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











Base de datos
Intervalo de año de publicación
1.
Cancer Lett ; 558: 216059, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36813650

RESUMEN

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Editor-in-Chief and Authors. Following concerns raised in the public domain, the authors contacted the journal to request the retraction of the article. Sections of panels from various figures appear similar to each other, particularly panels from Figs. 3G, 5B and 3G and 5F, 3F, S4D, S5D, S5C and S10C, as well as S10E.

2.
Cell Death Dis ; 13(9): 835, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-36171198

RESUMEN

Intersectin 1 (ITSN1) contains two isoforms: ITSN1-S and ITSN1-L, which are highly regulated by alternative splicing. Our previous results showed that the two isoforms of ITSN1 displayed opposite functions: ITSN1-S promoted glioma development, while ITSN1-L exerted an inhibitory role in glioma progression. In this study, our transcriptome analysis using a large glioma cohort indicated that the ratio of ITSN1-S/ITSN1-L was positively correlated with glioma grading and poor prognosis. We identified the RNA-binding protein polypyrimidine tract-binding protein 1 (PTBP1) as an ITSN1 pre-mRNA interaction protein through RNA pull-down assay and RNA immunoprecipitation assay. Knockdown of PTBP1 decreased the ratio of ITSN1-S/ITSN1-L. Minigene reporter assay and mutation analyses further confirmed PTBP1 targeted polypyrimidine sequences on ITSN1 exon 30 (TTGCACTTCAGTATTTT) and promoted the inclusion of ITSN1 exon 30. Subsequently, silencing PTBP1 inhibited glioma cell proliferation, migration, and invasion by down-regulating the ratio of ITSN1-S/ITSN1-L. Taken together, our study provides a novel mechanism that PTBP1 modulates the alternative splicing of ITSN1 and promotes glioma proliferation and motility by up-regulating the ratio of ITSN1-S/ITSN1-L, thereby highlighting that PTBP1 may be an attractive therapeutic target for gliomas.


Asunto(s)
Empalme Alternativo , Glioma , Proteínas Adaptadoras del Transporte Vesicular , Empalme Alternativo/genética , Movimiento Celular/genética , Glioma/genética , Glioma/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Isoformas de Proteínas/metabolismo , Precursores del ARN/metabolismo , Proteínas de Unión al ARN/metabolismo
3.
Front Genet ; 13: 866702, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35685435

RESUMEN

Background: YAP, coded by YAP1 gene, is critical in the Hippo pathway. It has been reported to be involved in the tumorigenesis and progression of several cancers. However, its roles on tumor cell proliferation in diverse cancers remain to be elucidated. And there is currently no clinically feasible drug that can directly target YAP in cancers. This research aimed to explore the regulatory mechanism of YAP in promoting tumor proliferation of multiple cancers, in order to find new strategies for inhibiting the overgrowth of YAP-driven cancers. Methods: We investigated the expression pattern of YAP1 in pan-cancer across numerous databases and our cohorts. First, univariate Cox regression analysis and survival analysis were used to evaluate the effect of YAP1 on the prognosis of cancer patients. Second, TIMER was used to explore the relationship between YAP1 expression and tumor cell proliferation. Third, functional and pathway enrichment was performed to search for targets of YAP involved in cell cycle in cancers. At last, GDSC and CCLE datasets were used to assess the correlation between SKP2 expression and MLN4924 IC50 values. Results: Differential expression analysis of multiple databases and qPCR validation showed that YAP1 was generally overexpressed in pan-cancers. Survival analysis revealed that YAP1 over-expression was significantly related to poor prognosis of patients with PAAD. The expression level of YAP1 was positively correlated with the proliferation in varieties of tumors. Further, SKP2 was confirmed as a target of YAP in promoting tumor cell proliferation. In addition, SKP2 expression was negatively correlated with MLN4924 IC50 values in almost all cancer types. Conclusion: YAP1 is frequently overexpressed in human cancers. YAP promoted tumor cell proliferation by up-regulating SKP2 expression in multiple cancers. The comprehensive pan-cancer analysis suggested that inhibition of Skp2 with MLN4924 might be an effective therapeutic strategy for attenuating tumor cell proliferation in YAP-driven cancers.

4.
Cancer Res ; 82(12): 2254-2268, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35413105

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic disease with few effective treatments. Here we show that the mitochondrial calcium uniporter (MCU) promotes PDAC cell migration, invasion, metastasis, and metabolic stress resistance by activating the Keap1-Nrf2 antioxidant program. The cystine transporter SLC7A11 was identified as a druggable target downstream of the MCU-Nrf2 axis. Paradoxically, despite the increased ability to uptake cystine, MCU-overexpressing PDAC demonstrated characteristics typical of cystine-deprived cells and were hypersensitive to cystine deprivation-induced ferroptosis. Pharmacologic inhibitors of SLC7A11 effectively induced tumor regression and abrogated MCU-driven metastasis in PDAC. In patient-derived organoid models in vitro and patient-derived xenograft models in vivo, MCU-high PDAC demonstrated increased sensitivity to SLC7A11 inhibition compared with MCU-low tumors. These data suggest that MCU is able to promote resistance to metabolic stress and to drive PDAC metastasis in a cystine-dependent manner. MCU-mediated cystine addiction could be exploited as a therapeutic vulnerability to inhibit PDAC tumor growth and to prevent metastasis. SIGNIFICANCE: Elevated mitochondrial calcium uptake in PDAC promotes metastasis but exposes cystine addiction and ferroptosis sensitivity that could be targeted to improve pancreatic cancer treatment.


Asunto(s)
Canales de Calcio/metabolismo , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Calcio/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Cistina/metabolismo , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas
5.
J Cachexia Sarcopenia Muscle ; 12(3): 704-716, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33763996

RESUMEN

BACKGROUND: Malnutrition and muscle wasting are common features frequently observed in pancreatic ductal adenocarcinoma (PDAC) patients with cancer cachexia. They are associated with reduced survival and quality of life. Nutrition therapy is an important part of multimodal cancer care in PDAC. However, due to the complexity of nutrition assessment, only 30-60% of patients with nutritional risks receive nutritional treatment at present. It is important to identify biomarkers that may be used to improve management of PDAC-associated malnutrition. Serum insulin-like growth factor binding protein 2 (IGFBP2) has emerged as a potential serum biomarker in a variety of tumours. However, its association with malnutrition and muscle wasting in PDAC is unclear. METHODS: We evaluated the tumour IGFBP2 expression and serum IGFBP2 level in 98 PDAC patients using immunohistochemistry and enzyme-linked immunosorbent assay and analysed the correlation between them. Furthermore, we explored the relationship between IGFBP2 of both tumour and serum and nutritional status (Patient-Generated Subjective Global Assessment and skeletal muscle index). Pan02 IGFBP2 stable transfection cell lines, Pan02 PLV-IGFBP2 cells, and PLKO-IGFBP2 cells were injected subcutaneously into the flank of C57BL/6 mouse. Serum IGFBP2 levels, food intake, and body weight of these mice were measured. The degree of muscle atrophy is characterized by haematoxylin and eosin, Oil Red O, and Masson's trichrome staining. The mRNA and protein expression of several essential muscle-related signal proteins such as atrogin-1 and muscle RING finger 1 was measured. RESULTS: Among 98 patients, we found that tumour IGFBP2 expression is related to plasma IGFBP2 levels (rs  = 0.562, P < 0.001), and they significantly increased among patients with Patient-Generated Subjective Global Assessment ≥9 and correlated with overall survival. Moreover, serum IGFBP2 level is negatively correlated with skeletal muscle index (rs  = -0.600, P < 0.001) and Hounsfield units (rs  = -0.532, P < 0.001). In mice injected with Pan02 PLV-IGFBP2 cell, circulating IGFBP2 was elevated while body weight and food intake were decreased when compared with Pan02 PLV-Control group. These mice also exhibited significantly aggravated muscle fibre atrophy, lipid deposition, and increased collagen tissue, and the expression of mRNA and protein of atrogin-1 and muscle RING finger 1 in the gastrocnemius muscle is increased. Conversely, these symptoms were alleviated in the PLKO-IGFBP2 group. CONCLUSIONS: In the current study, there is a significant correlation between serum IGFBP2 levels, malnutrition, and muscle atrophy in PDAC. Our results suggested that serum IGFBP2 level might be a promising biomarker and intervention targets for PDAC-associated severe malnutrition and muscle wasting.


