Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Cancer ; 153(11): 1877-1884, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37163613

RESUMEN

Patients with metastatic colorectal cancer (mCRC) have poor long-term survival. Rechallenge with anti-epidermal growth factor receptor (anti-EGFR) based therapy has shown certain activity as late-line therapy. To further improve clinical outcomes, we evaluated the antitumor efficacy and safety of cetuximab in combination with camrelizumab and liposomal irinotecan in patients with RASwt mCRC pretreated with anti-EGFR-based therapy. Patients with RASwt mCRC who had received at least two prior systemic therapies, including anti-EGFR-based treatment in the metastatic or unresectable disease setting, were enrolled in cohort B. Patients were treated with cetuximab (500 mg/m2 ) and camrelizumab (200 mg) plus liposomal irinotecan (HR070803, 60 mg/m2 ) intravenously once every 2 weeks. The primary endpoint was the objective response rate (ORR) by RECIST v1.1. The secondary endpoints included disease control rate (DCR), progression-free survival (PFS), overall survival (OS) and safety. At the data cutoff (23 November 2022), 19 patients were enrolled in the two stages, and 16 were evaluable for efficacy analyses. The ORR was 25% (95% confidence interval [CI]: 10.2%-49.5%), and DCR was 75% (95% CI: 50.5%-89.8%). The median PFS and OS were 6.9 (95% CI: 2.6-11.2) and 15.1 (95% CI: 6.1-24.0) months, respectively. Grade 3 treatment-related adverse events (TRAEs) occurred in 15.8% (3/19) of patients. No grade ≥4 TRAEs were found in the safety population. Our study suggests that anti-EGFR retreatment therapy with cetuximab plus camrelizumab and liposomal irinotecan (HR070803) is a promising late-line treatment option with good antitumor activity and well-tolerated toxicity in RASwt mCRC patients.


Asunto(s)
Neoplasias del Colon , Neoplasias Colorrectales , Neoplasias Gastrointestinales , Neoplasias del Recto , Humanos , Cetuximab/efectos adversos , Irinotecán , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Recto/tratamiento farmacológico , Neoplasias Gastrointestinales/tratamiento farmacológico , Retratamiento , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Proteínas Proto-Oncogénicas p21(ras)/genética , Camptotecina
2.
Biol Proced Online ; 25(1): 6, 2023 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-36870954

RESUMEN

BACKGROUND: YTHDF2 is one of important readers of N6-methyladenosine (m6A) modification on RNA. Growing evidence implicates that YTHDF2 takes an indispensable part in the regulation of tumorigenesis and metastasis in different cancers, but its biological functions and underlying mechanisms remain elusive in gastric cancer (GC). AIM: To investigate the clinical relevance and biological function of YTHDF2 in GC. RESULTS: Compared with matched normal stomach tissues, YTHDF2 expression was markedly decreased in gastric cancer tissues. The expression level of YTHDF2 was inversely associated with gastric cancer patients' tumor size, AJCC classification and prognosis. Functionally, YTHDF2 reduction facilitated gastric cancer cell growth and migration in vitro and in vivo, whereas YTHDF2 overexpression exhibited opposite phenotypes. Mechanistically, YTHDF2 enhanced expression of PPP2CA, the catalytic subunit of PP2A (Protein phosphatase 2A), in an m6A-independent manner, and silencing of PPP2CA antagonized the anti-tumor effects caused by overexpression of YTHDF2 in GC cells. CONCLUSION: These findings demonstrate that YTHDF2 is down-regulated in GC and its down-regulation promotes GC progression via a possible mechanism involving PPP2CA expression, suggesting that YTHDF2 may be a hopeful biomarker for diagnosis and an unrevealed treatment target for GC.

3.
Small ; 19(24): e2207817, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36919945

RESUMEN

Both the uncoordinated Pb2+ and excess PbI2 in perovskite film will create defects and perturb carrier collection, thus leading to the open-circuit voltage (VOC ) loss and inducing rapid performance degradation of perovskite solar cells (PSCs). Herein, an additive of 3-aminothiophene-2-carboxamide (3-AzTca) that contains amide and amino and features a large molecular size is introduced to improve the quality of perovskite film. The interplay of size effect and adequate bonding strength between 3-AzTca and uncoordinated Pb2+ regulates the mineralization of PbI2 and generates low-dimensional PbI2 phase, thereby boosting the crystallization of perovskite. The decreased defect states result in suppressed nonradiative recombination and reduced VOC loss. The power conversion efficiency (PCE) of modified PSC is improved to 22.79% with a high VOC of 1.22 V. Moreover, the decomposition of PbI2 and perovskite films is also retarded, yielding enhanced device stability. This study provides an effective method to minimize the concentration of uncoordinated Pb2+ and improve the PCE and stability of PSCs.

4.
BMC Cancer ; 23(1): 1105, 2023 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-37957631

RESUMEN

BACKGROUND: Ubiquitin-specific protease 32 (USP32) is a highly conserved gene that promotes cancer progression. However, its role in hepatocellular carcinoma (HCC) is not well understood. The aim of this project is to explore the clinical significance and functions of USP32 in HCC. METHODS: The expression of USP32 in HCC was evaluated using data from TCGA, GEO, TISCH, tissue microarray, and human HCC samples from our hospital. Survival analysis, PPI analysis and GSEA analysis were performed to evaluate USP32-related clinical significance, key molecules and enrichment pathways. Using the ssGSEA algorithm and TIMER, we investigated the relationships between USP32 and immune infiltrates in the TME. Univariate and multivariate Cox regression analyses were then used to identify key USP32-related immunomodulators and constructed a USP32-related immune prognostic model. Finally, CCK8, transwell and colony formation assays of HCC cells were performed and an HCC nude mouse model was established to verify the oncogenic role of USP32. RESULTS: USP32 is overexpressed in HCC and its expression is an independent predictive factor for outcomes of HCC patients. USP32 is associated with pathways related to cell behaviors and cancer signaling, and its expression is significantly correlated with the infiltration of immune cells in the TME. We also successfully constructed a USP32-related immune prognostic model using 5 genes. Wet experiments confirmed that knockdown of USP32 could repress the proliferation, colony formation and migration of HCC cells in vitro and inhibit tumor growth in vivo. CONCLUSION: USP32 is highly expressed in HCC and closely correlates with the TME of HCC. It is a potential target for improving the efficacy of chemotherapy and developing new strategies for targeted therapy and immunotherapy in HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Ubiquitina Tiolesterasa , Animales , Humanos , Ratones , Adyuvantes Inmunológicos , Algoritmos , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Procesos Neoplásicos , Ubiquitina Tiolesterasa/genética
5.
Pharmacol Res ; 196: 106915, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37689128

RESUMEN

Ferroptosis has been suggested to play a potential role in cancer therapy as an iron-dependent programmed cell death mechanism distinct from other forms. Hepatocellular carcinoma (HCC) remains a great threat, with high mortality and limited therapeutic options. The induction of ferroptosis has emerged as a novel and promising therapeutic strategy for HCC. In the present study, we identified protein inhibitor of activated STAT3 (PIAS3) as a driver of ferroptosis in HCC using TMT-based quantitative proteomics and ferroptosis-related functional assays. Mechanistically, thioredoxin-interacting protein (TXNIP) was confirmed to be PIAS3 in promoting ferroptotic cell death, based on RNA-seq analysis. Knockdown of TXNIP degrades ferroptotic susceptibility caused by PIAS3-overexpression, whereas transfection-forced reexpression of TXNIP restores sensitivity to ferroptosis in PIAS3-downregulated cells. PIAS3 interacts with SMAD2/3 to activate transforming growth factor (TGF)-ß signaling, leading to increased TXNIP expression. Our study revealed the critical role of PIAS3 in ferroptosis and a novel actionable axis-PIAS3/TGF-ß/TXNIP that could govern ferroptotic sensitivity, paving the path for using ferroptosis as an efficient approach in HCC therapies.

6.
Pharm Res ; 40(3): 689-699, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36539669

RESUMEN

BACKGROUND: Gastric cancer (GC) has always been a great threat to human health due to its aggressiveness and lethality. Anlotinib, a novel multi-target tyrosine kinase inhibitor (TKI), has been certified its anti-tumor effects on various tumors. Nonetheless, there are few studies on applying anlotinib as a treatment for GC. The underlying mechanism of acquired resistance during anlotinib administration remains unclear. METHODS: We investigated the toxicologic effects of anlotinib on GC cells through CCK8, colony-forming, and flow cytometry assays in vitro and xenograft models in vivo. Anlotinib-resistant GC cells, AGS-R and MGC803-R, were generated and characterized by cell proliferation and apoptosis assays. The signaling pathways involved in anlotinib resistance were probed using Cignal™ Finder 10-Pathway Reporter Array. Western blot and dual-luciferase reporter assays were performed to confirm the relationships. The TGF-ß inhibitor LY364947 was introduced to demonstrate the importance of TGF-ß signaling in anlotinib resistance via a series of functional assays. RESULTS: Anlotinib suppressed cell growth and induced apoptosis in vitro and inhibited tumorigenesis and metastasis in vivo, while its anti-tumor effects were impaired in anlotinib-resistant cells. The results of dual-luciferase reporter assays and western blot indicated TGF-ß signaling was activated in anlotinib-resistant GC cells. LY364947 combined with Anlotinib exerted a better antineoplastic effect than monotherapy and considerably reversed the anlotinib resistance in GC. CONCLUSIONS: Our findings suggested that TGF-ß signaling may take a significant part in anlotinib resistance in GC. The suppression of TGF-ß signaling may be a possible and promising approach for the GC oncotherapy when combined with anlotinib.


Asunto(s)
Antineoplásicos , Quinolinas , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Quinolinas/farmacología , Quinolinas/uso terapéutico , Proliferación Celular , Factor de Crecimiento Transformador beta
7.
Mol Cancer ; 21(1): 118, 2022 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-35619132

RESUMEN

BACKGROUND: PIK3CA mutation and PTEN suppression lead to tumorigenesis and drug resistance in colorectal cancer (CRC). There is no research on the role of circular RNAs (circRNAs) in regulating PIK3CA mutation and MEK inhibitor resistance in CRC. METHODS: The expression of circLHFPL2 in PIK3CA-mutant and wild-type cells and tissues was quantified by RNA-sequencing and qRT-PCR. CCK-8 assay and colony formation assay were used to evaluate cell viability. Annexin V/PI staining was implemented to assess cell apoptosis. Luciferase assay, biotin-coupled microRNA capture, and RIP assay were used to validate the interaction among potential targets. Western blotting and qRT-PCR assays were used to evaluate the expression of involved targets. Xenograft tumor in a nude mouse model was used to explore the role of circRNAs in vivo. RESULTS: RNA sequencing defined downregulated expression of circLHFPL2 in both PIK3CAH1047R (HCT116) and PIK3CAE545K (DLD1) cells. CircLHFPL2 was also downregulated in PIK3CA-mutant CRC primary cells and tissues, which was correlated with poor prognosis. CircLHFPL2 was mainly localized in the cytoplasm and its downregulation was attributed to the PI3K/AKT signaling pathway activated by phosphorylating Foxo3a. CircLHFPL2 inhibited PI3KCA-Mut CRC progression both in vitro and in vivo. Furthermore, our work indicated that circLHFPL2 acts as a ceRNA to sponge miR-556-5p and miR-1322 in CRC cells and in turn modulate the expression of PTEN. Importantly, circLHFPL2 was able to overcome PIK3CA-mediated MEK inhibitor resistance in CRC cells. CONCLUSIONS: Downregulation of circLHFPL2 sustains the activation of the PI3K/AKT signaling pathway via a positive feedback loop in PIK3CA-mutant CRC. In addition, downregulation of circLHFPL2 leads to MEK inhibitor resistance in CRC. Therefore, targeting circLHFPL2 could be an effective approach for the treatment of CRC patients harboring oncogenic PIK3CA mutations.


Asunto(s)
Neoplasias Colorrectales , MicroARNs , Animales , Carcinogénesis , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I/genética , Neoplasias Colorrectales/patología , Regulación hacia Abajo , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/uso terapéutico , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Circular/genética
8.
Pharm Res ; 39(5): 867-876, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35578065

RESUMEN

BACKGROUND: Gastric cancer (GC) remains a significant health problem and carries with it substantial morbidity and mortality. Chidamide is a novel and orally administered histone deacetylase (HDAC) inhibitor and has been demonstrated its anti-tumor efficacy on different kinds of hematological and solid tumors. However, the underlying mechanism of chidamide resistance is still poorly characterized. METHODS: We established chidamide resistant GC cell lines, AGS ChiR and MGC803 ChiR and investigated the toxicologic effects through cell survival, colony formation and flow cytometry assays in vitro, and a subcutaneous xenograft model in vivo. RNA-sequence was then performed to screen chidamide resistance-associated genes between AGS and AGS ChiR cells. The role of Lymphocyte cytosolic protein 1 (LCP1) in chidamide resistance was explored by gain- and loss-of-function analyses. RESULTS: We found that chidamide significantly inhibited cell proliferation and induced the apoptosis in a concentration-dependent manner in wild-type GC cell lines as compared to chidamide resistant cell lines. The transcriptomic profiling, quantitative RT-PCR, and western blot data revealed that LCP1 was upregulated in AGS ChiR cells compared with parental cells. Overexpression of LCP1 conferred and knockdown of LCP1 attenuated the chidamide resistance of GC cells. Epigenetic derepression of LCP1 by chidamide may be a possible reason for the contribution of LCP1 to chidamide resistance. CONCLUSIONS: These findings illustrated that LCP1 may play a chidamide resistance role in GC, suggesting that LCP1 could be a potential target for the therapy of GC combined with chidamide.


Asunto(s)
Resistencia a Antineoplásicos , Inhibidores de Histona Desacetilasas , Proteínas de Microfilamentos , Neoplasias Gástricas , Aminopiridinas/farmacología , Apoptosis , Benzamidas/farmacología , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos/genética , Perfilación de la Expresión Génica , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Proteínas de Microfilamentos/genética , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética
9.
J Cell Mol Med ; 25(2): 1024-1034, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33277798

RESUMEN

Tetraspanin CD63 has been widely implicated in tumour progression of human malignancies. However, its role in the tumorigenesis and metastasis of hepatocellular carcinoma (HCC) remains unclear yet. In the present study, we aimed to investigate the specific function and underlying mechanisms of CD63 in HCC progression. CD63 expression in HCC tissues was detected using immunohistochemistry and quantitative real-time PCR analyses; effects of CD63 on HCC cell proliferation and migration were investigated by CCK-8 assay, colony formation assay, transwell assay and a xenograft model of nude mice. RNA-sequencing, bioinformatics analysis, dual-luciferase reporter assay and Western blot analysis were performed to explore the underlying molecular mechanisms. Results of our experiments showed that CD63 expression was frequently reduced in HCC tissues compared with adjacent normal tissues, and decreased CD63 expression was significantly associated with larger tumour size, distant site metastasis and higher tumour stages of HCC. Overexpression of CD63 inhibited HCC cell proliferation and migration, whereas knockdown of CD63 promoted these phenotypes. IL-6, IL-27 and STAT3 activity was regulated by CD63, and blockade of STAT3 activation impaired the promotive effects of CD63 knockdown on HCC cell growth and migration. Our findings identified a novel CD63-IL-6/IL-27-STAT3 axis in the development of HCC and provided a potential target for the diagnosis and treatment of this disease.


Asunto(s)
Carcinogénesis/patología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Factor de Transcripción STAT3/metabolismo , Tetraspanina 30/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación/patología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Transducción de Señal
10.
Exp Cell Res ; 389(1): 111894, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32035950

RESUMEN

FTO, a demethylase for N6-methyladenosine (m6A) modification, has been implicated in multiple tumors. However, its roles in esophageal squamous cell carcinoma (ESCC) remain uncovered. This study aims to evaluate the clinical relevance and functional roles in this disease. Through immunohistochemistry, qRT-PCR and Western blot analyses, we found FTO expression in ESCC tissues was stronger than that in adjacent normal tissues, and the survival curves displayed high FTO expression had a trend toward poor prognosis. Functionally, silencing of FTO inhibited ESCC cell growth and migration in CCK8, EdU, colony formation and transwell assays and FTO overexpression showed the opposite results. Furthermore, FTO was also required for the tumorigenicity of ESCC cells in nude mice. The data from RNA-seq analysis revealed that MMP13 expression was significantly affected by FTO knockdown. qRT-PCR and Western blot assays confirmed that MMP13 was positively regulated by FTO in both mRNA and protein levels. Additionally, the functional link between FTO and MMP13 was explored by CCK8 and transwell chamber approaches. These findings suggest that FTO is up-regulated and plays oncogenic roles in ESCC. MMP13 may function as a downstream target of FTO.


Asunto(s)
Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/fisiología , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/patología , Metaloproteinasa 13 de la Matriz/genética , Animales , Línea Celular Tumoral , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas de Esófago/genética , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Desnudos , Invasividad Neoplásica , Regulación hacia Arriba/genética
11.
Exp Cell Res ; 374(1): 12-18, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30408464

RESUMEN

Human phosphatidylinositol-4-phosphate adaptor protein-2 (FAPP2) is well-known to function as a cytoplasmic lipid transfer protein during vesicle maturation. However, the expression and role of FAPP2 in tumor remain elusive. In this study, data from immunohistochemical assays displayed that FAPP2 was remarkably upregulated (57.8%) in 90 cases of colon cancer samples in contrast to their corresponding adjacent tissues. Disruption of FAPP2 by CRISPR/Cas9 technique in colon cancer cells led to an attenuated effect on cell growth analyzed by CCK8 and colony formation assays. Meanwhile, the tumorigenicity of FAPP2 downregulated cells also decreased in nude mice model. Accordantly, CCK8 assays also indicated that FAPP2 overexpression could promote colon cancer cell growth. In addition, dual luciferase reporter assays and western blot analyses revealed that Wnt/ß-catenin signaling was involved in the FAPP2-regulated tumor cell growth. These findings suggest that FAPP2 could act as an oncogene in the regulation of tumor growth and may provide a new therapeutic target for human colon cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Vía de Señalización Wnt , Animales , Línea Celular Tumoral , Proliferación Celular , Humanos , Masculino , Ratones Desnudos , Transcripción Genética , Ensayo de Tumor de Célula Madre , Regulación hacia Arriba/genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Nanobiotechnology ; 17(1): 116, 2019 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-31767014

RESUMEN

Tumor cells with heterogeneity and diversity can express different markers. At present, positive separation of circulating tumor cells (CTC) taking EpCAM as the marker was used in most cases which could be one-sided, while this study successfully prepared four antibody-modified magnetic immunoliposomes (MIL) by using the self-assembled liposome with antibody derivatives. This study aims to explore the separation efficiency and clinical detection feasibility of single or combined use of MIL with multi-tumor markers on different tumors. Captured CTC were stained with CK-FITC, CD45-PE and DAPI, and fluorescence microscope was used for the observation, analysis and calculation. The result indicated that the CTC number positive rate in blood samples of four different magnetic balls on the same patient could be up to 87.5% in 32 patients with 14 different kinds tumors. While the effect of directly mixed separation by four kinds of magnetic balls was not satisfying. It suggested that the MIL of multi-tumor markers could be a powerful tool for CTC separation in application of tumor screening and prognosis.


Asunto(s)
Molécula de Adhesión Celular Epitelial/análisis , Nanopartículas de Magnetita/análisis , Neoplasias/diagnóstico , Células Neoplásicas Circulantes/patología , Animales , Biomarcadores de Tumor/análisis , Línea Celular Tumoral , Separación Celular/métodos , Humanos , Neoplasias/sangre
13.
IUBMB Life ; 70(2): 120-128, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29283497

RESUMEN

The serine-threonine kinase receptor-associated protein (STRAP) has been implicated in multiple human cancers. However, its expression and function are currently unclear and controversial in different tissue types. In the present study, we report that aberrant downregulation of STRAP in hepatocellular carcinoma (HCC) facilitated tumor cell growth and metastasis in a phosphatase and tensin homologue (PTEN)-dependent manner. Immunohistochemical analysis and quantitative real-time polymerase chain reaction results indicated that STRAP was frequently downregulated in HCC samples. Functionally, knockdown of STRAP by RNA inference in HCC cells promoted proliferation and migration in vitro and tumorigenicity and lung metastasis in vivo. Through detecting the expression of some tumor-related genes using western blot analysis, we found the tumor suppressor PTEN was decreased upon STRAP silencing. Further analyses demonstrated that silenced STRAP led to PTEN protein degradation. Immunohistochemical analysis revealed that STRAP expression was closely associated with PTEN expression in 30 cases of HCC samples. These findings strongly suggest that STRAP plays an inhibitory role in HCC via regulating PTEN expression and could be a potential therapeutic target for this disease. © 2017 IUBMB Life, 70(2):120-128, 2018.


Asunto(s)
Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Proteínas de Neoplasias/metabolismo , Fosfohidrolasa PTEN/metabolismo , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Movimiento Celular , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Ratones Endogámicos BALB C , Proteínas de Neoplasias/genética , Fosfohidrolasa PTEN/genética , Proteínas de Unión al ARN , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Opt Express ; 25(14): 15662-15675, 2017 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-28789080

RESUMEN

It is challenging in realizing high-performance transparent organic light-emitting diodes (OLEDs) with symmetrical light emission to both sides. Herein, an efficient transparent OLED with highly balanced white emission to both sides is demonstrated by integrating quasi-periodic nanostructures into the organic emitter and the metal-dielectric composite top electrode, which can simultaneously suppressing waveguide and surface plasmonic loss. The power efficiency and external quantum efficiency are raised to 83.5 lm W-1 and 38.8%, respectively, along with a bi-directional luminance ratio of 1.26. The proposed scheme provides a facile route for extending application scope of transparent OLEDs for future transparent displays and lightings.

15.
Opt Express ; 24(6): A674-81, 2016 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-27136885

RESUMEN

Flexible organic light-emitting diodes (OLEDs) are emerging as a leading technology for rollable and foldable display applications. For the development of high-performance flexible OLEDs on plastic substrate, we report a transparent nanocomposite electrode with superior mechanical, electrical, and optical properties, which is realized by integrating the nanoimprinted quasi-random photonic structures into the ultrathin metal/dielectric stack to collectively optimize the electrical conduction and light outcoupling capabilities. The resulting flexible OLEDs with green emission yield the enhanced device efficiency, reaching the maximum external quantum efficiency of 43.7% and luminous efficiency of 154.9 cd/A, respectively.

16.
Mol Cell Biochem ; 406(1-2): 31-41, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25907137

RESUMEN

Long non-coding RNAs (LncRNAs) have been reported that play important roles in the progression and metastasis of some carcinomas. In the present study, we identified a new LncRNA, FRLnc1, from a microarray analysis in which those LncRNAs were regulated by FOXM1, an oncogene widely studied in most malignancies. Quantitative real-time PCR (qRT-PCR) results in gastric cancer cell lines indicated FRLnc1 expression is positively correlated with FOXM1 level, supporting the microarray data. Furthermore, the RNA level of FRLnc1 is upregulated in 49% (20/41) of cancer samples compared with neighboring non-cancerous stomach tissues. The in vitro functional analyses demonstrated that FRLnc1 knockdown by RNA interference suppressed cell migration in MGC803 and AGS cells, whereas FRLnc1 overexpression promoted cell migration in BGC823 and SGC7901 cells. Moreover, FRLnc1 could enhance the distant metastasis of SGC7901 cells by tail vein injection approach in mice. We also identified TGFß1 and Twist as the downstream effectors of FRLnc1 in the regulation of cell migration by qRT-PCR analysis. Taken together, our findings suggest that FRLnc1 is involved in gastric cancer cell migration and for the first time set up the link between FOXM1 and LncRNA in cancer.


Asunto(s)
Movimiento Celular , Factores de Transcripción Forkhead/genética , Neoplasias Pulmonares/metabolismo , ARN Largo no Codificante/genética , Neoplasias Gástricas/metabolismo , Animales , Línea Celular Tumoral , Proteína Forkhead Box M1 , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/secundario , Masculino , Ratones Desnudos , Trasplante de Neoplasias , Proteínas Nucleares/metabolismo , Neoplasias Gástricas/patología , Factor de Crecimiento Transformador beta1/metabolismo , Proteína 1 Relacionada con Twist/metabolismo
17.
Tumour Biol ; 35(5): 4779-84, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24470138

RESUMEN

The aim of this study was to find estrogen receptor (ER) binding sites of estradiol (E2)-treated and control groups and discuss the roles of ER activation in the tumorigenesis and progression of various human cancers. The ER ChIP-seq data GSE19013 was downloaded from Gene Expression Omnibus database, including E2-treated data GSM470419 and control data GSM470418. MACS software was utilized to identify ER binding sites in two groups. R's ChIPpeakAnno was used to detect ER-regulated target genes. Motif finding was employed to analyze ER concordant transcription factors (TFs) in MCF7 cell. The Gene Ontology (GO) was used to conduct functional enrichment analysis. We identified 9,134 ER binding sites in E2 stimulation group and 1,969 in control group. GO enrichment analysis of target genes showed that ER-regulated target genes mainly participated in mRNA catabolic process, protein complex disassembly, and protein localization to organelle-related biology process; while in E2 stimulation group, the function of ER-regulated target genes sharply changed. The effect of E2 in MCF7 cell suggested that activated ER probably reacted with several TFs and then co-regulated related genes expression. Furthermore, several TFs, such as PAX6, SMAD3, and ESR2, had multiply cellular regulation function. Our results showed that E2 stimulates breast cancer cell growth through ER. This may infer the function of ER in occurrence and development of breast cancer. Together, our study would pave ways for discussing ER concordant TFs and studying other ER-recruited TFs.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Receptores de Estrógenos/metabolismo , Sitios de Unión , Estradiol/farmacología , Femenino , Humanos , Células MCF-7 , Elementos de Respuesta , Proteína smad3/fisiología
18.
J Cell Commun Signal ; 18(1): e12017, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38545256

RESUMEN

Gastric cancer (GC) is one of the most common solid cancers with high incidence and mortality worldwide. Chronic gastritis and consequent inflammatory microenvironment is known as a major cause leading to gastric carcinogenesis. Here we report that PIK3CD that encodes p110δ, a catalytic subunit of the class IA PI3Ks, is overexpressed and tumorigenic in GC and associated with tumor inflammatory microenvironment. By investigating the data from TCGA database and our immunohistochemical staining and quantitative real-time PCR results from clinical samples, we found PIK3CD exhibits higher expression level in GC tissues compared with adjacent non-tumorous stomach tissues. Genetic silencing of PIK3CD in GC cells retards proliferation and migration in vitro and tumorigenicity and metastasis in vivo. In contrast, enhanced expression of PIK3CD promotes these phenotypes in vitro. Furthermore, pharmacological inhibition of PIK3CD could reduce GC cell viability and colony formation capacities. More importantly, we reveal a relevant mechanism that PIK3CD, but not PIK3CA and PIK3CB, is transcriptionally regulated by the pro-inflammatory IL2/JAK3/STAT5 axis and tumor-infiltrating immune cells such as lymphocytes. These observations may setup a new crosstalk between tumor inflammatory microenvironment, IL2/JAK3/STAT5 signaling and PI3K/AKT signaling. Targeting PIK3CD may be a promising therapy strategy for GC.

19.
Cancer Res ; 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38885324

RESUMEN

Cellular oxidative stress plays a key role in the development and progression of hepatocellular carcinoma (HCC). A better understanding of the processes that regulate reactive oxygen species (ROS) homeostasis could uncover improved strategies for treating HCC. Here, we identified WNK1 as an antioxidative factor and therapeutic target in HCC. In human HCC, WNK1 expression was increased and correlated with poor patient prognosis. WNK1 knockdown significantly inhibited cell proliferation and xenograft tumor growth. Mechanistically, WNK1 competed with NRF2 for binding to the partial Kelch domain of KEAP1, reducing NRF2 ubiquitination and promoting NRF2 accumulation and nuclear translocation to increase antioxidant response. WNK1 silencing increased H2O2-induced apoptosis and inhibited cell growth by elevating reactive oxygen species (ROS) levels, which could be rescued by treatment with the antioxidant N-acetylcysteine (NAC) and NRF2 activator tert-butylhydroquinone (tBHQ). Liver-specific WNK1 knockout mouse models of HCC substantiated that WNK1 promoted HCC development by regulating ROS levels. WNK463, an inhibitor of the WNK kinase family, suppressed HCC progression and altered the redox status. These findings suggest that WNK1 plays a critical role in HCC development and progression and that the WNK1-oxidative stress axis may be a promising therapeutic target for HCC.

20.
Adv Mater ; 35(6): e2208604, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36440601

RESUMEN

The charge carriers of single-junction solar cells can be fluently extracted and then collected by electrodes, leading to weak charge carrier accumulation and low energy loss (Eloss ). However, in tandem solar cells (TSCs), it is a considerable challenge to obtain a balance between the densities of the holes and electrons extracted from the two respective subcells to facilitate an efficient recombination in the interconnecting layer (ICL). Herein, a charge-carrier-dynamic management strategy for inorganic perovskite/organic TSCs is proposed, centered on the simultaneous regulation of the defect states of CsPbI1.9 Br1.1 perovskite in the front subcell and hole transport ability from the perovskite to ICL. The target hole density on the perovskite surface and the hole loss before reaching the ICL are significantly improved. As a result, the hole/electron density offset in the ICL can be effectively narrowed, leading to a balanced charge carrier recombination, which reduces the Eloss in TSCs. The resulting inorganic perovskite/organic 0.062-cm2 TSC exhibits a remarkable power conversion efficiency (PCE) of 23.17% with an ultrahigh open-circuit voltage (Voc ) of 2.15 V, and the PCE of the 1.004-cm2 device (21.69%) exhibited a weak size-dependence. This charge-carrier-dynamic management strategy can also effectively enhance the operational and ultraviolet-light stabilities of the TSCs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA