Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Theranostics ; 11(19): 9667-9686, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34646392

RESUMEN

The tumorous niche may drive the plasticity of heterogeneity and cancer stemness, leading to drug resistance and metastasis, which is the main reason of treatment failure in most cancer patients. The aim of this study was to establish a tumor microenvironment (TME)-based screening to identify drugs that can specifically target cancer stem cells (CSCs) and cancer-associated fibroblasts (CAFs) in the TME. Methods: Lung cancer patient-derived cancer cell and CAFs were utilized to mimic the TME and reproduce the stemness properties of CSCs in vitro and develop a high-throughput drug screening platform with phenotypical parameters. Limiting dilution assay, sphere-forming and ALDH activity assay were utilized to measure the cancer stemness characteristics. In vivo patient-derived xenograft (PDX) models and single-cell RNA sequencing were used to evaluate the mechanisms of the compounds in CSCs and CAFs. Results: The TME-based drug screening platform could comprehensively evaluate the response of cancer cells, CSCs and CAFs to different treatments. Among the 1,524 compounds tested, several drugs were identified to have anti-CAFs, anticancer and anti-CSCs activities. Aloe-emodin and digoxin both show anticancer and anti-CSCs activity in vitro and in vivo, which was further confirmed in the lung cancer PDX model. The combination of digoxin and chemotherapy improved therapeutic efficacy. The single-cell transcriptomics analysis revealed that digoxin could suppress the CSCs subpopulation in CAFs-cocultured cancer cells and cytokine production in CAFs. Conclusions: The TME-based drug screening platform provides a tool to identify and repurpose compounds targeting cancer cells, CSCs and CAFs, which may accelerate drug development and therapeutic application for lung cancer patients.


Asunto(s)
Reposicionamiento de Medicamentos/métodos , Células Madre Neoplásicas/efectos de los fármacos , Microambiente Tumoral/fisiología , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Proliferación Celular , Evaluación Preclínica de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Detección Precoz del Cáncer , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Neoplasias Pulmonares/patología , Células Madre Neoplásicas/metabolismo , Preparaciones Farmacéuticas
2.
Sci Rep ; 5: 16342, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26552835

RESUMEN

As chronic pain affects 115 million people and costs $600B annually in the US alone, effective noninvasive nonpharmacological remedies are desirable. The purpose of this study was to determine the efficacy and the generalisability of Noxipoint therapy (NT), a novel electrotherapy characterised by site-specific stimulation, intensity-and-submodality-specific settings and a immobilization period, for chronic neck and shoulder pain. Ninety-seven heavily pretreated severe chronic neck/shoulder pain patients were recruited; 34 and 44 patients were randomly allocated to different treatment arms in two patient-and-assessor-blinded, randomised controlled studies. The participants received NT or conventional physical therapy including transcutaneous electrical nerve stimulation (PT-TENS) for three to six 90-minute sessions. In Study One, NT improved chronic pain (-89.6%, Brief Pain Inventory, p < 0.0001, 95% confidence interval), function (+77.4%, range of motion) and quality of life (+88.1%) at follow-up (from 4 weeks to 5 months), whereas PT-TENS resulted in no significant changes in these parameters. Study Two demonstrated similar advantages of NT over PT-TENS and the generalisability of NT. NT-like treatments in a randomised rat study showed a similar reduction in chronic hypersensitivity (-81%, p < 0.01) compared with sham treatments. NT substantially reduces chronic neck and shoulder pain, restores function, and improves quality of life in a sustained manner.


Asunto(s)
Terapia por Estimulación Eléctrica , Dolor de Hombro/terapia , Estimulación Eléctrica Transcutánea del Nervio , Adulto , Animales , Dolor Crónico , Terapia por Estimulación Eléctrica/efectos adversos , Femenino , Estudios de Seguimiento , Humanos , Hiperalgesia/terapia , Hiperalgesia/veterinaria , Masculino , Persona de Mediana Edad , Mialgia/etiología , Calidad de Vida , Ratas , Resultado del Tratamiento
3.
BMC Infect Dis ; 14: 142, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24628789