Asunto(s)
Carcinoma Ductal Pancreático , Desnutrición , Neoplasias Pancreáticas , Animales , Biomarcadores , Peso Corporal , Carcinoma Ductal Pancreático/complicaciones , Carcinoma Ductal Pancreático/diagnóstico , Proteínas Portadoras , Humanos , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Desnutrición/diagnóstico , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético , Atrofia Muscular , Neoplasias Pancreáticas/complicaciones , Calidad de Vida
6.
Signal Transduct Target Ther ; 5(1): 38, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32300119

RESUMEN

High expression of PD-L1 marks the poor prognosis of pancreatic ductal adenocarcinomas (PDAC). However, the regulatory mechanism of PD-L1 remains elusive. We recently reported that cancer Forkhead box protein 3 (Cancer-FOXP3 or C-FOXP3) promoted immune evasion of PDAC by recruiting Treg cells into PDAC via upregulation of CCL5. In this study, we confirmed that PD-L1 was overexpressed in PDAC samples from two independent cohorts of patients with radical resection. Moreover, C-FOXP3 was colocalized and correlated with the expression of PD-L1 in tumor cells at the mRNA and protein levels, and this finding was confirmed by the The Cancer Genome Atlas (TCGA) database. Chromatin immunoprecipitation (ChIP) revealed that C-FOXP3 directly bound to the promoter region of PD-L1 in pancreatic cancer cells. Furthermore, overexpression of C-FOXP3 activated the luciferase reporter gene under the control of the PD-L1 promoter. However, mutation of the binding motif-a completely reversed the luciferase activity. In addition, C-FOXP3-induced upregulation of PD-L1 effectively inhibited the activity of CD8+ T cells. Based on our recent finding that the CCL-5 antibody achieved a better response to PDAC models with high C-FOXP3 levels, we further demonstrated that the PD-L1 antibody strengthened the antitumor effect of CCL-5 blockade in xenograft and orthotopic mouse models with high C-FOXP3 levels. In conclusion, C-FOXP3 directly activates PD-L1 and represents a core transcription factor that mediates the immune escape of PDAC. Combined blockade of PD-L1 and CCL-5 may provide an effective therapy for patients with PDAC that have high C-FOXP3 levels.


Asunto(s)
Adenocarcinoma/inmunología , Antígeno B7-H1/genética , Carcinoma Ductal Pancreático/inmunología , Quimiocina CCL5/genética , Factores de Transcripción Forkhead/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma/terapia , Anciano , Animales , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Linfocitos T CD8-positivos/inmunología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/terapia , Quimiocina CCL5/antagonistas & inhibidores , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Xenoinjertos , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratones , Persona de Mediana Edad , Pronóstico , Linfocitos T Reguladores/inmunología
7.
Cancer Lett ; 453: 10-20, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30922919

RESUMEN

Perineural invasion (PNI) is a typical pathological feature of pancreatic ductal adenocarcinoma (PDAC). PNI is associated with poor prognosis of PDAC patients, however, the underlying molecular mechanisms in the development of PNI have not been fully revealed. In this study, we found that the expression of GM-CSF and HIF-1α were dramatically increased in PDAC cells. The overexpression of HIF-1α and GM-CSF closely correlated with increased occurrence of PNI and cancer-related pain and shortened overall survival in PDAC patients. GM-CSF expression in PDAC cells was mediated by HIF-1α through direct binding to the hypoxia response element in the GM-CSF promoter. The activated HIF-1α can up-regulate GM-CSF expression and secretion, which promoted the migration of Schwann cells in tumor microenvironment. Furthermore, GM-CSF overexpression could rescue the inhibition of Schwann cells migration by HIF-1α knockdown. Moreover, HIF-1α inhibition with PX478 drastically reduced the expression level of GM-CSF and decreased the PNI in a PDAC xenograft mouse model. Overall, our results demonstrated that the HIF-1α/GM-CSF pathway is involved in the tumor-nerve interactions and promotes the occurrence of PNI. The blockade of this signal might help to inhibit PDAC progression.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Pancreáticas/patología , Células de Schwann/patología , Adulto , Anciano , Animales , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Movimiento Celular/fisiología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Estadificación de Neoplasias , Neoplasias Pancreáticas/metabolismo , Nervios Periféricos/patología , Células de Schwann/metabolismo , Regulación hacia Arriba
8.
Cancer Biol Med ; 15(1): 70-78, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29545970

RESUMEN

OBJECTIVE: Cystic lesions of the pancreas have been increasingly recognized. Some lesions exhibit benign behavior, while others have unequivocal malignant potential. Thus, accurate identification of malignancy in patients diagnosed with pancreatic cystic neoplasms (PCNs) remains a major challenge. The aim of this study was to define a combined criterion to better predict malignant lesions in patients with PCNs. METHODS: We retrospectively analyzed 165 patients who underwent resection of PCNs from October 2011 to May 2017. The relationship among malignancy and serum carbohydrate antigen 19-9 (CA19-9), preoperative neutrophil-to-lymphocyte ratio (NLR), and the presence of enhanced solid component on imaging was analyzed. RESULTS: NLR before surgery in patients with malignant PCNs (2.81±2.14) was significantly higher than that in patients diagnosed with pancreatic neuroendocrine tumor (1.90±0.69, P=0.013) or healthy volunteers (1.40±0.48; P<0.001). Serum CA19-9 ≥39 U/mL, NLR >1.976 and presence of enhanced solid component were independent predictors of PCN malignancy. A combined criterion meeting any two or more of the three elements including CA19-9 ≥39 U/mL, NLR >1.976, and presence of enhanced solid component on computed tomography imaging is an indicator with a high positive predictive value of 80.5% and a high negative predictive value of 87.9%, and thus, represents a highly accurate test (86.1%). CONCLUSIONS: The new combined criterion is an effective predictor of tumor malignancy in patients with PCNs.

9.
Mol Med Rep ; 17(2): 2543-2548, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29207108

RESUMEN

The aim of the present study was to screen the potential osteosarcoma (OS)­associated genes and to obtain additional insight into the pathogenesis of OS. Transcriptional profile (ID: GSE28256) and copy number variations (CNV) profile were downloaded from Gene Expression Omnibus database. Differentially expressed genes (DEGs) between MET proto­oncogene­transformed human primary osteoblast (MET­HOB) samples and the control samples were identified using the Linear Models for Microarray Data package. Subsequently, CNV areas and CNVs were identified using cut­off criterion of >30%­overlap within the cases using detect_cnv.pl in PennCNV. Genes shared in DEGs and CNVs were obtained and discussed. Additionally, the Database for Annotation, Visualization and Integrated Discovery was used to identify significant Gene Ontology (GO) functions and pathways in DEGs with P<0.05. A total of 1,601 DEGs were screened out in MET­HOBs and compared with control samples, including 784 upregulated genes, such as E2F transcription factor 1 (E2F1) and 2 (E2F2) and 817 downregulated genes, such as retinoblastoma 1 (RB1) and cyclin D1 (CCND1). DEGs were enriched in 344 GO terms, such as extracellular region part and extracellular matrix and 14 pathways, including pathways in cancer and extracellular matrix­receptor interaction. Additionally, 239 duplications and 439 deletions in 678 genes from 1,313 chromosome regions were detected. A total of 12 genes were identified to be CNV­driven genes, including cadherin 18, laminin subunit α 1, spectrin ß, erythrocytic, ciliary rootlet coiled­coil, rootletin pseudogene 2, ß­1,4-N-acetyl-galactosaminyltransferase 1, G protein regulated inducer of neurite outgrowth 1, EH domain binding protein 1­like 1, growth factor independent 1, cathepsin Z, WNK lysine deficient protein kinase 1, glutathione S­transferase mu 2 and microsomal glutathione S­transferase 1. Therefore, cell cycle­associated genes including E2F1, E2F2, RB1 and CCND1, and cell adhesion­associated genes, such as CDH18 and LAMA1 may be used as diagnosis and/or therapeutic markers for patients with OS.


Asunto(s)
Transformación Celular Neoplásica/genética , Variaciones en el Número de Copia de ADN , Osteoblastos/metabolismo , Transcriptoma , Línea Celular Transformada , Biología Computacional/métodos , Perfilación de la Expresión Génica , Ontología de Genes , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Osteoblastos/patología , Proteínas Proto-Oncogénicas c-met/genética
10.
Cancer Lett ; 402: 61-70, 2017 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-28576749

RESUMEN

KRAS mutation is the most common genetic event in pancreatic cancer. Whereas KRAS itself has proven difficult to inhibit, agents that target key downstream signals of KRAS, such as RAF, are possibly effective for pancreatic cancer treatment. Because selective BRAF inhibitors paradoxically induce downstream signaling activation, a pan-RAF inhibitor, LY3009120 is a better alternate for KRAS-mutant tumor treatment. Here we explored a new combinational strategy using a YAP inhibitor and LY3009120 in pancreatic cancer treatment. We found that reduced YAP expression closely correlates with longer relapse-free and overall survival of patients. Stable knockdown of YAP significantly inhibited pancreatic cancer cell proliferation and tumor growth. In addition, LY3009120 exhibited a dramatically enhanced antitumor effect in combination with YAP knockdown. YAP depletion blocks the activation of a parallel AKT signal pathway after LY3009120 treatment. Finally, combination with a YAP inhibitor, verteporfin, significantly enhanced the antitumor efficacy of LY3009120. Collectively, our results demonstrate that genetic or pharmacological inhibition of YAP can increase sensitivity to LY3009120 in pancreatic cancer through blocking compensatory activation of a parallel AKT signal pathway, thereby validating a combinatorial approach for treating KRAS-mutant pancreatic cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Mutación , Neoplasias Pancreáticas/tratamiento farmacológico , Compuestos de Fenilurea/farmacología , Fosfoproteínas/antagonistas & inhibidores , Porfirinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas p21(ras)/genética , Pirimidinas/farmacología , Quinasas raf/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Carcinoma Ductal Pancreático/enzimología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Pruebas Genéticas , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Ratones Desnudos , Persona de Mediana Edad , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Fenotipo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Análisis de Matrices Tisulares , Factores de Transcripción , Transfección , Carga Tumoral/efectos de los fármacos , Verteporfina , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Señalizadoras YAP , Quinasas raf/metabolismo
11.
Cancer Lett ; 393: 113-124, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28153790

RESUMEN

Stem cell factor (SCF) is a multifunctional cytokine responsible for tumorigenesis and progression. In this study, we report that increased expression of SCF in hepatocellular carcinoma (HCC) patients is highly associated with metastasis and poor prognosis. SCF inhibition with RNAi inhibited HCC cell migration, invasion in vitro, and reduced intrahepatic metastases burden and significantly prolonged survival in a HCC xeograft mouse model. SCF depletion in HCC xeograft decreased the expression of vimentin and increase the expression of E-cadherin, implicating a role for SCF in epithelial-mesenchymal transition. Our data further demonstrated that HCC cells secreted soluble SCF to promote HUVECs angiogenesis. The overexpression of SCF in HCC is regulated by hypoxic conditions through a selective HIF-2α-dependent mechanism. Knocking-down HIF-2α significantly decreased expression of SCF. Chromatin immunoprecipitation and luciferase assay demonstrated that HIF-2α directly induce the transcription of SCF gene through the hypoxia response element in SCF promoter. In conclusion, we demonstrate that the hypoxia microenvironment in HCC up-regulates SCF expression, which in turn promotes angiogenesis and HCC metastasis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/secundario , Movimiento Celular , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Factor de Células Madre/metabolismo , Animales , Antígenos CD , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cadherinas/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/mortalidad , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidad , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Neovascularización Patológica , Neovascularización Fisiológica , Comunicación Paracrina , Pronóstico , Interferencia de ARN , Transducción de Señal , Factor de Células Madre/genética , Análisis de Supervivencia , Factores de Tiempo , Transcripción Genética , Activación Transcripcional , Transfección , Hipoxia Tumoral , Regulación hacia Arriba , Vimentina/metabolismo
12.
Cancer Lett ; 378(2): 87-96, 2016 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-27212442

RESUMEN

Arsenic trioxide (ATO) has been selected as a promising treatment not only in leukemia but also in solid tumors. Previous studies showed that the cytotoxicity of ATO mainly depends on the induction of reactive oxygen species. However, ATO has only achieved a modest effect in pancreatic ductal adenocarcinoma, suggesting that the existing radical scavenging proteins, such as hypoxia inducible factor-1, attenuate the effect. The goal of this study is to investigate the effect of combination treatment of ATO plus PX-478 (hypoxia-inducible factor-1 inhibitor) and its underlying mechanism. Here, we showed that PX-478 robustly strengthened the anti-growth and pro-apoptosis effect of ATO on Panc-1 and BxPC-3 pancreatic cancer cells in vitro. Meanwhile, in vivo mouse xenograft models also showed the synergistic effect of ATO plus PX-478 compared with any single agent. Further studies showed that the anti-tumor effect of ATO plus PX-478 was derived from the reactive oxygen species-induced apoptosis. We next confirmed that Hypoxia-inducible factor-1 cleared reactive oxygen species by its downstream target, forkhead box O transcription factors, and this effect may justify the strategy of ATO plus PX-478 in the treatment of pancreatic cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Arsenicales/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Compuestos de Mostaza/farmacología , Óxidos/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Fenilpropionatos/farmacología , Animales , Apoptosis/efectos de los fármacos , Trióxido de Arsénico , Sitios de Unión , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones Desnudos , Estrés Oxidativo/efectos de los fármacos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Regiones Promotoras Genéticas , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Activación Transcripcional , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA