Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nanomedicine ; 42: 102538, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35183762

RESUMEN

Non-alcoholic steatohepatitis (NASH) is the major form of chronic liver disease in adults; however, there are no approved drugs for NASH. In this study, we designed the PNM-G-PV method, in which gelatin nanoparticles (G) are loaded with pioglitazone and vitamin E (G-PV) and then encapsulated by the surfaces of platelet-neutrophil hybrid membranes (PNM). Inherited from the natural source cells, the PNM show immune evading ability due to the surface marker comprising a number of "do not eat me" signals and has dual inflammatory enrichment capabilities due to specific surface adhesion molecules. By functionalizing the gelatin nanoparticle biomimetic surfaces, PNM-G can enhance the targeting to inflammatory sites and enrich liver tissue. The high expression of matrix metalloproteinase-9 (MMP-9) at the NASH site enables the gelatin nanoparticles to intelligently respond to degradation and then release vitamin E and pioglitazone for drug treatment. We performed an in vivo analysis of these nanoparticles to monitor changes in triacylglycerol metabolism in liver tissues and assessed the therapeutic efficacy of PNM-G-PV in a NASH rat model. The results showed that PNM-G-PV exhibited better therapeutic efficacy than therapies using G-PV or PV alone. This work explores a new biomedical use of PNM-G-PV and a promising NASH treatment protocol based on a new drug delivery system.


Asunto(s)
Nanopartículas , Enfermedad del Hígado Graso no Alcohólico , Animales , Gelatina , Hígado/metabolismo , Neutrófilos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Pioglitazona/uso terapéutico , Ratas , Vitamina E
2.
Exp Cell Res ; 383(2): 111542, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31381879

RESUMEN

Hepatocellular carcinoma(HCC) is a malignant tumor with high mortality due to lack of early diagnostic methods and effective treatments, and the molecular mechanisms are intricate and remain unclear. In the present study, the role of macrophage receptor with collagenous structure (MARCO) in tumor advancement of HCC was investigated. We examined expression level of MARCO in HCC samples, corresponding adjacent nontumor tissues and six hepatoma cell lines by polymerase chain reaction and immunohistochemistry (IHC). Clinical information of HCC patients was also analyzed. The role of MARCO involved in HCC progression via multiple functional experiments in vitro and in vivo was investigated. Bioinformatics analysis was conducted to further explore biological functions of MARCO. We found MARCO was suggestively down-regulated in HCC and associated with favorable prognosis, and MARCO upregulation oppressed tumor cell migration and invasion. Besides, overexpression of MARCO not only promoted apoptosis of hepatoma cells but also suppressed proliferation in vivo and in vitro. Furthermore, gene set enrichment analysis (GSEA) analysis suggested that MARCO may be related to the P53 signaling pathway, and this prediction was confirmed in this study as well. In sum, our study indicated that MARCO was involved in HCC progression and it can be defined as a novel probable biomarker as well as treatment target for HCC.


Asunto(s)
Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Receptores Inmunológicos/genética , Anciano , Animales , Biomarcadores de Tumor/genética , Carcinoma Hepatocelular/genética , Estudios de Casos y Controles , Movimiento Celular/genética , Proliferación Celular/genética , Células Cultivadas , Conjuntos de Datos como Asunto , Progresión de la Enfermedad , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad
3.
Pain Manag Nurs ; 20(1): 39-46, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29779795

RESUMEN

OBJECTIVE: The objective of this study was to compare the effects produced by yoga nidra and relaxation music for pain management in patients undergoing colonoscopy. A quasiexperimental design was used. METHODS: In total, 144 patients who were scheduled to undergo colonoscopy were assigned to three different treatment groups. Group 1 was a no treatment control group, group 2 was delivered relaxing music, and group 3 was delivered a yoga nidra recording. The primary outcome was pain score. Secondary treatment efficacy measures were an overall patient satisfaction score, a willingness to repeat the procedure score, and a perceived colonoscope insertion difficulty score. Secondary objective treatment effect measures were systolic and diastolic blood pressure and total procedure duration. RESULTS: The patients' perceptions of pain and the endoscopist's perceived colonoscope insertion difficulty were significantly reduced by both the music and the yoga nidra recording (p < .05). Overall patient satisfaction was significantly improved by both the music and the yoga nidra recording (p < .05). Patients' willingness to repeat the procedure and the total procedure duration were significantly improved and reduced, respectively, by the yoga nidra recording (p < .05), but there were no significant differences compared to the music group. There were no statistically significant differences among the three groups in terms of blood pressure. CONCLUSIONS: Both the yoga nidra recording and the relaxation music helped reduce the pain participants undergoing colonoscopy experienced. The yoga nidra recording was the most successful intervention among the three groups.


Asunto(s)
Colonoscopía/métodos , Manejo del Dolor/métodos , Yoga/psicología , Adulto , China , Colonoscopía/normas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Manejo del Dolor/normas , Satisfacción del Paciente
4.
J Cell Mol Med ; 22(5): 2569-2579, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29516682

RESUMEN

This study aimed to explore the effects of CLIC1 gene silencing on proliferation, migration, invasion and apoptosis of human gallbladder cancer (GBC). GBC and normal gallbladder tissues were extracted for the detection of mRNA and protein expressions of CLIC1. GBC-SD and NOZ cells in the logarithmic growth phase were selected to conduct the experiment. Three different siRNA recombined expression vectors were established using CLIC1 as a target at different sites. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blotting were, respectively, used to detect the CLIC1 mRNA and protein expressions. MTT assay was performed to detect the cell proliferation. Flow cytometry was applied to measure the cell apoptosis and cell cycle distribution. The variations of cell migration and invasion were evaluated using Transwell assay. GBC tissues showed higher CLIC1 mRNA and protein expressions than normal gallbladder tissues. The CLIC1 mRNA and protein expressions in the CLIC1 siRNA group were significantly lower than those in the NC and blank groups. Compared with the NC and blank groups, the CLIC1 siRNA group showed a significant decrease in cell proliferation but an obvious increase in apoptosis rate in GBC cells. Besides, in the CLIC1 siRNA group, cell percentage in G0/G1 and G2/M phase was gradually increased but decreased in S phases. The migration and invasion abilities in GBC cells were significantly lower than those in the NC and blank groups. Our study demonstrates that CLIC1 gene silencing could promote apoptosis and inhibit proliferation migration and invasion of GBC cells.


Asunto(s)
Apoptosis/genética , Movimiento Celular/genética , Canales de Cloruro/genética , Neoplasias de la Vesícula Biliar/genética , Neoplasias de la Vesícula Biliar/patología , Silenciador del Gen , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Canales de Cloruro/metabolismo , Vesícula Biliar/metabolismo , Vesícula Biliar/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo
5.
Nanotechnology ; 29(8): 084002, 2018 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-29339567

RESUMEN

Recently, red blood cell (RBC) membrane-coated nanoparticles have attracted much attention because of their excellent immune escapability; meanwhile, gold nanocages (AuNs) have been extensively used for cancer therapy due to their photothermal effect and drug delivery capability. The combination of the RBC membrane coating and AuNs may provide an effective approach for targeted cancer therapy. However, few reports have shown the utilization of combining these two technologies. Here, we design erythrocyte membrane-coated gold nanocages for targeted photothermal and chemical cancer therapy. First, anti-EpCam antibodies were used to modify the RBC membranes to target 4T1 cancer cells. Second, the antitumor drug paclitaxel (PTX) was encapsulated into AuNs. Then, the AuNs were coated with the modified RBC membranes. These new nanoparticles were termed EpCam-RPAuNs. We characterized the capability of the EpCam-RPAuNs for selective tumor targeting via exposure to near-infrared irradiation. The experimental results demonstrate that EpCam-RPAuNs can effectively generate hyperthermia and precisely deliver the antitumor drug PTX to targeted cells. We also validated the biocompatibility of the EpCam-RAuNs in vitro. By combining the molecularly modified targeting RBC membrane and AuNs, our approach provides a new way to design biomimetic nanoparticles to enhance the surface functionality of nanoparticles. We believe that EpCam-RPAuNs can be potentially applied for cancer diagnoses and therapies.

6.
Tumour Biol ; 37(1): 1079-89, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26271667

RESUMEN

LncRNA has provided an important new perspective regarding gene regulation. Both the expression and activation of EGFR have been proven to be under the tight control of the GHR pathway. EGFR-AS1 has been found to inhibit the expression of EGFR. GHR-siRNA and EGFR-AS1-siRNA were transfected into HCC cell lines, and a series of WB, q-PCR, and IF experiments was conducted to evaluate whether EGFR-AS1 participated in the regulation of GHR and EGFR. We found that impeded expression of GHR decreased the expression of EGFR and EGFR-AS1 in vivo and in vitro. Then, it was verified that EGFR and EGFR-AS1 were relatively upregulated in HCC tissue, and they were significantly related to some clinical characteristics and patient prognosis. Furthermore, EGFR-AS1 was determined to promote HCC development by improving the ability of invasion and proliferation of HCC cells in vitro, and it was also found to affect the cell cycle. Our study identified that EGFR-AS1 may promote HCC genesis and development. EGFR-AS1 may act as a prognostic factor in HCC. More importantly, we observed that the inhibition of EGFR-AS1 in HCC cells significantly impeded cell proliferation and invasion in vivo, which might provide a potential possibility for targeted therapy of HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Proteínas Portadoras/genética , Receptores ErbB/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/genética , ARN Largo no Codificante/genética , Animales , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Perfilación de la Expresión Génica , Humanos , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Pronóstico , Carga Tumoral , Regulación hacia Arriba
7.
J Biol Chem ; 289(47): 32639-55, 2014 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-25271158

RESUMEN

Patients with chronic hepatitis B usually exhibit a low response to treatment with interferon α (IFN-α). An alternative approach to increase the response rate of IFN-α might be to immunologically stimulate the host with glucocorticoids (GCs) before treatment with IFN-α, but the underlying mechanism remains unclear. We hypothesized that the GCs enhance IFN signaling by inducing S-adenosylmethionine (AdoMet) when hepatitis B virus (HBV) replication was effectively suppressed by IFN-α. Here, we investigated the effect of GCs and IFN-α on AdoMet production and methionine adenosyltransferase 1A (MAT1A) expression in vitro. Furthermore, we determined whether post-transcriptional regulation is involved in HBV-repressed MAT1A expression and AdoMet production induced by dexamethasone (Dex). We found that AdoMet homeostasis was disrupted by Dex and that Dex directly regulated MAT1A expression by enhancing the binding of the glucocorticoid receptor (GR) to the glucocorticoid-response element (GRE) of the MAT1A promoter. HBV reduced AdoMet production by increasing methylation at GRE sites within the MAT1A promoter. The X protein of hepatitis B virus led to hypermethylation in the MAT1A promoter by recruiting DNA methyltransferase 1, and it inhibited GR binding to the GRE in the MAT1A promoter. Dex could increase an antiviral effect by inducing AdoMet production via a positive feedback loop when HBV is effectively suppressed by IFN-α, and the mechanism that involves Dex-induced AdoMet could increase STAT1 methylation rather than STAT1 phosphorylation. These findings provide a possible mechanism by which GC-induced AdoMet enhances the antiviral activity of IFN-α by restoring STAT1 methylation in HBV-infected cells.


Asunto(s)
Glucocorticoides/farmacología , Virus de la Hepatitis B/efectos de los fármacos , Interferón-alfa/farmacología , S-Adenosilmetionina/metabolismo , Factor de Transcripción STAT1/metabolismo , Antivirales/farmacología , Línea Celular , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/efectos de los fármacos , Dexametasona/farmacología , Expresión Génica/efectos de los fármacos , Células Hep G2 , Virus de la Hepatitis B/fisiología , Humanos , Immunoblotting , Metionina Adenosiltransferasa/genética , Metionina Adenosiltransferasa/metabolismo , Metilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Receptores de Glucocorticoides/metabolismo , Elementos de Respuesta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
8.
Nat Commun ; 15(1): 1170, 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38326322

RESUMEN

SAP05, a secreted effector by the obligate parasitic bacteria phytoplasma, bridges host SPL and GATA transcription factors (TFs) to the 26 S proteasome subunit RPN10 for ubiquitination-independent degradation. Here, we report the crystal structures of SAP05 in complex with SPL5, GATA18 and RPN10, which provide detailed insights into the protein-protein interactions involving SAP05. SAP05 employs two opposing lobes with an acidic path and a hydrophobic path to contact TFs and RPN10, respectively. Our crystal structures, in conjunction with mutagenesis and degradation assays, reveal that SAP05 targets plant GATAs but not animal GATAs dependent on their direct salt-bridged electrostatic interactions. Additionally, SAP05 hijacks plant RPN10 but not animal RPN10 due to structural steric hindrance and the key hydrophobic interactions. This study provides valuable molecular-level information into the modulation of host proteins to prevent insect-borne diseases.


Asunto(s)
Factores de Transcripción , Ubiquitina , Ubiquitina/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitinación
9.
Hepatol Res ; 43(4): 374-83, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23072598

RESUMEN

AIM: Methionine adenosyltransferase 1A (MAT1A) is inactivated in HCC and may be stimulated by an epigenetic change involving promoter hypermethylation in hepatocarcinogenesis. However, the possible clinical impact and prognosis of this inactivation have not been investigated. METHODS: We studied the methylation status of the CpG sites in the promoter region and the mRNA and protein expression of MAT1A in HCC and corresponding adjacent non-tumor tissues using methylation-specific polymerase chain reaction, reverse transcription polymerase chain reaction and immunohistochemistry techniques. RESULTS: MAT1A promoter methylation was significantly higher in HCC than that in adjacent non-tumor tissues (P < 0.0001). Bisulfite sequencing showed that the four CpG sites were hypermethylated in HCC while hypomethylation was found in the corresponding adjacent non-tumor tissues. Furthermore, MAT1A methylation was significantly associated with protein expression (P = 0.022). Low expression of MAT1A was correlated with larger tumor size, higher tumor-node-metastasis stage, positive hepatitis B surface antigen status and high α-fetoprotein (AFP) serum levels (P < 0.05). MAT1A promoter methylation was also correlated with high AFP serum level (P < 0.05). In univariate survival analysis, low expression of MAT1A was significantly associated with shortened patient survival (P < 0.001). Furthermore, in multivariate analysis, MAT1A expression was found as an independent prognostic factor (P = 0.016). CONCLUSION: Our observations suggest that hypermethylation of the MAT1A promoter may be one of the events in the development of HCC. Low expression of MAT1A is likely involved in the progression of the tumor and was found to be an independent factor for poor prognosis of patients with HCC.

10.
Nat Commun ; 14(1): 2474, 2023 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-37120596

RESUMEN

The ribosome-associated quality-control (RQC) pathway degrades aberrant nascent polypeptides arising from ribosome stalling during translation. In mammals, the E3 ligase Pirh2 mediates the degradation of aberrant nascent polypeptides by targeting the C-terminal polyalanine degrons (polyAla/C-degrons). Here, we present the crystal structure of Pirh2 bound to the polyAla/C-degron, which shows that the N-terminal domain and the RING domain of Pirh2 form a narrow groove encapsulating the alanine residues of the polyAla/C-degron. Affinity measurements in vitro and global protein stability assays in cells further demonstrate that Pirh2 recognizes a C-terminal A/S-X-A-A motif for substrate degradation. Taken together, our study provides the molecular basis underlying polyAla/C-degron recognition by Pirh2 and expands the substrate recognition spectrum of Pirh2.


Asunto(s)
Mamíferos , Ubiquitina-Proteína Ligasas , Animales , Ubiquitina-Proteína Ligasas/metabolismo , Proteolisis , Mamíferos/metabolismo
11.
J Biol Chem ; 286(19): 17168-80, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21247894

RESUMEN

The X protein (HBx) of hepatitis B virus (HBV) is involved in the development of hepatocellular carcinoma (HCC), and methionine adenosyltransferase 2A (MAT2A) promotes the growth of liver cancer cells through altering S-adenosylmethionine homeostasis. Thus, we speculated that a link between HBx and MAT2A may contribute to HCC development. In this study, the effects of HBx on MAT2A expression and cell apoptosis were investigated, and the molecular mechanism by which HBx and MAT2A regulate tumorigenesis was evaluated. Results from immunohistochemistry analyses of 37 pairs of HBV-associated liver cancer tissues/corresponding peritumor tissues showed that HBx and MAT2A are highly expressed in most liver tumor tissues. Our in vitro results revealed that HBx activates MAT2A expression in a dose-dependent manner in hepatoma cells, and such regulation requires the cis-regulatory elements NF-κB and CREB on the MAT2A gene promoter. Electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) further demonstrated that HBx facilitates the binding of NF-κB and CREB to MAT2A gene promoter. In addition, overexpression of HBx or MAT2A inhibits cell apoptosis, whereas knockdown of MAT2A expression stimulates apoptosis in hepatoma cells. Furthermore, we demonstrated that HBx reduces MAT1A expression and AdoMet production but enhances MAT2ß expression. Thus, we proposed that HBx activates MAT2A expression through NF-κB and CREB signaling pathways to reduce AdoMet production, inhibit hepatoma cell apoptosis, and perhaps enhance HCC development. These findings should provide new insights into our understanding how the molecular mechanisms underline the effects of HBV infection on the production of MAT2A and the development of HCC.


Asunto(s)
Apoptosis , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Regulación Viral de la Expresión Génica , Virus de la Hepatitis B/metabolismo , Metionina Adenosiltransferasa/genética , Transactivadores/metabolismo , Carcinoma Hepatocelular/virología , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica/métodos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/virología , Metionina Adenosiltransferasa/metabolismo , FN-kappa B/metabolismo , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal , Proteínas Reguladoras y Accesorias Virales
12.
Int J Biol Sci ; 18(7): 2932-2948, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35541917

RESUMEN

Long noncoding RNAs (lncRNAs) play an important role in the progression of hepatocellular carcinoma (HCC). Linc01612 is a novel lncRNA that function remains unknown in the progression of cancers, including HCC. In this study, we discovered that Linc01612 is significantly down-regulated in HCC tissues than in non-tumor tissues and correlated with poor prognosis. Linc01612 mainly localizes in the cytoplasm and functions as a tumor suppressor by repressing the growth and metastasis of hepatoma cells in vitro and in vivo. Mechanistically, in p53-expressing hepatoma cells, Linc01612 acts as a competitive endogenous RNA and promotes the expression of activation transcription factor 3 (ATF3) by sponging microRNA-494 (miR-494), which in turn inhibits MDM2-mediated ubiquitination of p53 and activates the p53 pathway. Furthermore, in p53-null hepatoma cells, Linc01612 exerts its biological functions by physically interacting with Y-box binding protein 1 protein (YBX1) and promoting the ubiquitin-mediated degradation of YBX1. Interestingly, the Linc01612-YBX1 signaling pathway is also present in p53-expressing hepatoma cells. In conclusion, our study indicated that Linc01612 is a functional lncRNA in HCC and Linc01612 may serve as a potential diagnostic biomarker and therapeutic target for HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroARNs , ARN Largo no Codificante , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Hepáticas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Factor de Transcripción 3/genética , Factor de Transcripción 3/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinación/genética , Proteína 1 de Unión a la Caja Y/genética , Proteína 1 de Unión a la Caja Y/metabolismo
13.
Front Oncol ; 12: 916457, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35814451

RESUMEN

Intraductal papillary neoplasm of the bile duct (IPNB) is a rare bile duct tumor characterized by intraductal papillary or villous neoplasms covered by neoplastic epithelium with fine fibrovascular stalks in the dilated bile ducts (1). Its true etiology remains unknown. Herein, we report two cases of IPNB that underwent surgical resection. The first case was a 66-year-old male who complained of upper abdominal pain for three years. We found obstruction of the common bile duct and dilation of the intrahepatic and extrahepatic bile ducts after MRCP. Laparoscopic hepatic segmentectomy (S2, S3, S4), resection of the common bile duct, cholecystectomy, and hepaticojejunostomy were performed. The second case was a 67-year-old male with asymptomatic dilation of the intrahepatic duct. The patient underwent robot-assisted laparoscopic hepatic segmentectomy (S5, S6, S7, S8), resection of the common bile duct, hepaticojejunostomy and cholecystectomy.

14.
Int J Biol Sci ; 18(1): 82-95, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34975319

RESUMEN

HCC has remained one of the challenging cancers to treat, owing to the paucity of drugs targeting the critical survival pathways. Considering the cancer cells are deficient in DNase activity, the increase of an autonomous apoptisis endonuclease should be a reasonable choice for cancer treatment. In this study, we investigated whether DNASE1L3, an endonuclease implicated in apoptosis, could inhibit the progress of HCC. We found DNASE1L3 was down-regulated in HCC tissues, whereas its high expression was positively associated with the favorable prognosis of patients with HCC. Besides, serum DNASE1L3 levels were lower in HCC patients than in healthy individuals. Functionally, we found that DNASE1L3 inhibited the proliferation of tumor cells by inducing G0/G1 cell cycle arrest and cell apoptosis in vitro. Additionally, DNASE1L3 overexpression suppressed tumor growth in vivo. Furthermore, we found that DNASE1L3 overexpression weakened glycolysis in HCC cells and tissues via inactivating the rate-limiting enzymes involved in PTPN2-HK2 and CEBPß-p53-PFK1 pathways. Finally, we identified the HBx to inhibit DNASE1L3 expression by up-regulating the expression of ZNF384. Collectively, our findings demonstrated that DNASE1L3 could inhibit the HCC progression through inducing cell apoptosis and weakening glycolysis. We believe DNASE1L3 could be considered as a promising prognostic biomarker and therapeutic target for HCC.


Asunto(s)
Apoptosis , Carcinoma Hepatocelular/metabolismo , Desoxirribonucleasas/metabolismo , Glucólisis , Neoplasias Hepáticas/metabolismo , Animales , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Regulación hacia Abajo , Xenoinjertos , Hexoquinasa/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Fosfofructoquinasa-1/metabolismo
15.
Zhonghua Wai Ke Za Zhi ; 49(4): 346-50, 2011 Apr 01.
Artículo en Zh | MEDLINE | ID: mdl-21612702

RESUMEN

OBJECTIVE: To evaluate and compare the efficiency and safety of laparoscopic surgery (LS) and open surgery (OS) in the treatment of colorectal carcinoma. METHODS: Randomized controlled trials on laparoscopic surgery and open surgery for colorectal carcinoma from January 2000 to October 2010 were searched in the databases of EMbase, PubMed, Cochrane Library, Sciencedirect, Springer, VIP, CNKI, CBMdisc. The methodological quality was assessed according to the standard of Cochrane systematic review. For homogeneous studies, RevMan5.0 software was used for meta-analysis. RESULTS: A total of 13 RCTs involving 4603 patients were included in this study, and among those 6 were multi-center randomized controlled trials. The meta-analysis showed that: the operation time of the LS group was longer than that of the OS group (WMD = 38.91, 95%CI: 33.89 - 43.93, P < 0.001), the blood loss (WMD = -138.14, 95%CI: -195.79 - -80.50, P < 0.001) and the length of hospital stay (WMD = 2.91, 95%CI: -4.65 - -1.17, P = 0.001) of the LS group was less than those in OS group. There was no significant differences between the two groups in the number of dissected lymph nodes (WMD = -0.62, 95%CI: -1.47 - 0.23, P = 0.150). There was no significant differences between the two groups in terms of the postoperative complications (30 days) (RR = 0.78, 95%CI: 0.59 - 1.01, P = 0.06). There was no significant differences between the two groups in 3-year overall survival (RR = 1.00, 95%CI: 0.96 - 1.04, P = 0.970). There was no significant differences between the two groups in 5-year overall survival (RR = 1.03, 95%CI: 0.99 - 1.08, P = 0.140). There was no significant differences between the two groups in 5-year overall recurrence (RR = 0.89, 95%CI: 0.74 - 1.07, P = 0.200). CONCLUSIONS: Laparoscopic surgery for colorectal carcinoma is a safe and effective therapy as open surgery in the short term or long term outcomes. It could be an acceptable alternative to open surgery for colorectal carcinoma.


Asunto(s)
Neoplasias Colorrectales/cirugía , Laparoscopía , Humanos , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento
16.
Front Surg ; 8: 678853, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34336917

RESUMEN

Background: Neuroendocrine tumors are heterogeneous malignancies that originate from the neuroendocrine system. Previous studies show that this cancer type mainly localizes in the gastrointestinal tract and often metastasizes to the liver. Primary liver neuroendocrine tumors are very rare and primary hepatic neuroendocrine tumors (PHNET) with concurrent hepatocellular carcinoma (HCC) are extremely rare. To the best of our knowledge, only few PHNET cases have been identified, making their diagnosis difficult. Here, we report the biggest ever reported and "deceiving" lesion of a mixed neuroendocrine-non-neuroendocrine neoplasm in the liver, aiming to raise awareness and improve treatment of the disease. Case Presentation: Here, we report a preoperative misdiagnosed case that presented with hepatocellular carcinoma clinical features and no extrahepatic tumors. Postoperative pathology confirmed that it was a mixed neuroendocrine-non-neuroendocrine neoplasm. The patient was then referred for etoposide and cisplatin-based chemotherapy. No disease recurrence was observed at the 6-month follow-up. Conclusion: We report a very rare and easily misdiagnosed case and we speculate that there were "undifferentiated cells" undergoing neuroendocrine and hepatocellular carcinoma differentiation, during which some hepatocellular carcinoma cells express neuroendocrine features. We recommend proper surgery and postoperative platinum-based chemotherapy in the management of this disease.

17.
Aging (Albany NY) ; 13(7): 9874-9899, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33744849

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most challenging and aggressive cancers with limited treatment options because of tumor heterogeneity. Tumor angiogenesis is a hallmark of HCC and is necessary for tumor growth and progression. DNA damage stress and its associated deoxyribonuclease1-like 3 (DNASE1L3) are involved in HCC progression. Here, we explored the influence mechanism of DNASE1L3 on tumor angiogenesis under DNA damage stress in vitro and in vivo. DNASE1L3 was found downregulated and negatively correlated with poor prognosis of resectable and unresectable HCC patients. The tissue microarray of HCC revealed the negative association between DNASE1L3 and cancer vasculature invasion. Mechanistically, DNASE1L3 was found to relieve cytoplasmic DNA accumulation under DNA damage stress in HCC cell lines, in turn cell senescence and senescence-associated secretory phenotype were arrested via the p53 and NF-κB signal pathway, and hence, tumor angiogenesis was impaired. Furthermore, we found that DNASE1L3 excised these functions by translocating to the nucleus and interacting with H2BE under DNA damage stress using co-immunoprecipitation and fluorescence resonance energy transfer assay. In conclusion, DNASE1L3 inhibits tumor angiogenesis via impairing the senescence-associated secretory phenotype in response to DNA damage stress.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Senescencia Celular/genética , Endodesoxirribonucleasas/metabolismo , Neoplasias Hepáticas/metabolismo , Neovascularización Patológica/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Proliferación Celular/genética , Daño del ADN/fisiología , Endodesoxirribonucleasas/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Pronóstico , Tasa de Supervivencia
18.
Cancer Lett ; 513: 75-89, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-33957185

RESUMEN

Emerging evidence has shown that aberrant expression of lncRNA-TP53TG1 plays important roles in various malignancies. However, the biological functions of lncRNA-TP53TG1 in hepatocarcinogenesis, as well as the underlying mechanisms, remain largely unknown. Here, we assessed whether lncRNA-TP53TG1 plays a key role in the progression of hepatocellular carcinoma (HCC). The expression of lncRNA-TP53TG1 was significantly decreased in HCC tissues and cells. Decreased expression of lncRNA-TP53TG1 was associated with aggressive clinical phenotypes and a poor prognosis. Ectopic expression of lncRNA-TP53TG1 inhibited hepatoma cell proliferation and migration in vitro and in vivo, whereas lncRNA-TP53TG1 knockdown exerted the opposite effects. Furthermore, lncRNA-TP53TG1 played an important role in slowing the epithelial-mesenchymal transition (EMT) process in HCC. Mechanistically, lncRNA-TP53TG1 physically interacted with PRDX4 and promoted its ubiquitin-mediated degradation, resulting in the inactivation of the WNT/ß-catenin signaling pathway in hepatoma cells. Our findings demonstrate a novel mechanism by which lncRNA-TP53TG1 exerts its tumor-suppressive effects through the WNT/ß-catenin signaling pathway in a PRDX4-mediated manner in HCC. Based on these results, lncRNA-TP53TG1 potentially represents a prognostic indicator and therapeutic target for patients with HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Peroxirredoxinas/metabolismo , ARN Largo no Codificante/metabolismo , beta Catenina/metabolismo , Animales , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Metástasis de la Neoplasia , Pronóstico , Estudios Retrospectivos
19.
RSC Adv ; 10(12): 7194-7205, 2020 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-35493895

RESUMEN

Developing new drugs for cancer treatment remains a challenging task. Herein, cancer cell membrane biomimetic ferrous ion-doped metal-organic framework nanoparticles (ZIF-8 nanoparticles) combined with dihydroartemisinin (DHA) have been designed for targeted cancer treatment with low toxicity and side effects. The biomimetic nanomaterials (CDZs) have excellent homologous targeting ability and can accumulate in tumor tissues. In an acid tumor microenvironment, ferrous ions and DHA could be released with the degradation of materials. DHA, an ancient Chinese medicine, combines with ferrous ions to produce a powerful anti-tumor effect. In human liver cancer models, about 90.8% of tumor growth was suppressed. In addition, the nanomaterial has no obvious toxic and side effects in vivo and is a highly effective and low-toxicity anti-tumor drug with a strong clinical application value.

20.
Front Genet ; 11: 153, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32180800

RESUMEN

Hepatocellular carcinoma (HCC) remains hard to diagnose early and cure due to a lack of accurate biomarkers and effective treatments. Hence, it is necessary to explore the tumorigenesis and tumor progression of HCC to discover new biomarkers for clinical treatment. We performed weighted gene co-expression network analysis (WGCNA) to explore hub genes that have high correlation with clinical information. In this study, we found 13 hub genes (GTSE1, PLK1, NCAPH, SKA3, LMNB2, SPC25, HJURP, DEPDC1B, CDCA4, UBE2C, LMNB1, PRR11, and SNRPD2) that have high correlation with histologic grade in HCC by analyzing TCGA LIHC dataset. All of these 13 hub genes could be used to effectively distinguish high histologic grade from low histologic grade of HCC through analysis of the ROC curve. The overall survival and disease-free survival information showed that high expression of these 13 hub genes led to poor prognosis. Meanwhile, these 13 hub genes had significantly different expression in HCC tumor and non-tumor tissues. We downloaded GSE6764, which contains corresponding clinical information, to validate the expression of these 13 hub genes. At the same time, we performed quantitative real-time PCR to validate the differences in the expression tendencies of these 13 hub genes between HCC tumor tissues and non-tumor tissues and high histologic grade and low histologic grade. We also explored mutation and methylation information of these 13 hub genes for further study. In summary, 13 hub genes correlated with the progression and prognosis of HCC were discovered by WGCNA in our study, and these hub genes may contribute to the tumorigenesis and tumor progression of HCC.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA