Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biochim Biophys Acta ; 1843(2): 387-97, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24184161

RESUMEN

High-level expression of vascular endothelial growth factor (VEGF)-C is associated with chemoresistance and adverse prognosis in acute myeloid leukemia (AML). Our previous study has found that VEGF-C induces cyclooxygenase-2 (COX-2) expression in AML cell lines and significant correlation of VEGF-C and COX-2 in bone marrow specimens. COX-2 has been reported to mediate the proliferation and drug resistance in several solid tumors. Herein, we demonstrated that the VEGF-C-induced proliferation of AML cells is effectively abolished by the depletion or inhibition of COX-2. The expression of endothelin-1 (ET-1) rapidly increased following treatment with VEGF-C. We found that ET-1 was also involved in the VEGF-C-mediated proliferation of AML cells, and that recombinant ET-1 induced COX-2 mRNA and protein expressions in AML cells. Treatment with the endothelin receptor A (ETRA) antagonist, BQ 123, or ET-1 shRNAs inhibited VEGF-C-induced COX-2 expression. Flow cytometry and immunoblotting revealed that VEGF-C induces S phase accumulation through the inhibition of p27 and the upregulation of cyclin E and cyclin-dependent kinase-2 expressions. The cell-cycle-related effects of VEGF-C were reversed by the depletion of COX-2 or ET-1. The depletion of COX-2 or ET-1 also suppressed VEGF-C-induced increases in the bcl-2/bax ratio and chemoresistance against etoposide and cytosine arabinoside in AML cells. We also demonstrated VEGF-C/ET-1/COX-2 axis-mediated chemoresistance in an AML xenograft mouse model. Our findings suggest that VEGF-C induces COX-2-mediated resistance to chemotherapy through the induction of ET-1 expression. Acting as a key regulator in the VEGF-C/COX-2 axis, ET-1 represents a potential target for ameliorating resistance to chemotherapy in AML patients.


Asunto(s)
Ciclooxigenasa 2/biosíntesis , Resistencia a Antineoplásicos , Endotelina-1/metabolismo , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/patología , Factor C de Crecimiento Endotelial Vascular/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Inducción Enzimática/efectos de los fármacos , Humanos , Masculino , Ratones , Regulación hacia Arriba/efectos de los fármacos , Factor C de Crecimiento Endotelial Vascular/farmacología
2.
Hepatology ; 59(3): 974-85, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24114941

RESUMEN

UNLABELLED: Leukocyte cell-derived chemotoxin 2 (LECT2) has been shown to act as a tumor suppressor in hepatocellular carcinoma (HCC). However, the underlying mechanism has not yet been completely defined. Here, we employ a LECT2-affinity column plus liquid chromatography coupled with tandem mass spectrometry to identify LECT2-binding proteins and found that MET receptor strongly interacted with LECT2 protein. Despite the presence of hepatocyte growth factor, the LECT2 binding causes an antagonistic effect to MET receptor activation through recruitment of protein tyrosine phosphatase 1B. The antagonistic effect of LECT2 on MET activation also mainly contributes to the blockage of vascular invasion and metastasis of HCC. Furthermore, serial deletions and mutations of LECT2 showed that the HxGxD motif is primarily responsible for MET receptor binding and its antagonistic effects. CONCLUSION: These findings reveal a novel, specific inhibitory function of LECT2 in HCC by the direct binding and inactivation of MET, opening a potential avenue for treating MET-related liver cancer.


Asunto(s)
Carcinoma Hepatocelular/patología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias Hepáticas/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Carcinoma Hepatocelular/metabolismo , Células Hep G2 , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Neoplasias Hepáticas/metabolismo , Invasividad Neoplásica/patología , Fosforilación/fisiología , Unión Proteica/fisiología , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-met/química
3.
Ann Surg Oncol ; 22 Suppl 3: S1556-65, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25613390