RESUMEN

BACKGROUND: The pandemic potential of avian influenza A/H5N1 should not be overlooked, and the continued development of vaccines against these highly pathogenic viruses is a public health priority. METHODS: This open-label extension booster study followed a Phase III study of 1206 adults who had received two 3.75 µg doses of primary AS03A-adjuvanted or non-adjuvanted H5N1 split-virus vaccine (A/Vietnam/1194/2004; clade 1) (NCT00449670). The aim of the extension study was to evaluate different timings for heterologous AS03A-adjuvanted booster vaccination (A/Indonesia/5/2005; clade 2.1) given at Month 6, 12, or 36 post-primary vaccination. Immunogenicity was assessed 21 days after each booster vaccination and the persistence of immune responses against the primary vaccine strain (A/Vietnam) and the booster strain (A/Indonesia) was evaluated up to Month 48 post-primary vaccination. Reactogenicity and safety were also assessed. RESULTS: After booster vaccination given at Month 6, HI antibody responses to primary vaccine, and booster vaccine strains were markedly higher with one dose of AS03A-H5N1 booster vaccine in the AS03A-adjuvanted primary vaccine group compared with two doses of booster vaccine in the non-adjuvanted primary vaccine group. HI antibody responses were robust against the primary and booster vaccine strains 21 days after boosting at Month 12 or 36. At Month 48, in subjects boosted at Month 6, 12, or 36, HI antibody titers of ≥1:40 against the booster strain persisted in 39.2%, 61.2%, and 95.6% of subjects, respectively. Neutralizing antibody responses and cell-mediated immune responses also showed that AS03A-H5N1 heterologous booster vaccination elicited robust immune responses within 21 days of boosting at Month 6, 12, or 36 post-primary vaccination. The booster vaccine was well tolerated, and no safety concerns were raised. CONCLUSIONS: In Asian adults primed with two doses of AS03A-adjuvanted H5N1 pandemic influenza vaccine, strong cross-clade anamnestic antibody responses were observed after one dose of AS03A-H5N1 heterologous booster vaccine given at Month 6, 12, or 36 after priming, suggesting that AS03A-adjuvanted H5N1 vaccines may provide highly flexible prime-boost schedules. Although immunogenicity decreased with time, vaccinated populations could potentially be protected for up to three years after vaccination, which is likely to far exceed the peak of the a pandemic.


Asunto(s)
Esquemas de Inmunización , Inmunización Secundaria , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Polisorbatos/administración & dosificación , Escualeno/administración & dosificación , Escualeno/inmunología , alfa-Tocoferol/administración & dosificación , alfa-Tocoferol/inmunología , Adulto , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Pueblo Asiatico , Reacciones Cruzadas/inmunología , Combinación de Medicamentos , Femenino , Humanos , Vacunas contra la Influenza/efectos adversos , Masculino , Persona de Mediana Edad , Polisorbatos/efectos adversos , Escualeno/efectos adversos , Adulto Joven , alfa-Tocoferol/efectos adversos
4.
J Thorac Oncol ; 7(6): 993-1000, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22525557

RESUMEN

INTRODUCTION: Tumor tissue is often not obtainable or suitable for molecular-based epidermal growth factor receptor (EGFR) mutational analysis in advanced non-small-cell lung cancer (NSCLC). This retrospective and single-institution study was conducted to evaluate the role of effusion immunocytochemistry using two EGFR mutant-specific antibodies for the detection of relevant EGFR mutations in NSCLC, along with the selection of candidates for first-line therapy with EGFR tyrosine kinase inhibitors (TKIs). METHODS: Immunocytochemistry using two antibodies binding specifically to the major forms of mutant EGFR, L858R, and E746-A750 deletion (delE746-A750), was performed on cell blocks of malignant pleural effusion (MPE) from 78 patients with lung adenocarcinoma, who received first-line EGFR TKIs. The yield of EGFR-mutation detection and prediction of response rate and progression-free survival to TKI treatment by immunocytochemistry were compared with those by clinical characteristics and EGFR sequencing using cell-derived RNA from MPEs. RESULTS: Of the 78 MPE samples, direct sequencing using cell-derived RNA identified L858R mutation in 42 cases, deletions in exon 19 in 12 cases (delE746-A750 in eight cases), other types of mutations in three cases, and wild-type EGFR in 21 cases. Effusion immunocytochemistry with these two mutant-specific antibodies exhibited a sensitivity of 71% and 88% and a specificity of 86% and 96% for identifying predefined L858R and delE746-A750 mutations, respectively. Effusion immunocytochemistry provided a superior prediction of tumor response and progression-free survival to first-line EGFR TKIs than did clinical characteristics like sex and smoking status. Patients whose effusion immunocytochemistry showed a reaction to either of the two antibodies had a comparable TKI response rate (67% versus 72%) to those with EGFR mutations assessed by direct sequencing from cell-derived RNA. CONCLUSIONS: Effusion immunocytochemistry could be introduced into clinical practice to identify more NSCLC patients likely to have benefit from first-line TKI treatment, especially for those without adequate tissue for molecular-based EGFR analysis.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Receptores ErbB/genética , Inmunohistoquímica/métodos , Neoplasias Pulmonares/tratamiento farmacológico , Mutación , ARN Neoplásico/genética , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma del Pulmón , Anciano , Anilidas/uso terapéutico , Análisis Mutacional de ADN/métodos , Receptores ErbB/antagonistas & inhibidores , Estudios de Seguimiento , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Pirimidinas/uso terapéutico , Estudios Retrospectivos
5.
PLoS One ; 6(8): e23756, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21858220

RESUMEN

BACKGROUND: Non-small cell lung cancer (NSCLC) patients with L858R or exon 19 deletion mutations in epidermal growth factor receptor (EGFR) have good responses to the tyrosine kinase inhibitor (TKI), gefitinib. However, patients with wild-type EGFR and acquired mutation in EGFR T790M are resistant to gefitinib treatment. Here, we showed that curcumin can improve the efficiency of gefitinib in the resistant NSCLC cells both in vitro and in vivo models. METHODS/PRINCIPAL FINDINGS: After screening 598 herbal and natural compounds, we found curcumin could inhibit cell proliferation in different gefitinib-resistant NSCLC cell lines; concentration-dependently down-regulate EGFR phosphorylation through promoting EGFR degradation in NSCLC cell lines with wild-type EGFR or T790M EGFR. In addition, the anti-tumor activity of gefitinib was potentiated via curcumin through blocking EGFR activation and inducing apoptosis in gefitinib-resistant NSCLC cell lines; also the combined treatment with curcumin and gefitinib exhibited significant inhibition in the CL1-5, A549 and H1975 xenografts tumor growth in SCID mice through reducing EGFR, c-MET, cyclin D1 expression, and inducing apoptosis activation through caspases-8, 9 and PARP. Interestingly, we observed that the combined treatment group represented better survival rate and less intestinal mucosal damage compare to gefitinib-alone therapy. We showed that curcumin attenuated the gefitinib-induced cell proliferation inhibition and apoptosis through altering p38 mitogen-activated protein kinase (MAPK) activation in intestinal epithelia cell. CONCLUSIONS/SIGNIFICANCE: Curcumin potentiates antitumor activity of gefitinib in cell lines and xenograft mice model of NSCLC through inhibition of proliferation, EGFR phosphorylation, and induction EGFR ubiquitination and apoptosis. In addition, curcumin attenuates gefitinib-induced gastrointestinal adverse effects via altering p38 activation. These findings provide a novel treatment strategy that curcumin as an adjuvant to increase the spectrum of the usage of gefitinib and overcome the gefitinib inefficiency in NSCLC patients.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Curcumina/farmacología , Receptores ErbB/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Curcumina/administración & dosificación , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Gefitinib , Humanos , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Intestinos/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones SCID , Fosforilación/efectos de los fármacos , Quinazolinas/administración & dosificación , Quinazolinas/farmacología , Ubiquitinación/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Formos Med Assoc ; 110(12): 780-6, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22248833

RESUMEN

BACKGROUND/PURPOSE: A multicenter study (NCT00449670) conducted across Taiwan, Singapore, Hong Kong and Thailand evaluated the safety and manufacturing consistency of four formulations of an AS03(A)-adjuvanted H5N1 vaccine in terms of immune response against the vaccine-homologous strain (A/Vietnam/1194/2004). This manuscript presents data from the Taiwanese population. METHODS: A total of 400 individuals, aged 18-60 years, were randomized into six groups (2:2:2:2:1:1 ratio) to receive two doses (21 days apart) of one of the four adjuvanted formulations (H5N1-AS03(A)-groups) or one of the two nonadjuvanted formulations (H5N1-DIL-groups). Blood samples collected before vaccination (Day 0) and 21 days after each vaccine dose were analyzed using hemagglutination inhibition (HI) assay. Adverse events were recorded. RESULTS: All four AS03(A)-adjuvanted formulations induced comparable immune responses against the A/Vietnam/1194/2004 strain; following the second dose, immune response in terms of HI antibodies was higher in the H5N1-AS03(A)-groups {seroprotection rate=91.6% [95% confidence interval (CI): 87.9-94.4]; geometric mean titer (GMT)=177.6 (95% CI: 153.2-206.0)} compared with the H5N1-DIL-groups [seroprotection rates=5.0% (95% CI: 1.4-12.3); GMT=6.3 (95% CI: 5.4-7.4)]. Immune response against the heterologous A/Indonesia/05/2005 strain was also stronger in the H5N1-AS03(A)-groups [seroprotection rate=45.6% (95% CI: 40.0-51.4); GMT=20.5 (95% CI: 17.8-23.7)] compared with the H5N1-DIL groups [seroprotection rate=0.0% (95% CI: 0.0-4.5); GMT=5.0 (95% CI: 5.0-5.0)]. The overall reactogenicity profile of the adjuvanted formulations was clinically acceptable. CONCLUSION: The AS03(A)-adjuvanted H5N1 influenza vaccine formulations induced stronger immune response against the vaccine-homologous and heterologous strains than the nonadjuvanted formulations. The AS03(A)-adjuvanted H5N1 vaccine demonstrated a good immunogenicity and an acceptable safety profile in the Taiwanese population.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Adolescente , Adulto , Anticuerpos Antivirales/sangre , Método Doble Ciego , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Vacunas contra la Influenza/efectos adversos , Masculino , Persona de Mediana Edad , Taiwán , Tocoferoles/administración & dosificación
7.
J Formos Med Assoc ; 108(2): 112-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19251546

RESUMEN

BACKGROUND/PURPOSE: To study the clinical features, diagnostic processes, timing of antibiotic administration and outcomes of patients with severe community-acquired septic meningitis at an emergency department (ED), who required intensive care unit (ICU) admission. METHODS: From January 1993 to December 2005, the medical records of patients admitted to the ICU with a diagnosis of community-acquired septic meningitis were reviewed. The clinical characteristics, including causative pathogens, treatment course, and outcomes were collected and analyzed. RESULTS: A total of 40 patients were included, with an overall in-hospital mortality rate of 77.5%. The most common pathogen was Klebsiella pneumoniae (n=20, 50%), followed by Streptococcus pneumoniae (n=6, 15%), and Cryptococcus neoformans (n=5, 12.5%). There was a mean duration of 8.9 hours between ED arrival and initiation of antibiotic therapy. Effective antibiotics were administered for a mean period of 23.8 hours after arrival. Time delay from ED arrival to ICU admission was correlated with time delay of effective antibiotics administration, head computed tomography, and cerebrospinal fluid study (r=0.32, 0.47, and 0.53, respectively; p=0.05, 0.006, and 0.001, respectively). Earlier ICU admission was demonstrated in survivors as compared with those who died (11.1 vs. 38.0 hours, p=0.01). CONCLUSION: Severe septic meningitis remains a disease with high mortality and morbidity. Expeditious diagnostic processes with early appropriate antibiotic treatment and ICU admission at the ED are important in improving the quality of care and patient outcome.


Asunto(s)
Infecciones Comunitarias Adquiridas/terapia , Servicio de Urgencia en Hospital/estadística & datos numéricos , Unidades de Cuidados Intensivos/estadística & datos numéricos , Meningitis/terapia , Choque Séptico/terapia , Adulto , Anciano , Anciano de 80 o más Años , Antibacterianos/administración & dosificación , Estudios de Cohortes , Infecciones Comunitarias Adquiridas/diagnóstico , Infecciones Comunitarias Adquiridas/microbiología , Infecciones Comunitarias Adquiridas/mortalidad , Esquema de Medicación , Medicamentos Herbarios Chinos , Femenino , Mortalidad Hospitalaria , Humanos , Masculino , Meningitis/complicaciones , Meningitis/diagnóstico , Meningitis/mortalidad , Persona de Mediana Edad , Evaluación de Procesos y Resultados en Atención de Salud/estadística & datos numéricos , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Choque Séptico/diagnóstico , Choque Séptico/microbiología , Choque Séptico/mortalidad , Factores de Tiempo , Adulto Joven
8.
Mol Cancer Ther ; 7(11): 3527-38, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19001436

RESUMEN

Tanshinones are the major bioactive compounds of Salvia miltiorrhiza Bunge (Danshen) roots, which are used in many therapeutic remedies in Chinese traditional medicine. We investigated the anticancer effects of tanshinones on the highly invasive human lung adenocarcinoma cell line, CL1-5. Tanshinone I significantly inhibited migration, invasion, and gelatinase activity in macrophage-conditioned medium-stimulated CL1-5 cells in vitro and also reduced the tumorigenesis and metastasis in CL1-5-bearing severe combined immunodeficient mice. Unlike tanshinone IIA, which induces cell apoptosis, tanshinone I did not have direct cytotoxicity. Real-time quantitative PCR, luciferase reporter assay, and electrophoretic mobility shift assay revealed that tanshinone I reduces the transcriptional activity of interleukin-8, the angiogenic factor involved in cancer metastasis, by attenuating the DNA-binding activity of activator protein-1 and nuclear factor-kappaB in conditioned medium-stimulated CL1-5 cells. Microarray and pathway analysis of tumor-related genes identified the differentially expressed genes responding to tanshinone I, which may be associated with the Ras-mitogen-activated protein kinase and Rac1 signaling pathways. These results suggest that tanshinone I exhibits anticancer effects both in vitro and in vivo and that these effects are mediated at least partly through the interleukin-8, Ras-mitogen-activated protein kinase, and Rac1 signaling pathways. Although tanshinone I has a remarkable anticancer action, its potential anticoagulant effect should be noted and evaluated.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Fenantrenos/farmacología , Abietanos , Animales , Apoptosis , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones SCID , Modelos Biológicos , Metástasis de la Neoplasia/patología , ARN Mensajero/metabolismo
9.
J Formos Med Assoc ; 106(2 Suppl): S1-6, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17493889

RESUMEN

Leptospirosis and scrub typhus coinfections have been reported in up to 41% of agricultural workers with acute leptospirosis in Thailand, but only sporadically in Taiwan. Because of the nonspecific clinical presentations, it is difficult to differentiate patients with coinfections from leptospirosis alone. However, failure to identify coinfection may lead to mortality if inappropriate antibiotics are used. We report a 31-year-old man coinfected with leptospirosis and scrub typhus, which manifested as diffuse alveolar hemorrhage and acute renal failure mimicking pulmonary-renal syndrome. The patient was treated by early plasma exchange and a 7-day course of moxifloxacin therapy. Both pulmonary hemorrhage and hypoxemia resolved substantially on the 4th day of hospitalization. He had a complete recovery from the disease after 6 weeks of hospitalization.


Asunto(s)
Hemorragia/terapia , Leptospirosis/complicaciones , Enfermedades Pulmonares/terapia , Intercambio Plasmático/métodos , Tifus por Ácaros/complicaciones , Adulto , Antibacterianos/uso terapéutico , Hemorragia/etiología , Humanos , Enfermedades Pulmonares/etiología , Masculino
10.
Cancer Res ; 67(10): 4816-26, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17510411

RESUMEN

HLJ1 is a novel tumor and invasion suppressor that inhibits tumorigenesis and cancer metastasis. However, the mechanism of HLJ1 activation is currently unclear. Here, we identify an enhancer segment in the HLJ1 gene at -2,125 to -1,039 bp upstream of the transcription start site. A 50-bp element between -1,492 and -1,443 bp is the minimal enhancer segment, which includes the activator protein 1 (AP-1) site (-1,457 to -1,451 bp), an essential regulatory domain that binds the transcriptional factors FosB, JunB, and JunD. Chromatin immunoprecipitation assays confirm that these AP-1 family members bind to a specific site in the HLJ1 enhancer segment in vivo. Overexpression of either YY1 at promoter or AP-1 at enhancer results in a 3-fold increase in the transcriptional activity of HLJ1. We propose a novel mechanism whereby expression of the tumor suppressor, HLJ1, is up-regulated via enhancer AP-1 binding to promoter YY1 and the coactivator, p300, through DNA bending and multiprotein complex formation. The combined expression of AP-1 and YY1 enhances HLJ1 expression by more than five times and inhibits in vitro cancer cell invasion. Elucidation of the regulatory mechanism of HLJ1 expression may facilitate the development of personalized therapy by inhibiting cancer cell proliferation, angiogenesis, and metastasis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Proteínas del Choque Térmico HSP40/genética , Factor de Transcripción AP-1/genética , Factor de Transcripción YY1/genética , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Secuencia de Bases , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Elementos de Facilitación Genéticos , Proteínas del Choque Térmico HSP40/biosíntesis , Proteínas del Choque Térmico HSP40/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-jun/biosíntesis , Proteínas Proto-Oncogénicas c-jun/genética , Factor de Transcripción AP-1/metabolismo , Activación Transcripcional , Transfección , Regulación hacia Arriba , Factor de Transcripción YY1/biosíntesis , Factor de Transcripción YY1/metabolismo
11.
Expert Opin Ther Targets ; 10(2): 253-66, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16548774

RESUMEN

The multifunctional transcription factor Yin Yang 1 (YY1) is a complex protein that has been shown to play pivotal roles in development, differentiation, cellular proliferation and apoptosis. It can act as a transcriptional repressor, an activator, or an initiator element binding protein that directs and initiates transcription of numerous cellular and viral genes. Because the expression and function of YY1 are known to be intimately associated with cell-cycle progression, the physiological significance of YY1 activity has recently been applied to models of cancer biology. Several lines of evidence imply that YY1 expression and/or activation is associated with tumourigenesis, in addition to its regulatory roles in normal biological processes. However, controversial results also raised and indicated that further studies are still needed to piece all of the seemingly contradictory data into a complete picture. On the basis of YY1 regulations and functions, novel drugs and specific treatment strategies may be developed with new therapeutic applications for tumour patients in the future.


Asunto(s)
Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Factor de Transcripción YY1/metabolismo , Animales , Humanos , Neoplasias/genética , Factor de Transcripción YY1/genética , Factor de Transcripción YY1/fisiología
12.
Oncogene ; 24(25): 4081-93, 2005 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-15782117

RESUMEN

By using microarray and an invasion/metastasis lung cell line model, we identified the DnaJ-like heat shock protein 40, HLJ1, and found that the expression of HLJ1 correlates negatively with cancer cell invasion ability. Overexpression of HLJ1 can suppress cancer cell invasion in vitro. We further characterize the putative promoter region and investigate the transcriptional regulations of human HLJ1. A serial deletion of the 1.2 kb at the 5'-flanking region of the human HLJ1 gene was subcloned into a vector containing reporter gene and transfected into human lung adenocarcinoma cell line CL1-0, followed by luciferase activity assay. The results indicated that the region from -232 to +176 could drive the basal transcriptional activity of the HLJ1 gene. Sequence analysis of the HLJ1 gene promoter region showed absence of a TATA box, but identified an inverted CCAAT box and four YY1 transcriptional factor-binding sites, which may be important in the regulation of HLJ1 expression. Co-transfection of the YY1 and HLJ1 basal promoter regions, site-directed mutagenesis, and electrophoretic mobility shift assay confirmed that YY1 could upregulate HLJ1 basal promoter activity. Furthermore, we also demonstrated that overexpression of YY1 in CL1-0 cells can increase HLJ1 expression and reduce cell invasive capability. The reduction of cancer cell invasive ability is, at least in part, through upregulation of E-cadherin expression. The increase in HLJ1 and E-cadherin expression, as well as the suppression of invasion ability, can be reversed specifically by HLJ1 siRNA.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Choque Térmico/genética , Neoplasias Pulmonares/genética , Factores de Transcripción/metabolismo , Regiones no Traducidas 5'/genética , Adenocarcinoma , Secuencia de Bases , Línea Celular Tumoral , Factores de Unión al ADN Específico de las Células Eritroides , Regulación Neoplásica de la Expresión Génica , Proteínas del Choque Térmico HSP40 , Humanos , Neoplasias Pulmonares/patología , Datos de Secuencia Molecular , Invasividad Neoplásica , Metástasis de la Neoplasia/prevención & control , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , Proteínas Represoras/metabolismo , Proteínas Supresoras de Tumor/genética , Factor de Transcripción YY1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA