagem Celular Tumoral , Citoesqueleto/efeitos dos fármacos , Citotoxicidade Imunológica/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/metabolismo , Decitabina/farmacologia , Inibidores Enzimáticos/farmacologia , Epigênese Genética/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Sinapses Imunológicas/efeitos dos fármacos , Marcação por Isótopo , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/genética , Subpopulações de Linfócitos/efeitos dos fármacos , Subpopulações de Linfócitos/metabolismo , Masculino , Camundongos Endogâmicos NOD , Fosfotirosina/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sobrevida , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
EBioMedicine ; 57: 102846, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32629386

RESUMO

BACKGROUND: Aberrant fucosylation plays a critical role in lung cancer progression. Nevertheless, the key fucosyltransferase with prognostic significance in lung cancer patients, the enzyme's intracellular targets, and complex molecular mechanisms underlying lung cancer metastasis remain incompletely understood. METHODS: We performed a large-scale transcriptome-clinical correlation to identify major fucosyltransferases with significant prognostic values. Invasion, migration, cell adhesion assays were performed using lung cancer cells subject to genetic manipulation of FUT4 levels. Genome-wide RNA-seq and immunoprecipitation-mass spectrometry were used to characterize major cellular processes driven by FUT4, as well as profiling its intracellular protein targets. We also performed lung homing and metastasis assays in mouse xenograft models to determine in vivo phenotypes of high FUT4-expressing cancer cells. FINDINGS: We show that FUT4 is associated with poor overall survival in lung adenocarcinoma patients. High FUT4 expression promotes lung cancer invasion, migration, epithelial-to-mesenchymal transition, and cell adhesion. FUT4-mediated aberrant fucosylation markedly activates multiple cellular processes, including membrane trafficking, cell cycle, and major oncogenic signaling pathways. The effects are independent of receptor tyrosine kinase mutations. Notably, genetic depletion of FUT4 or targeting FUT4-driven pathways diminishes lung colonization and distant metastases of lung cancer cells in mouse xenograft models. INTERPRETATION: We propose that FUT4 can be a prognostic predictor and therapeutic target in lung cancer metastasis. Our data provide a scientific basis for a potential therapeutic strategy using targeted therapy in a subset of patients with high FUT4-expressing tumors with no targetable mutations.


Assuntos
Adenocarcinoma de Pulmão/genética , Carcinogênese/genética , Fucosiltransferases/genética , Glicoproteínas/genética , Adenocarcinoma de Pulmão/patologia , Animais , Adesão Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica/genética , Xenoenxertos , Humanos , Camundongos , Metástase Neoplásica , Transdução de Sinais/genética , Transcriptoma/genética
6.
Medicine (Baltimore) ; 96(31): e7640, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28767579

RESUMO

BACKGROUND: It has been proposed that the transient receptor potential (TRP) channel Melastatin 8 (TRPM8) is a cold-sensing TRP channel. However, its presence and its role in the nasal cavity have not yet been fully studied. METHODS: Immunohistology was used to study TRPM8 receptors in both the nasal mucosa tissue and the primary cultures of human nasal cells. Cells from primary cultures were immunostained with antibodies to TRPM8, mucin, cytokeratin (CK)-14, CK-18, and vimentin. Western blotting and real-time polymerase chain reaction (PCR) were used to determine the physiological role of TRPM8 in mucus production in the nasal cavity, with and without its agonist and antagonist. RESULTS: The TRPM8 is clearly present in the epithelium, mucous glands, and vessels. No obvious TRPM8-immunoreactive cells were detected in the connective tissue. Immunostaining of cytospin preparations showed that epithelial cells test positive for CK-14, CK-18, TRPM8, and mucin 5AC (MUC5AC). Fibroblastic cells are stained negative for TRPM8. Secreted mucins in the cultured supernatant are detected after exposure to menthol and moderate cooling to 24°C. Both induce a statistically significant increase in the level of MUC5AC mRNA and mucin production. BCTC, a TRPM8 antagonist, has a statistically significant inhibitory effect on MUC5AC mRNA expression and MUC5AC protein production that is induced by menthol and moderate cooling to 24°C. CONCLUSIONS: The study demonstrates that TRPM8 is present in the nasal epithelium. When it is activated by moderate cooling to 24°C or menthol, TRPM8 induces the secretion of mucin. This study shows that TRPM8 channels are important regulators of mucin production. Therefore, TRPM8 antagonists could be used to treat refractory rhinitis.


Assuntos
Temperatura Baixa , Células Epiteliais/metabolismo , Mucosa Nasal/metabolismo , Canais de Cátion TRPM/metabolismo , Western Blotting , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Imuno-Histoquímica , Queratina-14/metabolismo , Queratina-18/metabolismo , Masculino , Mentol/farmacologia , Mucina-5AC/metabolismo , Mucosa Nasal/citologia , Mucosa Nasal/efeitos dos fármacos , Pirazinas/farmacologia , Piridinas/farmacologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fármacos do Sistema Sensorial/farmacologia , Canais de Cátion TRPM/antagonistas & inibidores
8.
Eur Arch Otorhinolaryngol ; 274(2): 845-853, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27623823

RESUMO

Both glucocorticoids and H1-antihistamines are widely used on patients with airway diseases. However, their direct effects on airway epithelial cells are not fully explored. Therefore, we use the primary culture of human nasal epithelial cells (HNEpC) to delineate in vitro mucosal responses to above two drugs. HNEpC cells were cultured with/without budesonide and azelastine. The growth rate at each group was recorded and measured as population double time (PDT). The histamine1-receptor (H1R), muscarinic1-receptor (M1R) and M3R were measured using immunocytochemistry and western blotting after 7-days treatment. Then, we used histamine and methacholine to stimulate the mucus secretion from HNEpC and observed the MUC5AC expression in culture supernatants. Concentration-dependent treatment-induced inhibition of HNEpC growth rate was observed. Cells incubated with azelastine proliferated significantly slower than that with budesonide and the combined use of those drugs led to significant PDT prolong. The immunocytochemistry showed the H1R, M1R and M3R were obviously located in the cell membrane without apparent difference after treatment. However, western blotting showed that budesonide can significantly up-regulate the H1R, M1R and M3R level while azelastine had opposite effects. Histamine and methacholine stimulated MUC5AC secretion was greater in cells treated with budesonide but was lesser in those treated with azelastine, as compared to controls. Our data suggest that both budesonide and azelastine can significantly inhibit HNEpC proliferation, and therefore, be helpful in against airway remodeling. Long-term use of budesonide might amplify histamine signaling and result in airway hyperreactivity to stimulants by enhancing H1R, M1R and M3R expression while azelastine can oppose this effect. Therefore, combined use of those two drugs in patients with chronic inflammatory airway diseases may be an ideal option.


Assuntos
Budesonida/farmacologia , Células Epiteliais/efeitos dos fármacos , Glucocorticoides/farmacologia , Antagonistas não Sedativos dos Receptores H1 da Histamina/farmacologia , Histamina/metabolismo , Mucosa Nasal/efeitos dos fármacos , Ftalazinas/farmacologia , Biomarcadores/metabolismo , Western Blotting , Células Cultivadas , Humanos , Imuno-Histoquímica , Mucosa Nasal/citologia , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
9.
Nat Commun ; 7: 13867, 2016 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-28004751

RESUMO

Hypoxia is a major driving force of cancer invasion and metastasis. Here we show that death domain-associated protein (Daxx) acts to negatively regulate hypoxia-induced cell dissemination and invasion by inhibiting the HIF-1α/HDAC1/Slug pathway. Daxx directly binds to the DNA-binding domain of Slug, impeding histone deacetylase 1 (HDAC1) recruitment and antagonizing Slug E-box binding. This, in turn, stimulates E-cadherin and occludin expression and suppresses Slug-mediated epithelial-mesenchymal transition (EMT) and cell invasiveness. Under hypoxic conditions, stabilized hypoxia-inducible factor (HIF)-1α downregulates Daxx expression and promotes cancer invasion, whereas re-expression of Daxx represses hypoxia-induced cancer invasion. Daxx also suppresses Slug-mediated lung cancer metastasis in an orthotopic lung metastasis mouse model. Using clinical tumour samples, we confirmed that the HIF-1α/Daxx/Slug pathway is an outcome predictor. Our results support that Daxx can act as a repressor in controlling HIF-1α/HDAC1/Slug-mediated cancer cell invasion and is a potential therapeutic target for inhibition of cancer metastasis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Histona Desacetilase 1/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Proteínas Nucleares/metabolismo , Fatores de Transcrição da Família Snail/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/secundário , Linhagem Celular Tumoral , Proteínas Correpressoras , Transição Epitelial-Mesenquimal , Histona Desacetilase 1/química , Histona Desacetilase 1/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/química , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Pulmonares/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Modelos Biológicos , Chaperonas Moleculares , Invasividade Neoplásica/prevenção & controle , Proteínas Nucleares/química , Proteínas Nucleares/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transdução de Sinais , Fatores de Transcrição da Família Snail/química , Fatores de Transcrição da Família Snail/genética , Transcriptoma , Hipóxia Tumoral/fisiologia , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
10.
Am J Rhinol Allergy ; 29(4): e112-6, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26163239

RESUMO

BACKGROUND: The transient receptor potential channel melastatin 8 (TRPM8) has been proposed to be a cold receptor. However, its distribution and physiologic role in the nose is not yet fully explored. OBJECTIVE: We investigated the expression of TRPM8 in human nasal mucosa and its function when using the TRPM8 agonist. METHODS: Immunohistochemistry was used to study TRPM8 receptors in the nasal mucosa from patients with and those without allergic rhinitis (AR). By using isometric contraction studies, we also tested the effectiveness of the TRPM8 agonist menthol on nasal mucosa. Changes in nasal mucosal contractility in response to the application of the adrenergic agent methoxamine were also measured. We explored the effect of menthol on electrical field stimulation (EFS) induced nasal mucosal contractions. RESULTS: TRPM8 immunoreactivity was present principally in the nasal cilia, epithelium, and subepithelium around the glands. Except for nerve fibers, no obvious TRPM8-immunoreactive cells were detected in connective tissues. The immunoreactivity revealed no significant difference between patients with AR and those without AR. Adding menthol had a negligible effect on the basal tension of the nasal mucosa, but higher doses of menthol had a significant spasmolytic effect on nasal mucosa precontracted with methoxamine. Menthol inhibited the spike contraction induced by EFS, even at low doses. CONCLUSIONS: The finding of the TRPM8 immunoreactivity underlines the important physiologic role of the nose in temperature regulation, both in patients with allergy and those without allergy. Isometric contraction studies demonstrate the role of TRPM8 in regulating nasal patency and airway resistance. The antiadrenergic effect of menthol showed an effect apparently opposite that of clinical observations, that we usually feel decongested after menthol inhalation. The underlying mechanisms deserve further investigation, and the TRPM8 antagonists deserve consideration for treatment of rhinitis in a therapeutic trial.


Assuntos
Biomarcadores/metabolismo , Temperatura Baixa , Mucosa Nasal/metabolismo , Rinite/metabolismo , Canais de Cátion TRPM/metabolismo , Humanos , Imuno-Histoquímica , Mucosa Nasal/inervação , Rinite/tratamento farmacológico , Canais de Cátion TRPM/antagonistas & inibidores
11.
Respir Res ; 15: 106, 2014 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-25186164

RESUMO

BACKGROUND: Chronic pulmonary obstructive disease (COPD) has become the fourth leading cause of death worldwide. Cigarette smoking induces neutrophil elastase (NE) and contributes to COPD, but the detailed mechanisms involved are not fully established. In an animal model of pulmonary emphysema, there are increased expressions of placenta growth factor (PlGF) and lung epithelial (LE) cell apoptosis. This study hypothesized that excessive NE may up-regulate PlGF and that PlGF-induced LE apoptosis mediates the pathogenesis of pulmonary emphysema. METHODS: Human bronchial epithelial cells, BEAS-2B, and primary mouse type II alveolar epithelial cells were treated with NE. The PlGF promoter activity was examined by luciferase activity assay, while PlGF expression and secretion were evaluated by RT-PCR, Western blotting, and ELISA. Both cell lines were treated with PlGF to evaluate its effects and the downstream signaling pathways leading to LE cell apoptosis. PlGF knockout and wild-type mice were instilled with NE to determine the roles of PlGF and its downstream molecules in NE-promoted mice pulmonary apoptosis and emphysema phenotype. RESULTS: The transcriptional factor, early growth response gene-1, was involved in the NE-promoted PlGF promoter activity, and the expression and secretion of PlGF mRNA and protein in LE cells. PlGF-induced LE cell apoptosis and NE-induced mice pulmonary apoptosis and emphysema were mediated by the downstream c-Jun N-terminal kinase (JNK) and protein kinase C (PKC)δ signaling pathways. CONCLUSION: The NE-PlGF-JNK/PKCδ pathway contributes to the pathogenesis of LE cell apoptosis and emphysema. PlGF and its downstream signaling molecules may be potential therapeutic targets for COPD.


Assuntos
Células Epiteliais Alveolares/enzimologia , Apoptose , Elastase de Leucócito/metabolismo , Proteínas da Gravidez/metabolismo , Enfisema Pulmonar/enzimologia , Transdução de Sinais , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/imunologia , Células Epiteliais Alveolares/patologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Elastase de Leucócito/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Crescimento Placentário , Proteínas da Gravidez/genética , Proteínas da Gravidez/farmacologia , Regiões Promotoras Genéticas , Proteína Quinase C-delta/metabolismo , Enfisema Pulmonar/genética , Enfisema Pulmonar/imunologia , Enfisema Pulmonar/patologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção , Regulação para Cima
12.
Autophagy ; 10(9): 1509-21, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24988221

RESUMO

Chronic obstructive pulmonary disease (COPD) is a devastating disease, which is associated with increasing mortality and morbidity. Therefore, there is a need to clearly define the COPD pathogenic mechanism and to explore effective therapies. Previous studies indicated that cigarette smoke (CS) induces autophagy and apoptosis in lung epithelial (LE) cells. Excessive ELANE/HNE (elastase, neutrophil elastase), a factor involved in protease-antiprotease imbalance and the pathogenesis of COPD, causes LE cell apoptosis and upregulates the expression of several stimulus-responsive genes. However, whether or not elastase induces autophagy in LE cell remains unknown. The level of PGF (placental growth factor) is higher in COPD patients than non-COPD controls. We hypothesize that elastase induces PGF expression and causes autophagy in LE cells. In this study, we demonstrated that porcine pancreatic elastase (PPE) induced PGF expression and secretion in LE cells in vitro and in vivo. The activation of MAPK8/JNK1 (mitogen-activated protein kinase 8) and MAPK14/p38alpha MAPK signaling pathways was involved in the PGF mediated regulation of the TSC (tuberous sclerosis complex) pathway and autophagy in LE cells. Notably, PGF-induced MAPK8 and MAPK14 signaling pathways mediated the inactivation of MTOR (mechanistic target of rapamycin), the upregulation of MAP1LC3B/LC3B (microtubule-associated protein 1 light chain 3 ß) and the increase of autophagosome formation in mice. Furthermore, the PPE-induced autophagy promotes further apoptosis in vitro and in vivo. In summary, elastase-induced autophagy promotes LE cell apoptosis and pulmonary emphysema through the upregulation of PGF. PGF and its downstream MAPK8 and MAPK14 signaling pathways are potential therapeutic targets for the treatment of emphysema and COPD.


Assuntos
Autofagia , Enfisema/metabolismo , Células Epiteliais/efeitos dos fármacos , Pulmão/metabolismo , Elastase Pancreática/metabolismo , Proteínas da Gravidez/metabolismo , Autofagia/fisiologia , Linhagem Celular , Células Epiteliais/metabolismo , Humanos , Pulmão/patologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Fator de Crescimento Placentário , Doença Pulmonar Obstrutiva Crônica/metabolismo
13.
Ann Surg Oncol ; 19(1): 336-43, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21516372

RESUMO

BACKGROUND: Survivin has multiple functions during the progression of cancer. However, the role of survivin in the progression and metastasis of hepatocellular carcinoma (HCC) remains unknown. MATERIALS AND METHODS: Survivin expression in HCC cells (Mahlavu and Hep3B) was assessed using reverse transcription real-time PCR and Western blot analyses. In addition, survivin expression in HCC cells was manipulated using small interfering RNA (siRNA) or overexpression and proliferation and transwell migration assays were performed to monitor the effect of manipulated survivin expression on the growth rate and migratory ability of the transfected cells. RESULTS: Among the HCC cell lines tested, we found high endogenous expression of survivin mRNA and protein in Mahlavu cells. After silencing survivin expression in Mahlavu cells, there was a dramatic decrease in the cell growth rate and an increase in the metastatic potential of the cells. Overexpression of survivin in Hep3B cells suppressed the ability of the cell to migrate. The mechanism of enhanced cell migration caused by decreased survivin expression is mediated through the downregulation of glucose-regulated protein 78 (GRP78) and the upregulation of the epithelial-mesenchymal transition (EMT) marker, vimentin. CONCLUSIONS: Survivin may mediate metastasis in HCC. The knockdown of survivin expression may enhance cancer metastasis through the downregulation of GRP78 and upregulation of vimentin expression.


Assuntos
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Movimento Celular , Transição Epitelial-Mesenquimal , Proteínas de Choque Térmico/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Apoptose , Western Blotting , Carcinoma Hepatocelular/genética , Adesão Celular , Proliferação de Células , Chaperona BiP do Retículo Endoplasmático , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Proteínas de Choque Térmico/antagonistas & inibidores , Proteínas de Choque Térmico/genética , Humanos , Técnicas Imunoenzimáticas , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Proteínas Inibidoras de Apoptose/genética , Neoplasias Hepáticas/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Survivina , Células Tumorais Cultivadas , Vimentina/genética , Vimentina/metabolismo
14.
J Oral Pathol Med ; 40(7): 560-6, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21226887

RESUMO

BACKGROUNDS: MicroRNAs are small noncoding RNAs involved in posttranscriptional gene regulation, which play an important role in both physiological functioning and pathological progression. The miR-221/miR-222 microRNA family has been shown to be related to the neoplastic process in a number of different types of cancers; nevertheless, its function in oral squamous cell carcinoma (OSCC) remained uncertain. MATERIALS AND METHODS: Paired OSCC and matched noncancerous oral mucosa were examined for miR-221/miR-222 expression using quantitative reverse-transcription PCR. Ectopic expression of miR-221/miR-222 by lentiviral infection was investigated to explore its in vitro and in vivo impact on the oncogenic phenotype and the expression of various target genes. The expression of Cip/Kip cell cycle regulator p27 in tumors was analyzed with immunohistochemistry. RESULTS: The expression levels of miR-221 and miR-222 were highly correlated in OSCC. Increased miR-221/miR-222 expression was found in 40% of OSCC tissues. The ectopic expression of miR-221 or of miR-222 increased growth and anchorage-independent colony formation of OSCC cell lines. It also resulted in an increase in the tumorigenesis of an OSCC cell line in nude mice. Western blot analysis suggested that p27 and p57 might be the targets of miR-221/miR-222. p27 expression was reversely associated with the miR-221 and miR-222 expression level in OSCC tissues. CONCLUSIONS: Our findings suggested that increased miR-221/miR-222 expression was associated with the OSCC cell growth.


Assuntos
Carcinoma de Células Escamosas/patologia , MicroRNAs/análise , Neoplasias Bucais/patologia , Alphapapillomavirus/isolamento & purificação , Animais , Carcinoma de Células Escamosas/virologia , Estudos de Casos e Controles , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p27/análise , Inibidor de Quinase Dependente de Ciclina p57/análise , Progressão da Doença , Vetores Genéticos/genética , Humanos , Lentivirus/genética , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Mucosa Bucal/patologia , Neoplasias Bucais/virologia , Transplante de Neoplasias , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Autophagy ; 6(6): 725-37, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20523123

RESUMO

Areca (betel) chewing was tightly linked to oral tumorigenesis in Asians. Areca nut was a recently confirmed group I carcinogen and a popular addictive substance used by Asians. Meanwhile, the pathogenetic impact of areca on oral epithelial cells was still unclear. This study investigated the association between the induction of autophagy by areca nut extract (ANE) and the molecular regulation underlying this induction in oral cancer cells. Oral cancer cells were treated with ANE to incite the signaling changes underlying phenotypic alterations. The NFkappaB activation and reactive oxygen species (ROS) genesis were induced by ANE and the NFkappaB activation could be the basis of the ROS genesis. Furthermore, p38 activation and upregulation of MKP-1 phosphatase occurred following ANE treatment. These effects can be inhibited by ROS blockers. ANE treatment induced autophagy among oral cancer cells, which was characterized by LC3-II accumulation, genesis of autophagosomes and the appearance of EGFP-LC3 puncta. This induction was mediated through the activation of p38, MKP-1 and HIF-1alpha. Knockdown of ANE-modulated HIF-1alpha expression reduced autophagy. Blockage of ANE-induced autophagy increased the proportion of oral cancer cells undergoing apoptotic death. This study identified for the first time that ANE modulates a signaling cascade that induces HIF-1alpha expression in oral cancer cells. The eventual induction of autophagy was beneficial to cell survival from ANE-induced apoptosis.


Assuntos
Areca/química , Autofagia/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Bucais/patologia , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/farmacologia , Regulação para Cima/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Fosfatase 1 de Especificidade Dupla/metabolismo , Ativação Enzimática/efeitos dos fármacos , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Neoplasias Bucais/enzimologia , NF-kappa B/metabolismo , Nozes/química , Fagossomos/efeitos dos fármacos , Fagossomos/metabolismo , Fagossomos/ultraestrutura , Fitoterapia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Oral Oncol ; 45(12): 1032-6, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19666237

RESUMO

Oral squamous cell carcinoma (OSCC) in Asians is highly associated with the abuse of areca (betel) chewing. There are several hundred million Asians who chew areca and are therefore at high risk of OSCC. Aberrance in cyclin D1 (CCND1) and/or cortactin (CTTN), which are localized on 11q13, seems to be critical events for the development of oral carcinogenesis. This study identified amplifications of CCND1 and CTTN by quantitative (Q)-PCR analysis in 50% and 45% of OSCC samples, respectively. Co-amplification of both genes was identified in 20% of tumors. Higher CTTN expression was associated with nodal metastasis of the OSCC, while the amplification of CCND1 was identified in 28% of oral brushed samples from areca chewers, who form a high risk group for OSCC. This study confirms the importance of alterations in CCND1 and CTTN with respect to areca-associated OSCC, and demonstrates that there is an early occurrence of amplification of these genes in the risk population. The non-invasive brushing sampling method coupling with Q-PCR analysis needs to be validated for use as an early detection system for gene copy changes, which should aid oral cancer prevention.


Assuntos
Areca , Carcinoma de Células Escamosas/genética , Cortactina/genética , Ciclina D1/genética , Neoplasias Bucais/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Areca/efeitos adversos , Carcinoma de Células Escamosas/induzido quimicamente , Variações do Número de Cópias de DNA , Feminino , Amplificação de Genes , Humanos , Masculino , Mastigação , Pessoa de Meia-Idade , Mucosa Bucal/química , Neoplasias Bucais/induzido quimicamente , Reação em Cadeia da Polimerase/métodos
17.
J Oral Pathol Med ; 38(1): 79-86, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19192053

RESUMO

BACKGROUND: Areca nut chewing is associated with an increase in the incidence of oral neoplastic or inflammatory diseases. Aberrations in matrix metalloprotease (MMP) expression are associated with the pathogenesis of oral diseases. This study investigated the potential effects of areca nut extract (ANE) on human gingival fibroblasts and the consequential impacts on inflammatory pathogenesis. METHODS: Analyses of senescence marker, cell viability, changes of the cell cycle, and cell granularity in gingival fibroblasts together with an assessment of the invasiveness of polymorphonuclear (PMN) leukocytes after treatment with the supernatant of ANE-treated gingival fibroblasts were performed to characterize the phenotypic impacts. Western blotting and gelatin zymography were used to assay the expression and activity of MMP-2. RESULTS: Chronic subtoxic (<10 microg/ml) ANE treatment resulted in premature growth arrest, appearance of senescence-associated beta-galactosidase activity and various other senescence-associated phenotypes in gingival fibroblasts. Gingival fibroblasts established from older individuals had a higher propensity to become ANE-induced senescent gingival fibroblasts. An activation of MMP-2 was identified in senescent cells. PMN leukocytes treated with the supernatant of ANE-induced senescent cells exhibited a significant increase in invasiveness, which was abrogated by both a MMP-2 blocker and a MMP-2 nullifying antibody. CONCLUSIONS: This study provides evidence whereby MMP-2 secreted from ANE-induced senescent gingival fibroblasts would facilitate the invasiveness of PMN leukocytes, which could be associated with the oral inflammatory process in areca chewers.


Assuntos
Areca , Fibroblastos/efeitos dos fármacos , Gengiva/efeitos dos fármacos , Metaloproteinase 2 da Matriz/metabolismo , Infiltração de Neutrófilos/fisiologia , Neutrófilos/fisiologia , Extratos Vegetais/farmacologia , Anticorpos , Biomarcadores/análise , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Grânulos Citoplasmáticos/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fibroblastos/enzimologia , Gengiva/citologia , Gengiva/enzimologia , Humanos , Ácidos Hidroxâmicos/farmacologia , Inibidores de Metaloproteinases de Matriz , Neutrófilos/enzimologia , Fenótipo
18.
Carcinogenesis ; 27(6): 1273-84, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16474177

RESUMO

Around 200-600 million Asians chew areca (also called betel), which contains a mixture of areca nut and other ingredients. Epidemiological evidences indicated that areca use is tightly linked to oral carcinogenesis. This study investigated the effects of ripe areca nut extract (ANE) on cultured normal human oral keratinocyte (NHOK). Acute subtoxic ANE treatment inhibited DNA synthesis and induced cell cycle arrest at G1 phase in early passage (< 4th passage) cells. This was accompanied by a slight increase in the sub-G1 cellular fraction. O6-Methylguanine-DNA methyltransferase (MGMT), Hsp27 and p38MAPK was upregulated. p16 and p21 were remarkably upregulated early and declined afterwards. In contrast, the increase of dephosphorylated Rb seemed to be secondary to the episodes of p16 and p21 upregulation. To simulate the chronic areca exposure in vivo, constant ANE treatment in serial NHOK culture was performed. It resulted in a significant decrease in the population doubling, increase in senescence-associated beta-galactosidase (SA-beta-Gal) and decrease in cell proliferation in NHOK of late passages (> or = 4th passage). Induction of senescence-associated phenotypes, G2/M accumulation and genomic instability following long-term ANE treatment were also observed in a low-grade oral carcinoma cell. ANE-treated NHOK also had a higher nuclear factor-kappaB (NF-kappaB) fraction and a lower cytosolic IkappaBalpha level relative to the control in late passages. Moreover, electrophoretic mobility shift assay (EMSA) indicated that ANE treatment shifted the NF-kappaB complex from high mobility position to lower mobility position in late-passaged NHOK. ANE treatment also upregulated IL-6 and cyclooxygenase-2 (COX-2) mRNA expressions in late-passaged NHOK. In summary, our findings suggest that ANE induces the cell cycle arrest at G1/S phase and the occurrence of senescence-associated phenotypes of NHOK. The upregulation of p38MAPK, p16, p21, NF-kappaB, IL-6 and COX-2 are likely to participate in the control of these impacts.


Assuntos
Areca/metabolismo , Fase G1 , Queratinócitos/citologia , Extratos Vegetais/metabolismo , Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Ciclo-Oxigenase 2/metabolismo , Gengiva/citologia , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/biossíntese , Humanos , Proteínas I-kappa B/metabolismo , Interleucina-6/metabolismo , Queratinócitos/metabolismo , Chaperonas Moleculares , Inibidor de NF-kappaB alfa , Proteínas de Neoplasias/biossíntese , Fenótipo , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...