Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Microbiol ; 20(1): 75, 2020 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-32245419

RESUMO

BACKGROUND: Oral microbiota is not only important for maintaining oral health but also plays a role in various oral diseases. However, studies regarding microbiome changes in oral lichen planus (OLP) are very limited. To the best of our knowledge, there has been only two studies investigating salivary microbiome changes in OLP. Therefore, the purpose of this study was to identify the characteristic microbial profile in the saliva of OLP patients, with or without erosive lesions, and compare that with recurrent aphthous ulcer (RAU), a common oral immunological disorder that also shows multiple erosive/ulcerative lesions. Whole saliva samples were collected from 20 patients with OLP (erosive E, n = 10 and non-erosive NE, n = 10), 10 patients with RAU (U) and 10 healthy controls (C). DNA was extracted from the saliva samples, and the 16S rDNA gene V4 hypervariable region was analyzed using Illumina sequencing. RESULTS: We obtained 4949 operational taxonomic units (OTUs) from the V4 region in all saliva samples. Community composition analysis showed a clear decreased relative abundance of genera Streptococcus and Sphingomonas in saliva from RAU patients when compared to the other three groups. Relative abundance of Lautropia and Gemella were higher in E group, whereas relative abundance of Haemophilus and Neisseria were higher in NE group when compared to C group. Abiotrophia and Oribacterium were higher in OLP (combining E and NE groups), while Eikenella and Aggregatibacter were lower when compared to C group. There was statistically significance in α-diversity between E and RAU groups(p < 0.05). Significant differences in ß-diversity were detected in bacteria between E and C; NE and C; as well as E and NE groups. The LDA effect size algorithm identified the g_Haemophilus might be the potential biomarker in NE group. CONCLUSIONS: We found that salivary microbiome in erosive OLP was significantly different from that found in RAU; and these changes may be related to the underlying disease process rather than presence of ulcerative/erosive lesions clinically. In addition, our findings in bacterial relative abundance in OLP were significantly different from the previously reported findings, which points to the need for further research in salivary microbiome of OLP.


Assuntos
Bactérias/classificação , Disbiose/microbiologia , Líquen Plano Bucal/microbiologia , RNA Ribossômico 16S/genética , Análise de Sequência de DNA/métodos , Estomatite Aftosa/diagnóstico , Algoritmos , Bactérias/genética , Bactérias/isolamento & purificação , Estudos de Casos e Controles , DNA Bacteriano/genética , DNA Ribossômico/genética , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Filogenia , Saliva/microbiologia
2.
Neurotox Res ; 27(1): 55-68, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25048111

RESUMO

The accumulation of a large amount of amyloid-ß (Aß42) in brain neurons is one of the debilitating characteristics of Alzheimer's disease. In this study, we determined the effects of peroxisome proliferator-activated receptor alpha (PPARα) activation on neuronal degeneration using a model of Aß42-induced cytotoxicity. We found that 0.5 µM Aß42 induced DNA damage and apoptosis in NT2N cells after 6 h of treatment. Co-treatment of Aß42-treated cells with Wy14643, a PPARα ligand, significantly increased cell viability after 24 h compared with cells treated with Aß42 alone. There were no differences in the protein levels of caspase-3, Bcl-2/Bax or p53 between cells treated with Aß42 alone and those treated with both Aß42 and Wy14643. However, the addition of Wy14643 significantly suppressed the Aß42-induced upregulation of Endo G and AIF protein levels. Immunohistochemical analyses further demonstrated that Wy14643 reduced the expression of Endo G and AIF translocated from the cytoplasm into the nucleus, which occurred concomitantly with the decrease in DNA damage in Aß42-treated cells. Our data clearly show that PPARα activation prevents DNA damage and neuronal cell apoptosis by decreasing the expression and translocation of AIF/Endo G to the nucleus in a caspase-3- and p53-independent pathway in the NT2N cell model. This role of PPARα in promoting neuron survival suggests a possible clinical application in treating Aß42-associated neurotoxicity in Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Fator de Indução de Apoptose/metabolismo , Dano ao DNA/fisiologia , Endodesoxirribonucleases/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , PPAR alfa/metabolismo , Fragmentos de Peptídeos/toxicidade , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Humanos , PPAR alfa/agonistas , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos
3.
Nucleic Acids Res ; 42(13): 8777-88, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24990372

RESUMO

The RstA/RstB system is a bacterial two-component regulatory system consisting of the membrane sensor, RstB and its cognate response regulator (RR) RstA. The RstA of Klebsiella pneumoniae (kpRstA) consists of an N-terminal receiver domain (RD, residues 1-119) and a C-terminal DNA-binding domain (DBD, residues 130-236). Phosphorylation of kpRstA induces dimerization, which allows two kpRstA DBDs to bind to a tandem repeat, called the RstA box, and regulate the expression of downstream genes. Here we report the solution and crystal structures of the free kpRstA RD, DBD and DBD/RstA box DNA complex. The structure of the kpRstA DBD/RstA box complex suggests that the two protomers interact with the RstA box in an asymmetric fashion. Equilibrium binding studies further reveal that the two protomers within the kpRstA dimer bind to the RstA box in a sequential manner. Taken together, our results suggest a binding model where dimerization of the kpRstA RDs provides the platform to allow the first kpRstA DBD protomer to anchor protein-DNA interaction, whereas the second protomer plays a key role in ensuring correct recognition of the RstA box.


Assuntos
Proteínas de Bactérias/química , DNA Bacteriano/química , Proteínas de Ligação a DNA/química , Klebsiella pneumoniae/genética , Regiões Promotoras Genéticas , Proteínas de Bactérias/metabolismo , DNA Bacteriano/metabolismo , Proteínas de Ligação a DNA/metabolismo , Modelos Moleculares , Ligação Proteica , Multimerização Proteica , Estrutura Terciária de Proteína , Termodinâmica
4.
J Bacteriol ; 195(20): 4726-34, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23955005

RESUMO

Iron is essential for pathogen survival, virulence, and colonization. Feo is suggested to function as the ferrous iron (Fe(2+)) transporter. The enterobacterial Feo system is composed of 3 proteins: FeoB is the indispensable component and is a large membrane protein likely to function as a permease; FeoA is a small Src homology 3 (SH3) domain protein that interacts with FeoB; FeoC is a winged-helix protein containing 4 conserved Cys residues in a sequence suitable for harboring a putative iron-sulfur (Fe-S) cluster. The presence of an iron-sulfur cluster on FeoC has never been shown experimentally. We report that under anaerobic conditions, the recombinant Klebsiella pneumoniae FeoC (KpFeoC) exhibited hyperfine-shifted nuclear magnetic resonance (NMR) and a UV-visible (UV-Vis) absorbance spectrum characteristic of a paramagnetic center. The electron paramagnetic resonance (EPR) and extended X-ray absorption fine structure (EXAFS) results were consistent only with the [4Fe-4S] clusters. Substituting the cysteinyl sulfur with oxygen resulted in significantly reduced cluster stability, establishing the roles of these cysteines as the ligands for the Fe-S cluster. When exposed to oxygen, the [4Fe-4S] cluster degraded to [3Fe-4S] and eventually disappeared. We propose that KpFeoC may regulate the function of the Feo transporter through the oxygen- or iron-sensitive coordination of the Fe-S cluster.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Klebsiella pneumoniae/metabolismo , Absorciometria de Fóton , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Proteínas Ferro-Enxofre/classificação , Proteínas Ferro-Enxofre/genética , Klebsiella pneumoniae/genética , Espectroscopia de Ressonância Magnética , Oxirredução
5.
Am J Chin Med ; 41(3): 683-96, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23711149

RESUMO

Oral cancer is a common malignancy associated with high morbidity and mortality. While p38 MAPK is reported to be involved in different cellular activities such as proliferation and differentiation, reports rarely define the roles of the individual members of the p38 MAPK family in cancer. We used two unique cell lines developed by our lab representing chemically induced oral cancer cells (T28) and non-tumor cells (N28) obtained from tissues surrounding the induced cancer as a model to screen out whether p38 MAPK is involved in the malignant transformation processes. The results suggest an association between p38ß not p38α and oral cancer development. Additionally, the anti-cancer activity of thymoquinone (TQ) was screened out and we found evidences suggesting that the anti-tumor activity of TQ may be attributed to the downregulation of p38ß MAPK.


Assuntos
Apoptose/efeitos dos fármacos , Benzoquinonas/farmacologia , Proteína Quinase 11 Ativada por Mitógeno/antagonistas & inibidores , Neoplasias Bucais/tratamento farmacológico , Neoplasias de Células Escamosas/tratamento farmacológico , Nigella sativa/química , Extratos Vegetais/farmacologia , Animais , Anticarcinógenos/farmacologia , Anticarcinógenos/uso terapêutico , Antineoplásicos Fitogênicos/farmacologia , Antineoplásicos Fitogênicos/uso terapêutico , Benzoquinonas/uso terapêutico , Linhagem Celular , Linhagem Celular Tumoral , Regulação para Baixo , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Mucosa Bucal/citologia , Mucosa Bucal/efeitos dos fármacos , Neoplasias Bucais/metabolismo , Neoplasias de Células Escamosas/metabolismo , Fitoterapia , Extratos Vegetais/uso terapêutico
6.
Thyroid ; 23(7): 879-84, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23421548

RESUMO

BACKGROUND: The possibility of an association of Graves' disease (GD) with subsequent cancers has been previously reported. METHODS: Our study used the Taiwanese National Health Insurance Research Database (NHIRD), which identified 5025 newly diagnosed GD patients from 1997 to 2010, and 20,100 frequency matched non-GD patients. The risk of developing cancer for GD patients was measured using the Cox proportional hazard model. RESULTS: The incidence of developing cancer in the GD cohort was 4.92 per 1000 person-years and was 1.37-fold higher than in the comparison cohort (p<0.001). Compared with patients aged 20-34 years, older age groups demonstrated a higher risk of developing cancer (35-49 years: hazard ratio (HR)=4.15; 50-64 years: HR=7.39;≥65 years: HR=13.4). After adjusting for sex, age, and comorbidities, the HR for developing breast cancer and thyroid cancer was 1.58- and 10.4-fold higher for patients with GD. Furthermore, the incidence rates (IRR) were the highest in the first three years: 2.06 [confidence interval (CI)=1.87-2.27] and 15.6 [CI=13.9-17.5] in breast cancer and thyroid cancer with GD respectively. Specifically, a 16-fold hazard of developing thyroid cancer was present in the first three years in the GD cohort compared to the non-GD cohort [CI=7.95-32.1]. CONCLUSIONS: GD patients have a higher risk of cancer, particularly thyroid and breast cancer sequent within six and three years respectively. Strategies for preventing thyroid and breast cancer are proposed.


Assuntos
Neoplasias da Mama/etiologia , Doença de Graves/complicações , Neoplasias da Glândula Tireoide/etiologia , Adulto , Idoso , Neoplasias da Mama/epidemiologia , Comorbidade , Feminino , Doença de Graves/epidemiologia , Humanos , Incidência , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Neoplasias/epidemiologia , Neoplasias/etiologia , Modelos de Riscos Proporcionais , Fatores de Risco , Taiwan/epidemiologia , Neoplasias da Glândula Tireoide/epidemiologia
7.
Biomol NMR Assign ; 7(1): 85-8, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22481468

RESUMO

Bacterial cells often use two-component signal transduction systems to regulate genes in response to environmental stimuli. The RstA/RstB system is a two-component regulatory system consisting of the membrane sensor, RstB, and its cognate response regulator RstA. The RstA of Klebsiella pneumoniae consists of a N-terminal receiver domain (NRD, residues 1-119) and a C-terminal DNA-binding domain (DBD, residues 130-236). Phosphorylation of the response regulator induces a conformational change in the regulatory domain of RstA, which results in activation of the effector domain to regulate the downstream genes, including the ferrous iron transport system (Feo), at low-pH condition. Here we report the (1)H, (13)C and (15)N resonance assignments and secondary structure identification of the DBD of RstA from K. pneumoniae as a first step for unraveling the structural and functional relationship of the RstA/RstB two component system.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , DNA/metabolismo , Klebsiella pneumoniae , Ressonância Magnética Nuclear Biomolecular , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
8.
J Investig Med ; 60(8): 1209-13, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23076161

RESUMO

BACKGROUND: The peroxisome proliferator-activated receptor alpha (PPARA) and apolipoprotein E (APOE) proteins are reported to be correlated with lipid metabolism, cardiovascular disease, and breast cancer. METHODS: We screened APOE and PPARA (S24F and V227A) polymorphisms in 306 breast cancer patients and 300 noncancer controls and determined the relationship between their genetic polymorphisms and breast cancer risk. Interactions with clinical characteristics were also examined. RESULTS: We found that the risk of breast cancer was associated with APOE genotypes (P = 0.014) but not with PPARA S24F or V227A genotypes. The combined effects of F24/APOE genotypes (P = 0.003) on breast cancer risk were more significant than the individual effect of APOE genotypes (P = 0.014). F24/ε4 carriers had a higher tendency to develop breast cancer than F24/ε3 carriers (P = 0.013), and this effect is stronger than with individual ε4 carriers (P = 0.029). In addition, both F24/ε4 and V227/ε4 carriers were significantly enriched in the human epidermal growth factor receptor 2/neu negative status. CONCLUSIONS: These findings suggest that the APOE ε4 genotype plays a major role in the prediction of breast cancer, but the PPARA F24 mutation enhances this outcome. The combined effects of F24/ε4 genotypes are positively associated with risk of breast cancer.


Assuntos
Apolipoproteínas E/genética , Povo Asiático/genética , Neoplasias da Mama/genética , Variação Genética/genética , PPAR alfa/genética , Polimorfismo Genético/genética , Adulto , Idoso , Neoplasias da Mama/etnologia , Feminino , Humanos , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores de Risco , Taiwan/etnologia
9.
Toxicol Appl Pharmacol ; 263(3): 360-7, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22820424

RESUMO

Baicalein is a flavonoid, known to have anti-inflammatory and anti-cancer effects. As an aryl hydrocarbon receptor (AhR) ligand, baicalein at high concentrations blocks AhR-mediated dioxin toxicity. Because AhR had been reported to play a role in regulating the cell cycle, we suspected that the anti-cancer effect of baicalein is associated with AhR. This study investigated the molecular mechanism involved in the anti-cancer effect of baicalein in oral cancer cells HSC-3, including whether such effect would be AhR-mediated. Results revealed that baicalein inhibited cell proliferation and increased AhR activity in a dose-dependent manner. Cell cycle was arrested at the G1 phase and the expression of CDK4, cyclin D1, and phosphorylated retinoblastoma (pRb) was decreased. When the AhR was suppressed by siRNA, the reduction of pRb was partially reversed, accompanied by a decrease of cell population at G1 phase and an increase at S phase, while the reduction of cyclin D1 and CDK4 did not change. This finding suggests that the baicalein activation of AhR is indeed associated with the reduction of pRb, but is independent of the reduction of cyclin D1 and CDK4. When cells were pre-treated with LiCl, the inhibitor of GSK-3ß, the decrease of cyclin D1 was blocked and the reduction of pRb was recovered. The data indicates that in HSC-3 the reduction of pRb is both mediated by baicalein through activation of AhR and facilitation of cyclin D1 degradation, which causes cell cycle arrest at the G1 phase, and results in the inhibition of cell proliferation.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Flavanonas/farmacologia , Neoplasias Bucais/tratamento farmacológico , Receptores de Hidrocarboneto Arílico/metabolismo , Antineoplásicos Fitogênicos/administração & dosagem , Carcinoma de Células Escamosas/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina D1/efeitos dos fármacos , Ciclina D1/metabolismo , Relação Dose-Resposta a Droga , Flavanonas/administração & dosagem , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Bucais/patologia , Fosforilação/efeitos dos fármacos , Proteína do Retinoblastoma/metabolismo
10.
Toxicol Appl Pharmacol ; 259(3): 293-301, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22273977

RESUMO

Environmental cigarette smoke has been suggested to promote lung adenocarcinoma progression through aryl hydrocarbon receptor (AhR)-signaled metabolism. However, whether AhR facilitates metabolic activation or detoxification in exposed adenocarcinoma cells remains ambiguous. To address this question, we have modified the expression level of AhR in two human lung adenocarcinoma cell lines and examined their response to an extract of cigarette sidestream smoke particulates (CSSP). We found that overexpression of AhR in the CL1-5 cell line reduced CSSP-induced ROS production and oxidative DNA damage, whereas knockdown of AhR expression increased ROS level in CSSP-exposed H1355 cells. Oxidative stress sensor Nrf2 and its target gene NQO1 were insensitive to AhR expression level and CSSP treatment in human lung adenocarcinoma cells. In contrast, induction of AhR expression concurrently increased mRNA expression of xenobiotic-metabolizing genes CYP1B1, UGT1A8, and UGT1A10 in a ligand-independent manner. It appeared that AhR accelerated xenobiotic clearing and diminished associated oxidative stress by coordinate regulation of a set of phase I and II metabolizing genes. However, the AhR-signaled protection could not shield cells from constant oxidative stress. Prolonged exposure to high concentrations of CSSP induced G0/G1 cell cycle arrest via the p53-p21-Rb1 signaling pathway. Despite no effect on DNA repair rate, AhR facilitated the recovery of cells from growth arrest when CSSP exposure ended. AhR-overexpressing lung adenocarcinoma cells exhibited an increased anchorage-dependent and independent proliferation when recovery from exposure. In summary, our data demonstrated that AhR protected lung adenocarcinoma cells against CSSP-induced oxidative stress and promoted post-exposure clonogenicity.


Assuntos
Adenocarcinoma/metabolismo , Neoplasias Pulmonares/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Fumaça/efeitos adversos , Poluição por Fumaça de Tabaco/efeitos adversos , Hidrocarboneto de Aril Hidroxilases/genética , Linhagem Celular Tumoral , Proliferação de Células , Citocromo P-450 CYP1B1 , Dano ao DNA/efeitos dos fármacos , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glucuronosiltransferase/genética , Humanos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Xenobióticos/metabolismo
12.
Toxicol In Vitro ; 22(8): 1832-9, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18801422

RESUMO

The objective was to investigate the regulation of glutathione (GSH) turnover in porcine aortic endothelial cells (PAECs) treated with sodium arsenite (NaAsO(2)), arsenic trioxide (As(2)O(3)) or sodium arsenate (Na(2)HAsO(4)) up to 72 hr at 0, 1, 5, and 10 microM, respectively. Intracellular GSH and glutathione disulfide (GSSG) contents, as well as the activities and mRNA levels of glutamate-cysteine lyase (GCL; gamma-glutamylcysteine synthetase) and gamma-glutamyl transpeptidase (GGT), were examined. The trivalent arsenic compounds increased GSH and GSSG contents in PAECs. An increase in GCL activity was observed at 24hr whereas an increase in GCL mRNA level was observed at 72 hr. The increase in GGT activity was only observed at 72 hr. In addition, a tendency of increase in GGT mRNA level was observed. Na(2)HAsO(4) treatment did not affect GSH content and the turnover-related enzymes. A differential GSH modulation in PAECs by trivalent arsenic compounds was found. The regulatory mechanism responsible for the As(2)O(3)-induced GSH increase is related to the GSH-turnover enzymes, GCL and GGT, while that for the NaAsO(2)-induced GSH increase may not be related to expression of GSH-turnover enzymes.


Assuntos
Arseniatos/toxicidade , Arsenitos/toxicidade , Glutationa/efeitos dos fármacos , Óxidos/toxicidade , Compostos de Sódio/toxicidade , Animais , Aorta Torácica/citologia , Aorta Torácica/efeitos dos fármacos , Trióxido de Arsênio , Arsenicais , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Glutamato-Cisteína Ligase/efeitos dos fármacos , Glutamato-Cisteína Ligase/metabolismo , Glutationa/metabolismo , Dissulfeto de Glutationa/efeitos dos fármacos , Dissulfeto de Glutationa/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Suínos , Fatores de Tempo , gama-Glutamiltransferase/efeitos dos fármacos , gama-Glutamiltransferase/metabolismo
13.
Toxicol Sci ; 89(1): 205-13, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16237193

RESUMO

Both benzo[a]pyrene (BaP) and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) are potent ligands of aryl hydrocarbon receptors (AhR). Although animal studies indicate that both compounds induce pathological changes in the peripheral lung, the specific cell type involved remains unclear. Clara cells, expressing Clara cell specific protein (CCSP) and abundant in cytochrome P450, are nonciliated bronchiolar epithelial cells in the peripheral lung. Here we explore the hypothesis that CCSP-positive Clara cells are highly responsive to AhR ligands and are the primary cell type involved in BaP- and TCDD-induced toxicities. The responsiveness to AhR ligands was evaluated by measuring the respective mRNA and protein levels of cytochrome P450 1A1 (CYP1A1) and 1B1 (CYP1B1) using real-time RT-PCR and immunocytochemistry assays. Two in vitro models were used: primary cultures of human small airway epithelial (SAE) cells and rat lung slice cultures. In the presence of calcium, human SAE cells differentiated into CCSP-positive cells. BaP- and TCDD-induced mRNA and protein levels of CYP1A1 and CYP1B1 levels were significantly elevated in CCSP-positive cell cultures. Similarly, AhR mRNA and protein levels were increased in CCSP-positive cell cultures, as determined by real-time RT-PCR and Western blot analysis. When rat lung slice cultures were treated with BaP or TCDD for 24 h, CYP1A1 and CYP1B1 proteins were strongly induced in Clara cells. These results indicate that, in the peripheral lung of both rats and humans, CCSP-positive cells (Clara cells) may be more sensitive to AhR ligands than other cell types.


Assuntos
Hidrocarboneto de Aril Hidroxilases/biossíntese , Citocromo P-450 CYP1A1/biossíntese , Pulmão/enzimologia , Mucosa Respiratória/enzimologia , Uteroglobina/metabolismo , Animais , Hidrocarboneto de Aril Hidroxilases/genética , Benzo(a)pireno/farmacologia , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1B1 , Indução Enzimática/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Expressão Gênica/efeitos dos fármacos , Humanos , Pulmão/citologia , Pulmão/efeitos dos fármacos , Masculino , Dibenzodioxinas Policloradas/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Mucosa Respiratória/citologia , Mucosa Respiratória/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Toxicol Appl Pharmacol ; 195(1): 83-91, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14962508

RESUMO

Epidemiological studies on 2,3,7,8,-tetrachlorodibenzo-p-dioxin (TCDD) exposure indicated high incidences of pulmonary dysfunctions and lung cancer. Animal studies also demonstrated lung cancer development in female, but not in male, rats exposed to TCDD. Such effects, however, have not been reported in polychlorinated biphenyls (PCB) exposure. In our present study, we have investigated the effects of TCDD and PCB126, with or without cotreatment with 17 beta-estradiol (E2), on a human bronchial epithelial cell line BEAS-2B. We found that treatment with either TCDD or PCB126 alone reduced cell numbers as well as thymidine incorporation. Cell death, however, was only detected in PCB126-, but not TCDD-, treated cultures. The TCDD-induced cell reduction, therefore, could not be contributed to cell death. Meanwhile, because TCDD- and PCB126-enhanced CYP1A1 and CYP1B1 expressions were significantly reduced by the AhR antagonist and CYP1 inhibitor alpha-naphthoflavone (ANF), this indicated that the effects of TCDD and PCB126 were AhR and cytochrome p450 1 dependent. We also found that while E2 itself did not alter CYP1A1 and CYP1B1 expressions, cotreatment of E2 with TCDD or PCB126 would significantly enhance TCDD-, but not PCB126-, induced toxicity. We further demonstrated that in the presence of E2, 1 nM TCDD increased the production of E2 metabolites, 2-methoxyestradiol (2-MeOE2) and 4-methoxyestradiol (4-MeOE2). PCB126, however, only increased 2-MeOE2 formation without significant induction of 4-MeOE2. We believe that these metabolites, especially 4-MeOE2, interacted with TCDD to further suppress cell growth. Our data provided the first demonstration on the enhancement of TCDD-induced toxicity in human lung cells via interaction with estrogen.


Assuntos
Brônquios/citologia , Células Epiteliais/efeitos dos fármacos , Estradiol/análogos & derivados , Estradiol/metabolismo , Bifenilos Policlorados/toxicidade , Dibenzodioxinas Policloradas/toxicidade , 2-Metoxiestradiol , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Contagem de Células , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...