Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Sci Rep ; 13(1): 10508, 2023 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-37380717

RESUMEN

A fundamental goal in cancer-associated genome sequencing is to identify the key genes. Protein-protein interactions (PPIs) play a crucially important role in this goal. Here, human reference interactome (HuRI) map was generated and 64,006 PPIs involving 9094 proteins were identified. Here, we developed a physical link and co-expression combinatory network construction (PLACE) method for genes of interest, which provides a rapid way to analyze genome sequencing datasets. Next, Kaplan‒Meier survival analysis, CCK8 assays, scratch wound assays and Transwell assays were applied to confirm the results. In this study, we selected single-cell sequencing data from patients with hepatocellular carcinoma (HCC) in GSE149614. The PLACE method constructs a protein connection network for genes of interest, and a large fraction (80%) of the genes (screened by the PLACE method) were associated with survival. Then, PLACE discovered that transmembrane protein 14B (TMEM14B) was the most significant prognostic key gene, and target genes of TMEM14B were predicted. The TMEM14B-target gene regulatory network was constructed by PLACE. We also detected that TMEM14B-knockdown inhibited proliferation and migration. The results demonstrate that we proposed a new effective method for identifying key genes. The PLACE method can be used widely and make outstanding contributions to the tumor research field.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/genética , Pronóstico , Neoplasias Hepáticas/genética , Mapeo Cromosómico , Análisis de Secuencia de ARN
2.
Med Oncol ; 39(5): 51, 2022 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-35150321

RESUMEN

BACKGROUND: Non-small cell lung carcinoma (NSCLC) accounts for the majority of lung cancer which is one of the most common cancer types and results in high percentage of cancer-related deaths. Although NSCLC patients have been benefiting from the existing standard treatments, more candidate biomarkers for effective diagnosis and targets for therapy are still required to be uncovered. The expression pattern and biological function of Excision repair cross-complementation group 6 like (ERCC6L) in NSCLC are ill-investigated. METHODS: We performed bioinformatic analyses in NSCLC patients with lung adenocarcinoma (LUAD) or lung squamous cell carcinoma (LUSC), respectively. Patient survival determination and meta-analysis were carried out to check the clinical significance of ERCC6L. Datamining was also performed to evaluate the ERCC6L mRNA and protein expression levels in patients with LUAD and the correlation with immune cell infiltration. In silico prediction indicated the potential interacting proteins and correlated pathways of ERCC6L in LUAD. Loss-of-function studies were performed to determine the role of ERCC6L in LUAD cells. RESULTS: Here, we found that ERCC6L is upregulated in patients with LUAD and LUSC and is strongly associated with poor outcomes of LUAD, but not LUSC, patients. In addition, ERCC6L mRNA and protein were shown to be more expressed in patients with advanced stages of LUAD. Finally, functional analyses reveal the promoting effects of ERCC6L on LUAD cell survival, migration and invasion. CONCLUSIONS: Cohort data analysis and experimental validation shed light on the promising prognostic and therapeutic application of ERCC6L in LUAD, but maybe not LUSC, patients.


Asunto(s)
Adenocarcinoma del Pulmón/patología , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Escamosas/patología , ADN Helicasas/genética , Neoplasias Pulmonares/patología , Células A549 , Adenocarcinoma del Pulmón/genética , Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Células Escamosas/genética , Biología Computacional , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Pronóstico , ARN Mensajero/genética , Tasa de Supervivencia
3.
J Cancer Res Clin Oncol ; 147(2): 499-505, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33225417

RESUMEN

PURPOSE: There is not much progress in the treatment for lung squamous cell carcinoma LSCC in the past few years. Rapamycin Rapa, an inhibitor of mammalian target of rapamycin mTOR, has exhibited antitumor efficacy in a variety of malignant tumors. It has recently been reported that Rapamycin can induce autophagy signaling pathway in lung cancer and Glypican-3GPC3 can promote the growth of hepatocellular carcinoma by stimulating canonical Wnt signaling pathway. The aim of this study is to investigate the mechanisms of rapamycin's antitumor efficacy in relation to GPC3/Wnt/ß-catenin pathway and autophagy in LSCC. METHODS: SK-MES-1 cells, a LSCC cell line, were treated with various concentrations of rapamycin with or without Glypican-3 GPC3-targeting siRNA. SK-MES-1 cell proliferation was determined by MTT assay. Protein expression levels of GPC3, ß-catenin, Beclin-1 were checked via western blotting. We established the xenograft mice model to investigate the suppression effect of rapamycin on LSCC. In addition, we further testified the metabolism protein of autophagy process using the xenograft tumor tissue. RESULTS: Rapamycin could inhibit the SK-MES-1 cell proliferation in a concentration-dependent manner both in vitro and in vivo by decreasing the GPC3 expression and downregulating the glypican-3/Wnt/ß-catenin signaling pathway. In addition, we found that GPC3 silencing can activate the glypican-3/Wnt/ß-catenin pathway and autophagy, which contribute to the suppression of tumor growth both in vitro and in vivo. CONCLUSION: Rapamycin suppresses the growth of lung cancer through down-regulating glypican-3/Wnt/ß-catenin signaling, which mediates with activation of autophagy. This study suggests GPC3 is a new promising target for rapamycin in the treatment of lung cancer.


Asunto(s)
Autofagia/efectos de los fármacos , Carcinoma de Células Escamosas/tratamiento farmacológico , Glipicanos/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Sirolimus/farmacología , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Femenino , Glipicanos/fisiología , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/fisiología , Vía de Señalización Wnt/fisiología , beta Catenina/fisiología
4.
Gene ; 754: 144859, 2020 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-32535049

RESUMEN

DNA topoisomerases essentially remove topological strains generated during DNA replication, transcription, DNA repair, and other cytogenetic processes. However, distinct expression level and prognostic significance of individual topoisomerase isoforms in gastric cancer (GC) remain largely unexplored. In this study, we utilized Oncomine and Kaplan-Meier plotter database to detect the mRNA expression level of individual topoisomerase isoforms as well as assess their prognostic significance in GC patients. With the exception of TOP3B and TOP2B, levels of all topoisomerase isoforms were found to be elevated in GC patients when compared to the normal tissues. Elevated expression of TOP1 and TOP1MT was relevant to longer overall survival (OS) in GC and gastric intestinal type adenocarcinoma (GITA) patients, but not in diffuse gastric adenocarcinoma (DFA) patients. Increased expression of TOP2A and TOP2B was related to better OS in GC, as well as in GITA and DFA patients. In contrast, increased expression TOP3A and TOP3B was associated with shorter OS in GC, as well as in GITA and DFA patients. We also applied the Tumor IMmune Estimation Resource (TIMER) tool to assess the correlations between distinct topoisomerase isoforms and the infiltrating immune cell landscape. Furthermore, we found that down-regulating the expression of TOP3A by shRNA significantly inhibited the proliferation and colony formation in GC cells compared to control shRNA treated cells. Thus our study lays the framework for utilizing topoisomerases in better understanding the complexity and heterogeneity of GC and for developing strategies for novel customized therapy in GC patients.


Asunto(s)
Adenocarcinoma/patología , Biomarcadores de Tumor/metabolismo , ADN-Topoisomerasas de Tipo II/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Neoplasias Gástricas/patología , Adenocarcinoma/enzimología , Biomarcadores de Tumor/genética , ADN-Topoisomerasas de Tipo II/genética , Precursores Enzimáticos , Perfilación de la Expresión Génica , Humanos , Proteínas de Unión a Poli-ADP-Ribosa/genética , Pronóstico , Neoplasias Gástricas/enzimología , Tasa de Supervivencia
5.
Oncotarget ; 8(32): 52866-52876, 2017 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-28881778

RESUMEN

There are limited strategies for the treatment of hepatocellular carcinoma (HCC). In this study, we prepared a Bispecific T cell engager (BiTE) targeting Glypican 3 (GPC3) and CD3. The GPC3/CD3 BiTE was prepared by fusing the single-chain variable fragment (scFv) of the humanized anti-GPC3 antibody (9F2) with the scFv of the anti-CD3 antibody (OKT3). The in vitro and in vivo cytotoxic activities of the GPC3/CD3 BiTE were evaluated against various HCC cell lines. The GPC3/CD3 BiTE could efficiently mediate the T cell killing of GPC3-positive HCC in vitro, which was dependent on GPC3 expression on the surface of HCC cells. Moreover, our study indicates that, in the presence of the GPC3/CD3 BiTE, T cells could efficiently destroy GPC3-positive human HCC cells in vitro and in vivo. Additionally, our study further proved that GPC3 is not expressed in normal tissues. Thus, GPC3 may be a cancer-specific antigen. Collectively, these findings suggest that this anti-GPC3 BiTE might be a promising anti-tumor reagent for patients with GPC3-positive HCC.

6.
Cancer Immunol Immunother ; 66(4): 475-489, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28035433

RESUMEN

Adoptive immunotherapy leveraging chimeric antigen receptor-modified T (CAR-T) cells holds great promise for the treatment of cancer. However, tumor-associated antigens often have low expression levels in normal tissues, which can cause on-target, off-tumor toxicity. Recently, we reported that GPC3-targeted CAR-T cells could eradicate hepatocellular carcinoma (HCC) xenografts in mice. However, it remains unknown whether on-target, off-tumor toxicity can occur. Therefore, we proposed that dual-targeted CAR-T cells co-expressing glypican-3 (GPC3) and asialoglycoprotein receptor 1 (ASGR1) (a liver tissue-specific protein)-targeted CARs featuring CD3ζ and 28BB (containing both CD28 and 4-1BB signaling domains), respectively, may have reduced on-target, off-tumor toxicity. Our results demonstrated that dual-targeted CAR-T cells caused no cytotoxicity to ASGR1+GPC3- tumor cells, but they exhibited a similar cytotoxicity against GPC3+ASGR1- and GPC3+ASGR1+ HCC cells in vitro. We found that dual-targeted CAR-T cells showed significantly higher cytokine secretion, proliferation and antiapoptosis ability against tumor cells bearing both antigens than single-targeted CAR-T cells in vitro. Furthermore, the dual-targeted CAR-T cells displayed potent growth suppression activity on GPC3+ASGR1+ HCC tumor xenografts, while no obvious growth suppression was seen with single or double antigen-negative tumor xenografts. Additionally, the dual-targeted T cells exerted superior anticancer activity and persistence against single-targeted T cells in two GPC3+ASGR1+ HCC xenograft models. Together, T cells carrying two complementary CARs against GPC3 and ASGR1 may reduce the risk of on-target, off-tumor toxicity while maintaining relatively potent antitumor activities on GPC3+ASGR1+ HCC.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Carcinoma Hepatocelular/terapia , Inmunoterapia Adoptiva/métodos , Neoplasias Hepáticas/terapia , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/fisiología , Animales , Receptor de Asialoglicoproteína/inmunología , Carcinoma Hepatocelular/inmunología , Línea Celular Tumoral , Proliferación Celular , Citocinas/metabolismo , Glipicanos/inmunología , Humanos , Neoplasias Hepáticas/inmunología , Activación de Linfocitos , Ratones , Ratones SCID , Especificidad de Órganos , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión/genética , Especificidad del Receptor de Antígeno de Linfocitos T , Linfocitos T/trasplante , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Oncotarget ; 7(17): 24752-65, 2016 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-27029073

RESUMEN

There are still unmet medical needs for the treatment of glioblastoma (GBM), the most frequent and aggressive brain tumor worldwide. EGFRvIII, overexpressed in approximately 30% of GBM, has been regarded as a potential therapeutic target. In this study, we demonstrated that CH12, an anti-EGFRvIII monoclonal antibody, could significantly suppress the growth of EGFRvIII+ GBM in vivo; however, PTEN deficiency in GBM reduced the efficacy of CH12 by attenuating its effect on PI3K/AKT/mTOR pathway. To overcome this problem, CH12 was combined with the mTOR inhibitor rapamycin, leading to a synergistic inhibitory effect on EGFRvIII+PTEN- GBM in vivo. Mechanistically, the synergistic antitumor effect was achieved via attenuating EGFR and PI3K/AKT/mTOR pathway more effectively and reversing the STAT5 activation caused by rapamycin treatment. Moreover, the combination therapy suppressed angiogenesis and induced cancer cell apoptosis more efficiently. Together, these results indicated that CH12 and rapamycin could synergistically suppress the growth of EGFRvIII+PTEN- GBM, which might have a potential clinical application in the future.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Receptores ErbB/inmunología , Glioblastoma/tratamiento farmacológico , Sirolimus/farmacología , Animales , Antibacterianos/administración & dosificación , Antibacterianos/farmacología , Anticuerpos Monoclonales/administración & dosificación , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Sinergismo Farmacológico , Glioblastoma/enzimología , Glioblastoma/patología , Humanos , Ratones , Fosfohidrolasa PTEN/metabolismo , Sirolimus/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Oncotarget ; 7(3): 2496-507, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26684028

RESUMEN

There are unmet medical needs for patients with lung squamous cell carcinoma (LSCC). Therefore, in this study, we explored the antitumor potential of third-generation glypican 3 (GPC3)-redirected chimeric antigen receptor (CAR)-engineered T lymphocytes (CARgpc3 T cells) in tumor models of LSCC. First, we demonstrated by immunohistochemistry (IHC) that GPC3 was expressed in 66.3% of LSCC samples and in 3.3% of lung adenocarcinoma (LAD) samples but not in normal lung tissues. In the presence of GPC3-positive LSCC cells, CARgpc3 T cells were highly activated and increased in number. CARgpc3 T cells could specifically lyse GPC3-positive LSCC cells in vitro. In two established LSCC xenograft models, CARgpc3 T cells could almost completely eliminate the growth of GPC3-positive cells. Additionally, the CARgpc3 T cells were able to persist in vivo and efficiently infiltrate the cancerous tissues. Taken together, these findings indicate that CARgpc3 T cells might be a novel potential therapeutic agent for the treatment of patients with LSCC.


Asunto(s)
Carcinoma de Células Escamosas/terapia , Glipicanos/inmunología , Inmunoterapia Adoptiva , Neoplasias Pulmonares/terapia , Linfocitos T Citotóxicos/inmunología , Animales , Apoptosis , Western Blotting , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Proliferación Celular , Citometría de Flujo , Glipicanos/metabolismo , Humanos , Técnicas para Inmunoenzimas , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Receptores de Antígenos de Linfocitos T/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Oncotarget ; 6(36): 38840-53, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26474285

RESUMEN

Although Trastuzumab, an anti-HER2 antibody, benefits certain patients with HER2-overexpressing breast cancer, de novo or acquired trastuzumab resistance remains a haunting issue. EGFRvIII, co-expressing with HER2 in some breast tumors, indicates a poor clinical prognosis. However, the role of EGFRvIII in the function of trastuzumab is not clear. Here, we demonstrated that EGFRvIII overexpression contributed to de novo trastuzumab resistance and the feedback activation of STAT3 caused by trastuzumab also resulted in acquired resistance in EGFRvIII(+)HER2(+) breast cancers. CH12, a highly effective anti-EGFRvIII monoclonal antibody that preferentially binds to EGFRvIII, significantly suppressed the growth of EGFRvIII+HER2(+) breast cancer cells in vitro and in vivo. Importantly, CH12 in combination with trastuzumab had a synergistic inhibitory effect on EGFRvIII(+)HER2(+) breast cancers in vitro and in vivo via attenuating the phosphorylation of EGFR and HER2 and their downstream signal pathways more effectively and reversing STAT3 feedback activation. Moreover, the combination therapy suppressed angiogenesis and induced cell apoptosis significantly. Together, these results suggested a synergistic efficacy of the combination of trastuzumab with CH12 against EGFRvIII(+)HER2(+) breast cancers, which might be a potential clinical application in the future.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Receptores ErbB/inmunología , Receptor ErbB-2/metabolismo , Trastuzumab/farmacología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Receptores ErbB/biosíntesis , Femenino , Humanos , Ratones , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Trastuzumab/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA