Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Phytomedicine ; 129: 155548, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38583347

RESUMEN

BACKGROUND: Oral leukoplakia (OLK), characterized by abnormal epithelial hyperplasia, is the most common precancerous oral mucosa lesion and is closely related to oxidative stress. Cucurbitacin B (CuB), a tetracyclic triterpenoid molecule derived from plants, has shown promising anti-proliferative and antioxidant effects in preclinical studies. However, whether CuB can play an antiproliferative role in OLK by regulating oxidative stress remains elusive. PURPOSE: To investigate the role of CuB in inhibiting the malignant progression of oral leukoplakia and to further explore its underlying mechanisms of action. METHODS: In vitro, the effect of CuB on the proliferation, migration, apoptosis, and cell cycle of OLK cells DOK was detected. The core genes and key pathways of OLK and CuB were analyzed in the transcriptome database, by using immunofluorescence, qRT-PCR, and Western blot to evaluate the expression levels of the ferroptosis markers ROS, GSH, MDA, Fe2+, and marker genes SLC7A11, GPX4, and FTH1. Immunohistochemistry of human tissue was performed to investigate the expression of the SLC7A11. In vivo, the model of OLK was established in C57BL/6 mice and the biosafety of CuB treatment for OLK was further evaluated. RESULTS: CuB substantially suppressed the proliferation of DOK cells. Bioinformatics analysis showed that the core targets of OLK crossing with CuB include SLC7A11 and that the essential pathways involve ROS and ferroptosis. In vitro experiments indicated that CuB might promote ferroptosis by down-regulating the expression of SLC7A11. We observed a gradual increase in SLC7A11 expression levels during the progression from normal oral mucosa to oral leukoplakia with varying degrees of epithelial dysplasia. In vivo experiments demonstrated that CuB inhibited the malignant progression of OLK by promoting ferroptosis in OLK mice and exhibited a certain level of biosafety. CONCLUSION: This study demonstrated for the first time that CuB could effectively inhibit the malignant progression of OLK by inducing ferroptosis via activating the SLC7A11/ mitochondrial oxidative stress pathway. These findings indicate that CuB could serve as the lead compound for the future development of anti-oral leukoplakia drugs.

2.
Hereditas ; 161(1): 10, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38414024

RESUMEN

BACKGROUND: The novel form of regulatory cell death, cuproptosis, is characterized by proteotoxicity, which ultimately leads to cell death. Its targeting has emerged as a promising therapeutic approach for oral squamous cell carcinoma (OSCC). Long noncoding RNAs (lncRNAs) participate in epigenetic regulation and have been linked to the progression, prognosis, and treatment of OSCC. Thus, this study aimed to identify new cuproptosis-related lncRNAs (CRLs), establish predictive models for clinical prognosis, immune response, and drug sensitivity, and provide novel insights into immune escape and tumor drug resistance. METHODS: The present study screened eight CRLs (THAP9-AS1, STARD4-AS1, WDFY3-AS2, LINC00847, CDKN2A-DT, AL132800.1, GCC2-AS1, AC005746.1) using Lasso Cox regression analysis to develop an eight-CRL prognostic model. Patients were categorized into high- and low-risk groups using risk scores. To evaluate the predictive ability of the model, Kaplan-Meier analysis, ROC curves, and nomograms were employed. Furthermore, the study investigated the differences in immune function and anticancer drug sensitivity between the high- and low-risk groups. To validate the expression of CRLs in the model, OSCC cell lines were subjected to quantitative real-time fluorescence PCR (qRT-PCR). RESULTS: The results of the study showed that the high-risk group had a shorter overall survival (OS) time in OSCC patients. Cox regression analysis demonstrated that the high-risk score was an independent risk factor for a poor prognosis. The validity of the model was confirmed using ROC curve analysis, and a nomogram was developed to predict the prognosis of OSCC patients. Furthermore, patients in the high-risk group with high TMB had a poorer prognosis. Patients in the low-risk group responded better to immunotherapy than those in the high-risk group. Additionally, the risk scores were significantly associated with drug sensitivity in OSCC patients. Finally, the findings of qRT-PCR supported the reliability of the proposed risk model. CONCLUSION: The study identified and established the 8-CRL model, which represents a novel pathway of lncRNA regulation of cuproptosis in OSCC. This model provides guidance for the prognosis and treatment of OSCC and offers a new insight into immune escape and tumor drug resistance.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , ARN Largo no Codificante , Humanos , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/terapia , Carcinoma de Células Escamosas de Cabeza y Cuello , ARN Largo no Codificante/genética , Epigénesis Genética , Reproducibilidad de los Resultados , Neoplasias de la Boca/genética , Neoplasias de la Boca/terapia , Pronóstico , Biomarcadores , Inmunoterapia , Biología Computacional , Apoptosis
3.
Mol Divers ; 2023 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-37615817

RESUMEN

Oral squamous cell carcinoma (OSCC) is a malignant tumor with a high incidence and poor prognosis. Cucurbitacin B (CuB) is a tetracyclic triterpenoid small-molecule compound extracted from plants, such as Cucurbitaceae and Brassicaceae, which has powerful anticancer effects. However, the effect and mechanism of CuB on OSCC remain unclear. Within the framework of the current study, network pharmacology was used to analyze the relationship between CuB and OSCC. The network pharmacology analysis showed that CuB and OSCC share 134 common targets; among them, PIK3R1, SRC, STAT3, AKT1, and MAPK1 are the key targets. The molecular docking analysis showed that CuB binds five target proteins. The results of the enrichment analysis showed that CuB exerted effects on OSCC through various pathways; of these pathways, PI3K-AKT was the most important pathway. The results of the in vitro cell experiments showed that CuB could inhibit the proliferation and migration of SCC25 and CAL27 cells, block the cell cycle in the G2 phase, induce cell apoptosis, and regulate the protein expression of the PI3K-AKT signaling pathway. The results of the in vivo animal experiments showed that CuB could inhibit 4NQO-induced oral cancer in mice. Therefore, network pharmacology, molecular docking, cell experiments, and animal experiments showed that CuB could play a role in OSCC by regulating multiple targets and pathways.

4.
Phytomedicine ; 103: 154195, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35667260

RESUMEN

BACKGROUND: Oral leukoplakia (OLK), an uncharacterized pathological condition that occurs as a white patch in the oral mucosa, is the most common precancerous condition. Scutellaria baicalensis Georgi (SBG) is a medicinal plant with a wide range of pharmacological effects. Increased evidence shows that SBG has potential therapeutic effects on OLK. However, the therapeutic mechanisms of SBG against OLK have not yet been completely elucidated. PURPOSE: This study aimed to clarify the active components and multi-target mechanisms of SBG against OLK via network pharmacology, molecular docking and experimental evaluations. STUDY DESIGN AND METHODS: The active components and related targets of SBG were screened by the TCMSP database and Swiss Target Prediction database. Potential therapeutic targets of OLK were collected using the GeneCards and OMIM databases. Then, we established protein-protein interaction (PPI), compound-target-disease (C-T-D), and compound-target-pathway (C-T-P) networks by Cytoscape to identify the main components, core targets, and pharmacological pathways of SBG against OLK via applying data mining techniques and topological parameters. Metascape database was utilized for GO and KEGG pathway analysis. Molecular docking techniques were used to estimate the binding force between the components and the hub genes. Subsequently, a series of in vitro experiments, specifically CCK-8 assay, clone formation assay, wound healing assay, flow cytometry, RT-qPCR and western blotting were conducted for further verification. RESULTS: There were 25 active components and 31 related target genes in SBG against OLK. PPI analysis showed that Akt1, VEGFA, EGFR, HIF1A and PTGS2 shared the highest centrality among all target genes. KEGG pathway analysis found that PI3K-Akt signaling pathway may occupy core status in the anti-OLK system. Molecular docking results showed that the main active components of SBG had a strong binding affinity to the hub genes. In vitro experiments showed that the leading component baicalein may inhibit proliferation, block cells in the S phase, induce DOK cell apoptosis, and downregulate the mRNA expression of 5 hub genes by inhibiting PI3K/Akt signaling pathway activation. CONCLUSION: The most predominant component of SBG against OLK was baicalein and the key pathway was PI3K/Akt. The main components and hub genes had robust binding abilities. In vitro experiments showed that baicalein could inhibit the proliferation of DOK cells, induce apoptosis, block the cell cycle, and inhibit the mRNA expression level of the hub genes by inhibiting the PI3K/Akt pathway.


Asunto(s)
Medicamentos Herbarios Chinos , Scutellaria baicalensis , Medicamentos Herbarios Chinos/farmacología , Leucoplasia Bucal , Simulación del Acoplamiento Molecular , Farmacología en Red , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , ARN Mensajero , Scutellaria baicalensis/química
5.
Artículo en Inglés | MEDLINE | ID: mdl-34335829

RESUMEN

BACKGROUND: Scutellaria baicalensis Georgi (SBG) has been widely shown to induce apoptosis and inhibit invasion and migration of various cancer cells. Increased evidence shows that SBG may be useful to treat oral squamous cell carcinoma (OSCC). However, the biological activity and possible mechanisms of SBG in the treatment of OSCC have not been fully elucidated. This study aimed to clarify the bioactive component and multitarget mechanisms of SBG against OSCC using network pharmacology and molecular docking. METHODS: Traditional Chinese Medicine Systems Pharmacology (TCMSP) database was used to predict the active components in SBG, and putative molecular targets of SBG were identified using the Swiss Target Prediction database. OSCC-related targets were screened by GeneCards, Online Mendelian Inheritance in Man (OMIM), and Therapeutic Target Database (TTD). Then, we established protein-protein interaction (PPI), compound-target-disease (C-T-D), and compound-target-pathway (C-T-P) networks by Cytoscape to identify the main components, core targets, and pharmacological pathways of SBG against OSCC via applying data mining techniques and topological parameters. Metascape database was utilized for Gene Ontology (GO) and pathway enrichment analysis. The potential interaction of the main components with core targets was revealed by molecular docking simulation, and for the correlation between core targets and OSCC prognosis analysis, the Kaplan-Meier Plotter online database was used. RESULTS: There were 25 active compounds in SBG and 86 genes targeted by OSCC. A total of 141 signaling pathways were identified, and it was found that the PI3K-Akt signaling pathway may occupy core status in the anti-OSCC system. GO analysis revealed that the primary biological processes were related to apoptosis, proliferation, and migration. Molecular docking results confirmed that core targets of OSCC had a high affinity with the main compounds of SBG. CONCLUSION: Our study demonstrated multicomponent, multitarget, and multipathway characteristics of SBG in the treatment of OSCC and provided a foundation for further drug development research.

6.
BMC Microbiol ; 20(1): 75, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32245419

RESUMEN

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.


Asunto(s)
Bacterias/clasificación , Disbiosis/microbiología , Liquen Plano Oral/microbiología , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN/métodos , Estomatitis Aftosa/diagnóstico , Algoritmos , Bacterias/genética , Bacterias/aislamiento & purificación , Estudios de Casos y Controles , ADN Bacteriano/genética , ADN Ribosómico/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Filogenia , Saliva/microbiología
7.
Neurotox Res ; 27(1): 55-68, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25048111

RESUMEN

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.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Factor Inductor de la Apoptosis/metabolismo , Daño del ADN/fisiología , Endodesoxirribonucleasas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , PPAR alfa/metabolismo , Fragmentos de Péptidos/toxicidad , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Humanos , PPAR alfa/agonistas , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos
8.
Nucleic Acids Res ; 42(13): 8777-88, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24990372

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/química , ADN Bacteriano/química , Proteínas de Unión al ADN/química , Klebsiella pneumoniae/genética , Regiones Promotoras Genéticas , Proteínas Bacterianas/metabolismo , ADN Bacteriano/metabolismo , Proteínas de Unión al ADN/metabolismo , Modelos Moleculares , Unión Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , Termodinámica
9.
J Bacteriol ; 195(20): 4726-34, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23955005

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Hierro-Azufre/metabolismo , Klebsiella pneumoniae/metabolismo , Absorciometría de Fotón , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica/fisiología , Proteínas Hierro-Azufre/clasificación , Proteínas Hierro-Azufre/genética , Klebsiella pneumoniae/genética , Espectroscopía de Resonancia Magnética , Oxidación-Reducción
10.
Am J Chin Med ; 41(3): 683-96, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23711149

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzoquinonas/farmacología , Proteína Quinasa 11 Activada por Mitógenos/antagonistas & inhibidores , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de Células Escamosas/tratamiento farmacológico , Nigella sativa/química , Extractos Vegetales/farmacología , Animales , Anticarcinógenos/farmacología , Anticarcinógenos/uso terapéutico , Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/uso terapéutico , Benzoquinonas/uso terapéutico , Línea Celular , Línea Celular Tumoral , Regulación hacia Abajo , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Mucosa Bucal/citología , Mucosa Bucal/efectos de los fármacos , Neoplasias de la Boca/metabolismo , Neoplasias de Células Escamosas/metabolismo , Fitoterapia , Extractos Vegetales/uso terapéutico
11.
Thyroid ; 23(7): 879-84, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23421548

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/etiología , Enfermedad de Graves/complicaciones , Neoplasias de la Tiroides/etiología , Adulto , Anciano , Neoplasias de la Mama/epidemiología , Comorbilidad , Femenino , Enfermedad de Graves/epidemiología , Humanos , Incidencia , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Neoplasias/epidemiología , Neoplasias/etiología , Modelos de Riesgos Proporcionales , Factores de Riesgo , Taiwán/epidemiología , Neoplasias de la Tiroides/epidemiología
12.
Biomol NMR Assign ; 7(1): 85-8, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22481468

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , ADN/metabolismo , Klebsiella pneumoniae , Resonancia Magnética Nuclear Biomolecular , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
13.
J Investig Med ; 60(8): 1209-13, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23076161

RESUMEN

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.


Asunto(s)
Apolipoproteínas E/genética , Pueblo Asiatico/genética , Neoplasias de la Mama/genética , Variación Genética/genética , PPAR alfa/genética , Polimorfismo Genético/genética , Adulto , Anciano , Neoplasias de la Mama/etnología , Femenino , Humanos , Persona de Mediana Edad , Estudios Retrospectivos , Factores de Riesgo , Taiwán/etnología
14.
Toxicol Appl Pharmacol ; 263(3): 360-7, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22820424

RESUMEN

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.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Flavanonas/farmacología , Neoplasias de la Boca/tratamiento farmacológico , Receptores de Hidrocarburo de Aril/metabolismo , Antineoplásicos Fitogénicos/administración & dosificación , Carcinoma de Células Escamosas/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D1/efectos de los fármacos , Ciclina D1/metabolismo , Relación Dosis-Respuesta a Droga , Flavanonas/administración & dosificación , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias de la Boca/patología , Fosforilación/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo
15.
Toxicol Appl Pharmacol ; 259(3): 293-301, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22273977

RESUMEN

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.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias Pulmonares/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Humo/efectos adversos , Contaminación por Humo de Tabaco/efectos adversos , Hidrocarburo de Aril Hidroxilasas/genética , Línea Celular Tumoral , Proliferación Celular , Citocromo P-450 CYP1B1 , Daño del ADN/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glucuronosiltransferasa/genética , Humanos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Xenobióticos/metabolismo
17.
Toxicol In Vitro ; 22(8): 1832-9, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18801422

RESUMEN

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.


Asunto(s)
Arseniatos/toxicidad , Arsenitos/toxicidad , Glutatión/efectos de los fármacos , Óxidos/toxicidad , Compuestos de Sodio/toxicidad , Animales , Aorta Torácica/citología , Aorta Torácica/efectos de los fármacos , Trióxido de Arsénico , Arsenicales , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Glutamato-Cisteína Ligasa/efectos de los fármacos , Glutamato-Cisteína Ligasa/metabolismo , Glutatión/metabolismo , Disulfuro de Glutatión/efectos de los fármacos , Disulfuro de Glutatión/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Porcinos , Factores de Tiempo , gamma-Glutamiltransferasa/efectos de los fármacos , gamma-Glutamiltransferasa/metabolismo
18.
Toxicol Appl Pharmacol ; 220(3): 271-7, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17397890

RESUMEN

Epidemiological studies indicated that people exposed to dioxins were prone to the development of lung diseases including lung cancer. Animal studies demonstrated that 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) increased liver tumors and promoted lung metaplasia in females. Metabolic changes in 17beta-estradiol (E(2)) resulted from an interaction between TCDD and E(2) could be associated with gender difference. Previously, we reported that methoxylestradiols (MeOE(2)), especially 4-MeOE(2), accumulated in human lung cells (BEAS-2B) co-treated with TCDD and E(2). In the present study, we demonstrate unique accumulation of 4-MeOE(2), as a result of TCDD/E(2) interaction and revealed its bioactivity in human lung epithelial cell line (H1355). 4-Methoxyestradiol treatment significantly decreased cell growth and increased mitotic index. Elevation of ROS and SOD activity, with a concomitant decrease in the intracellular GSH/GSSG ratio, was also detected in 4-MeOE(2)-treated cells. Quantitative comet assay showed increased oxidative DNA damage in the 4-MeOE(2)-treated H1355 cells, which could be significantly reduced by the anti-oxidant N-acetylcysteine (NAC). However, inhibition of cell growth and increase in mitotic arrest induced by 4-MeOE(2) were unaffected by NAC. We concluded that 4-MeOE(2) accumulation resulting from TCDD and E(2) interaction would contribute to the higher vulnerability on lung pathogenesis in females when exposed to TCDD.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Estradiol/análogos & derivados , Estrés Oxidativo/efectos de los fármacos , Acetilcisteína/farmacología , Línea Celular , Proliferación Celular/efectos de los fármacos , Ensayo Cometa , Daño del ADN , Relación Dosis-Respuesta a Droga , Células Epiteliales/metabolismo , Células Epiteliales/patología , Estradiol/metabolismo , Estradiol/farmacología , Femenino , Depuradores de Radicales Libres/farmacología , Disulfuro de Glutatión/metabolismo , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Índice Mitótico , Modelos Biológicos , Dibenzodioxinas Policloradas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Huso Acromático/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Tubulina (Proteína)/efectos de los fármacos
19.
Arch Toxicol ; 80(6): 310-8, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16328441

RESUMEN

Survivin is a member of the inhibitors of apoptosis protein (IAP) family and is highly expressed in various cancer cells. However, the molecular mechanisms regulating survivin expression remain unclear. In this study, we investigated the role of mitogen-activated protein kinases (MAPKs) in regulating survivin in the human lung adenocarcinoma cell line H1355 in response to arsenic trioxide (As(3+)). Our data indicated that As(3+) induced cytotoxicity accompanied by down-regulation of survivin, cleavage of Poly ADP-ribosyl polymerase (PARP) and activations of MAPKs, including ERK1/2, p38 and c-jun N-terminal kinase (JNK). We found that blockage of p38 or JNK activation attenuated the As(3+)-induced survivin down-regulation and PARP cleavage with significant reversal of cell viability, however, by only 5-8%. On the other hand, the MEK inhibitor PD098059 or the ubiquitin-proteasome inhibitor MG-132 exhibited little effect on survivin down-regulation and PARP cleavage induced by As(3+). In this study, we demonstrated that As(3+) could down-regulate survivin via activations of p38 and JNK in an ubiquitin-proteasome independent pathway and lead to cytotoxicity and apoptosis in the human lung adenocarcinoma cell line H1355.


Asunto(s)
Adenocarcinoma/metabolismo , Antineoplásicos/toxicidad , Neoplasias Pulmonares/metabolismo , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas de Neoplasias/biosíntesis , Óxidos/toxicidad , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Apoptosis/efectos de los fármacos , Trióxido de Arsénico , Arsenicales , Línea Celular Tumoral/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Antagonismo de Drogas , Humanos , Proteínas Inhibidoras de la Apoptosis , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Survivin
20.
Toxicol Sci ; 89(1): 205-13, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16237193

RESUMEN

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.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/biosíntesis , Citocromo P-450 CYP1A1/biosíntesis , Pulmón/enzimología , Mucosa Respiratoria/enzimología , Uteroglobina/metabolismo , Animales , Hidrocarburo de Aril Hidroxilasas/genética , Benzo(a)pireno/farmacología , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1B1 , Inducción Enzimática/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Expresión Génica/efectos de los fármacos , Humanos , Pulmón/citología , Pulmón/efectos de los fármacos , Masculino , Dibenzodioxinas Policloradas/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Mucosa Respiratoria/citología , Mucosa Respiratoria/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...