Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Cell Physiol ; 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38742685

RESUMEN

N6-methyladenosine (m6A) methylation has been widely regarded in numerous biological functions including CR. Nonetheless, the molecular process of m6A methylation behind CR in non-small cell lung cancer (NSCLC) has no apparent significance. We identified in this study that the expression of FTO alpha-ketoglutarate dependent dioxygenase (FTO) was downregulated in CR NSCLC tissues and cells in vivo and in vitro. Additionally, RIP-seq indicated that loss of FTO contributed to the elevated m6A methylation at 5'-untranslated region of RNAs which were closely connected with tumor resistance and malignancy, and FTO exerted to exclude the recruitment of eIF3A to these target genes in CR NSCLC. Moreover, FTO-enriched transcripts displayed a reduced translational capability in CR NSCLC compared to the regular NSCLC cells. Finally, we also identified RNA binding motif protein 5 (RBM5) that could specially interact with FTO in regular NSCLC compared to CR NSCLC. Deficiency of RBM5 resulted in the abnormal recognition of transcripts by FTO, and led to the translation silencing of genes associated with CR such as ATP7A, ERCC1, CD99, CDKN3, XRCC5, and NOL3. Taken together, our data characterized FTO as a novel translation regulator and revealed the molecular mechanism on gene translation through the synergistic effects with RBM5 and m6A methylation in CR NSCLC cells.

2.
J Exp Clin Cancer Res ; 43(1): 117, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38641672

RESUMEN

BACKGROUND: Cholangiocarcinoma (CCA) is a highly malignant cancer of the biliary tract with poor prognosis. Further mechanistic insights into the molecular mechanisms of CCA are needed to develop more effective target therapy. METHODS: The expression of the histone lysine acetyltransferase KAT2B in human CCA was analyzed in human CCA tissues. CCA xenograft was developed by inoculation of human CCA cells with or without KAT2B overexpression into SCID mice. Western blotting, ChIP-qPCR, qRT-PCR, protein immunoprecipitation, GST pull-down and RNA-seq were performed to delineate KAT2B mechanisms of action in CCA. RESULTS: We identified KAT2B as a frequently downregulated histone acetyltransferase in human CCA. Downregulation of KAT2B was significantly associated with CCA disease progression and poor prognosis of CCA patients. The reduction of KAT2B expression in human CCA was attributed to gene copy number loss. In experimental systems, we demonstrated that overexpression of KAT2B suppressed CCA cell proliferation and colony formation in vitro and inhibits CCA growth in mice. Mechanistically, forced overexpression of KAT2B enhanced the expression of the tumor suppressor gene NF2, which is independent of its histone acetyltransferase activity. We showed that KAT2B was recruited to the promoter region of the NF2 gene via interaction with the transcription factor SP1, which led to enhanced transcription of the NF2 gene. KAT2B-induced NF2 resulted in subsequent inhibition of YAP activity, as reflected by reduced nuclear accumulation of oncogenic YAP and inhibition of YAP downstream genes. Depletion of NF2 was able to reverse KAT2B-induced reduction of nuclear YAP and subvert KAT2B-induced inhibition of CCA cell growth. CONCLUSIONS: This study provides the first evidence for an important tumor inhibitory effect of KAT2B in CCA through regulation of NF2-YAP signaling and suggests that this signaling cascade may be therapeutically targeted for CCA treatment.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Animales , Humanos , Ratones , Neoplasias de los Conductos Biliares/genética , Conductos Biliares Intrahepáticos/metabolismo , Línea Celular Tumoral , Proliferación Celular , Colangiocarcinoma/patología , Regulación Neoplásica de la Expresión Génica , Genes de la Neurofibromatosis 2 , Histonas/metabolismo , Lisina/metabolismo , Ratones SCID , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/metabolismo
3.
J Exp Clin Cancer Res ; 42(1): 263, 2023 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-37817227

RESUMEN

BACKGROUND: RNA N6-Methyladenosine (m6A) modification is implicated in the progression of human cancers including cholangiocarcinoma (CCA). METTL16 is recently identified as a new RNA methyltransferase responsible for m6A modification, although the role of METTL16 in CCA has not yet been examined. The current study aims to investigate the effect and mechanism of the RNA methyltransferase METTL16 in CCA. METHODS: The expression of METTL16 in CCA was examined by analyzing publicly available datasets or by IHC staining on tumor samples. siRNA or CRISPR/Cas9-mediated loss of function studies were performed in vitro and in vivo to investigate the oncogenic role of METTL16 in CCA. MeRIP-Seq was carried out to identify the downstream target of METTL16. ChIP-qPCR, immunoprecipitation, and immunoblots were used to explore the regulation mechanisms for METTL16 expression in CCA. RESULTS: We observed that the expression of METTL16 was noticeably increased in human CCA tissues. Depletion of METTL16 significantly inhibited CCA cell proliferation and decreased tumor progression. PRDM15 was identified as a key target of METTL16 in CCA cells. Mechanistically, our data showed that METTL16 regulated PRDM15 protein expression via YTHDF1-dependent translation. Accordingly, we observed that restoration of PRDM15 expression could rescue the deficiency of CCA cell proliferation/colony formation induced by METTL16 depletion. Our subsequent analyses revealed that METTL16-PRDM15 signaling regulated the expression of FGFR4 in CCA cells. Specifically, we observed that PRDM15 protein was associated with the FGFR4 promoter to regulate its expression. Furthermore, we showed that the histone acetyltransferase p300 cooperated with the transcription factor YY1 to regulate METTL16 gene expression via histone H3 lysine 27 (H3K27) acetylation in CCA cells. CONCLUSIONS: This study describes a novel METTL16-PRDM15-FGFR4 signaling axis which is crucial for CCA growth and may have important therapeutic implications. We showed that depletion of METTL16 significantly inhibited CCA cell proliferation and decreased tumor progression.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Humanos , Metiltransferasas/genética , Metiltransferasas/metabolismo , Línea Celular Tumoral , Colangiocarcinoma/patología , ARN Interferente Pequeño , Conductos Biliares Intrahepáticos/metabolismo , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Proteínas de Unión al ADN , Factores de Transcripción/genética
4.
J Clin Transl Hepatol ; 11(2): 369-381, 2023 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-36643033

RESUMEN

Background and Aims: Emerging evidence suggests that RNA-binding motif (RBM) proteins are involved in hepatocarcinogenesis and act either as oncogenes or tumor suppressors. The objective of this study was to investigate the role of RBM34, an RBM protein, in hepatocellular carcinoma (HCC). Methods: We first examined the expression of RBM34 across cancers. The correlation of RBM34 with clinicopathological features and the prognostic value of RBM34 for HCC was then investigated. Functional enrichment analysis of RBM34-related differentially expressed genes (DEGs) was performed to explore its biological function. RNA sequencing (RNA-seq) was applied to identify downstream genes and pathways affected upon RBM34 knockout. The correlation of RBM34 with immune characteristics was also analyzed. The oncogenic function of RBM34 was examined in in vitro and in vivo experiments. Results: RBM34 was highly expressed in hepatocellular carcinoma and correlated with poor clinicopathological features and prognosis. RBM34 was positively associated with tumor immune cell infiltration, biomarkers of immune cells, and immune checkpoint expression. A positive correlation was also observed between RBM34, T cell exhaustion, and regulatory T cell marker genes. Knockout of RBM34 significantly inhibited cell proliferation, migration, and xenograft tumor growth, and sensitized HCC cells to sorafenib treatment. RBM34 inhibition reduced FGFR2 expression and affected PI3K-AKT pathway activation in HCC cells. Conclusions: Our study suggests that RBM34 may serve as a new prognostic marker and therapeutic target of HCC.

5.
J Cell Mol Med ; 26(18): 4859-4871, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35989423

RESUMEN

RNA-binding Motif Protein39 (RBM39) is identified as a splicing factor and transcription coactivator. Despite mounting evidence that RBM39 plays a critical role in the development of specific malignancies, no systematic pan-cancer investigation of RBM39 has been conducted. As a result, we set out to investigate RBM39's prognostic significance and putative immunological activities in 33 different cancers. Based on TCGA and CCLE, GTEx, cBioportal and HPA, we used a series of bioinformatics approaches to explore the potential oncogenic role of RBM39, including analysis of the expression of the pan-cancer species RBM39, the prognostic relationship between RBM39 expression and overall survival (OS), disease-specific survival (DSS) and progression-free interval (PFI), the relationship between RBM39 expression and clinical phenotype, analysis of the relationship between RBM39 expression and tumour mutational burden (TMB), microsatellite instability (MSI), DNA methylation and immune cell infiltration. Our results showed that RBM39 is overexpressed in most cancers. RBM39 was positively or negatively correlated with the prognosis of different tumours. RBM39 expression was associated with TMB and MSI in 9 and 12 cancer types. In addition, RBM39 expression was associated with DNA methylation in almost all tumours. There are eight tumours were screened for further study, including BRCA, COAD, HNSC, LIHC, LUSC, SKCM, STAD, UCEC. In the screed tumours, RBM39 was found to be negatively correlated with the infiltration of most immune cells. In addition, the correlation with RBM39 expression varied by immune cell subtype. Based on RBM39's role in tumorigenesis and tumour immunity, we suggest it can serve as a surrogate prognostic marker.


Asunto(s)
Neoplasias , Proteínas de Unión al ARN , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Humanos , Inestabilidad de Microsatélites , Neoplasias/patología , Pronóstico , Motivos de Unión al ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
6.
J Zhejiang Univ Sci B ; 22(9): 774-781, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34514757

RESUMEN

Until now, there has been a lack of standard and effective treatments for patients with recurrent malignant tumors or abdominal and pelvic malignancies with extensive invasion (Morris, 2000). Generally, these patients face problems such as inability to undergo surgery or chemotherapy resistance (Combs et al., 2016). Re-radiotherapy has achieved a prominent place in the treatment of patients who have received radiotherapy previously and developed in-field recurrences (Straube et al., 2018). However, re-radiotherapy is very complicated, requiring comprehensive consideration of appropriate radiation dose, interval from first radiotherapy, boundary of the radiotherapy target area, and damage to surrounding normal tissues (Straube et al., 2019). In other words, it is necessary to focus on the protection of surrounding normal tissues while maximizing the efficacy of re-radiotherapy in such patients.


Asunto(s)
Neoplasias Abdominales/radioterapia , Neoplasias Pélvicas/radioterapia , Animales , Daño del ADN , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Dosificación Radioterapéutica
7.
Med Oncol ; 38(5): 57, 2021 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-33835288

RESUMEN

The small nuclear ribonucleoprotein polypeptides B And B' (SNRPB) is a core component of spliceosome and plays a key role in pre-mRNA splicing. Emerging evidence suggests that it involves in the development of several types of cancer. Our previous study has demonstrated SNRPB is highly expressed in non-small-cell lung cancer (NSCLC) and functions as an oncogene. However, whether SNRPB contributes to cisplatin resistance in NSCLC is still unknown. In this study, we found that SNRPB negatively regulates cisplatin resistance in NSCLC cells. Knocking out of SNRPB could significantly decrease cisplatin-induced cell growth inhibition, cell cycle arrest and apoptosis in H1299 cells. However, enforced expression of SNRPB in H460 cells can markedly promote cisplatin-induced cell growth inhibition, cell cycle arrest and apoptosis. Our results also indicate that overexpression of SNRPB enhances the inhibitory effects of cisplatin on H460 cell-mediated xenograft tumors. Our results suggest that SNRPB may be a prediction marker for NSCLC patients in response to cisplatin-based chemotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Cisplatino/uso terapéutico , Neoplasias Pulmonares/metabolismo , Proteínas Nucleares snRNP/biosíntesis , Animales , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones Endogámicos BALB C , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
8.
Onco Targets Ther ; 13: 10001-10012, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33116585

RESUMEN

PURPOSE: DNA methylation plays an important role in regulating gene expression. Methyl-CpG-binding domain (MBD) proteins recognize and bind to methylated DNA, which mediate gene silencing by the interaction with deacetylases and histone methyltransferases. MBD2 has been reported in various human cancers; however, its clinical implication and potential regulatory role in renal cell carcinoma (RCC) have not been elaborated. MATERIALS AND METHODS: In the study, we estimated the expression and prognostic value of MBD2 in RCC cell lines and tissues by Western blotting and immunohistochemistry. The associations of MBD2 expression and pathological characters and survival in RCC patients were performed using χ2 and Kaplan-Meier survival analysis, respectively. Univariate and multivariable Cox regression analyses suggested the independent predictors in RCC prognosis. The functional role of MBD2 in RCC progression was assessed by in vitro cell experiments. In addition, we identified the MBD2-mediated alterations of protein-related proliferation and EMT markers in RCC cells after MBD2 overexpression and knockdown. RESULTS: We found that the protein levels of MBD2 were upregulated in RCC cells and tissues. High MBD2 expression was related to TNM stage and predicted poorer survival in RCC. Enforced expression of MBD2 significantly promoted the proliferation, cycle progress, invasion and migration of RCC cells in vitro. However, downregulating MBD2 remarkably weakened the above cell functions. Mechanistically, the promotive effect of MBD2 overexpression may be regulated by its effects onp21, p53 and Cyclin D1 expression and EMT process. CONCLUSION: These results indicated that MBD2confers an oncogenic function in the malignant progression of RCC. MBD2 could be served as a meaningful prognostic biomarker and a latent therapeutic target in RCC patients.

9.
Artif Cells Nanomed Biotechnol ; 48(1): 305-314, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31858836

RESUMEN

Combining DNA damage repair inhibitors and chemotherapeutic agents is an emerging strategy in cancer treatment. In this study, we engineered the polycation nanoparticle (NP), which co-encapsulated DNA damage repair inhibitor Dbait and chemotherapeutic drug Docetaxel (Dtxl), using H1 nanopolymer (folate--polyethylenimine600-cyclodextrin), and the size of H1/Dbait/Dtxl was about 117 nm. We demonstrated that H1/Dbait/Dtxl enhanced the efficiency of radio-chemotherapy in prostate cancer cells by CCK-8 assay and colony-forming assay. Importantly, the improvement of radio-chemotherapy of H1/Dbait/Dtxl in prostate cancer was also validated in vivo, and the NP did not have a high toxicity profile. The results of immunohistochemistry and western blot supported that the improved therapeutic efficacy was through inhibiting DNA damage repair signalling pathway. Our study supports further investigations using NP to co-deliver DNA damage repair inhibitors and chemotherapeutics to improve the therapeutic efficacy of cancer.


Asunto(s)
Antineoplásicos , Quimioradioterapia , Docetaxel , Nanopartículas , Neoplasias de la Próstata , Antineoplásicos/química , Antineoplásicos/farmacología , Daño del ADN , Docetaxel/química , Docetaxel/farmacología , Humanos , Masculino , Nanopartículas/química , Nanopartículas/uso terapéutico , Células PC-3 , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia
10.
Cell Death Dis ; 10(9): 667, 2019 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-31511502

RESUMEN

SNRPB is a core component of spliceosome and plays a major role in regulating alternative splicing of the pre-mRNA. However, little is known about its role in cancer to date. In this study, we observe that SNRPB is overexpressed in NSCLC and correlated with poor prognosis in patients with NSCLC. We demonstrate that SNRPB promotes NSCLC tumorigenesis both in vitro and in vivo. Mechanistically, we reveal that RAB26 is a critical target of SNRPB. Suppression of SNRPB leads to retention of intron seven in the RAB26 mRNA and reduced RAB26 mRNA through activation of nonsense-mediated RNA decay (NMD). Moreover, forced expression of RAB26 partially restores the decreased tumorigenicity in NSCLC cells with SNRPB depletion. Our study unveils a novel role of SNRPB in facilitating NSCLC tumorigenesis via regulation of RAB26 expression and proposes that the SNRPB/RAB26 pathway may offer a therapeutic vulnerability in NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Proteínas Nucleares snRNP/metabolismo , Empalme Alternativo/genética , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/secundario , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Humanos , Intrones , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Degradación de ARNm Mediada por Codón sin Sentido/genética , Trasplante Heterólogo , Proteínas de Unión al GTP rab/genética , Proteínas Nucleares snRNP/genética
11.
J Cell Mol Med ; 23(5): 3520-3529, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30821058

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can induce apoptosis in cancer cells while sparing normal cells, thereby leading to the development of TRAIL receptor agonists for cancer treatment. However, these agonist-based therapeutics exhibit little clinical benefits due to the lack of biomarkers to predict whether patients are responsive to the treatment, as well as determine the resistance of cancer cells to TRAIL-based agonists. Our previous study has demonstrated that ISG12a enhances TRAIL-induced apoptosis and might serve as a biomarker to predict the TRAIL response. The downstream mechanism by which ISG12a augments TRAIL-induced apoptosis remains to be elucidated. In this study, we found that ISG12a was localized in the mitochondria and nucleus and augmented TRAIL-induced apoptosis through intrinsic apoptotic pathway. In addition, ISG12a interacted with NR4A1 and promoted its nuclear-to-cytoplasm translocation. Upon translocate to cytoplasm, NR4A1 targeted mitochondria and induced Bcl2 conformational change, thereby exposing its BH3 domain. Moreover, TRAIL treatment can induce NR4A1 expression through the activation of NF-κB in TRAIL-resistant Huh7 hepatoma cells. Knockdown of NR4A1 could overcome TRAIL resistance. However, in TRAIL-sensitive LH86 liver cancer cells, TRAIL activated the Jun N-terminal kinases signalling pathway. Overall, these results showed that both ISG12a and its interaction partner NR4A1 are involved in TRAIL-mediated apoptosis in hepatoma cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Apoptosis/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células HEK293 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Proteínas de la Membrana/genética , Mitocondrias/genética , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
12.
Mol Cancer Ther ; 18(2): 323-334, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30401695

RESUMEN

Renal carcinoma presents a rapid progression in patients with high metastasis with no effective therapeutic strategy. In this study, we designed a folate-grafted PEI600-CyD (H1) nanoparticle-mediated DNA vaccine containing an adjuvant of absent in melanoma 2 (AIM2) and a tumor-specific antigen of carbonic anhydrase IX (CAIX) for renal carcinoma therapy. Mice bearing subcutaneous human CAIX (hCAIX)-Renca tumor were intramuscularly immunized with H1-pAIM2/pCAIX, H1-pCAIX, H1-pAIM2, or Mock vaccine, respectively. The tumor growth of hCAIX-Renca was significantly inhibited in H1-pAIM2/pCAIX vaccine group compared with the control group. The vaccine activated CAIX-specific CD8+ T-cell proliferation and CTL responses, and enhanced the induction of multi-functional CD8+ T cells (expressing TNF-α, IL-2, and IFN-γ). CD8+ T-cell depletion resulted in the loss of anti-tumor activity of H1-pAIM2/pCAIX vaccine, suggesting that the efficacy of the vaccine was dependent on CD8+ T-cell responses. Lung metastasis of renal carcinoma was also suppressed by H1-pAIM2/pCAIX vaccine treatment accompanied with the increased percentages of CAIX-specific multi-functional CD8+ T cells in the spleen, tumor, and bronchoalveolar lavage as compared with H1-pCAIX vaccine. Similarly, the vaccine enhanced CAIX-specific CD8+ T-cell proliferation and CTL responses. Therefore, these results indicated that H1-pAIM2/pCAIX vaccine exhibits the therapeutic efficacy of anti-renal carcinoma by enhancing tumor-specific multi-functional CD8+ T-cell responses. This vaccine strategy could be a potential and promising approach for the therapy of primary solid or metastasis tumors.


Asunto(s)
Antígenos de Neoplasias/administración & dosificación , Linfocitos T CD8-positivos/inmunología , Anhidrasa Carbónica IX/administración & dosificación , Proteínas de Unión al ADN/administración & dosificación , Neoplasias Renales/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Vacunas de ADN/administración & dosificación , Animales , Antígenos de Neoplasias/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/farmacología , Anhidrasa Carbónica IX/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Proteínas de Unión al ADN/farmacología , Humanos , Inyecciones Intramusculares , Neoplasias Renales/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Ratones , Nanopartículas , Vacunas de ADN/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Cell Mol Med ; 22(11): 5670-5681, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30160343

RESUMEN

Renal cell carcinoma (RCC) is a high metastasis tumour with less effective treatment available currently. Absent in melanoma 2 (AIM2) as a tumour suppressor might be used as a potential therapeutic target for RCC treatment. Here, we found that AIM2 expression was significantly decreased in RCC patient specimens and renal carcinoma cell lines (786-O and OSRC-2). To establish a safe and effective AIM2 gene delivery system, we formed the nanoparticles consisting of a folate grafted PEI600-CyD (H1) nanoparticle-mediated AIM2 gene (H1/pAIM2) as an effective delivery agent. Delivery of H1/pAIM2 in renal carcinoma cells could remarkably increase the expression of AIM2, and subsequently decrease cell proliferation, migration, and invasion as well as enhance cell apoptosis. In order to evaluate the therapeutic efficacy of AIM2 in vivo, H1/pAIM2 nanoparticles were injected intratumorally into 786-O-xenograft mice. Administration of H1/pAIM2 nanoparticles could inhibit the tumour growth as evidenced by reduced tumour volume and weight. Furthermore, Blockade of inflammasome activation triggered by H1/pAIM2 nanoparticles using inflammasome inhibitor YVAD-CMK abrogated the anti-tumoral activities of H1/AIM2. These results indicated the therapeutic effect of H1/pAIM2 nanoparticles was mainly attributable to its capability to enhance the inflammasome activation. H1/AIM2 nanoparticles might act as an efficient therapeutic approach for RCC treatment.


Asunto(s)
Carcinoma de Células Renales/terapia , Proteínas de Unión al ADN/genética , Técnicas de Transferencia de Gen , Nanopartículas/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/administración & dosificación , Proteínas de Unión al ADN/química , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Xenoinjertos , Humanos , Inflamasomas/genética , Ratones , Nanopartículas/química , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología
14.
Pancreatology ; 18(3): 328-333, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29525378

RESUMEN

OBJECTIVE: To explore the molecular mechanisms of celecoxib-induced pancreatic cancer suppression in vivo and in vitro. METHODS: The anti-pancreatic cancer activities of celecoxib (0, 20, 60 and 100 µmol/L) were investigated by cell viability and migration of Panc-1 and Bxpc-3 cells in vitro. The expression of L1CAM in pancreatic cancer and adjacent tissues was compared using immunohistochemistry. The expressions of L1CAM, STAT3, p-STAT3, NF-κB, p-NF-κB were determined by western blotting, and cell invasive ability was determined by wound healing assay in L1CAM-silenced and over-expressed Panc-1and Bxpc-3 cells. RESULTS: The expression of L1CAM in pancreatic carcinoma was stronger than that in the adjacent tissues and L1CAM could increase the growth and invasion of pancreatic cancer cells. Over-expression of L1CAM activated the STAT3/NF-κB signaling pathway in Panc-1 and Bxpc-3 pancreatic cancer cells and celecoxib inhibited their viability and the expressions of STAT3, p-STAT3, NF-κB, p-NF-κB as well as full length L1CAM in a concentration dependent manner. CONCLUSIONS: L1CAM was highly expressed in pancreatic cancer tissue and positively correlated with age, TNM staging and tumor differentiation. L1CAM activated the STAT/NF-κB signaling pathway and celecoxib could inhibit the activity of L1CAM, STAT3 and the NF-κB signaling pathway resulting in decreased growth and invasion of pancreatic cancer cells.


Asunto(s)
Antígeno CD56/antagonistas & inhibidores , Celecoxib/farmacología , Proliferación Celular/efectos de los fármacos , Inhibidores de la Ciclooxigenasa 2/farmacología , Metástasis de la Neoplasia/prevención & control , Neoplasias Pancreáticas/prevención & control , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción ReIA/antagonistas & inhibidores , Antígeno CD56/genética , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , Inmunohistoquímica , Invasividad Neoplásica/genética , Neoplasias Pancreáticas/patología , Plásmidos/genética , Factor de Transcripción STAT3/genética , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/genética , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/genética
15.
Nanomedicine ; 13(4): 1309-1321, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28185938

RESUMEN

Androgen receptor (AR) plays a crucial role in the development and progression of prostate cancer (PCa). PCa patients typically receive androgen deprivation therapy; nonetheless, these patients eventually develop castration and radiation resistance. We hypothesized that we could further improve radiotherapeutic efficacy of hormone-independent PCa (HIPC) by silencing AR. In this study, nanoparticle (NP) AR-shRNA was formulated using folate-targeted H1 nanopolymer. We demonstrated that NP AR-shRNA enhances PCa radiosensitivity as indicated by the inhibition of cell growth, increased apoptosis, and increased cell cycle arrest in AR-dependent HIPC in vitro. The radiosensitizing effect of NP AR-shRNA could be validated in vivo, as NP AR-shRNA significantly suppressed tumor growth and prolonged the survival of HIPC tumor-bearing mice. Analysis at the molecular level revealed that NP AR-shRNA inhibits DNA damage repair signaling pathways. Our study supports further investigation of NP AR-shRNA for the improvement of radiotherapy efficacy in HIPC.


Asunto(s)
Antagonistas de Receptores Androgénicos/uso terapéutico , Ácido Fólico/química , Nanopartículas/química , Neoplasias de la Próstata/radioterapia , ARN Interferente Pequeño/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones Desnudos , Tolerancia a Radiación , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Biosci ; 42(3): 491-499, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29358562

RESUMEN

Hypoxia plays a key role in tumour cell survival, invasion, and metastasis. An increasing number of studies have attempted to characterize the tumour response to hypoxia and to identify predictive markers of disease. Here we show that hypoxia increases tumour cell invasion and migration by the modulation of Rab11, an important molecule for vesicular trafficking. In our study, we found that Rab11, together with the activation of Rac1, could stimulate invasion and migration of cervical cancer cell lines HeLa/SiHa in hypoxia. Activation of Rac1 activity by hypoxia seems to be central to carcinoma invasion. We also found that these effects could be related to the integrin αvß3. In addition, we studied the molecular pathway for this process. Our results showed that in cervical cancer cell lines HeLa/SiHa, Rac1 activation in hypoxia could stimulate invasion and migration, and this process was mediated by integrin αvß3-mediated FAK and PI3K phosphorylation. Furthermore, hypoxia induced a dramatic increase in αvß3 integrin surface expression, and this increase is dependent on Rab11. In conclusion, our study might provide a new mechanism for the effect of hypoxia on stimulating cervical carcinoma invasion.


Asunto(s)
Quinasa 1 de Adhesión Focal/genética , Regulación Neoplásica de la Expresión Génica , Integrina alfaVbeta3/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas de Unión al GTP rab/genética , Proteína de Unión al GTP rac1/genética , Hipoxia de la Célula , Línea Celular Tumoral , Movimiento Celular , Femenino , Quinasa 1 de Adhesión Focal/metabolismo , Células HeLa , Humanos , Integrina alfaVbeta3/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Transporte de Proteínas , Transducción de Señal , Proteínas de Unión al GTP rab/metabolismo , Proteína de Unión al GTP rac1/metabolismo
17.
J Virol ; 90(15): 6832-45, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27194766

RESUMEN

UNLABELLED: Interferons (IFNs) restrict various kinds of viral infection via induction of hundreds of IFN-stimulated genes (ISGs), while the functions of the majority of ISGs are broadly unclear. Here, we show that a high-IFN-inducible gene, ISG12a (also known as IFI27), exhibits a nonapoptotic antiviral effect on hepatitis C virus (HCV) infection. Viral NS5A protein is targeted specifically by ISG12a, which mediates NS5A degradation via a ubiquitination-dependent proteasomal pathway. K374R mutation in NS5A domain III abrogates ISG12a-induced ubiquitination and degradation of NS5A. S-phase kinase-associated protein 2 (SKP2) is identified as an ubiquitin E3 ligase for NS5A. ISG12a functions as a crucial adaptor that promotes SKP2 to interact with and degrade viral protein. Moreover, the antiviral effect of ISG12a is dependent on the E3 ligase activity of SKP2. These findings uncover an intriguing mechanism by which ISG12a restricts viral infection and provide clues for understanding the actions of innate immunity. IMPORTANCE: Upon virus invasion, IFNs induce numerous ISGs to control viral spread, while the functions of the majority of ISGs are broadly unclear. The present study shows a novel antiviral mechanism of ISGs and elucidated that ISG12a recruits an E3 ligase, SKP2, for ubiquitination and degradation of viral protein and restricts viral infection. These findings provide important insights into exploring the working principles of innate immunity.


Asunto(s)
Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Hepatitis C/prevención & control , Proteínas de la Membrana/farmacología , Transducción de Señal/efectos de los fármacos , Ubiquitina/metabolismo , Proteínas no Estructurales Virales/metabolismo , Células Cultivadas , Hepatitis C/metabolismo , Hepatitis C/virología , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/virología , Humanos , Proteolisis , Ubiquitinación
18.
Cancer Lett ; 379(2): 213-9, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27108065

RESUMEN

Anti-angiogenesis-related therapies have become the standard care for patients with advanced hepatocellular carcinoma (HCC), as HCC is a highly vascularized solid tumor. Unfortunately, only modest and limited efficacies are observed. Emerging evidence have attributed to the limited efficacy to the presence of cancer stem cells (CSCs) in the tumor. CSCs predominantly drives angiogenesis via releasing proangiogenic factors and exosomes. They have the ability to resistant intratumoral hypoxia via autophagy or by directly forming the tubular structure to obtain blood. On the other hand, the vascular niche in tumor microenvironment also releases growth factors via juxtacrine and paracrine mechanisms to support the growth of CSCs and maintain its stemness features. This positive feedback loop between angiogenesis and CSCs exists in liver tumor microenvironment that is responsible for the development and poor prognosis of HCC. In this review, we summarize recent advances in our understanding of the crosstalks between angiogenesis and CSCs, and their interactions in liver tumor microenvironment and their purpose that an effective anti-angiogenic therapy should also target CSCs for HCC treatment.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Células Madre Neoplásicas/metabolismo , Neovascularización Patológica , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Retroalimentación Fisiológica , Humanos , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Transducción de Señal , Microambiente Tumoral
19.
Mol Med Rep ; 13(2): 1788-94, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26718033

RESUMEN

Mesenchymal stem cell (MSC)-like cells have been isolated from various types of tumor. It has previously been reported that MSCs are involved in tumorigenesis and its prognosis. The aim of the present study was to isolate and compare MSC-like cells from human gastric cancer (GC) and its metastatic deposits in ovarian tissue. MSC-like cells were isolated from human gastric cancer (hGC-MSCs) and the corresponding ovarian metastatic tissues (hGCOM-MSCs) from 40 patients. The characteristics of hGC-MSCs and hGCOM-MSCs, including their morphology, surface antigens, specific gene expression and differentiation potential, were similar to those of MSCs derived from human bone marrow (hBM-MSCs) but different from GC cells. In conclusion, the present study demonstrated that MSC-like cells could be isolated from GC tissue and its ovarian metastatic tissues. The existence of MSC-like cells in GC tissues and its ovarian metastatic tissues suggests that they may be a potential target for cancer therapy, and provides an experimental foundation for investigating their role in the initiation and progression of ovarian metastasis of GC.


Asunto(s)
Separación Celular/métodos , Células Madre Mesenquimatosas/citología , Neoplasias Ováricas/secundario , Neoplasias Gástricas/patología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Biomarcadores de Tumor/metabolismo , Ciclo Celular , Diferenciación Celular , Proliferación Celular , Forma de la Célula , Femenino , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunofenotipificación , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Osteogénesis/genética , Neoplasias Ováricas/genética , Neoplasias Gástricas/genética
20.
Exp Cell Res ; 333(2): 316-326, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25724899

RESUMEN

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide. Interferon-alpha (IFN-α) has recently been recognized to harbor therapeutic potential in the prevention and treatment of HCC, but it remains controversial as to whether IFN-α exerts direct cytotoxicity against HCC. Cyclooxygenase-2 (COX-2) is overexpressed in HCC and is considered to play a role in hepatocarcinogenesis. Therefore, we aimed to elucidate the combined effect of a COX-2 inhibitor, celecoxib, and IFN-α on in vitro growth suppression of HCC using the hepatoma cell line HLCZ01 and the in vivo nude mouse xenotransplantation model using HLCZ01 cells. Treatment with celecoxib and IFN-α synergistically inhibited cell proliferation in a dose- and time-dependent manner. Apoptosis was identified by 4׳,6-diamidino-2-phenylindole dihydrochloride and fluorescent staining. IFN-α upregulated the expression of TRAIL, while celecoxib increased the expression of TRAIL receptors. The combined regimen with celecoxib and IFN-α reduced the growth of xenotransplanted HCCs in nude mice. The regulation of IFN-α- and COX-2 inhibitor-induced cell death is impaired in a subset of TRAIL-resistant cells. The molecular mechanisms of HCC cells resistant to TRAIL-induced apoptosis were explored using molecular biological and immunological methods. Interferon-α and the COX-2 inhibitor celecoxib synergistically increased TRAIL-induced apoptosis in hepatocellular carcinoma. These data suggest that IFN-α and celecoxib may offer a novel role with important implications in designing new therapeutics for TRAIL-resistant tumors.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Interferón-alfa/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Pirazoles/farmacología , Sulfonamidas/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Animales , Carcinoma Hepatocelular/patología , Caspasas/metabolismo , Celecoxib , Línea Celular Tumoral , Inhibidores de la Ciclooxigenasa 2/farmacología , Sinergismo Farmacológico , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...