Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Gene Med ; 26(1): e3609, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37849429

RESUMEN

BACKGROUND: Liver cancer, a common malignancy within the digestive system, presents with a particularly grim prognosis. Within the immune microenvironment, the role of natural killer (NK) cells in liver cancer remains unclear. METHODS: We sourced data on clinical parameters and gene expressions for liver cancer patients from The Cancer Genome Atlas Program database and carried out all analyses using R software and its relevant codes. RESULTS: In our research, we delved into the genes intertwined with NK cells in hepatocellular carcinoma (HCC). Leveraging the QUANTISEQ and MCPCOUNTER algorithms to quantify NK cells, we spotlighted genes vital to the recruitment of NK cells. Among these genes, GDE1, WDFY3, DNAJB14, PKD2, DGAT2, SGMS2 and MKNK2 showed a strong correlation with patient outcomes. We also mapped out the single-cell expression trajectories of these genes within the HCC milieu. From our findings, SGMS2 emerged as a key gene warranting further scrutiny. Our in-depth analysis of SGMS2 shed light on its influence over specific biological pathways, its contribution to the immune landscape and its role in genomic instability within HCC. Drawing from this, we formulated a predictive model rooted in SGMS2-associated genes. This model showcased remarkable precision across both training and validation cohorts. CONCLUSIONS: Overall, our investigation underscored the profound implications of SGMS2, a gene pivotal to NK cell infiltration, in the landscape of HCC, thereby positioning it as a potential linchpin in oncological strategies.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Células Asesinas Naturales/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Microambiente Tumoral/genética
2.
J Cell Mol Med ; 22(6): 3246-3258, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29602239

RESUMEN

Toll-like receptor 4 (TLR4) which acts as a receptor for lipopolysaccharide (LPS) has been reported to be involved in carcinogenesis. However, the regulatory mechanism of it has not been elucidated. Herein, we demonstrate that TLR4 promotes the malignant growth of liver cancer stem cells. Mechanistically, TLR4 promotes the expression of histone-lysine N-methyltransferase (SUV39 h2) and increases the formation of trimethyl histone H3 lysine 9-heterochromatin protein 1-telomere repeat binding factor 2 (H3K9me3-HP1-TRF2) complex at the telomeric locus under mediation by long non coding RNA urothelial cancer-associated 1 (CUDR). At the telomeric locus, this complex promotes binding of POT1, pPOT1, Exo1, pExo1, SNM1B and pSNM1B but prevents binding of CST/AAF to telomere, thus controlling telomere and maintaining telomere length. Furthermore, TLR4 enhances interaction between HP1α and DNA methyltransferase (DNMT3b), which limits RNA polymerase II deposition on the telomeric repeat-containing RNA (TERRA) promoter region and its elongation, thus inhibiting transcription of TERRA. Ultimately, TLR4 enhances the telomerase activity by reducing the interplay between telomerase reverse transcriptase catalytic subunit (TERT) and TERRA. More importantly, our results reveal that tri-complexes of HP1 isoforms (α, ß and γ) are required for the oncogenic action of TLR4. This study elucidates a novel protection mechanism of TLR4 in liver cancer stem cells and suggests that TLR4 can be used as a novel therapeutic target for liver cancer.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Neoplasias Hepáticas/genética , Telomerasa/genética , Receptor Toll-Like 4/genética , Línea Celular Tumoral , Homólogo de la Proteína Chromobox 5 , ADN (Citosina-5-)-Metiltransferasas/genética , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Isoformas de Proteínas/genética , ARN Largo no Codificante/genética , Telómero/genética , Homeostasis del Telómero/genética , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Factores de Transcripción/genética , ADN Metiltransferasa 3B
3.
Clin Lab ; 61(5-6): 537-47, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26118187

RESUMEN

BACKGROUND: Wnt5a is involved in the activation of human hepatic stellate cells (HSC) and related with the occurrence of liver fibrosis. As the function and mechanism that Wnt5a mediates HSC activation remains unclear, we sought to investigate them. METHODS: Wnt5a levels were determined in the HSC cell line LX-2 after lipopolysaccharide (LPS) and TNF-α stimulation. HSC cells showing stable and efficient overexpression or featuring knockdown of Wnt5a were constructed by a lentivirus system. Regulation of cytokine and collagen expressions were confirmed by quantitative PCR or ELISA in stable LX-2 cell lines showing Wnt5a overexpression or knockdown. Proliferation was determined by 5-ethynyl-2'-deoxyuridine labeling. Relevant signaling pathways were identified using specific protein antibodies. RESULTS: LPS and TNF-α induced Wnt5a expression in LX-2 cells. Compared with control cells, an increase in IL-10, IL-6, COL1, and COL3 secretion in a stable LX-2 cell line showing Wnt5a overexpression was observed. Knockdown of Wnt5a obviously reduced the production of IL-1ß, IL-6, COL1, and COL3. Wnt5a overexpression promoted LX-2 proliferation, while Wnt5a knockdown dramatically inhibited cell proliferation. Compared with the effects of Wnt5a knockdown cells, Wnt5a-overexpressing cells triggered the phosphorylation of c-Jun N-terminal kinase (JNK) and ß-catenin, which leads to ß-catenin degradation and inactivates the canonical Wnt/ß-catenin pathway. CONCLUSIONS: Wnt5a regulates inflammatory cytokine and collagen production and cell proliferation, which is independent of the canonical Wnt signaling pathway. The results reveal a new signaling mechanism in HSC activation and could provide a new strategy for hepatic fibrosis treatment.


Asunto(s)
Proliferación Celular , Colágeno/metabolismo , Citocinas/metabolismo , Células Estrelladas Hepáticas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Wnt/metabolismo , Línea Celular , Humanos , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Transducción de Señal , Proteína Wnt-5a , beta Catenina/metabolismo
4.
Biochem Biophys Res Commun ; 453(3): 533-8, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25285629

RESUMEN

Pancreatic cancer remains fatal to the fast majority of affected patients. Activation of phosphoinositide-3 kinase (PI3K)-AKT-mammalian target of rapamycin (mTOR) pathway plays an important role in pancreatic cancer progression and chemo-resistance. In the present study, we examined the activity of GDC-0980, a novel class I PI3K/mTOR kinase inhibitor, against pancreatic cancer cells in vitro. GDC-0980 inhibited AKT-mTOR activation and pancreatic cancer cell (PANC-1 and Capan-1 lines) survival. In both cancer cell lines, GDC-0980 simultaneously activated apoptosis and autophagy, the latter was detected by p62 degradation, Beclin-1 upregulation and light chain 3B (LC3B) conversion from a cytosolic (LC3B-I) to a membrane-bound (LC3B-II) form. Autophagy inhibitors including 3-methyladenine, hydroxychloroquine, NH4Cl and bafilomycin A1 enhanced apoptosis and cytotoxicity by GDC-0980, such an effect was reversed by caspase inhibitors (z-VAD-FMK and z-ITED-FMK). Furthermore, knockdown of LC3B or Beclin-1 through siRNA increased GDC-0980-induced anti-pancreatic cancer cell activity. Thus, inhibition of autophagy sensitizes GDC-0980-induced anti-pancreatic cancer activity, suggesting a novel therapeutic strategy for GDC-0980 sensitization.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Neoplasias Pancreáticas/patología , Pirimidinas/farmacología , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Línea Celular Tumoral , Citometría de Flujo , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Neoplasias Pancreáticas/metabolismo
5.
Clin Lab ; 60(1): 29-35, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24600972

RESUMEN

BACKGROUND: Wnt-induced secreted protein-1 (WISP-1/CCN4) is a member of the CCN family growth factors, and its role in liver fibrosis is largely unknown. METHODS: For in vitro, hepatic stellate cells (HSCs) were isolated from Sprague-Dawley rats. Expression of WISP-1 during progressive activation of cultured rat HSCs was analyzed by qRT-PCR. The effects of TNF-a and TGF-beta1 on WISP-1 expression were analyzed in stellate cell lines HSC-T6 and LX-2. The effect of exogenous WISP-1 protein on LX-2 proliferation was examined. For in vivo, expressions of WISP-1 in fibrotic liver of a carbon tetrachloride (CCl4)-induced fibrosis rat model were analyzed by qRT-PCR and immunohistochemistry. RESULTS: In vitro, WISP-1 was increasingly expressed during progressive activation of cultured rat HSCs. WISP-1 was significantly induced in HSC-T6 cells by TNF-a and in LX-2 cells by TGF-beta1. Recombinant WISP-1 protein promoted LX-2 proliferation in a dose-dependent manner. In vivo, both mRNA and protein expression levels of WISP-1 were increased significantly in experimental hepatic fibrosis model. CONCLUSIONS: Our results showed the upregulation of WISP-1 in both in vitro and in vivo liver fibrosis models, and WISP-1 stimulated the proliferation of HSCs in vitro. These results may be helpful to elucidate the exact role of WISP-1 in liver fibrogenesis.


Asunto(s)
Proteínas CCN de Señalización Intercelular/metabolismo , Tetracloruro de Carbono/toxicidad , Cirrosis Hepática/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Secuencia de Bases , Proteínas CCN de Señalización Intercelular/genética , Células Cultivadas , Cartilla de ADN , Inmunohistoquímica , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/genética , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
6.
Clin Invest Med ; 37(1): E38-46, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24502810

RESUMEN

PURPOSE: Esophageal varices are a dangerous complication of liver cirrhosis. The development of cost effective, noninvasive means for prediction of large esophageal varices could reduce the use of upper gastrointestinal endoscopy in variceal screening and also provide an alternative way to confirm the results of conventional endoscopic diagnosis. Previously proposed predictive models are neither sensitive nor specific. METHODS: A retrospective study based on a group of 104 liver cirrhosis patients was performed. Multiple statistical approaches were used to evaluate the association of large esophageal varices with 20 individual and six compound clinical laboratory variables. A new predictive model was developed. RESULTS: Univariate analysis suggested that eight out of 26 variables were significantly associated with large esophageal varices. Further stepwise logistic regression eventually identified three variables (hemoglobin level, portal vein diameter and the ratio of platelet count/spleen diameter) that contributed significantly to the final regression model. Receiver operating characteristic (ROC) curve analysis showed that this new regression model achieved 77.8% and 72% of diagnostic sensitivity and specificity, respectively, for the prediction of large esophageal varices. In our study group, its diagnostic accuracy (AUROC=0.814) was found to be significantly higher than six predictive models previously published. CONCLUSIONS: No single variable offers self-sufficient predictive function for large esophageal varices. A comprehensive model using multiple variables significantly improves the predictive accuracy in screening the most at risk patients with potential variceal hemorrhage.


Asunto(s)
Várices Esofágicas y Gástricas/diagnóstico , Várices Esofágicas y Gástricas/etiología , Cirrosis Hepática/complicaciones , Anciano , Várices Esofágicas y Gástricas/sangre , Femenino , Hemoglobinas/metabolismo , Humanos , Cirrosis Hepática/sangre , Cirrosis Hepática/patología , Modelos Logísticos , Masculino , Persona de Mediana Edad , Modelos Biológicos , Análisis Multivariante , Recuento de Plaquetas , Vena Porta/patología , Valor Predictivo de las Pruebas , Curva ROC , Estudios Retrospectivos , Factores de Riesgo , Bazo/patología
7.
J Chin Med Assoc ; 86(9): 806-817, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37501238

RESUMEN

BACKGROUND: Aberrant glycosylation performed by glycosyltransferases is a leading cause of gastric cancer (GC). Protein O-fucosyltransferase 1 (POFUT1) expression is increased in GC specimens and cells. In this study, the biological effects and mechanisms of POFUT1 underlying the development of GC were investigated. METHODS: POFUT1 downregulated and upregulated GC cells were established. The effects of POFUT1 on cell proliferation, metastasis and apoptosis were examined using cell counting kit-8 (CCK8) assay, transwell assay, and flow cytometry. Subcutaneous xenograft tumor models were established followed by immunohistochemistry staining of resected tumors. Facilitating modulators and transcription factors were detected by western blot, immunofluorescence, luciferase reporter assay, and co-immunoprecipitation. RESULTS: POFUT1 played a pro-oncogenic role both in vivo and in vitro, which promoted proliferation and metastasis, as well as inhibited apoptosis in GC cells. POFUT1 promoted Cyclin D3 expression and inhibited the expression of apoptotic proteins, such as Bcl-2-associated X protein (Bax) and cleaved caspase 3, facilitating tumor growth. Moreover, POFUT1 accelerated matrix metalloproteases expression and attenuated E-cadherin expression, contributing to GC metastasis. In addition, POFUT1 expression promoted the expression and nuclear translocation of Notch1 intracellular domain (NICD1) and ß-catenin and inhibited ß-catenin phosphorylation degradation, accompanied by the activation of recombination signal binding protein-Jκ (RBP-J) and T-cell factor (TCF) transcription factors, respectively. It is notable that parafibromin integrated NICD1 and ß-catenin, enabling the concerted activation of Wnt and Notch signaling targeted proteins. CONCLUSION: These observations indicated that POFUT1 promoted GC development through activation of Notch and Wnt signaling pathways, which depended on the parafibromin-NICD1-ß-catenin complex. This work provides new evidence for the further diagnosis and treatment of GC.


Asunto(s)
Neoplasias Gástricas , Vía de Señalización Wnt , Humanos , Neoplasias Gástricas/patología , beta Catenina/metabolismo , Proliferación Celular , Línea Celular Tumoral , Movimiento Celular
9.
Molecules ; 17(9): 10267-75, 2012 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-22926307

RESUMEN

Oxidative stress is involved in the development and progression of disease. Because sodium aescinate has been reported to have immunity enhancing and antioxidative effects, we investigated its activity by employing a hepatocellular carcinoma (HCC) mouse model. Sixty BALB/c mice were randomly divided into four groups, including a 1.4 mg/kg treated group (n = 15), a 2.8 mg/kg treated group (n = 15), an untreated hepatocellular carcinoma control group (n = 15) and a normal control group (n = 15). After H22 cells were cultured for one week, we collected 2 × 106 cells and injected them subcutaneously as 0.2 mL cell suspensions in sterile saline into the right shoulder region of every mouse. The animals were monitored for changes in activity, physical condition and body weight during the experiment. The next day after injection of H22 cells, animals in these test groups received one intraperitoneal injection of drug or physiological saline for 13 days. Results showed that in the sodium aescinate injection liquid (SAIL)-treated HCC mice, serum interleukin-1 beta (IL-1ß), interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ), Gamma-glutamyltransferase (γ-GT), alanine transaminase (ALT), aspartate transaminase (AST) and alkaline phosphatase (ALP) levels were significantly decreased compared with normal control mice. In addition, treatment with sodium aescinate injection liquid significantly decreased blood and liver malondialdehyde (MDA) levels, increased glutathione (GSH) levels, and antioxidant enzyme [superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-Px)] activities in a dose-dependent manner. We conclude that sodium aescinate injection liquid can decrease oxidative injury and enhance immunity functions in HCC mice.


Asunto(s)
Antioxidantes/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Factores Inmunológicos/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Compuestos de Sodio/farmacología , Alanina Transaminasa/metabolismo , Animales , Aspartato Aminotransferasas/metabolismo , Humanos , Interferón gamma/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/análisis , Ratones , Ratones Endogámicos BALB C , Distribución Aleatoria , Compuestos de Sodio/administración & dosificación , Superóxido Dismutasa/metabolismo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/análisis , Ensayos Antitumor por Modelo de Xenoinjerto , gamma-Glutamiltransferasa/metabolismo
10.
J Breath Res ; 17(1)2022 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-36347037

RESUMEN

The spread of coronavirus disease 2019 (COVID-19) results in an increasing incidence and mortality. The typical diagnosis technique for severe acute respiratory syndrome coronavirus 2 infection is reverse transcription polymerase chain reaction, which is relatively expensive, time-consuming, professional, and suffered from false-negative results. A reliable, non-invasive diagnosis method is in urgent need for the rapid screening of COVID-19 patients and controlling the epidemic. Here we constructed an intelligent system based on the volatile organic compound (VOC) biomarkers in human breath combined with machine learning models. The VOC profiles of 122 breath samples (65 of COVID-19 infections and 57 of controls) were identified with a portable gas chromatograph-mass spectrometer. Among them, eight VOCs exhibited significant differences (p< 0.001) between the COVID-19 and the control groups. The cross-validation algorithm optimized support vector machine (SVM) model was employed for the prediction of COVID-19 infection. The proposed SVM model performed a powerful capability in discriminating COVID-19 patients from healthy controls, with an accuracy of 97.3%, a sensitivity of 100%, a specificity of 94.1%, and a precision of 95.2%, and anF1 score of 97.6%. The SVM model was also compared with other common machine models, including artificial neural network,k-nearest neighbor, and logistic regression, and demonstrated obvious superiority in the prediction of COVID-19 infection. Furthermore, user-friendly software was developed based on the optimized SVM model. The developed intelligent platform based on breath analysis provides a new strategy for the point-of-care screening of COVID and shows great potential in clinical application.


Asunto(s)
COVID-19 , Compuestos Orgánicos Volátiles , Humanos , Pruebas Respiratorias/métodos , Compuestos Orgánicos Volátiles/análisis , Máquina de Vectores de Soporte , Biomarcadores/análisis
11.
J Oncol ; 2021: 9919494, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34194499

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) is a common malignant cancer worldwide. CXCL5 has a role in inhibiting cell viability and metastasis in many tumors. In the present study, we investigated the role of CXCL5 in HCC and explored the underlying mechanism. Material and Methods. RT-qPCR and western blot were performed to evaluate the mRNA and protein levels of CXCL5. CCK-8 and transwell assay were applied to measure the proliferative and invasive abilities. Meanwhile, the Kaplan-Meier method was used to assess the survival of HCC patients. RESULTS: CXCL5 was upregulated in HCC tissues, which predicted a shorter overall survival in HCC. CXCL5 was a target gene of miR-577, and its expression was mediated by miR-577 in HCC. Knockdown of CXCL5 suppressed HuH-7 cell proliferation, invasion, and EMT and inhibited the NF-κB signaling pathway in cells. Moreover, knockdown of CXCL5 inhibited the xenograft growth of HuH-7 cells. CONCLUSION: Overexpression of CXCL5 predicts poor prognosis in HCC patients. Knockdown of CXCL5 inhibits cell proliferation and invasion through the NF-κB signaling pathway in HCC. The newly identified role of the CXCL5/miR-577/NF-κB axis provides novel insights into the targeted therapy of HCC.

12.
Cell Death Discov ; 7(1): 274, 2021 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-34608122

RESUMEN

Tryptophan metabolism is an essential regulator of tumor immune evasion. However, the effect of tryptophan metabolism on cancer cells remains largely unknown. Here, we find that tumor cells have distinct responses to tryptophan deficiency in terms of cell growth, no matter hepatocellular carcinoma (HCC) cells, lung cancer cells, or breast cancer cells. Further study shows that ERRFI1 is upregulated in sensitive HCC cells, but not in resistant HCC cells, in response to tryptophan deficiency, and ERRFI1 expression level positively correlates with HCC patient overall survival. ERRFI1 knockdown recovers tryptophan deficiency-suppressed cell growth of sensitive HCC cells. In contrast, ERRFI1 overexpression sensitizes resistant HCC cells to tryptophan deficiency. Moreover, ERRFI1 induces apoptosis by binding PDCD2 in HCC cells, PDCD2 knockdown decreases the ERRFI1-induced apoptosis in HCC cells. Thus, we conclude that ERRFI1-induced apoptosis increases the sensitivity of HCC cells to tryptophan deficiency and ERRFI1 interacts with PDCD2 to induce apoptosis in HCC cells.

13.
Theranostics ; 11(12): 6006-6018, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33897895

RESUMEN

Objectives: Sorafenib is the only FDA-approved first-line target drug for HCC patients. However, sorafenib merely confers 3-5 months of survival benefit with less than 30% of HCC patients sensitive to sorafenib therapy. Thus, it's necessary to develop a sensitizer for hepatocellular carcinoma (HCC) to sorafenib. Methods: The principal component analysis, gene ontology, and KEGG analysis are utilized following RNA-sequencing. The mass spectrometry analysis following immunoprecipitation is performed to discover the phosphatase targets. Most importantly, both the cell line-derived xenograft (CDX) and the patient-derived xenograft (PDX) mouse model are used to determine the effect of 3-HAA on sorafenib-resistant HCC in vivo. Results: In nude mice carrying HCC xenograft, tumor growth is inhibited by sorafenib or 3-HAA alone. When used in combination, the treatment particularly prevents the xenograft from growing. Combined treatment also suppresses the growth of sorafenib-resistant (≥30mg/kg) PDXs. In a set of mechanistic experiments, we find enhanced AKT activation and decreased apoptotic cells in de novo and acquired sorafenib-resistant HCC cells and tissues. 3-HAA decreases AKT phosphorylation and increases the apoptosis of HCC in both cultured cells and mouse xenografts by upregulation of phosphatases PPP1R15A/DUSP6. PPP1R15A/PPP1α directly reduces Akt phosphorylation while DUSP6 decreases Akt activity through inhibiting PDK1. The AKT activator abolishes 3-HAA inhibition of HCC growth in vitro and in mice. Conclusion: This study demonstrates that 3-HAA sensitizes HCC cells to sorafenib by upregulation of phosphatases, suggesting it as a promising molecule for HCC therapy.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Quinurenina/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Monoéster Fosfórico Hidrolasas/metabolismo , Sorafenib/farmacología , Regulación hacia Arriba/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Onco Targets Ther ; 13: 9159-9167, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32982304

RESUMEN

PURPOSE: Hepatocellular carcinoma (HCC) is the most common primary liver tumor and the third greatest cause of cancer-related death worldwide. Programmed cell death 4 (PDCD4) was reported as a potential tumor-suppressor in hepatocarcinogenesis. However, relatively little is known about mechanisms that regulate PDCD4 expression in HCC. The aim of the present study is to investigate the expression of PDCD4 and miR-182 in human HCC cell lines and clinical HCC specimens and determine whether PDCD4 is a direct target of miR-182 in HCC cell lines. MATERIALS: The expression of miR-182 and PDCD4 in human HCC cell lines and HCC tissues were examined using qRT-PCR and Western blot method. Transwell and wound healing assays were carried out to explore the influence of miR-182 on hepatoma cells migration. A luciferase reporter assay was conducted to confirm target association. RESULTS: In our research, we found that PDCD4 was downregulated, whereas miR-182 was upregulated in liver cancer cell lines and HCC tissues. Transwell and wound healing assays illustrated that miR-182 contributed to migration activities of liver cancer cell lines. Loss or increase of miR-182 can lead to a negative expression of PDCD4 protein level. The luciferase reporter assay showed that PDCD4 is a direct target of miR-182. CONCLUSION: All these findings suggest that miR-182 may act as an oncogenic role in liver cancer cells by directly and negatively regulating expression of PDCD4.

15.
Anal Chim Acta ; 1095: 179-184, 2020 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-31864620

RESUMEN

Abnormal expression of specific microRNAs (miRNAs) is associated with the occurrence, development and prognosis of many diseases. In this study, a miRNA detection method based on exponential amplification reaction (EXPAR) and triplex DNA mediated aggregation of gold nanoparticles (AuNPs) was established. Specifically, one class of AuNPs is conjugated with an EXPAR probe, on which there is a complementary sequence of the target miRNA. The EXPAR reaction is triggered and duplex DNA is formed on the surface of AuNPs when the target miRNA exists. Then, single DNA probe on another class of AuNPs interacts with the duplex DNA to form triplex DNA, leading to the aggregation of the two classes of AuNPs, which could be quantified by UV-vis. The proposed method is highly selective and can afford a detection limit of 0.23 fM. Notably, all the ingredients needed for the analysis can pre-add to a tube and only 30 min is needed for the whole detection process. The method is simple, fast and with considerable selectivity and accuracy, so a great potential for this method is expected to meet the need of point-of-care testing of miRNA.


Asunto(s)
Colorimetría/métodos , ADN/química , Nanopartículas del Metal/química , MicroARNs/análisis , Línea Celular Tumoral , ADN/genética , Sondas de ADN/química , Sondas de ADN/genética , Oro/química , Humanos , Límite de Detección , MicroARNs/genética , Mutación , Técnicas de Amplificación de Ácido Nucleico , Conformación de Ácido Nucleico , Hibridación de Ácido Nucleico
16.
Acta Biomater ; 117: 361-373, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33007481

RESUMEN

Peptide modified nanoparticles have emerged as powerful tools for enhanced cancer diagnosis and novel treatment strategies. Here, human programmed death-ligand 1 (PD-L1) peptides were used for the first time for the modification of gold nanoprisms (GNPs) to enhance targeting efficiency. A multifunctional nanoprobe was developed that the GNPs@PEG/Ce6-PD-L1 peptide (GNPs@PEG/Ce6-P) was used for imaging-guided photothermal/photodynamic therapy by using the targeting effect of PD-L1. Both confocal imaging and flow cytometry experiments demonstrated a remarkable affinity of the as-prepared nanoprobes GNPs@PEG/Ce6-P to lung cancer cells (HCC827), which have a high PD-L1 expression. Subsequent in vitro and in vivo experiments further demonstrated that the nanoprobes GNPs@PEG/Ce6-P not only allowed for real-time visualization via fluorescence (FL) imaging and photoacoustic (PA) imaging, but also served as phototherapy agents for synergistic photothermal therapy (PTT) and photodynamic therapy (PDT). Furthermore, treatments on human lung cancer cells-derived tumors demonstrated that the nanoprobes GNPs@PEG/Ce6-P could significantly suppress tumor growth through PTT and PDT from GNPs and Ce6, respectively. In conclusion, the as-prepared new nanoprobes show promising potential for nanomedicine with remarkable targeting ability for dual-mode imaging and enhanced PDT and PTT effects on lung cancer.


Asunto(s)
Neoplasias Pulmonares , Fotoquimioterapia , Porfirinas , Antígeno B7-H1 , Línea Celular Tumoral , Clorofilidas , Oro , Humanos , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/tratamiento farmacológico , Fármacos Fotosensibilizantes/uso terapéutico , Porfirinas/farmacología , Medicina de Precisión
17.
World J Gastroenterol ; 14(42): 6546-50, 2008 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-19030210

RESUMEN

AIM: To investigate the prognostic value of the model for end-stage liver disease (MELD) and three new MELD-based models combination with serum sodium in decompensated cirrhosis patients-the MELD with the incorporation of serum sodium (MELD-Na), the integrated MELD (iMELD), and the MELD to sodium (MESO) index. METHODS: A total of 166 patients with decompensated cirrhosis were enrolled into the study. MELD, MELD-Na, iMELD and MESO scores were calculated for each patient following the original formula on the first day of admission. All patients were followed up at least 1 year. The predictive prognosis related with the four models was determined by the area under the receiver operating characteristic curve (AUC) of the four parameters. Kaplan-Meier survival curves were made using the cut-offs identified by means of receiver operating characteristic (ROC). RESULTS: Out of 166 patients, 38 patients with significantly higher MELD-Na (28.84 +/- 2.43 vs 14.72 +/- 0.60), iMELD (49.04 +/- 1.72 vs 35.52 +/- 0.67), MESO scores (1.59 +/- 0.82 vs 0.99 +/- 0.42) compared to the survivors died within 3 mo (P < 0.001). Of 166 patients, 75 with markedly higher MELD-Na (23.01 +/- 1.51 vs 13.78 +/- 0.69), iMELD (44.06 +/- 1.19 vs 34.12 +/- 0.69), MESO scores (1.37 +/- 0.70 vs 0.93 +/- 0.40) than the survivors died within 1 year (P < 0.001). At 3 mo of enrollment, the iMELD had the highest AUC (0.841), and was followed by the MELD-Na (0.766), MESO (0.723), all larger than MELD (0.773); At 1 year, the iMELD still had the highest AUC (0.783), the difference between the iMELD and MELD was statistically significant (P < 0.05). Survival curves showed that the three new models were all clearly discriminated the patients who survived or died in short-term as well as intermediate-term (P < 0.001). CONCLUSION: Three new models, changed with serum sodium (MELD-Na, iMELD, MESO) can exactly predict the prognosis of patients with decompensated cirrhosis for short and intermediate period, and may enhance the prognostic accuracy of MELD. The iMELD is better prognostic model for outcome prediction in patients with decompensated cirrhosis.


Asunto(s)
Indicadores de Salud , Cirrosis Hepática/diagnóstico , Fallo Hepático/etiología , Modelos Biológicos , Sodio/sangre , Progresión de la Enfermedad , Humanos , Estimación de Kaplan-Meier , Cirrosis Hepática/sangre , Cirrosis Hepática/complicaciones , Cirrosis Hepática/mortalidad , Fallo Hepático/sangre , Fallo Hepático/mortalidad , Valor Predictivo de las Pruebas , Pronóstico , Curva ROC , Estudios Retrospectivos , Medición de Riesgo , Índice de Severidad de la Enfermedad , Factores de Tiempo
18.
Cell Cycle ; 17(16): 1949-1966, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30300116

RESUMEN

Long non-coding RNAs (lncRNAs) have been shown to play a significant role in the progression of many cancers, including pancreatic cancer (PC). However, the biological function and regulatory mechanisms of lncRNAs in PC remains largely unclear. The aim of this study was to identify and evaluate the potential functions of lncRNAs in PC and reveal the underlying mechanisms of their effects. Screening of published microarray data (GEO accession Nos. GSE16515 and GSE32688), revealed lncRNA AFAP1-AS1 to be one of the most upregulated lncRNAs in PC tissues. High expression of AFAP1-AS1 was correlated with advanced stages, tumor size and lymph node metastasis, as well as with poorer overall survival in patients with PC. Functionally, knockdown of AFAP1-AS1 by transfection with siRNA inhibited the proliferative and invasive capacities of PaCa-2 and SW1990 PC cells, promoted apoptosis of PC cells in vitro, and impaired in-vivo tumorigenicity. In particular, it was hypothesized that AFAP1-AS1 may act as a competitive endogenous RNA (ceRNA), effectively becoming a sink for miR-133a whose expression was found to be downregulated in PC tissues and cell lines, and which was negatively correlated with the expression of AFAP1-AS1. We also found that the IGF1R oncogene which is an important regulator of MEK/ERK signaling pathway, was positively regulated by AFAP1-AS1 through ameliorating miR-133a-mediated IGF1R repression in PC tissues. Moreover, we demonstrated that knockdown of IGF1R by transfection with si-IGF1R suppressed cell proliferation, invasion and migration of PaCa-2 and SW1990 PC cells, suggesting that IGF1R may function as an oncogene in PC cells. Further investigations revealed that miR-133a reversed the biological effects of AFAP1-AS1 on PC cells. Collectively, the findings provide new evidence that AFAP1-AS1 could regulate the progression of pancreatic cancer by acting as a ceRNA, and suggest it has potential for use as both a biomarker for the early detection PC and for the development of individualized therapies for PC.


Asunto(s)
MicroARNs/metabolismo , Oncogenes , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , ARN Largo no Codificante/metabolismo , Receptores de Somatomedina/genética , Regulación hacia Arriba/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones Desnudos , MicroARNs/genética , Persona de Mediana Edad , Modelos Biológicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Receptor IGF Tipo 1
19.
Gene ; 665: 67-73, 2018 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-29709639

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most lethal and prevalent cancers worldwide and has recently become the second most common cause of cancer-related deaths in men of developing countries. Guanine nucleotide-binding protein (G protein) has been reported to be associated with the early process of HCC. In our previous study, GNAO1, one of members of G protein, was found to be down-regulated in HCC. Thus, the present study aimed to throw light upon the mechanism of the abnormal expression of GNAO1 in HCC. First, qPCR results from two HCC cell lines (SMMC-7721 and QGY-7703) confirmed the down-expression of GNAO1, followed by the validation of the methylation status of the promoter region by bisulfite sequence PCR (BSP). Moreover, 5-Aza-2'-deoxycytidine (DAC) with Trichostatin A (TSA) treatment made it much clear that GNAO1 transcription was inhibited by promoter hypermethylation, contributing to its low expression. It was further revealed that the silencing effect was regulated by methyltransferase 1 (DNMT1), and was further enhanced by transforming growth factor ß (TGF-ß). In addition, the up-regulation of GNAO1 with the help of recombinant plasmid was also found to accelerate cell apoptosis, confirmed by flow cytometry and western blotting analysis. All these results above indicated that the promoter hypermethylation of GNAO1 might play an important role in HCC, suggesting that it might be used as a promising biomarker for HCC diagnosis and targeted therapy.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Metilación de ADN , ADN de Neoplasias/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/biosíntesis , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/metabolismo , Regiones Promotoras Genéticas , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN de Neoplasias/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Proteínas de Neoplasias/genética
20.
Cell Death Dis ; 9(3): 253, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29449541

RESUMEN

Maternally expressed gene 3 (MEG3) encodes an lncRNA which is suggested to function as a tumor suppressor and has been showed to involve in a variety of cancers. Herein, our findings demonstrate that MEG3 inhibits the malignant progression of liver cancer cells in vitro and in vivo. Mechanistically, MEG3 promotes the expression and maturition of miR122 which targets PKM2. Therefore, MEG3 decreases the expression and nuclear location of PKM2 dependent on miR122. Furthermore, MEG3 also inhibits CyclinD1 and C-Myc via PKM2 in liver cancer cells. On the other hand, MEG3 promotes ß-catenin degradation through ubiquitin-proteasome system dependent on PTEN. Strikingly, MEG3 inhibits ß-catenin activity through PKM2 reduction and PTEN increase. Significantly, we also found that excessive ß-catenin abrogated the effect of MEG3 in liver cancer. In conclusion, our study for the first time demonstrates that MEG3 acts as a tumor suppressor by negatively regulating the activity of the PKM2 and ß-catenin signaling pathway in hepatocarcinogenesis and could provide potential therapeutic targets for the treatment of liver cancer.


Asunto(s)
Proteínas Portadoras/metabolismo , Proliferación Celular , Neoplasias Hepáticas/enzimología , Proteínas de la Membrana/metabolismo , Fosfohidrolasa PTEN/metabolismo , ARN Largo no Codificante/metabolismo , Hormonas Tiroideas/metabolismo , beta Catenina/metabolismo , Animales , Proteínas Portadoras/genética , Línea Celular Tumoral , Estabilidad de Enzimas , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Proteínas de la Membrana/genética , Ratones Endogámicos BALB C , Ratones Desnudos , Fosfohidrolasa PTEN/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , ARN Largo no Codificante/genética , Transducción de Señal , Hormonas Tiroideas/genética , Carga Tumoral , beta Catenina/genética , Proteínas de Unión a Hormona Tiroide
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA