Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
HPB (Oxford) ; 2024 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-39054211

RESUMO

BACKGROUND: To evaluate long-term quality of life and survival in pancreatic ductal adenocarcinoma (PDAC) patients after pancreatoduodenectomy with extended lymphadenectomy (PDEL) and identify candidates. METHODS: Patients with resectable PDAC with ≥1 examined lymph node (LN) during pancreatoduodenectomy (PD), and were divided into the PD with standard lymphadenectomy (PDSL) and PDEL groups. Perioperative data, long-term quality of life and survival were compared, and the prognostic effect of LNs ± in every peripancreatic station were analysed. RESULTS: Screening 446 PDAC patients, 237 and 126 were included in the PDSL and PDEL groups, respectively. The PDEL group showed a longer operation time, greater intraoperative blood loss, severe diarrhoea, a higher incidence of grade III complications. Notably, the PDEL patients experienced significant relief from low back pain and diarrhoea, with an obvious survival advantage (p = 0.037), especially in patients with preoperative tumor contact with vascular and pathological N0; however, LNs+ in any station (No. 8p, 12, 14, or 16) were associated with a poorer prognosis. The vascular reconstruction, T and N stage were independent risk factors for survival. CONCLUSION: PDEL can relieve symptoms and prolong the survival of PDAC patients with acceptable complications, and EL should be performed regardless of preoperative LN enlargement.

2.
Food Chem ; 444: 138667, 2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-38335686

RESUMO

Iron-copper nanozymes (Fe-Cu NZs) with good peroxidase activity were prepared through hydrothermal method by using copper nitrate as copper source, iron acetate as iron source and 2, 5-dihydroxyterephthalic acid as organic ligand. Upon oxidation of the colourless TMB to light blue products by Fe-Cu NZs, the addition of Norfloxacin (NOR) resulted in a colour change to dark blue. The absorbance of the system correlated linearly with NOR concentration in the range of 3.3 µM to 66 µM, and the detection limit (LOD) was 0.386 µM. A rapid colourimetric assay for the determination of NOR in food matrices was developed, with a detection time of only one minute. Additionally, the assay facilitated the simultaneous catalytic degradation of NOR via Fe-Cu NZs. The primary degradation mechanism of NOR was identified as the transformation of the quinolone ring and the cleavage of the C9 = C10 double bond, which was substantiated by high-performance liquid chromatography (HPLC).


Assuntos
Norfloxacino , Quinolonas , Ferro/química , Cobre/química , Antioxidantes , Colorimetria/métodos , Peróxido de Hidrogênio
3.
Drug Resist Updat ; 73: 101027, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38290407

RESUMO

AIMS: Pancreatic cancer (PC) is a highly metastatic malignant tumor of the digestive system. Drug resistance frequently occurs during cancer treatment process. This study aimed to explore the link between chemoresistance and tumor metastasis in PC and its possible molecular and cellular mechanisms. METHODS: A Metastasis and Chemoresistance Signature (MCS) scoring system was built and validated based on metastasis- and chemoresistance-related genes using gene expression data of PC, and the model was applied to single-cell RNA sequencing data. The influence of linker histone H1.2 (H1-2) on PC was explored through in vitro and in vivo experiments including proliferation, invasion, migration, drug sensitivity, rescue experiments and immunohistochemistry, emphasizing its regulation with c-MYC signaling pathway. RESULTS: A novel MCS scoring system accurately predicted PC patient survival and was linked to chemoresistance and epithelial-mesenchymal transition (EMT) in PC single-cell RNA sequencing data. H1-2 emerged as a significant prognostic factor, with its high expression indicating increased chemoresistance and EMT. This upregulation was mediated by c-MYC, which was also found to be highly expressed in PC tissues. CONCLUSION: The MCS scoring system offers insights into PC chemoresistance and metastasis potential. Targeting H1-2 could enhance therapeutic strategies and improve PC patient outcomes.


Assuntos
Histonas , Neoplasias Pancreáticas , Humanos , Histonas/genética , Histonas/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Proto-Oncogênicas c-myc/uso terapêutico , Linhagem Celular Tumoral , Transdução de Sinais , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Proliferação de Células , Regulação Neoplásica da Expressão Gênica
5.
Front Oncol ; 13: 1138238, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37182151

RESUMO

Background: Many studies have reported that N6-methyladenosine (m6A) modification plays a critical role in the epigenetic regulation of organisms and especially in the pathogenesis of malignant diseases. However, m6A research has mainly focused on methyltransferase activity mediated by METTL3, and few studies have focused on METTL16. The aim of this study was to investigate the mechanism of METTL16, which mediates m6A modification, and its role in pancreatic adenocarcinoma (PDAC) cell proliferation. Methods: Clinicopathologic and survival data were retrospectively collected from 175 PDAC patients from multiple clinical centers to detect the expression of METTL16. CCK-8, cell cycle, EdU and xenograft mouse model experiments were used to evaluate the proliferation effect of METTL16. Potential downstream pathways and mechanisms were explored via RNA sequencing, m6A sequencing, and bioinformatic analyses. Regulatory mechanisms were studied through methyltransferase inhibition, RIP, MeRIP‒qPCR assays. Results: We found that METTL16 expression was markedly downregulated in PDAC, and multivariate Cox regression analyses revealed that METTL16 was a protective factor for PDAC patients. We also demonstrated that METTL16 overexpression inhibited PDAC cell proliferation. Furthermore, we identified a METTL16-p21 signaling axis, with downregulation of METTL16 resulting in inhibition of CDKN1A (p21). Additionally, METTL16 silencing and overexpression experiments highlighted m6A modification alterations in PDAC. Conclusions: METTL16 plays a tumor-suppressive role and suppresses PDAC cell proliferation through the p21 pathway by mediating m6A modification. METTL16 may be a novel marker of PDAC carcinogenesis and target for the treatment of PDAC.

6.
Oncologist ; 28(11): e1031-e1042, 2023 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-37159555

RESUMO

AIM: Tumor metabolism plays an important role in tumorigenesis and tumor progression. This study evaluated the potential association of tumor cell metabolism and immune cell tumor infiltration with the clinical course of hepatocellular carcinoma (HCC). METHODS: Gene-wise normalization and principal component analysis were performed to evaluate the metabolic system. A tumor microenvironment score system of tumor immune cell infiltration was constructed to evaluate its association with metabolic subtypes. Finally, we analyzed the impact of metabolism and immune cell infiltration on the clinical course of HCC. RESULTS: A total of 673 HCC patients were categorized into cholesterogenic (25.3%), glycolytic (14.6%), mixed (10.4%), and quiescent (49.8%) types based on glycolysis and cholesterol biosynthesis gene expression. The subgroups including the glycolytic genotyping expression (glycolytic and mixed types) showed a higher mortality rate. The glycolytic, cholesterogenic, and mixed types were positively correlated with M0 macrophage, resting mast cell, and naïve B-cell infiltration (P = .013, P = .019, and P = .006, respectively). In TCGA database, high CD8+ T cell and low M0 macrophage infiltration were associated with prolonged overall survival (OS, P = .0017 and P < .0001, respectively). Furthermore, in glycolytic and mixed types, patients with high M0 macrophage infiltration had a shorter OS (P = .03 and P = .013, respectively), and in quiescent type, patients with low naïve B-cell infiltration had a longer OS (P = .007). CONCLUSIONS: Tumor metabolism plays a prognostic role and correlates with immune cell infiltration in HCC. M0 macrophage and CD8+ T cell appear to be promising prognostic biomarker for HCC. Finally, M0 macrophages may represent a useful immunotherapeutic target in patients with HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Linfócitos T CD8-Positivos , Imunidade , Progressão da Doença , Microambiente Tumoral
7.
Cancer Med ; 12(10): 11651-11671, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37041721

RESUMO

BACKGROUND: Growing evidences suggest that circular RNAs (circRNAs) are important factors in cancer progression. Nevertheless, the role of circRNAs in the progression of pancreatic ductal adenocarcinoma (PDAC) remains unclear. METHODS: CircPTPRA was identified based on our previous circRNA array data analysis. Wound healing, transwell, and EdU assays were performed to investigate the effect of circPTPRA on the migration, invasion, and proliferation of PDAC cells in vitro. RNA pull-down, fluorescence in situ hybridization (FISH), RNA immunoprecipitation (RIP), and dual-luciferase reporter assays were conducted to verify the binding of circPTPRA with miR-140-5p. Subcutaneous xenograft model was constructed for in vivo experiment. RESULTS: CircPTPRA was significantly upregulated in PDAC tissues and cells compared to normal controls. Moreover, circPTPRA overexpression was positively correlated with lymph node invasion and worse prognosis in PDAC patients. In addition, overexpression of circPTPRA promoted PDAC migration, invasion, proliferation, and epithelial-mesenchymal transition (EMT) in vitro and in vivo. Mechanistically, circPTPRA upregulates LaminB1 (LMNB1) expression by sponging miR-140-5p and ultimately promotes the progression of PDAC. CONCLUSIONS: This study revealed that circPTPRA plays an important role in the progression of PDAC by sponging miR-140-5p. It can be explored as a potential prognostic marker and therapeutic target for PDAC.


Assuntos
Carcinoma Ductal Pancreático , MicroRNAs , Neoplasias Pancreáticas , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Circular/genética , RNA Circular/metabolismo , Hibridização in Situ Fluorescente , Neoplasias Pancreáticas/patologia , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Movimento Celular/genética , Neoplasias Pancreáticas
8.
Cancer Cell Int ; 22(1): 415, 2022 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-36539807

RESUMO

BACKGROUND: MicroRNAs (miRNAs), as an indispensable type of non-coding RNA (ncRNA), participate in diverse biological processes. However, the specific regulatory mechanism of certain miRNAs in pancreatic ductal adenocarcinoma (PDAC) remains unclear. METHODS: The expression of miR-194-5p in PDAC tissue microarray and cell lines were detected by RNA-scope and real-time quantitative PCR (RT-qPCR). The function of proliferation and migration carried by miR-194-5p in vitro and vivo was observed by several functional experiments. Informatics methods and RNA sequencing data were applied to explore the target of miR-194-5p and the upstream circular RNA (circRNA) of miR-194-5p. RNA-binding protein immunoprecipitation (RIP) assay and dual-luciferase reporter assay confirmed the relationships between miR-194-5p and SOCS2 or miR-194-5p and circPVRL3. The proliferation and migration abilities of SOCS2 and circPVRL3 were accessed by rescue experiments. RESULTS: In this study, we aimed to clarify the molecular mechanisms of miR-194-5p, which has critical roles during PDAC progression. We found that the expression of miR-194-5p was significantly upregulated in PDAC tissue compared to tumor-adjacent tissue and was highly related to age and nerve invasion according to RNAscope and RT‒qPCR. Overexpression of miR-194-5p accelerated the cell cycle and enhanced the proliferation and migration processes according to several functional experiments in vitro and in vivo. Specifically, circPVRL3, miR-194-5p, and SOCS2 were confirmed to work as competing endogenous RNAs (ceRNAs) according to informatics methods, RIP, and dual-luciferase reporter assays. Additionally, the rescue experiments confirmed the relationship among miR-194-5p, circPVRL3, and SOCS2 mRNA. Finally, the circPVRL3/miR-194-5p/SOCS2 axis activates the PI3K/AKT signaling pathway to regulate the proliferation and metastasis of PDAC. CONCLUSION: Our findings indicated that an increase of miR-194-5p caused by circPVRL3 downregulation stimulates the PI3K/AKT signaling pathway to promote PDAC progression via the circPVRL3/miR-194-5p/SOCS2 axis, which suggests that the circPVRL3/miR-194-5p/SOCS2 axis may be a potential therapeutic target for PDAC patients.