RESUMEN

BACKGROUND: Emerging evidence suggests that G9a, a histone methyltransferase, is involved in tumor progression and metastasis. However, the functional significance of G9a in endometrial carcinogenesis has not been defined. METHODS: The differential expression of G9a in cancer and normal tissues was assessed using an array of 28 paired samples. Tissue specimens from 94 patients with endometrial cancer who underwent primary surgery were immunohistochemically evaluated for G9a and E-cadherin expression. To assess the biologic role of G9a in endometrial cancer, G9a was either stably knocked down or knocked down using a tetracycline-controllable system in endometrial cancer cells, followed by functional assays. RESULTS: Increased G9a expression was identified in endometrial cancer tissues, and its expression was specifically correlated with deep myometrial invasion. Cell invasiveness was inhibited by an RNAi-mediated knockdown of G9a in invasive endometrial cancer cells in vitro and in vivo. An important mediator of G9a-induced tumor invasion is the epigenetic silencing of E-cadherin. Knockdown of G9a restored E-cadherin expression by reducing H3K9me2 levels and decreasing CDH1 promoter DNA methyltransferase recruitment. Knockdown of RNAi-mediated E-cadherin substantially relieved the invasion suppression imposed by G9a suppression. A significant negative correlation between G9a and E-cadherin expression was observed in endometrial cancer (Spearman's rho, -0.27; P = 0.02). CONCLUSIONS: This study provides the first clear evidence that G9a contributes to endometrial cancer progression. Mechanistic investigations suggest that E-cadherin repression mediates the effects of G9a. Targeting G9a-mediated epigenetic pathway dysregulation may be a therapeutic strategy for endometrial cancers.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Cadherinas/antagonistas & inhibidores , Neoplasias Endometriales/patología , Regulación Neoplásica de la Expresión Génica , Antígenos de Histocompatibilidad/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Miometrio/patología , Animales , Apoptosis , Biomarcadores de Tumor/genética , Western Blotting , Cadherinas/genética , Cadherinas/metabolismo , Movimiento Celular , Proliferación Celular , Inmunoprecipitación de Cromatina , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Epigénesis Genética , Femenino , Antígenos de Histocompatibilidad/genética , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Miometrio/metabolismo , Clasificación del Tumor , Invasividad Neoplásica , Estadificación de Neoplasias , Pronóstico , Regiones Promotoras Genéticas , Reacción en Cadena en Tiempo Real de la Polimerasa , Estudios Retrospectivos , Tasa de Supervivencia , Células Tumorales Cultivadas
4.
Ann Surg Oncol ; 22(13): 4335-43, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25820997

RESUMEN

BACKGROUND: To assess the correlations and functions of complement C1r/C1s, Uegf, Bmp1 domain-containing protein-1 (CDCP1) in identifying colorectal cancer (CRC) patients who are at high risk for metastasis. METHODS: Tumor specimens from 101 patients were analyzed by real-time polymerase chain reaction to detect CDCP1 expression. CDCP1 expression plasmids and shRNA were used to knock down CDCP1 expression in this study to investigate migratory and invasive abilities by Boyden chambers. The mRNA expression profiles in shCDCP1 transfectants were compared to those in control cells by conducting microarray analysis. Its downstream effectors were also invested in this study. RESULTS: CRC patients with a high CDCP1 expression had a statistically significant lower overall survival and disease-free survival compared to those exhibiting low CDCP1 expression. In vitro, knock-down CDCP1 expression significantly decreased migratory and invasive abilities in HCT116. Aberrant expression of CDCP1 increased cancer cell migration and invasion. By using integrated genomics, we identified ROCK1 (rho-associated, coiled-coil-containing protein kinase 1 pseudogene 1) as a downstream effector in CDCP1-mediated migration and as an invasion mediator. Clinically, ROCK1 and CDCP1 mRNA expression exhibited a strong positive correlation in CRC patient samples. CONCLUSIONS: Our results implicated CDCP1 as a key regulator of CRC migration and invasion, and suggest that it is a useful prognostic factor for patients with CRC. Improved identification of a high-risk subset of early metastatic patients may guide indications of individualized treatment in clinical practice.


Asunto(s)
Antígenos CD/genética , Biomarcadores de Tumor/genética , Moléculas de Adhesión Celular/antagonistas & inhibidores , Moléculas de Adhesión Celular/genética , Movimiento Celular , Neoplasias Colorrectales/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Recurrencia Local de Neoplasia/patología , Anciano , Antígenos de Neoplasias , Adhesión Celular , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Femenino , Estudios de Seguimiento , Humanos , Metástasis Linfática , Masculino , Invasividad Neoplásica , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/mortalidad , Estadificación de Neoplasias , Pronóstico , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Células Tumorales Cultivadas
5.
Am J Physiol Renal Physiol ; 307(5): F581-92, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24920753

RESUMEN

Emerging data have suggested that acute kidney injury (AKI) is often incompletely repaired and can lead to chronic kidney disease (CKD), which is characterized by tubulointerstitial inflammation and fibrosis. However, the underlying mechanisms linking AKI to CKD remain obscure. The present study aimed to investigate the role of cysteine-rich protein 61 (Cyr61) after unilateral kidney ischemia-reperfusion injury (IRI) in mice. After IRI, increased expression of Cyr61 was detected, predominately in the proximal tubular epithelium. This was confirmed by in vitro experiments, which showed that hypoxia stimulated Cyr61 expression in cultured proximal tubular epithelial cells. The proinflammatory property of Cyr61 was indicated by its ability to upregulate monocyte chemoattractant protein-1 and IL-6. Additionally, we found elevated urinary Cyr61 excretion in patients with AKI. Notably, treatment of mice with an anti-Cyr61 antibody attenuated the upregulation of kidney monocyte chemoattractant protein-1, IL-6, IL-1ß, and macrophage inflammatory protein-2 and reduced the infiltration of F4/80-positive macrophages on days 7 and 14 after IRI. In addition, blockade of Cyr61 reduced the mRNA expression of collagen, transforming growth factor-ß, and plasminogen activator inhibitor-I as well as the degree of collagen fibril accumulation, as evaluated by picrosirius red staining, and levels of α-smooth muscle actin proteins by day 14. Concurrently, in the treated group, peritubular microvascular density was more preserved on day 14. We conclude that Cyr61 blockade inhibits the triad of inflammation, interstitial fibrosis, and capillary rarefaction after severe ischemic AKI. The results of this study expand the knowledge of the mechanisms underlying the AKI-to-CKD transition and suggest that Cyr61 is a potential therapeutic target.


Asunto(s)
Lesión Renal Aguda/complicaciones , Proteína 61 Rica en Cisteína/antagonistas & inhibidores , Riñón/patología , Nefritis/etiología , Nefritis/prevención & control , Daño por Reperfusión/complicaciones , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Anticuerpos Antiidiotipos/farmacología , Células Cultivadas , Quimiocina CCL2/metabolismo , Proteína 61 Rica en Cisteína/efectos de los fármacos , Proteína 61 Rica en Cisteína/inmunología , Modelos Animales de Enfermedad , Fibrosis/etiología , Fibrosis/metabolismo , Fibrosis/prevención & control , Hipoxia/metabolismo , Técnicas In Vitro , Interleucina-6/metabolismo , Riñón/metabolismo , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Masculino , Ratones , Ratones Endogámicos ICR , Nefritis/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Serpina E2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
6.
Mol Cancer ; 13: 172, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25027955

RESUMEN

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is a common cancer worldwide. Emerging evidence indicates that alteration of epigenetics might be a key event in HNSCC progression. Abnormal expression of histone methyltransferase G9a, which contributes to transcriptional repression of tumor suppressors, has been implicated in promoting cancerous malignancies. However, its role in HNSCC has not been previously characterized. In this study, we elucidate the function of G9a and its downstream mechanism in HNSCC. METHODS: We investigated the clinical relevance of G9a in HNSCC using immunohistochemistry (IHC) staining. In vitro cell proliferation and tumorigenesis ability of G9a-manipulated HNSCC cells were examined with MTT assays, clonogenic assays, and soft agar assays. We examined different routes of cell death in HNSCC cells induced by G9a-depletion or enzymatic inhibition by immunoblot, flow cytometry, fluorescent and transmission electron microscopy analysis. Specific targets of G9a were identified by affymetrix microarray and quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Lastly, functions of G9a in vivo were confirmed with a xenograft tumor model. RESULTS: G9a expression is positively correlated to proliferation marker Ki-67 and to poor prognosis in HNSCC patients. Genetic or pharmacological inhibition of G9a reduced cell proliferation without inducing necrosis or apoptosis. Instead, autophagic cell death was the major consequence, and our investigation of mechanisms suggested it is mediated via the dual specificity phosphatase-4 (DUSP4) dependent ERK inactivation pathway. An orthotopic tumor model further confirmed the growth inhibiting effect and induction of autophagy that followed suppression of G9a. CONCLUSIONS: In this study, we provide evidence that G9a confers the survival advantage of HNSCC. Genetic or pharmacological inhibition of G9a induces autophagic cell death; this finding provides a basis for new therapeutic targets for treating HNSCC.


Asunto(s)
Carcinoma de Células Escamosas/genética , Fosfatasas de Especificidad Dual/genética , Neoplasias de Cabeza y Cuello/genética , Antígenos de Histocompatibilidad/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Animales , Autofagia/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/genética , Fosfatasas de Especificidad Dual/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/patología , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Humanos , Sistema de Señalización de MAP Quinasas/genética , Ratones , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Cancer ; 13: 189, 2014 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-25115793

RESUMEN

BACKGROUND: Ovarian cancer (OCa) peritoneal metastasis is the leading cause of cancer-related deaths in women with limited therapeutic options available for treating it and poor prognosis, as the underlying mechanism is not fully understood. METHOD: The clinicopathological correlation of G9a expression was assessed in tumor specimens of ovarian cancer patients. Knockdown or overexpression of G9a in ovarian cancer cell lines was analysed with regard to its effect on adhesion, migration, invasion and anoikis-resistance. In vivo biological functions of G9a were tested by i.p. xenograft ovarian cancer models. Microarray and quantitative RT-PCR were used to analyze G9a-regulated downstream target genes. RESULTS: We found that the expression of histone methyltransferase G9a was highly correlated with late stage, high grade, and serous-type OCa. Higher G9a expression predicted a shorter survival in ovarian cancer patients. Furthermore, G9a expression was higher in metastatic lesions compared with their corresponding ovarian primary tumors. Knockdown of G9a expression suppressed prometastatic cellular activities including adhesion, migration, invasion and anoikis-resistance of ovarian cancer cell lines, while G9a over-expression promoted these cellular properties. G9a depletion significantly attenuated the development of ascites and tumor nodules in a peritoneal dissemination model. Importantly, microarray and quantitative RT-PCR analysis revealed that G9a regulates a cohort of tumor suppressor genes including CDH1, DUSP5, SPRY4, and PPP1R15A in ovarian cancer. Expression of these genes was also inversely correlated with G9a expression in OCa specimens. CONCLUSION: We propose that G9a contributes to multiple steps of ovarian cancer metastasis and represents a novel target to combat this deadly disease.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Antígenos de Histocompatibilidad/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Neoplasias Peritoneales/secundario , Animales , Anoicis , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones Endogámicos NOD , Ratones SCID , Análisis Multivariante , Invasividad Neoplásica , Neoplasias Ováricas/genética , Neoplasias Peritoneales/genética , Pronóstico , Transducción de Señal/genética
8.
Ann Surg Oncol ; 21 Suppl 4: S601-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24770678

RESUMEN

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is a deadly disease with a poor prognosis. The single nucleotide polymorphisms (SNPs) involved in microRNA (miRNA) functions are potential biomarkers for prognosis of various human cancers. We investigated the association of the miRNA-related SNPs with the prognosis of ESCC. METHODS: A total of 504 patients with ESCC were enrolled. The genotypes of 18 miRNA-related SNPs were analyzed from the genomic DNA of peripheral leukocytes and were correlated with the prognosis of patients randomly assigned to a training set (n = 129) or an independent replication set (n = 375). RESULTS: In the training group, only the rs4919510 SNP of the mir-608 gene was significantly associated with clinical outcome (CG vs. GG, hazard ratio [HR] 0.47, 95 % confidence interval [CI] 0.27-0.82, P = 0.008 for death, HR 0.47, 95 % CI 0.29-0.77, P = 0.002 for recurrence). The association for overall survival was confirmed in an independent replication group (CG vs. GG, HR 0.74, 95 % CI 0.56-0.97, P = 0.031 for death). Two other SNPs, rs14035 of RAN and rs7813 of GEMIN4, showed a borderline significant association with the prognosis of ESCC. In a combined analysis, we demonstrated the cumulative effect of the mir-608, RAN, and GEMIN4 polymorphisms on the clinical outcome of ESCC (HR 1.40, 95 % CI 1.18-1.67, P trend < 0.001 for mortality, HR 1.30, 95 % CI 1.10-1.53, P trend = 0.002 for recurrence). The cumulative effect was more evident in patients receiving concurrent chemoradiotherapy. CONCLUSIONS: The hereditary genetic polymorphisms of mir-608, RAN, and GEMIN4 can serve as predictors for clinical outcome in ESCC patients treated with concurrent chemoradiotherapy.


Asunto(s)
Carcinoma de Células Escamosas/genética , Neoplasias Esofágicas/genética , MicroARNs/genética , Recurrencia Local de Neoplasia/genética , Ribonucleoproteínas Nucleares Pequeñas/genética , Proteína de Unión al GTP ran/genética , Anciano , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/terapia , Terapia Combinada , Supervivencia sin Enfermedad , Neoplasias Esofágicas/mortalidad , Neoplasias Esofágicas/terapia , Esofagectomía , Femenino , Genotipo , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Antígenos de Histocompatibilidad Menor , Polimorfismo de Nucleótido Simple , Estudios Retrospectivos , Tasa de Supervivencia
9.
Cancer Cell ; 9(3): 209-23, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16530705

RESUMEN

Flt-4, a VEGF receptor, is activated by its specific ligand, VEGF-C. The resultant signaling pathway promotes angiogenesis and/or lymphangiogenesis. This report provides evidence that the VEGF-C/Flt-4 axis enhances cancer cell mobility and invasiveness and contributes to the promotion of cancer cell metastasis. VEGF-C/Flt-4-mediated invasion and metastasis of cancer cells were found to require upregulation of the neural cell adhesion molecule contactin-1 through activation of the Src-p38 MAPK-C/EBP-dependent pathway. Examination of tumor tissues from various types of cancers revealed high levels of Flt-4 and VEGF-C expression that correlated closely with clinical metastasis and patient survival. The VEGF-C/Flt-4 axis, through upregulation of contactin-1, may regulate the invasive capacity in different types of cancer cells.


Asunto(s)
Adenocarcinoma/patología , Neoplasias Pulmonares/patología , Invasividad Neoplásica/patología , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adenocarcinoma/metabolismo , Animales , Moléculas de Adhesión Celular Neuronal/metabolismo , Movimiento Celular , Contactina 1 , Contactinas , Femenino , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Neoplasias Pulmonares/metabolismo , Metástasis Linfática/patología , Masculino , Ratones , Persona de Mediana Edad , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Biomed Chromatogr ; 28(9): 1284-93, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24691749

RESUMEN

A two-dimensional (2D) hydrophilic interaction liquid chromatography (HILIC) and reverse-phase (RP) liquid chromatography (LC) system coupled with triple-quadrupole mass spectrometry (MS) was developed to comprehensively profile ceramides and phosphatidylcholine in extracted biological samples. Briefly, the 2D HILIC-RPLC system used a silica HILIC column operated in the first dimension to distinguish the lipid classes and a BEH C18 column operated in the second dimension to separate the lipid species of the same class. The regression linearity of each lipid was satisfactory in both systems; however, the absolute matrix effect factor was reduced in 2D LC-MS/MS system. Limits of detection of 2D LC-MS/MS system were 2- to 3-fold lower compared with one-dimensional RPLC-MS/MS. The recovery from the sample ranged from 84.5 to 110%. To summarize, the developed method was proven to be accurate and producible, as relative standard deviations remained <20% at three spiked levels. The efficiency of this newly developed system was applied to measure changes of lipids in the liver of mice after naphthalene treatment. Orthogonal projection to latent structures-discriminant analysis discriminated the lipids from control and the treatment group. We concluded that 2D LC-MS/MS is a promising method to assist lipidomic studies of complex biological samples.


Asunto(s)
Ceramidas/análisis , Cromatografía Liquida/métodos , Hígado/metabolismo , Fosfatidilcolinas/análisis , Espectrometría de Masas en Tándem/métodos , Animales , Ceramidas/química , Ceramidas/metabolismo , Límite de Detección , Modelos Lineales , Hígado/química , Hígado/efectos de los fármacos , Masculino , Metabolómica , Ratones , Ratones Endogámicos ICR , Naftalenos/toxicidad , Fosfatidilcolinas/química , Fosfatidilcolinas/metabolismo , Reproducibilidad de los Resultados
11.
Biomacromolecules ; 14(9): 3183-91, 2013 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-23909559

RESUMEN

Oral cancer, a subtype of head and neck cancer, is one of the leading causes of cancer death and is difficult to detect in the early stages. Improved methods of detecting primary oral lesions during endoscopy would significantly improve cancer survival rates. Here we report a high-performance nanoparticle for photodynamic detection of oral cancer. Succinate-modified chitosan (SCHI) is physically complexed with folic-acid-modified chitosan to form nanoparticles with a high drug loading efficiency and to improve drug release in the cellular lysosome. The z-average diameter and zeta potential of the prepared nanoparticles (fSCN) were 110.0 nm and 18.6 mV, respectively, enough to keep the nanoparticles stable in aqueous suspension without aggregating. When loaded with 5-aminolaevulinic acid (5-ALA; 72.8% loading efficiency) in the prepared fSCNA, there were no significant differences between the fSCN and fSCNA in particle size or zeta potential. Moreover, the fSCNA nanoparticles were readily engulfed by oral cancer cells via folate-receptor-mediated endocytosis. The release of loaded 5-ALA in the lysosome was promoted by the reduced attraction intensity between chitosan and 5-ALA via the deprotonated SCHI molecules, which resulted in a higher accumulation of intracellular protoporphyrin IX (PpIX) for photodynamic detection. These results demonstrate that N-succinyl-chitosan-incorporated and folic-acid-conjugated chitosan nanoparticles are an excellent vector for oral-specific delivery of 5-ALA for fluorescent endoscopic detection.


Asunto(s)
Quitosano/análogos & derivados , Quitosano/química , Neoplasias de la Boca/diagnóstico , Nanopartículas , Animales , Línea Celular Tumoral , Femenino , Fluoresceínas/química , Fluoresceínas/metabolismo , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Ácido Fólico/química , Transportadores de Ácido Fólico/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Lisosomas/metabolismo , Ratones , Ratones Endogámicos ICR , Ratones SCID , Neoplasias de la Boca/metabolismo , Nanopartículas/química , Nanopartículas/metabolismo , Trasplante de Neoplasias , Imagen Óptica , Tamaño de la Partícula , Protoporfirinas/metabolismo , Anhídridos Succínicos/química
13.
Cancer ; 118(23): 5757-67, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22736399

RESUMEN

BACKGROUND: Annexin A1 (AnxA1) has been well-known as a glucocorticoid-regulated anti-inflammatory protein, and it is implicated in tumorigenesis in a tumor type-specific pattern. However, the role of AnxA1 in gastric cancer (GC) is indeterminate, and the underlying mechanism is not clear. The purpose of this study was to evaluate the prognostic significance and associated mechanism of AnxA1 in GC. METHODS: Immunohistochemical staining was employed to analyze 118 GC patients. Both AnxA1 gain-of-function and loss-of-function approaches were performed in GC cells. Western blotting and reverse-transcription polymerase chain reaction were used for assessment of the AnxA1 regulation mechanism in GC cells. An intraperitoneal inoculation model in severe combined immunodeficient mice was used for an in vivo assay. RESULTS: High AnxA1 expression was significantly associated with peritoneal metastasis (P = .009) and serosal invasion (P = .044). Cox multivariate analysis showed that high AnxA1 expression was an independent risk factor for poor overall survival in GC patients (P = .037). AnxA1 expression positively correlated with invasiveness of human GC cells both in vitro and in vivo. AnxA1 could regulate the GC cell invasion through the formyl peptide receptor (FPR)/extracellular signal-regulated kinase/integrin beta-1-binding protein pathway, and all 3 FPRs (FPR1 through FPR3) were involved in the regulation process. CONCLUSIONS: High AnxA1 expression was associated with more serosal invasion, more peritoneal metastasis, and poorer overall survival in GC patients. The current study demonstrated a novel mechanism involving FPRs, extracellular signal-regulated kinases 1 and 2, and integrin beta-1-binding protein 1 by which AnxA1 regulated GC cell invasion.


Asunto(s)
Anexina A1/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de la Membrana/fisiología , Receptores de Formil Péptido/fisiología , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Proteínas Adaptadoras Transductoras de Señales , Adulto , Anciano , Animales , Anexina A1/análisis , Femenino , Células HEK293 , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos NOD , Persona de Mediana Edad , Invasividad Neoplásica , Fosforilación
14.
Org Biomol Chem ; 9(6): 1917-26, 2011 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-21283915

RESUMEN

An efficient soluble polymer-supported method has been developed for the parallel synthesis of substituted benzimidazole linked benzoxazoles using focused microwave irradiation. The key step involves the amidation of 4-hydroxy-3-nitrobenzoic acid with polymer-immobilized o-phenylenediamine. Application of mild acidic conditions promoted the ring closure to furnish the benzimidazole ring. After hydrogenation of the nitro-group to amine, the resulted polymer conjugates underwent efficient ring closure with various alkyl, aryl and heteroaryl isothiocyanates to generate the polymer-bound benzimidazolyl benzoxazoles. The polymer-bound compounds were finally cleaved from the support to furnish benzimidazole linked benzoxazole derivatives. The efficacy of the resultant angular bis-heterocyclic library was studied against vascular endothelial growth factor receptor (VEGFR-3). The preliminary screening of these novel compounds exhibits moderate to high inhibition (IC(50) = 0.56-1.42 µM). This protocol provides an easy access to novel angular bis-heterocycles which have potential for the discovery of novel leads for targeted cancer therapeutics.


Asunto(s)
Bencimidazoles/química , Benzoxazoles/síntesis química , Polímeros/química , Inhibidores de Proteínas Quinasas/síntesis química , Receptor 3 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Línea Celular Tumoral , Humanos , Concentración 50 Inhibidora , Microondas , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/farmacología
15.
J Immunol ; 182(12): 7916-27, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19494316

RESUMEN

Thrombin plays an important role in lung inflammatory diseases. Thrombin can induce connective tissue growth factor (CTGF) expression in lung fibroblasts. However, little is known about the signaling pathway in thrombin-induced CTGF expression. In this study, we investigated the role of apoptosis signal-regulating kinase 1 (ASK1) in thrombin-induced CTGF expression in human lung fibroblasts. Thrombin caused a concentration- and time-dependent increase in CTGF expression in WI-38 cells and primary lung fibroblasts. Thrombin-induced CTGF expression and CTGF-luciferase activity were inhibited by a protease-activated receptor 1 antagonist (SCH79797), the dominant-negative mutants (DNs) of ASK1 and JNK1/2, and an AP-1 inhibitor (curcumin). Thrombin caused ASK1 Ser(967) dephosphorylation, the dissociation of ASK1 and 14-3-3, and a subsequent increase in ASK1 activity. Thrombin induced increases in JNK phosphorylation and kinase activity, which were attenuated by ASK1DN. Furthermore, SCH79797 diminished the thrombin-induced ASK1 and JNK activities. Thrombin-induced CTGF-luciferase activity was predominately controlled by the sequence -747 to -184 bp upstream of the transcription start site of the human CTGF promoter and was attenuated by transfection with the deleted AP-1 binding site construct. Thrombin caused increases in c-Jun phosphorylation, the formation of an AP-1-specific DNA-protein complex, and the recruitment of c-Jun to the CTGF promoter. Furthermore, thrombin-mediated AP-1 activation was inhibited by ASK1DN, JNK1/2DN, and SP600125. These results suggest for the first time that thrombin, acting through protease-activated receptor 1, activates the ASK1/JNK signaling pathway, which in turn initiates c-Jun/AP-1 activation and recruitment of c-Jun to the CTGF promoter and ultimately induces CTGF expression in human lung fibroblasts.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Pulmón/metabolismo , MAP Quinasa Quinasa Quinasa 5/metabolismo , Transducción de Señal , Factor de Transcripción AP-1/metabolismo , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/genética , Activación Enzimática , Fibroblastos , Humanos , Regiones Promotoras Genéticas/genética , Receptores Proteinasa-Activados/metabolismo , Transcripción Genética/genética
16.
Arch Toxicol ; 84(8): 651-60, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20464547

RESUMEN

Fibroblast growth factor (FGF)-9 belongs to the FGF family which modulate cell proliferation, differentiation, and motility. Benzo(a)pyrene is a polycyclic aromatic hydrocarbon (PAH) and ubiquitous environmental carcinogen present in automobile exhaust, cigarette smoke, and foods. The major purposes of this study were to explore the roles of FGF-9 in the benzo(a)pyrene-induced lung cancer invasion in vitro and the metastatic development of lung adenocarcinoma in human. The data of RT-PCR analysis indicated that treatments of human lung adenocarcinoma CL5 cells with benzo(a)pyrene and a PAH mixture motorcycle exhaust particulate (MEP) extracts increased FGF-9 mRNA expression. The increased expression was blocked by cotreatments with a p38 mitogen-activated protein kinase inhibitor SB202190 and an extracellular signal-regulated kinase inhibitor PD98059. The results of immunoblot analysis and Matrigel assay showed that benzo(a)pyrene and MEP extracts produced a concomitant induction of FGF-9 protein and invasive ability of CL5 cells. The benzo(a)pyrene- and MEP-induced invasion was suppressed by FGF-9 neutralizing antibodies. The results of immunohistochemistry analysis of human lung adenocarcinoma specimens showed that FGF-9 protein was detected in the adenocarcinoma cells but not in normal epithelium. FGF-9 staining intensity was positively correlated with status of disease and degree of lymph node metastasis in these lung adenocarcinomas. These present findings suggest that FGF-9 has potential roles in benzo(a)pyrene-induced CL5 cell invasion and human lung adenocarcinoma metastasis.


Asunto(s)
Adenocarcinoma/inducido químicamente , Benzo(a)pireno/toxicidad , Carcinógenos Ambientales/toxicidad , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Neoplasias Pulmonares/inducido químicamente , Adenocarcinoma/metabolismo , Adenocarcinoma/secundario , Adulto , Anciano , Femenino , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Invasividad Neoplásica
17.
Carcinogenesis ; 30(12): 2005-13, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19825968

RESUMEN

Vascular endothelial growth factor (VEGF)-C is recognized as a tumor lymphangiogenic factor based on the effects of activated VEGF-R3 on lymphatic endothelial cells. Many tumor cells express VEGF-R3 but the function of this receptor in tumor cells is largely unknown. It has been reported that the VEGF-C/VEGF-R3 axis is activated in subsets of leukemia patients. Herein, we have shown that VEGF-C induces angiogenic activity in the tube formation assay invitro and Matrigel plug assay in vivo by upregulating an angiogenic factor, cyclooxygenase-2 (COX-2), through VEGF-R3 in the human acute myeloid leukemia (AML) cell line, THP-1. COX-2 induction by VEGF-C was also observed in other VEGF-R3(+) human AML cell lines (U937 and HL60). Moreover, immunohistochemical analysis of bone marrow specimens of 37 patients diagnosed with AML revealed that VEGF-C expression in specimens was associated with the expression of COX-2 (P < 0.001). The manner by which signaling pathways transduced by VEGF-C is responsible for COX-2 upregulation was further investigated. Blocking the p42/44 mitogen-activated protein kinase (MAPK) pathway with the MAPK kinase inhibitor, PD 98059, failed to inhibit VEGF-C-mediated COX-2 expression. However, VEGF-C-induced COX-2 upregulation was effectively abolished by overexpression of dominant-negative c-Jun N-terminal kinase (JNK) or treatment with the JNK inhibitor, SP 600125. VEGF-C induced JNK-dependent nuclear translocation of c-Jun. Furthermore, chromatin immunoprecipitation and reporter assays revealed that VEGF-C enhanced c-Jun binding to the cyclic adenosine 3',5'-monophosphate-response element of the COX-2 promoter and induced COX-2 expression. In sum, the data herein highlight the pathogenic role of VEGF-C in leukemia via regulation of angiogenesis through upregulation of COX-2.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , MAP Quinasa Quinasa 4/metabolismo , Neovascularización Patológica , Factor de Transcripción AP-1/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Línea Celular Tumoral , Células Endoteliales/citología , Células HL-60 , Humanos , Inmunohistoquímica/métodos , Sistema de Señalización de MAP Quinasas , Células U937
18.
Carcinogenesis ; 30(2): 205-13, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18849299

RESUMEN

Stromal cell-derived factor 1alpha (SDF-1alpha) (CXCL12) has been observed to enhance tumor angiogenesis. However, the comprehensive role of SDF-1alpha (CXCL12)-CXCR4 interaction, exerted during angiogenesis, has not been well understood. We have previously demonstrated that human basal cell carcinoma (BCC) tissues and a BCC cell line (BCC-1/KMC) had significant expression of CXCR4, whose level was higher in invasive than in the non-invasive BCC types. Here, we observed that human BCC tissues with high expression levels of CXCR4 had higher vascularity. Further, among the 71 BCCs diagnosed between the years 2004-2005, BCCs with high CXCR4 expression had concomitantly higher microvessel density, as compared with those with low CXCR4 expression (P < 0.001). We found that SDF-1alpha induced angiogenic activity in human BCC cells, both in vitro and in vivo. SDF-1alpha significantly upregulated several angiogenesis-associated genes such as interferon-alpha-inducible protein 27, interleukin (IL)-6, bone morphogenetic protein (BMP)-6, SOCS2 and cyclooxygenase 2 (COX)-2 in human BCC cells. Among them, IL-6 was the earliest and highest upregulated gene whose induction was observed within 6 h of the commencement of SDF-1alpha-CXCR4 interaction. The mechanisms behind the SDF-1alpha-induced time and dose-dependent upregulation of messenger RNA expression and protein secretion of IL-6 were investigated. The transcriptional regulation of IL-6 by SDF-1alpha was mediated by phosphorylation of extracellular signal-related kinase 1/2 and activation of the nuclear factor-kappaB complex. The identification of the angiogenic profiles induced through SDF-1alpha-CXCR4 interactions in human BCC cells may contribute further insights into the mechanisms involved in the angiogenic potential of SDF-1alpha (CXCL12).


Asunto(s)
Carcinoma Basocelular/metabolismo , Quimiocina CXCL12/fisiología , Interleucina-6/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , FN-kappa B/metabolismo , Neovascularización Patológica/metabolismo , Neoplasias Cutáneas/metabolismo , Biomarcadores de Tumor/metabolismo , Carcinoma Basocelular/irrigación sanguínea , Carcinoma Basocelular/patología , Línea Celular Tumoral , Humanos , Neovascularización Patológica/patología , Fosforilación , Receptores CXCR4/metabolismo , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/patología , Regulación hacia Arriba
19.
Ann Surg Oncol ; 16(2): 524-33, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19050968

RESUMEN

The purpose of this study was to identify genes of interest for a subsequent functional and clinical cohort study using complementary (c)DNA microarrays. cDNA microarray hybridization was performed to identify differentially expressed genes between tumor and nontumor specimens in 30 gastric cancer patients. Subsequent functional studies of the selected gene were carried out, including cell cycle analysis, cell migration analysis, analyses of vascular endothelial growth factor (VEGF) and placenta growth factor (PlGF), and oligo-microarray studies using two pairs of stable cell lines of the selected gene. Another independent cohort study of 79 gastric cancer patients was conducted to evaluate the clinical significance of the selected gene in human gastric cancer. Calreticulin (CRT) was selected for further investigation. Two pairs of stable cell lines of CRT overexpression and CRT knockdown were constructed to perform functional studies. CRT enhanced gastric cancer cell proliferation and migration. Overexpressed CRT upregulated the expression and secretion of PlGF and VEGF. CRT had a reciprocal effect on connective tissue growth factor (CTGF) expression. Positive immunohistochemical staining of calreticulin was significantly correlated with high microvessel density (MVD) (p = 0.014), positive serosal invasion (p = 0.013), lymph node metastasis (p = 0.002), perineural invasion (p = 0.008), and poor patient survival (p = 0.0014). Multivariate survival analysis showed that CRT, MVD, and serosal invasion were independent prognosticators. We conclude that CRT overexpression enhances angiogenesis, and facilitates proliferation and migration of gastric cancer cells, which is in line with the association of CRT with MVD, tumor invasion, lymph node metastasis, and survival in gastric cancer patients.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Calreticulina/metabolismo , Neovascularización Patológica/metabolismo , Neoplasias Gástricas/irrigación sanguínea , Biomarcadores de Tumor/genética , Western Blotting , Calreticulina/genética , Ciclo Celular , Movimiento Celular , Proliferación Celular , Estudios de Cohortes , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Perfilación de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Metástasis Linfática , Masculino , Microcirculación , Persona de Mediana Edad , Estadificación de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor de Crecimiento Placentario , Proteínas Gestacionales/genética , Proteínas Gestacionales/metabolismo , Pronóstico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/patología , Tasa de Supervivencia , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
J Comb Chem ; 11(6): 1038-46, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19852442

RESUMEN

Pharmaceutically interesting, angular bis-benzimidazoles with three appendages have been synthesized successfully through a diversity-oriented approach with soluble support under microwave irradiation. Polymer immobilized o-phenylenediamine was selectively N-acylated with 2-chloro-3-nitrobenzoic acid in a primary aromatic amino moiety. The obtained amide was cyclized to benzimidazole in an acidic condition, and subsequently nucleophilic aromatic substitution with different amines was performed. Successive reduction, cyclization with various aldehydes and activated isothiocyanates yielded angular biheterocyclic benzimidazoles in good quantities. Reaction progress on polymer support was precisely monitored using the conventional proton NMR spectroscopy. Preliminary screening results showed some of these interesting compounds exhibited moderately to good inhibition against vascular endothelial growth factor receptor 3 (VEGFR-3), which is related to invasion and migration of cancer cells.


Asunto(s)
Bencimidazoles/síntesis química , Técnicas Químicas Combinatorias/métodos , Microondas , Bencimidazoles/química , Estructura Molecular , Estereoisomerismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA