Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Funct Integr Genomics ; 24(1): 8, 2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-38200280

RESUMEN

Members of the E26 transformation-specific (ETS) variant transcription factor family act as either tumor suppressors or oncogenic factors in numerous types of cancer. ETS variant transcription factor 7 (ETV7) participates in the development of malignant tumors, whereas its involvement in colorectal cancer (CRC) is less clear. In this study, The Cancer Genome Atlas (TCGA) and immunochemistry staining were applied to check the clinical relevance of ETV7 and interferon-induced protein with tetratricopeptide repeats 3 (IFIT3) in CRC patients. Overexpression and knockdown of ETV7 and IFIT3 were conducted by transfecting the cells with pCDNA3.1 plasmids and siRNAs, respectively. Western blotting was used to detect the protein expression of ETV7 in CRC cells. Cell Counting Kit-8, cell colony formation, and Transwell assays, as well as flow cytometry, were used to evaluate the proliferation, migration, cell cycle, and apoptosis of CRC cells. Furthermore, western blotting, RT-qPCR, and luciferase assay were used to explore the regulation of ETV7 on IFIT3. Rescue assay was used to investigate the significance of ETV7/IFIT3 axis on CRC progression. We found that ETV7 was upregulated in CRC tissues and cells. Overexpression of ETV7 stimulated the proliferation, migration, and cell cycle amplification, and reduced the apoptosis of CRC cells. Downregulation of ETV7 exerted the opposite effect on CRC cell progression. Moreover, we demonstrated that ETV7 stimulated the transcription activity, the mRNA and protein expression of IFIT3 in CRC cells. There was a positive correlation between ETV7 and IFIT3 in CRC patients. IFIT3 knockdown reversed the promotive effect exerted by overexpression of ETV7 on the amplification and migration of CRC cells. By contrast, overexpression of IFIT3 blocked the inhibitory effect of ETV7-targeting siRNA. In summary, ETV7 induces progression of CRC by activating the transcriptional expression of IFIT3. The EVT7/IFIT3 axis may be a novel target for CRC therapy.


Asunto(s)
Apoptosis , Neoplasias Colorrectales , Humanos , Regulación hacia Arriba , Regulación hacia Abajo , Apoptosis/genética , Neoplasias Colorrectales/genética , Proteínas Proto-Oncogénicas c-ets , Péptidos y Proteínas de Señalización Intracelular
2.
Acta Pharmacol Sin ; 44(12): 2525-2536, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37474643

RESUMEN

Heat shock protein family A member 8 (HSPA8) participates in the folding or degradation of misfolded proteins under stress and plays critical roles in cancer. In this study, we investigated the function of HSPA8 in the development of liver cancer. By analyzing the TCGA transcriptome dataset, we found that HSPA8 was upregulated in 134 clinical liver cancer tissue samples, and positively correlated with poor prognosis. IHC staining showed the nuclear and cytoplasmic localization of HSPA8 in liver cancer cells. Knockdown of HSPA8 resulted in a decrease in the proliferation of HepG2 and Huh-7 cells. ChIP-seq and RNA-seq analysis revealed that HSPA8 bound to the promoter of pleckstrin homology-like domain family A member 2 (PHLDA2) and regulated its expression. The transcription factor ETV4 in HepG2 cells activated PHLDA2 transcription. HSPA8 and ETV4 could interact with each other in the cells and colocalize in the nucleus. From a functional perspective, we demonstrated that HSPA8 upregulated PHDLA2 through the coactivating transcription factor ETV4 to enhance the growth of liver cancer in vitro and in vivo. From a therapeutic perspective, we identified both HSPA8 and PHDLA2 as novel targets in the treatment of HCC. In conclusion, this study demonstrates that HSPA8 serves as a coactivator of ETV4 and upregulates PHLDA2, leading to the growth of HCC, and is a potential therapeutic target in HCC treatment.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Neoplasias Hepáticas/genética , Factores de Transcripción/genética , Carcinoma Hepatocelular/genética , Proteínas de Choque Térmico , Regulación de la Expresión Génica , Proteínas Proto-Oncogénicas c-ets/genética
3.
J Med Virol ; 95(7): e28966, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37466313

RESUMEN

Viral immune evasion is crucial to the pathogenesis of hepatitis B virus (HBV) infection. However, the role of HBV in the modulation of innate immune evasion is poorly understood. A liver-specific histone acetyltransferase 1 (Hat1) knockout (KO) mouse model and HAT1 KO cell line were established. Immunohistochemistry staining, Western blot analysis, Southern blot analysis, Northern blot analysis, immunofluorescence assays, enzyme-linked immunosorbent assay, reverse transcription-quantitative polymerase chain reaction, and chromatin immunoprecipitation assays were performed in the livers of mouse models, primary human hepatocytes, HepG2-NTCP, and Huh7 and HepG2 cell lines. HBV-elevated HAT1 increased the expression of miR-181a-5p targeting cyclic GMP-AMP synthase (cGAS) messenger RNA 3' untranslated regions through modulating acetylation of H4K5 and H4K12 in vitro and in vivo, leading to the inability of cGAS-stimulator of interferon genes (STING) pathway and type I interferon (IFN-I) signaling. Additionally, HBV-elevated HAT1 promoted the expression of KPNA2 through modulating acetylation of H4K5 and H4K12 in the system, resulting in nuclear translocation of cGAS, HBx was responsible for the events by HAT1, suggesting that HBV-elevated HAT1 controls the cGAS-STING pathway and IFN-I signaling to modulate viral innate immune evasion. HBV confers innate immune evasion through triggering HAT1/acetylation of H4K5/H4K12/miR-181a-5p or KPNA2/cGAS-STING/IFN-I signaling. Our finding provides new insights into the mechanism by which HBV drives viral innate immune evasion.


Asunto(s)
Hepatitis B , MicroARNs , Ratones , Animales , Humanos , Virus de la Hepatitis B/genética , Evasión Inmune , Acetilación , Inmunidad Innata , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Histona Acetiltransferasas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , alfa Carioferinas/metabolismo
4.
Biomol Biomed ; 23(4): 605-615, 2023 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-36803544

RESUMEN

The Fc fragment of IgG binding protein (FCGBP) has been confirmed to play an important role in various cancers. However, the specific role of FCGBP in hepatocellular carcinoma (HCC) remains undefined. Thus, in this study, the enrichment analyses (Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and Gene Set Enrichment Analysis) of FCGBP in HCC and extensive bioinformatic analyses using data of clinicopathologic characteristics, genetic expression and alterations, and immune cell infiltration were perfomed. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to verify the expression of FCGBP in both HCC tissues and cell lines. The subsequent results confirmed thatFCGBP overexpression positively correlated with poor prognosis in patients with HCC. Additionally, FCGBP expression could effectively distinguish tumor tissues from normal tissues, which was verified by qRT-PCR. The result was further confirmed by using HCC cell lines. The time-dependent survival receiver operator characteristic curve exhibited the strong ability of FCGBP to predict survival in patients with HCC. Additionally, we revealed the strong relationship between FCGBP expression and a number of classic regulatory targets and classical oncogenic signaling pathways of tumors. Finally, FCGBP was involved in the regulation of immune infiltration in HCC. Therefore, FCGBP has potential value in the diagnosis, treatment, and prognosis of HCC and may be a potential biomarker or therapeutic target.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/genética , Proteínas Portadoras , Fragmentos Fc de Inmunoglobulinas , Inmunoglobulina G , Neoplasias Hepáticas/genética
5.
Cancer Med ; 12(3): 2312-2324, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36016484

RESUMEN

BACKGROUND: Liver transplantation (LT), resection (LR), and ablation (LA) are three curative-intent treatment options for patients with early hepatocellular carcinoma (HCC). We aimed to develop a prognostic calculator to compare the long-term outcomes following each of these therapies. METHODS: A total of 976 patients with HCC within the Milan criteria who underwent LT, LR, and LA between 2009 and 2019 from four institutions were evaluated. Multistate competing risks prediction models for recurrence-free survival (RFS), recurrence within the Milan criteria (RWM), and HCC-specific survival (HSS) were derived to develop a prognostic calculator. RESULTS: During a median follow-up of 51 months, 420 (43%) patients developed recurrence. In the multivariate analysis, larger tumor size, multinodularity, older age, male, higher alpha-fetoprotein (AFP), higher albumin-bilirubin (ALBI) grade, and the presence of portal hypertension were significantly associated with higher recurrence and decreased survival rates. The RFS and HSS were both significantly higher among patients treated by LT than by LR or LA and significantly higher between patients treated by LR than by LA (all p < 0.001). For multinodular HCC ≤3 cm, although LT had better RFS and HSS than LR or LA, LA was noninferior to LR. An online prognostic calculator was then developed based on the preoperative clinical factors that were independently associated with outcomes to evaluate RFS, RWM, and HSS at different time intervals for all three treatment options. CONCLUSIONS: Although LT resulted in the best recurrence and survival outcomes, LR and LA also offered durable long-term alternatives. This prognostic calculator is a useful tool for clinicians to guide an informed and personalized discussion with patients based on their tumor biology and liver function.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Trasplante de Hígado , Humanos , Masculino , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Hepatectomía/métodos , Trasplante de Hígado/métodos , Pronóstico , Estudios Retrospectivos , Recurrencia Local de Neoplasia/patología
6.
Int J Gen Med ; 15: 437-449, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35046705

RESUMEN

BACKGROUND: Hepatocellular carcinoma has been identified to be among the most prevalent malignancies in the world and has an unfavorable prognosis. Immune checkpoints perform an essential function in many biological processes and are associated with the survival of cancer patients. The function of immune checkpoints remains unknown. METHODS: We used bioinformatic methods to examine the prognostic value of immune checkpoints and the corresponding link to immune infiltration in HCC. qRT-PCR was used to validate the expression of immune checkpoints and their prognostic significance in HCC. RESULTS: The level of mRNA of SIGLEC15, PDCD1LG2, LAG3, PDCD1, CTLA4 as well as PDCD1LG2 was increased in HCC tissues as opposed to liver tissues. Immune checkpoints were shown to participate in the activation of the apoptotic pathway in HCC patients. The elevated expression of PDCD1 and PDCD1LG2 were shown to have a favorable recurrence-free survival (RFS), progression-free survival (PFS), disease-specific survival (DSS), and overall survival (OS). PDCD1, PDCD1LG2, and pT stage were independent variables that affect the HCC patients' prognoses as revealed by the multivariate and univariate analyses. A prediction nomogram indicated that the calibration plots for OS rates over three and five years had a stronger predictive performance in the TCGA HCC cohort in contrast with an ideal model. Positive correlations were observed between the PDCD1 and PDCD1LG2 expression and immune biomarkers, immune cells, chemokine receptors, as well as chemokines. CONCLUSION: The present research performed a thorough examination of the prognostic significance of immune checkpoints in HCC and its correlation with immune infiltration, which suggested that PDCD1 and PDCD1LG2 were prognostic biomarkers in HCC and related to the immune infiltration.

7.
Mol Med Rep ; 24(6)2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34676872

RESUMEN

The tumor suppressor gene adenomatous polyposis coli (APC) is frequently inactivated or absent in colorectal carcinoma (CRC). Loss­of­function of APC promotes the expression of ß­catenin, which is critical for CRC development. Since ß­catenin acts as an important transcription factor, blockage of ß­catenin may have side effects, including impairment of tissue homeostasis and regeneration, thus limiting the application of ß­catenin inhibitors for the treatment of patients with CRC. Therefore, identifying a novel substrate of APC/ß­catenin may provide essential clues to develop effective drugs. Small interfering RNA technology and lentivirus­mediated overexpression were performed for knockdown and overexpression of pleckstrin 2 (PLEK2) in CRC cells. Cell Counting Kit­8 and colony formation assays, and cell cycle analysis and cell apoptosis detection were used to detect the capacity of cell proliferation, cell cycle distribution and apoptosis. The present study demonstrated that the APC/ß­catenin signaling cascade transcriptionally activated PLEK2 in CRC cells. PLEK2 expression was markedly increased in CRC tissues. There was an inverse correlation between APC and PLEK2 expression in patients with CRC. In vitro, overexpression of PLEK2 increased the proliferation of CRC cells. Opposite results were observed in the cells with knockdown of PLEK2. Furthermore, PLEK2 promoted cell cycle progression and suppressed apoptosis. In summary, upregulation of PLEK2 contributed to CRC proliferation and colony formation activated by the APC/ß­catenin signal pathway. Targeting PLEK2 may be important for the treatment of patients with CRC with activation of the APC/ß­catenin signaling pathway.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Proteínas de la Membrana/efectos de los fármacos , beta Catenina/metabolismo , Poliposis Adenomatosa del Colon/genética , Proteína de la Poliposis Adenomatosa del Colon/genética , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Genes APC , Células HCT116 , Humanos , Proteínas de la Membrana/genética , ARN Interferente Pequeño , Regulación hacia Arriba , Vía de Señalización Wnt , beta Catenina/genética
8.
Peptides ; 135: 170422, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33144092

RESUMEN

Nonalcoholic steatohepatitis (NASH) is a global public health challenge. Overwhelmed oxidative stress and impaired autophagy play an important role in the progression of NASH. Chemerin is an adipokine that has attracted much attention in inflammation and metabolic diseases. This study aimed to examine the effects of chemerin in NASH and its association with oxidative stress and autophagy. In this study, chemerin was found to significantly ameliorate high-fat diet (HFD) induced NASH, marked by decreased serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), interleukin (IL)-1ß, IL-6, and tumor necrosis factor-α (TNF-α), decreased insulin resistance (IR) and leptin resistance (LR), and improved liver lesions. Besides, chemerin prevented enhanced oxidative stress in NASH mice by regulating the antioxidant defense system (MDA downregulation and upregulation of superoxide dismutase (SOD)). Moreover, chemerin contributed to the alleviation of NASH through autophagy activation (p62 downregulation, and upregulation of beclin-1 and LC3). Furthermore, these effects were related to increased phosphorylation of JAK2-STAT3 stimulated by chemerin, which could be inhibited by the CMKLR1 specific inhibitor α-NETA. In conclusion, excess chemerin highly probably ameliorated NASH by alleviating oxidative stress and promoting autophagy, the mechanism responsible for this process was related, at least in part, to the increased phosphorylation of JAK2-STAT3 stimulated by chemerin/CMKLR1. Rh-chemerin may represent promising therapeutic targets in the treatment of NASH.


Asunto(s)
Autofagia/genética , Quimiocinas/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Janus Quinasa 2/genética , Enfermedad del Hígado Graso no Alcohólico/genética , Factor de Transcripción STAT3/genética , Animales , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Humanos , Resistencia a la Insulina/genética , Leptina/metabolismo , Hígado/metabolismo , Hígado/patología , Ratones , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Estrés Oxidativo/genética , Transducción de Señal/genética
9.
Am J Transl Res ; 11(7): 4516-4523, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31396355

RESUMEN

Colon cancer is one of the most common malignancies worldwide, while the molecular mechanism remains largely unknown. miR-223-3p plays an important role in cancer development. Here, we found that miR-223-3p was up-regulated in 30 cases of colon cancer tissues as compared with their adjacent normal tissues. Lentivirus-mediated miR-223-3p over-expression promoted the proliferation, colony formation, migration and invasion of colon cancer cells. Inverse results were observed in miR-223-3p knockdown cells. Epithelial-mesenchymal transition (EMT) was regulated by miR-223-3p. In addition, cell apoptosis was suppressed and enhanced by miR-223-3p over-expression and knockdown, respectively. We further identified PRDM1, a tumor suppressor, was the target of miR-223-3p using microarray and luciferase assay. Our findings suggested that miR-223-3p acts as an oncogenic microRNA in colon cancer through regulating EMT and PRDM1.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...