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










Base de datos
Intervalo de año de publicación
2.
Cell Div ; 16(1): 8, 2021 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-34922589

RESUMEN

BACKGROUND: Heat shock factor 1 (HSF1) is the master regulator of the heat shock response and supports malignant cell transformation. Recent work has shown that HSF1 can access the promoters of heat shock proteins (HSPs) and allow HSP expression during mitosis. It also acts as a mitotic regulator, controlling chromosome segregation. In this study, we investigated whether the transactivation activity of HSF1 is required for the assembly of mitotic spindles. RESULTS: Our results showed that phosphorylation of HSF1 at serine 326 (S326) and its transactivation activity were increased during mitosis. Inhibition of the transactivation activity of HSF1 by KRIBB11 or CCT251263 during mitosis significantly increased the proportion of mitotic cells with abnormal spindles. It also hampered the reassembly of spindle microtubules after nocodazole treatment and washout by impeding the formation of chromosomal microtubule asters. Depletion of HSF1 led to defects in mitotic spindle assembly, subsequently attenuating cell proliferation and anchorage-independent cell growth (AIG). These HSF1 depletion-induced effects could be rescued by ectopically expressing wild-type HSF1 or a constitutively active mutant (∆202-316, caHSF1) but not the S326A or dominant negative (∆361-529, dnHSF1) mutants. In addition, overexpression of HSP70 partially reduced HSF1 depletion-induced spindle abnormalities. These results indicate that HSF1 may support cell proliferation and AIG by maintaining spindle integrity through its transactivation activity. Furthermore, inhibition of HSF1 transactivation activity by KRIBB11 or CCT251236 can enhance diverse anti-mitosis drug-induced spindle defects and cell death. CONCLUSIONS: The increased transactivation activity of HSF1 during mitosis appears to be required for accurate assembly of mitotic spindles, thereby supporting cell viability and probably AIG. In addition, inhibition of the transactivation activity of HSF1 may enhance the mitotic errors and cell death induced by anti-mitosis drugs.

3.
Front Cardiovasc Med ; 8: 738061, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34692790

RESUMEN

With an aging world population, risk stratification of community-based, elderly population is required for primary prevention. This study proposes a combined score developed using electrocardiographic (ECG) parameters and determines its long-term prognostic value for predicting risk of cardiovascular mortality. A cohort-study, conducted from December 2008 to April 2019, enrolled 5,380 subjects in Taiwan, who were examined, using three-serial-12-lead ECGs, and their health/demographic information were recorded. To understand the predictive effects of ECG parameters on overall-survival, Cox hazard regression analysis were performed. The mean age at enrollment was 69.04 ± 8.14 years, and 47.4% were males. ECG abnormalities, LVH [hazard ratio (HR) = 1.39, 95% confidence intervals (CI) = (1.16-1.67), P = 0.0003], QTc [HR = 1.31, CI = (1.07-1.61), P = 0.007] and PR interval [HR = 1.40, CI = (1.01-1.95), P = 0.04], were significantly associated with primary outcome all-cause death. Furthermore, LVH [HR = 2.37, CI = (1.48-3.79), P = 0.0003] was significantly associated with cardiovascular death, while PR interval [HR = 2.63, CI = (1.24- 5.57), P = 0.01] with unexplained death. ECG abnormality (EA) score was defined based on the number of abnormal ECG parameters for each patient, which was used to divide all patients into sub-groups. Competing risk survival analysis using EA score were performed by using the Gray's test, which reported that high-risk EA groups showed significantly higher cumulative incidence for all three outcomes. Prognostic models using the EA score as predictor were developed and a 10-fold cross validation design was adopted to conduct calibration and discrimination analysis, to establish the efficacy of the proposed models. Overall, ECG model could successfully predict people, susceptible to all three death outcomes (P < 0.05), with high efficacy. Statistically significant (P < 0.001) improvement of the c-indices further demonstrated the robustness of the prediction model with ECG parameters, as opposed to a traditional model with no EA predictor. The EA score is highly associated with increased risk of mortality in elderly population and may be successfully used in clinical practice.

4.
Circ Genom Precis Med ; 13(4): e002797, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32490690

RESUMEN

BACKGROUND: Brugada syndrome (BrS) is an oligogenic arrhythmic disease with increased risk of sudden cardiac arrest. Several BrS or ECG traits-related single-nucleotide polymorphisms (SNPs) were identified through previous genome-wide association studies in white patients. We aimed to validate these SNPs in BrS patients in the Taiwanese population, assessing the cumulative effect of risk alleles and the BrS-polygenic risk score in predicting cardiac events. METHODS: We genotyped 190 unrelated BrS patients using the TWB Array, and Taiwan Biobank was used as controls. SNPs not included in the array were imputed by IMPUTE2. Cox proportional hazards model was used to evaluate the associations between each particular SNP, the collective BrS-polygenic risk score, and clinical outcomes. RESULTS: Of the 88 previously reported SNPs, 22 were validated in Taiwanese BrS patients (P<0.05). Of the 22 SNPs, 2 (rs10428132 and rs9388451) were linked with susceptibility to BrS, 10 were SNPs previously reaching genome-wide significance, and 10 were SNPs associated with ECG traits. For the 3 most commonly reported SNPs, disease risk increased consistently with the number of risk alleles (odds ratio, 3.54; Ptrend=1.38×10-9 for 5 risk alleles versus 1). Similar patterns were observed in both SCN5A mutation+ (odds ratio, 3.66; Ptrend=0.049) and SCN5A mutation- (odds ratio, 3.75; Ptrend=8.54×10-9) subgroups. Furthermore, BrS patients without SCN5A mutations had more risk alleles than BrS patients with SCN5A mutations regardless of the range of polygenic risk scores. Three SNPs (rs4687718, rs7784776, and rs2968863) showed significant associations with the composite outcome (sudden cardiac arrest plus syncope, hazard ratio, 2.13, 1.48, and 0.41; P=0.02, 0.006, and 0.008, respectively). CONCLUSIONS: Our findings suggested that some SNPs associated with BrS or ECG traits exist across multiple populations. The cumulative risk of the BrS-related SNPs is similar to that in white BrS patients, but it appears to correlate with the absence of SCN5A mutations.


Asunto(s)
Síndrome de Brugada/genética , Estudio de Asociación del Genoma Completo , Adulto , Alelos , Pueblo Asiatico/genética , Síndrome de Brugada/patología , Estudios de Casos y Controles , Electrocardiografía , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Canal de Sodio Activado por Voltaje NAV1.5/genética , Oportunidad Relativa , Polimorfismo de Nucleótido Simple , Modelos de Riesgos Proporcionales , Sistema de Registros , Medición de Riesgo , Análisis de Secuencia de ADN , Taiwán
5.
Antioxidants (Basel) ; 9(1)2020 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-31936771

RESUMEN

Ultraviolet A (UVA)-irradiation induced reactive oxygen species (ROS) production mediates excessive melanogenesis in skin cells leading to pigmentation. We demonstrated the depigmenting and anti-melanogenic effects of Ectoine, a natural bacterial osmolyte, in UVA-irradiated human (HaCaT) keratinocytes, and the underlying molecular mechanisms were elucidated. HaCaT cells were pre-treated with low concentrations of Ectoine (0.5-1.5 µM) and assayed for various depigmenting and anti-melanogenic parameters. This pre-treatment significantly downregulated ROS generation, α-melanocyte-stimulating hormone (α-MSH) production, and proopiomelanocortin (POMC) expression in UVA-irradiated HaCaT cells. Also, antioxidant heme oxygenase-1 (HO-1), NAD(P)H dehydrogenase [quinone 1] (NQO-1), and γ-glutamate-cysteine ligase catalytic subunit (γ-GCLC) protein expressions were mediated via the nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) whose knockdown indeed impaired this effect signifying the importance of the Nrf2 pathway. Ectoine was mediating the activation of Nrf2 via the p38, protein kinase B (also known as AKT), protein kinase C (PKC), and casein kinase II protein kinase (CKII) pathways. The conditioned medium obtained from the Ectoine pre-treated and UVA-irradiated HaCaT cells downregulated the tyrosinase, tyrosinase-related protein-1 and -2 (TRP-1/-2), cyclic AMP (c-AMP) protein kinase, c-AMP response element-binding protein (CREB), and microphthalmia-associated transcription factor (MITF) expressions leading to melanoma B16F10 cells having inhibited melanin synthesis. Interestingly, this anti-melanogenic effect in α-MSH-stimulated B16F10 cells was observable only at 50-400 µM concentrations of Ectoine, signifying the key role played by Ectoine (0.5-1 µM)-treated keratinocytes in skin whitening effects. We concluded that Ectoine could be used as an effective topical natural cosmetic agent with depigmenting and anti-melanogenic efficacy.

6.
Front Pharmacol ; 10: 1107, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31680941

RESUMEN

Background: Aspirin is the most commonly used antiplatelet agent for the prevention of cardiovascular diseases. However, a certain proportion of patients do not respond to aspirin therapy. The mechanisms of aspirin non-response remain unknown. The unique metabolomes in platelets of patients with coronary artery disease (CAD) with aspirin non-response may be one of the causes of aspirin resistance. Materials and Methods: We enrolled 29 patients with CAD who were aspirin non-responders, defined as a study subject who were taking aspirin with a platelet aggregation time less than 193 s by PFA-100, and 31 age- and sex-matched patients with CAD who were responders. All subjects had been taking 100 mg of aspirin per day for more than 1 month. Hydrophilic metabolites from the platelet samples were extracted and analyzed by nuclear magnetic resonance (NMR). Both 1D 1H and 2D J-resolved NMR spectra were obtained followed by spectral processing and multivariate statistical analysis, such as partial least squares discriminant analysis (PLS-DA). Results: Eleven metabolites were identified. The PLS-DA model could not distinguish aspirin non-responders from responders. Those with low serum glycine level had significantly shorter platelet aggregation time (mean, 175.0 s) compared with those with high serum glycine level (259.5 s). However, this association became non-significant after correction for multiple tests. Conclusions: The hydrophilic metabolic profile of platelets was not different between aspirin non-responders and responders. An association between lower glycine levels and higher platelet activity in patients younger than 65 years suggests an important role of glycine in the pathophysiology of aspirin non-response.

7.
Oncotarget ; 6(6): 4239-52, 2015 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-25738362

RESUMEN

Oral squamous cell carcinoma (OSCC), which accounts for nearly 90% of head and neck cancers, is characterized by poor prognosis and a low survival rate. VEGF-A is the most established angiogenic factor involved in the angiogenic-regulated tumor progression. WISP-1/CCN4 is an extracellular matrix-related protein that belongs to the Cyr61, CTGF, Nov (CCN) family and regulates many biological functions, such as angiogenesis. Previous studies indicated the role of WISP-1 in tumor progression. However, the angiogenic property of WISP-1 in the cancer microenvironment has never been discussed. Here, we provide novel insights regarding the role of WISP-1 in the angiogenesis through promoting VEGF-A expression. In this study, the correlation of WISP-1 and VEGF-A was confirmed by IHC staining of specimens from patients with OSCC. In vitro results indicated that WISP-1 induced VEGF-A expression via the integrin αvß3/FAK/c-Src pathway, which transactivates the EGFR/ERK/HIF1-α signaling pathway in OSCC. This pathway in turn induces the recruitment of endothelial progenitor cells and triggers the neovascularization in the tumor microenvironment. Our in vivo data revealed that tumor-secreted WISP-1 promoted the angiogenesis through VRGF expression and increased angiogenesis-related tumor growth. Our study offers new information that highlights WISP-1 as a potential novel therapeutic target for OSCC.


Asunto(s)
Proteínas CCN de Señalización Intercelular/genética , Proteínas CCN de Señalización Intercelular/metabolismo , Carcinoma de Células Escamosas/irrigación sanguínea , Neoplasias de Cabeza y Cuello/irrigación sanguínea , Neoplasias de la Boca/irrigación sanguínea , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Xenoinjertos , Humanos , Masculino , Ratones , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello , Transfección
8.
Molecules ; 19(7): 8820-39, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24972270

RESUMEN

Increasing evidence suggests that inflammatory processes in the central nervous system that are mediated by microglial activation play a key role in neurodegeneration. Fisetin, a plant flavonol commonly found in fruits and vegetables, is frequently added to nutritional supplements due to its antioxidant properties. In the present study, treatment with fisetin inhibited microglial cell migration and ROS (reactive oxygen species) production. Treatment with fisetin also effectively inhibited LPS plus IFN-γ-induced nitric oxide (NO) production, and inducible nitric oxide synthase (iNOS) expression in microglial cells. Furthermore, fisetin also reduced expressions of iNOS and NO by stimulation of peptidoglycan, the major component of the Gram-positive bacterium cell wall. Fisetin also inhibited the enhancement of LPS/IFN-γ- or peptidoglycan-induced inflammatory mediator IL (interlukin)-1 ß expression. Besides the antioxidative and anti-inflammatory effects of fisetin, our study also elucidates the manner in fisetin-induced an endogenous anti-oxidative enzyme HO (heme oxygenase)-1 expression. Moreover, the regulatory molecular mechanism of fisetin-induced HO-1 expression operates through the PI-3 kinase/AKT and p38 signaling pathways in microglia. Notably, fisetin also significantly attenuated inflammation-related microglial activation and coordination deficit in mice in vivo. These findings suggest that fisetin may be a candidate agent for the development of therapies for inflammation-related neurodegenerative diseases.


Asunto(s)
Antiinflamatorios/farmacología , Flavonoides/farmacología , Microglía/inmunología , Fármacos Neuroprotectores/farmacología , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Flavonoles , Hemo-Oxigenasa 1/metabolismo , Lipopolisacáridos/farmacología , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos ICR , Microglía/efectos de los fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba
9.
Int J Mol Sci ; 15(1): 545-59, 2014 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-24398980

RESUMEN

Oral squamous cell carcinoma (OSCC) typically migrates and metastasizes. Interleukin-6 (IL-6) is a multifunctional cytokine associated with disease status and cancer outcomes. The effect of IL-6 on human OSCC cells, however, is unknown. Here, we showed that IL-6 increased cell migration and Intercellular adhesion molecule-1 (ICAM-1) expression in OSCC cells. Pretreatment of OSCC cells with IL-6R monoclonal antibody (mAb) significantly abolished IL-6-induced cell migration and ICAM-1 expression. By contrast, IL-6-mediated cell motility and ICAM-1 upregulation were attenuated by the Syk and c-Jun N-terminal kinase (JNK) inhibitors. Stimulation of OSCC cells with IL-6 promoted Syk and JNK phosphorylation. Furthermore, IL-6 enhanced AP-1 activity, and the IL-6R mAb, Syk inhibitor, or JNK inhibitor all reduced IL-6-mediated c-Jun phosphorylation, c-Jun binding to the ICAM-1 promoter, and c-Jun translocation into the nucleus. Our results indicate that IL-6 enhances the migration of OSCC cells by increasing ICAM-1 expression through the IL-6R receptor and the Syk, JNK, and AP-1 signal transduction pathways.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Interleucina-6/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Factor de Transcripción AP-1/metabolismo , Antracenos/farmacología , Anticuerpos Monoclonales/inmunología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Humanos , Molécula 1 de Adhesión Intercelular/química , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-6/antagonistas & inhibidores , Interleucina-6/genética , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Fosforilación/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptores de Interleucina-6/inmunología , Receptores de Interleucina-6/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
10.
PLoS One ; 8(10): e78022, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24205072

RESUMEN

Oral squamous cell carcinoma (OSCC) has a tendency to migrate and metastasize. WNT1-inducible signaling pathway protein 1 (WISP-1) is a cysteine-rich protein that belongs to the Cyr61, CTGF, Nov (CCN) family of matrix cellular proteins. The effect of WISP-1 on human OSCC cells, however, is unknown. Here, we showed that WISP-1 increased cell migration and intercellular adhesion molecule-1 (ICAM-1) expression in OSCC cells. Pretreatment of cells with integrin αvß3 monoclonal antibody (mAb) significantly abolished WISP-1-induced cell migration and ICAM-1 expression. On the other hand, WISP-1-mediated cell motility and ICAM-1 upregulation were attenuated by ASK1, JNK, and p38 inhibitor. Furthermore, WISP-1 also enhanced activator protein 1 (AP-1) activation, and the integrin αvß3 mAb, and ASK1, JNK, and p38 inhibitors reduced WISP-1-mediated AP-1 activation. Moreover, WISP-1 and ICAM-1 expression correlated with the tumor stage of patients with OSCC. Our results indicate that WISP-1 enhances the migration of OSCC cells by increasing ICAM-1 expression through the αvß3 integrin receptor and the ASK1, JNK/p38, and AP-1 signal transduction pathways.


Asunto(s)
Proteínas CCN de Señalización Intercelular/metabolismo , Carcinoma de Células Escamosas/metabolismo , Movimiento Celular/fisiología , MAP Quinasa Quinasa Quinasa 5/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Anticuerpos Monoclonales/farmacología , Proteínas CCN de Señalización Intercelular/genética , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Movimiento Celular/genética , Humanos , Integrina alfaVbeta3/antagonistas & inhibidores , Integrina alfaVbeta3/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , MAP Quinasa Quinasa Quinasa 5/genética , Proteínas Proto-Oncogénicas/genética , Transducción de Señal , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo
11.
Oral Oncol ; 49(12): 1103-12, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24070603

RESUMEN

OBJECTIVES: Perineural invasion is a prominent clinical feature of various cancers, which causes difficulty in curative resection. Glial cell-derived neurotrophic factor (GDNF), a potent neurotrophic factor, plays an important role in the invasive and metastatic behavior of various cancers. The aim of this study was to examine the role of GDNF on oral squamous cell carcinoma. MATERIALS AND METHODS: GDNF expression in tissue samples was analyzed by immunohistochemistry. Transwell assay, zymography, Western blot, reverse transcription-PCR, and electrophoretic mobility shift assay (EMSA) were carried out to assess the effects of GDNF on oral cancer cells. RESULTS: Human oral cancer tissues showed higher GDNF expression than that in normal tissues. We also found that application of human GDNF enhanced the cell migration ability of human oral cancers. Moreover, treatment with GDNF increased matrix metalloproteinase (MMP)-9 and MMP-13 expression in oral cancer. Inhibition of MMP-9 and MMP-13 in oral cancer cells by pharmacological inhibitors or neutralizing antibodies reduced GDNF-enhanced cell migration. Moreover, transfection with siRNA against MMP-13 inhibited GDNF-enhanced cell migration. Treatment with GDNF also increased ERK, p38 and JNK phosphorylation, and AP-1 DNA binding activity in human oral cancer cells. Inhibition of MAP kinase or AP-1 also reduced GDNF-induced oral cancer cell migration. In migration-prone sublines, oral cancer cells showed a higher migration ability than that of the original oral cancer cells. Surprisingly, the enhancement of cell migratory activity in migration-prone sublines was reduced by a GDNF-neutralizing antibody. Importantly, migration-prone sublines of oral cancer revealed higher GDNF expression. CONCLUSION: These results indicate a regulatory effect on cell migration by GDNF in oral squamous cancer.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Movimiento Celular/fisiología , Factores Neurotróficos Derivados de la Línea Celular Glial/metabolismo , Neoplasias de la Boca/metabolismo , Anticuerpos Neutralizantes/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Factores Neurotróficos Derivados de la Línea Celular Glial/efectos de los fármacos , Humanos , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Factor de Transcripción AP-1/antagonistas & inhibidores , Factor de Transcripción AP-1/metabolismo
12.
J Cell Biochem ; 113(6): 1977-86, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22253074

RESUMEN

Oral squamous cell carcinoma (OSCC) has a striking tendency to migrate and metastasize. Cysteine-rich 61 (Cyr61), from the CCN gene family, is a secreted and matrix-associated protein, which is involved in many cellular activities such as growth and differentiation. However, the effects of Cyr61 on human OSCC cells are largely unknown. In this study, we found that Cyr61 increased the migration and the expression of matrix metalloproteinases-3 (MMP)-3 in human OSCC cells. αvß5 or α6ß1 monoclonal antibody (mAb), focal adhesion kinase (FAK) inhibitor, and mitogen-activated protein kinase (MEK) inhibitors (PD98059 and U0126) inhibited the Cyr61-induced increase of the migration and MMP-3 up-regulation of OSCC cells. Cyr61 stimulation increased the phosphorylation of FAK, MEK, and extracellular signal-regulated kinase (ERK). In addition, NF-κB inhibitors suppressed the cell migration and MMP-3 expression enhanced by Cyr61. Moreover, Cyr61 increased NF-κB luciferase activity and binding of p65 to the NF-κB element on the MMP-3 promoter. Taken together, our results indicate that Cyr61 enhances the migration of OSCC cells by increasing MMP-3 expression through the αvß3 or α6ß1 integrin receptor, FAK, MEK, ERK, and NF-κB signal transduction pathway.


Asunto(s)
Proteína 61 Rica en Cisteína/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Neoplasias de la Boca/metabolismo , FN-kappa B/metabolismo , Anticuerpos Monoclonales/inmunología , Butadienos/farmacología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Flavonoides/farmacología , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Integrina alfa6beta1/inmunología , Integrina alfa6beta1/metabolismo , Metaloproteinasa 3 de la Matriz/genética , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias de la Boca/patología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/biosíntesis , Nitrilos/farmacología , Fosforilación , Regiones Promotoras Genéticas , Receptores de Vitronectina/inmunología , Receptores de Vitronectina/metabolismo , Factor de Transcripción ReIA/metabolismo
13.
Artículo en Inglés | MEDLINE | ID: mdl-21584270

RESUMEN

Type-2 ribosome-inactivating proteins, composed of a toxic A-chain and lectin-like B-chain, display various biological functions, including cytotoxicity and immunomodulation. We here cloned the lectin-like B-chain encoding fragment of a newly identified type-2 RIP gene, articulatin gene, from Viscum articulatum, into a bacterial expression vector to obtain nonglycosylated recombinant protein expressed in inclusion bodies. After purification and protein refolding, soluble refolded recombinant articulatin B-chain (rATB) showed lectin activity specific toward galactoside moiety and was stably maintained while stored in low ionic strength solution. Despite lacking glycosylation, rATB actively bound leukocytes with preferential binding to monocytes and in vitro stimulated PBMCs to release cytokines without obvious cytotoxicity. These results implicated such a B-chain fragment as a potential immunomodulator.

14.
Int Immunopharmacol ; 11(8): 948-54, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21352962

RESUMEN

Oral squamous cell carcinoma (SCC) has a striking tendency to migrate and metastasize. Cyclooxygenase (COX)-2, the inducible isoform of prostaglandin synthase, has been implicated in tumor metastasis. Connective tissue growth factor (CTGF), a secreted protein that binds to integrins, modulates the invasive behavior of certain human cancer cells. However, the effect of CTGF on migration activity and COX-2 expression in human oral cells is mostly unknown. Here we found that CTGF reduced the migration and expression of COX-2 in human oral cancer cells. αvß5 monoclonal antibody (mAb), phosphatidylinositol 3-kinase inhibitor (PI3K; Ly294002 and wortmannin) and Akt inhibitor reversed the CTGF-inhibited the migration and COX-2 down-regulation of oral cancer cells. CTGF stimulation decreased the phosphorylation of focal adhesion kinase (FAK), PI3K and Akt. In addition, c-Jun siRNA also antagonized the CTGF-inhibited migration and COX-2 expression. Moreover, CTGF decreased the binding of c-Jun to the AP-1 element on the COX-2 promoter. Taken together, our results indicated that CTGF inhibits the migration of oral cancer cells by decreasing COX-2 expression through the αvß5 integrin receptor, FAK, PI3K, Akt, c-Jun and AP-1 signal transduction pathways.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Factor de Crecimiento del Tejido Conjuntivo/farmacología , Ciclooxigenasa 2/biosíntesis , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Androstadienos/farmacología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Línea Celular Tumoral , Cromonas/farmacología , Factor de Crecimiento del Tejido Conjuntivo/antagonistas & inhibidores , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Ciclooxigenasa 2/genética , Regulación hacia Abajo , Quinasa 1 de Adhesión Focal/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes jun/genética , Humanos , Morfolinas/farmacología , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/genética , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Receptores de Vitronectina/inmunología , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/metabolismo , Wortmanina
15.
J Cell Physiol ; 226(6): 1573-82, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20945380

RESUMEN

We recently reported that peptidoglycan (PGN), a cell wall component of the Gram-positive bacterium, induces NF-κB activation and microglia activation. However, PGN-regulated AP-1 activation and cytokine expression in microglia remains unclear. This study investigated how PGN influences the signaling pathway involved in IL-6 production in microglia. IL-6 mRNA and protein level up-regulation were increased by PGN in a concentration- and time-dependent manner. In addition, PGN increased toll-like receptor-2 (TLR2) expression, but not TLR4 receptor up-regulation. Administration of TLR2 siRNA or TLR2 neutralized antibody effectively inhibited PGN-induced IL-6 expression. In contrast, PGN-induced IL-6 mRNA and protein up-regulation were attenuated by the SAPK/JNK (c-Jun N-terminal kinases) inhibitor SP600125. Treatment of microglia with PGN increased levels of JNK phosphorylation and c-Jun phosphorylation, and up-regulated of JNK kinase activity. Treatment of microglia with AP-1 inhibitors (Tanshinone IIA and curcumin) effectively reduced PGN-induced IL-6 expression. PGN also significantly increased c-Fos and phospho-c-Jun translocation to nucleus. In line with this, PGN also increased AP-1-DNA complexes formation, as determined by the electrophoretic mobility shift assay. Furthermore, PGN also increased IL-6 transcription activity determined by transfection with IL-6 promoter construct plasmid. Co-transfection with dominant negative mutant of JNK (DN-JNK), or treatment with SP600125, curcumin, or Tanshinone IIA effectively antagonized PGN-increased IL-6 transcription activity. Our data demonstrate that PGN-induced IL-6 expression is mediated by AP-1 activation through the TLR2 and JNK/c-Jun pathways in microglia.


Asunto(s)
Interleucina-6/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Microglía/metabolismo , Peptidoglicano/farmacología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Receptor Toll-Like 2/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Línea Celular , Cicloheximida/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-6/biosíntesis , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Microglía/efectos de los fármacos , Microglía/enzimología , Modelos Biológicos , Polimixina B/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor Toll-Like 2/genética
16.
J Cell Physiol ; 220(2): 418-26, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19334035

RESUMEN

CCL5 (previously called RANTES) is in the CC-chemokine family and plays a crucial role in the migration and metastasis of human cancer cells. On the other hand, the effect of CCL5 is mediated via CCR receptor. RT-PCR and flow cytometry studies demonstrated CCR5 but not CCR1 and CCR3 mRNA in oral cancer cell lines, especially higher in those with high invasiveness (SCC4) as compared with lower levels in HSC3 cells and SCC9 cells. Stimulation of oral cancer cells with CCL5 directly increased the migration and metalloproteinase-9 (MMP-9) production. MMP-9 small interfering RNA inhibited the CCL5-induced MMP-9 expression and thereby significantly inhibited the CCL5-induced cell migration. Activations of phospholipase C (PLC), protein kinase Cdelta (PKCdelta), and NF-kappaB pathways after CCL5 treatment was demonstrated, and CCL5-induced expression of MMP-9 and migration activity was inhibited by the specific inhibitor of PLC, PKCdelta, and NF-kappaB cascades. In addition, migration-prone sublines demonstrate that cells with increasing migration ability had more expression of MMP-9, CCL5, and CCR5. Taken together, these results indicate that CCL5/CCR5 axis enhanced migration of oral cancer cells through the increase of MMP-9 production.


Asunto(s)
Movimiento Celular/fisiología , Quimiocina CCL5/metabolismo , Neoplasias de la Boca , Receptores CCR5/metabolismo , Animales , Línea Celular Tumoral , Quimiocina CCL5/genética , Humanos , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , FN-kappa B/genética , FN-kappa B/metabolismo , Invasividad Neoplásica , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Regiones Promotoras Genéticas , Proteína Quinasa C-delta/genética , Proteína Quinasa C-delta/metabolismo , Receptores CCR5/genética , Transducción de Señal/fisiología
17.
Carcinogenesis ; 29(8): 1483-92, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18310089

RESUMEN

Oral squamous cell carcinoma (SCC) has a striking tendency to invade to bone. The chemokine stromal cell-derived factor-1 (SDF-1) is constitutively secreted by osteoblasts and plays a key role in homing of hematopoietic cells to the bone marrow. Interleukin (IL)-6 plays an important role in osteoclastogenesis. Herein, we found that SDF-1 alpha increased the secretion of IL-6 in cultured human SCC cells, as shown by reverse transcriptase-polymerase chain reaction and enzyme-linked immunosorbent assay. SDF-1 alpha also increased the surface expression of chemokine receptor 4 (CXCR4) in SCC cells. CXCR4-neutralizing antibody, CXCR4-specific inhibitor (AMD3100) or small interfering RNA against CXCR4 inhibited SDF-1 alpha-induced increase IL-6 production. The transcriptional regulation of IL-6 by SDF-1 alpha was mediated by phosphorylation of extracellular signal-regulated kinases (ERKs) and activation of the nuclear factor-kappa B (NF-kappaB) components p65 and p50. The binding of p65 and p50 to the NF-kappaB element on the IL-6 promoter was enhanced by SDF-1 alpha. In addition, IL-6 antibody antagonized the SCC-conditioned medium-increased osteoclastogenesis. These results suggested that SDF-1 alpha from osteoblasts could induce release of IL-6 in human SCC cells via activation of CXCR4, ERK and NF-kappaB pathway and thereby promote osteoclastogenesis.


Asunto(s)
Neoplasias Óseas/secundario , Huesos/fisiología , Quimiocina CXCL12/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Interleucina-6/metabolismo , Neoplasias de la Boca/patología , FN-kappa B/fisiología , Osteoblastos/fisiología , Receptores CXCR4/fisiología , Carcinoma de Células Escamosas , Diferenciación Celular , Línea Celular Tumoral , Humanos , Interleucina-6/genética , Osteoclastos/citología , Osteogénesis , Osteosarcoma , Ligando RANK/fisiología , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
In Vivo ; 21(6): 1003-9, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18210747

RESUMEN

The effects of coumarin on cell viability, cell cycle arrest and induction of apoptosis were investigated in human cervical cancer HeLa cells. Coumarin was cytotoxic with an IC50 of 54.2 microM, induced morphological changes, and caused G0/G1 arrest and apoptosis. The decreasing number of viable cells appeared to be due to induction of cell cycle arrest and apoptotic cell death, since coumarin induced morphologically apoptotic changes and internucleosomal DNA laddering fragmentation and increased the sub-G1 group. Coumarin affected the production of reactive oxygen species and Ca2+ concentration, and dose-dependently induced the depolarization of mitochondrial membrane potential. Also, coumarin treatment gradually decreased the expression of G0/G1-associated proteins which may have led to the G0/G1 arrest, and the anti-apoptotic proteins Bcl-2 and Bcl-xL, and increased the expression of the pro-apoptotic protein Bax. Coumarin decreased the mitochondrial membrane potential and promoted the release of cytochrome c and the activation of caspase-3 before leading to apoptosis. These results provide information on the mechanisms by which coumarin induces cell cycle arrest and apoptosis in human cervical cancer cells (HeLa).


Asunto(s)
Caspasa 2/metabolismo , Cumarinas/farmacología , Regulación hacia Abajo , Mitocondrias/efectos de los fármacos , FN-kappa B/metabolismo , Neoplasias del Cuello Uterino/patología , Calcio/metabolismo , Femenino , Células HeLa , Humanos , Potenciales de la Membrana , Mitocondrias/metabolismo , Mitocondrias/fisiología , Especies Reactivas de Oxígeno/metabolismo , Neoplasias del Cuello Uterino/metabolismo
19.
Curr Microbiol ; 51(4): 255-61, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16086103

RESUMEN

The effects of berberine on growth, arylamine N-acetyltransferase (NAT) activity, and gene expression in Salmonella Typhi (Typhi) were described. The growth inhibition of Typhi was determined by measuring absorbance by optical density (OD at 650 nm). The NAT activity was determined by measuring the levels of 2-aminofluorene (AF) and N-acetyl-2-aminofluorene (AAF) by high-performance liquid chromatography. The results demonstrated that 24-h berberine treatment decreased bacteria growth and amounts of AAF in Typhi. Western blotting and flow cytometry were used for examining the levels of NAT after bacteria were cotreated with or without various concentrations of berberine, and results indicated that berberine decreased the levels of NAT in Typhi. Polymerase chain reaction was used for examining the gene expression of NAT (mRNA NAT), and results indicated that berberine affects mRNA NAT1 expression in Typhi.


Asunto(s)
Arilamina N-Acetiltransferasa/metabolismo , Berberina/farmacología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Salmonella typhi/efectos de los fármacos , Berberis , Regulación hacia Abajo , ARN Mensajero/metabolismo , Salmonella typhi/genética , Salmonella typhi/metabolismo
20.
In Vivo ; 19(4): 683-8, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15999534

RESUMEN

The present studies were undertaken to analyze the factors regulating 3-hydroxycinnamic acid-induced apoptosis and cell cycle arrest. Treatment of human cervix HeLa cells with 3-hydroxycinnamic acid induced apoptosis and G0/G1-phase arrest. The percentage of apoptosis induced by 3-hydroxycinnamic acid in HeLa cells was increased with incubation time. The results also demonstrated that 3-hydroxycinnamic acid increased the expression of p53, caspase-3, Bax and cyclin B. These results demonstrated that 3-hydroxycinnamic acid induced apoptosis through p53- and caspase-3-dependent pathways.


Asunto(s)
Adenocarcinoma/patología , Apoptosis/efectos de los fármacos , Ácidos Cumáricos/farmacología , Fase G1/efectos de los fármacos , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Neoplasias del Cuello Uterino/patología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Adulto , Caspasa 3 , Caspasas/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclina B/metabolismo , Fragmentación del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Células HeLa/efectos de los fármacos , Células HeLa/metabolismo , Células HeLa/patología , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/metabolismo , Proteína X Asociada a bcl-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...