9.
Med Sci Monit ; 28: e938443, 2022 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-36404606

RESUMO

BACKGROUND Pancreaticoduodenectomy combined with revascularization (PDR) is the main surgical procedure for resectable pancreatic ductal adenocarcinoma (PDAC) with venous system invasion, but this procedure is discouraged in elderly patients because of physical complexity. Our aim was to explore the differences of perioperative and survival in patients of different ages who underwent PDR. MATERIAL AND METHODS We reviewed data from PDAC patients undergoing PDR from 2007 to 2018. Patients were subdivided into 3 groups according to age: <60 years, 60-70 years, and ≥70 years. Postoperative complications and long-term survival were compared among the 3 groups. RESULTS From 626 patients, 185 had en bloc venous resection who underwent PDR (103, 55, and 27 patients from young to elderly). Increasing age was linked to a higher prevalence of ICU management (P=0.035) and more serious complications (grade ≥III, P=0.043); overall mortality was 8.1% and did not significantly differ among age-matched groups. Further, there was no difference in overall survival (OS) or progression-free survival (PFS) based on age (<60, 60-70, ≥70, median OS were 9.7, 8.4 vs 9.1 months, respectively, P=0.787; median PFS were 6.9, 6.1 vs 8.4 months, respectively, P=0.603). However, patients <60 years whose tumors invaded the superior mesenteric vascular had better survival outcomes when compared with the other 2 groups (11.5 vs 8.4, 9.1 months, P=0.049). CONCLUSIONS The results show that age should not be considered an absolute contraindication for PDR, as elderly patients can achieve the same surgical efficacy and long-term survival prognosis.


Assuntos
Neoplasias Pancreáticas , Pancreaticoduodenectomia , Humanos , Idoso , Pessoa de Meia-Idade , Pancreaticoduodenectomia/métodos , Veia Porta/patologia , Pancreatectomia , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas
10.
Mol Cancer ; 21(1): 112, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35538494

RESUMO

BACKGROUND: Although gemcitabine has been considered as the first-line drug for advanced pancreatic cancer (PC), development of resistance to gemcitabine severely limits the effectiveness of this chemotherapy, and the underlying mechanism of gemcitabine resistance remains unclear. Various factors, such as ATP binding cassette (ABC) transporters, microRNAs and their downstream signaling pathways are included in chemoresistance to gemcitabine. This study investigated the potential mechanisms of microRNAs and ABC transporters related signaling pathways for PC resistance to gemcitabine both in vivo and in vitro. METHODS: Immunohistochemistry and Western blotting were applied to detect the expression of ABC transporters. Molecular docking analysis was performed to explore whether gemcitabine interacted with ABC transporters. Gain-of-function and loss-of-function analyses were performed to investigate the functions of hsa-miR-3178 in vitro and in vivo. Bioinformatics analysis, Western blotting and dual-luciferase reporter assay were used to confirm the downstream regulatory mechanisms of hsa-miR-3178. RESULTS: We found that P-gp, BCRP and MRP1 were highly expressed in gemcitabine-resistant PC tissues and cells. Molecular docking analysis revealed that gemcitabine can bind to the ABC transporters. Hsa-miR-3178 was upregulated in gemcitabine resistance PANC-1 cells as compared to its parental PANC-1 cells. Moreover, we found that hsa-miR-3178 promoted gemcitabine resistance in PC cells. These results were also verified by animal experiments. RhoB was down-regulated in gemcitabine-resistant PC cells and it was a downstream target of hsa-miR-3178. Kaplan-Meier survival curve showed that lower RhoB expression was significantly associated with poor overall survival in PC patients. Rescue assays demonstrated that RhoB could reverse hsa-miR-3178-mediated gemcitabine resistance. Interestingly, hsa-miR-3178 promoted gemcitabine resistance in PC by activating the PI3K/Akt pathway-mediated upregulation of ABC transporters. CONCLUSIONS: Our results indicate that hsa-miR-3178 promotes gemcitabine resistance via RhoB/PI3K/Akt signaling pathway-mediated upregulation of ABC transporters. These findings suggest that hsa-miR-3178 could be a novel therapeutic target for overcoming gemcitabine resistance in PC.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Desoxicitidina , MicroRNAs , Neoplasias Pancreáticas , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Proteína rhoB de Ligação ao GTP , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Linhagem Celular Tumoral , Desoxicitidina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Simulação de Acoplamento Molecular , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteína rhoB de Ligação ao GTP/metabolismo , Gencitabina , Neoplasias Pancreáticas
11.
Am J Transl Res ; 14(2): 788-797, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35273685

RESUMO

OBJECTIVE: To illustrate the role of LRIG1 in regulating the Notch signaling pathway and glioma cell proliferation, apoptosis and invasion. METHODS: The glioma cells (U373) were divided into control group, NC group and LRIG1 group. After transfection, the CCK-8 assay, Transwell assay, and Flow cytometry were used to explore the biological function of LRIG1 in glioma cells. At the end, Western blot was used to detect the expression of LRIG1, Notch1, Hes1, Bcl-2, and Bax. RESULTS: The LRIG1 expression in U373 cells was remarkably lower than that in normal glial cells (P=0.019). The LRIG1 expression in the LRIG1 group was successfully increased when compared with that in the control group (P=0.004). The cell viability of the LRIG1 group was significantly lower than that of the NC group and control group at 24 h, 48 h, and 72 h (P=0.040, 0.025; P=0.041, 0.041; P=0.035, 0.035) respectively. Increased LRIG1 expression level in glioma cells strongly inhibits cell migration in transwell experiment. Flow cytometry results indicated that the apoptosis rate of the LRIG1 group was critically higher than that of the NC group and control group (P=0.003; P=0.003). According to results of Western blot, the expression levels of Notch1, Hes1, Hes5, and Jagged1 in LRIG1 group were dramatically higher than that in NC group and control group (P=0.006, 0.013; P=0.025, 0.026; P=0.001, 0.004; P=0.025, 0.027; P=0.029, 0.021) reespectively. While Bax expression in LRIG1 group was lower than that of NC group and control group (P=0.018, 0.021). CONCLUSION: The up-regulation of LRIG1 can inhibit the proliferation and migration of glioma cells and promote apoptosis by regulating the Notch signaling pathway.

12.
Front Oncol ; 11: 628353, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33816264

RESUMO

Transmembrane protease serine 4 (TMPRSS4) is upregulated in various kinds of human cancers, including pancreatic cancer. However, its biological function in pancreatic ductal adenocarcinoma (PDAC) remains unclear. In the current study, real-time qPCR, immunohistochemical staining, Western blotting, and database (Cancer Genome Atlas and Gene Expression) analysis revealed remarkable overexpression of TMPRSS4 in PDAC tissue as compared to non-tumor tissue. The TMPRSS4 overexpression was associated with poor prognosis of PDAC patients. Moreover, multivariate analysis revealed that TMPRSS4 serves as an independent risk factor in PDAC. We performed gain-and loss-of-function analysis and found that TMPRSS4 promotes cellular proliferation and inhibits apoptosis of PDAC cells both in vitro and in vivo. Furthermore, we showed that TMPRSS4 might promote cell proliferation and inhibit apoptosis through activating ERK1/2 signaling pathway in pancreatic cancer cells. These findings were validated by using ERK1/2 phosphorylation inhibitor SCH772984 both in vitro and in vivo. Taken together, this study suggests that TMPRSS4 is a proto-oncogene, which promotes initiation and progression of PDAC by controlling cell proliferation and apoptosis. Our findings indicate that TMPRSS4 could be a promising prognostic biomarker and a therapeutic target for the treatment of pancreatic cancer.

13.
Aging (Albany NY) ; 12(22): 22840-22858, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33197892

RESUMO

Pancreatic cancer (PC) is a severe disease with the highest mortality rate among various cancers. It is urgent to find an effective and accurate way to predict the survival of PC patients. Gene set variation analysis (GSVA) was used to establish and validate a miRNA set-based pathway prognostic signature for PC (miPPSPC) and a mRNA set-based pathway prognostic signature for PC (mPPSPC) in independent datasets. An optimized miPPSPC was constructed by combining clinical parameters. The miPPSPC, optimized miPPSPC and mPPSPC were established and validated to predict the survival of PC patients and showed excellent predictive ability. Four metabolic pathways and one oxidative stress pathway were identified in the miPPSPC, whereas linoleic acid metabolism and the pentose phosphate pathway were identified in the mPPSPC. Key factors of the pentose phosphate pathway and linoleic acid metabolism, G6PD and CYP2C8/9/18/19, respectively, are related to the survival of PC patients according to our tissue microarray. Thus, the miPPSPC, optimized miPPSPC and mPPSPC can predict the survival of PC patients efficiently and precisely. The metabolic and oxidative stress pathways may participate in PC progression.


Assuntos
Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/genética , RNA Mensageiro/genética , Biomarcadores Tumorais/genética , Perfilação da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Prognóstico , Curva ROC
14.
Genomics ; 112(6): 5343-5355, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33189780

RESUMO

The extremely high proliferation rate of tumor cells contributes to pancreatic cancer (PC) progression. Runt-related transcription factor 1(RUNX1), a key factor in hematopoiesis that was correlated with tumor progression. However, the role of RUNX1 in PC proliferation was still unclear. We found that RUNX1 was significantly upregulated in PC tissues and its expression was negatively associated with prognosis of PC patients in a multicenter analysis according to immunohistochemical (IHC). RUNX1 downregulation in PC resulted in a significantly reduced cell proliferation rate, which was consistent with in vivo subcutaneous tumor formation assay results. RNA-seq and ChIP-seq results revealed that a portion of target genes, including HAP1, GPRC5B, PTPN21, VHL and EN2, were regulated by RUNX1, a finding successfully validated by ChIP-qPCR, qRT-PCR and Western blot. Subsequently, IHC and proliferation assays showed these target genes to be dysregulated in PC, affecting tumor growth. Our data suggest that RUNX1 plays an oncogenic role in tumor proliferation and is a potential prognostic biomarker and therapeutic target for PC.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Regulação Neoplásica da Expressão Gênica , Neoplasias Pancreáticas/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células , Sequenciamento de Cromatina por Imunoprecipitação , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Progressão da Doença , Feminino , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Prognóstico , RNA-Seq , Transcriptoma
15.
Cell Death Dis ; 11(6): 412, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32487998

RESUMO

Numerous long noncoding RNAs (lncRNAs) are aberrantly expressed in pancreatic cancer (PC); however, their functions and mechanisms in cancer progression are largely unknown. In this study, we identified a novel PC-associated lncRNA, RUNX1-IT1, that was significantly upregulated in PC patient samples from multiple centers and associated with poor prognosis. In vitro and in vivo, alterations in RUNX1-IT1 expression markedly affected PC proliferation, migration and invasion. RUNX1-IT1 contributed to the progression of PC by interacting with the adjacent gene RUNX1. Rescue experiments showed that RUNX1 reduced the cancer-promoting effect of RUNX1-IT1. RNA-seq analysis after silencing RUNX1-IT1 and RUNX1 highlighted alterations in the common target C-FOS. Mechanistically, we demonstrated that RUNX1-IT1 was a trans-acting factor that participated in the proliferation, migration and invasion of PC by recruiting RUNX1 to the C-FOS gene promoter. Furthermore, RUNX1-IT1 enhanced the transcription of the RUNX1 gene, indicating its potential as a cis-regulatory RNA involved in the upstream regulation of RUNX1. Overall, RUNX1-IT1 is a crucial oncogenic lncRNA that activates C-FOS expression by regulating and recruiting RUNX1 and is a potential prognostic biomarker and therapeutic target for PC.


Assuntos
Movimento Celular/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas c-fos/genética , RNA Longo não Codificante/metabolismo , Transcrição Gênica , Animais , Sequência de Bases , Linhagem Celular Tumoral , Proliferação de Células/genética , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica , Prognóstico , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Longo não Codificante/genética , Regulação para Cima/genética
16.
Mol Ther Nucleic Acids ; 19: 228-239, 2020 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-31846800

RESUMO

Treatment of pancreatic ductal adenocarcinoma (PDAC) remains a clinical challenge. There is an urgent need to develop novel strategies to enhance survival and improve patient prognosis. MicroRNAs (miRNAs) play critical roles as oncogenes or tumor suppressors in the regulation of cancer development and progression. In this study, we demonstrate that low expression of miR-15a is associated with poor prognosis of PDAC patients. miR-15a expression is reduced in PDAC while closely related miR-16 expression remains relatively unchanged. miR-15a suppresses several important targets such as Wee1, Chk1, Yap-1, and BMI-1, causing cell cycle arrest and inhibiting cell proliferation. Ectopic expression of miR-15a sensitizes PDAC cells to gemcitabine reducing the half maximal inhibitory concentration (IC50) more than 6.5-fold. To investigate the therapeutic potential of miR-15a, we used a modified miR-15a (5-FU-miR-15a) with uracil (U) residues in the guide strand replaced with 5-fluorouracil (5-FU). We demonstrated enhanced inhibition of PDAC cell proliferation by 5-FU-miR-15a compared to native miR-15a. In vivo we showed the therapeutic power of 5-FU-miR-15a alone or in combination with gemcitabine with near complete elimination of PDAC lung metastatic tumor growth. These results support the future development of 5-FU-miR-15a as a novel therapeutic agent as well as a prognostic biomarker in the clinical management of PDAC.

17.
FEBS J ; 286(24): 4889-4909, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31715067

RESUMO

Ubiquitin-conjugating enzyme 2C (UBE2C) is a core ubiquitin-conjugating enzyme in the ubiquitin-proteasome system that promotes cell cycle progression. Previous studies have indicated that UBE2C mediates tumorigenesis and progression in various cancers, but its role in pancreatic ductal adenocarcinoma (PDAC) remains unclear. This study elucidated the function of UBE2C in PDAC tumorigenesis and progression by determining UBE2C expression via real-time qPCR, western blotting and immunohistochemistry. The associations between UBE2C expression and clinicopathological characteristics and survival were assessed using a tissue microarray based on a multicentre PDAC cohort. We found that UBE2C was strongly expressed in PDAC patient tissues and was negatively associated with clinical stage, lymph node metastasis, perineural invasion and survival (all P < 0.05). Multivariate analysis revealed that high UBE2C expression is an independent risk factor for PDAC (P = 0.001). In the PDAC cell lines CFPAC-1 and Panc-1, silencing UBE2C suppressed cell proliferation by inducing G1/S arrest mediated by downregulation of cyclin D1. Furthermore, UBE2C knockdown decreased the migration of PDAC cells in vitro by downregulating epithelial-mesenchymal transition (EMT). RNA-seq analysis showed that upon silencing UBE2C in CFPAC-1 cells, cyclin D1 and vimentin were downregulated by approximately 3.5-fold and 2.6-fold, respectively, and the major enriched pathways were related to cell cycle progression. Experiments on tumour-bearing mice injected with CFPAC-1 cells indicated that UBE2C depletion significantly inhibits tumour growth in vivo. These results suggest that UBE2C is involved in the development and progression of PDAC by regulating cell proliferation and EMT. UBE2C is a novel potential therapeutic target for pancreatic cancer. DATABASE: Data are available in the GEO database under accession number GSE137172.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Neoplasias Pancreáticas/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Carcinoma Ductal Pancreático/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Proliferação de Células/fisiologia , Transição Epitelial-Mesenquimal/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Neoplasias Pancreáticas/genética , Modelos de Riscos Proporcionais , Enzimas de Conjugação de Ubiquitina/genética , Neoplasias Pancreáticas
18.
Onco Targets Ther ; 12: 5163-5175, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31456643

RESUMO

Purpose: Alkaloids are naturally occurring chemical compounds that are widely distributed in plants, and have pharmaceutical values and low toxicity. In recent years, some of them have been demonstrated to be promising therapeutic drug candidates for cancer treatment. Herein, we tried to explore the antitumor effect of dehydrocorydaline (DHC), a natural alkaloid isolated from Corydalis, on malignant melanoma. Methods: We treated two malignant metastatic melanoma cell lines, A375 and MV3, and a normal melanocyte cell line, PIG1, with various concentrations of DHC for set amounts of time, and detected cell proliferation, migration, and invasion by using MTT, BrdU, transwell, Western blot and soft agar assay in vitro and tumorigenicity in the xenografts in vivo. Results: Our results showed that DHC dramatically blocked cell proliferation and led to cell cycle arrest at G0/G1 phase and downregulated the expressions of cell cycle regulators CDK6 and Cyclin D1 in melanoma cells. However, DHC had little inhibitory effect on normal melanocyte cell line PIG-1. Meanwhile, DHC suppressed cell invasion and migration through modulating the epithelial-mesenchymal transition (EMT) markers including E-cadherin, vimentin, as well as ß-catenin. In addition, DHC also significantly attenuated tumor growth in vivo. The expressions of cell cycle-related and metastasis-related proteins were further confirmed by immunohistochemical staining in the xenografts. Importantly, MEK1/2-ERK1/2 cascade was inactivated after DHC treatment and ERK activator t-butylhydroquinone (tBHQ) treatment rescued DHC-induced cell proliferation inhibition. Conclusions: Our results indicated that DHC inhibited cell proliferation and migration/invasion via inactivating MAPK signaling, and showed that DHC might be a potential novel drug to treat malignant melanoma.

19.
Neuro Oncol ; 20(9): 1173-1184, 2018 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-29547938

RESUMO

Background: The cancer-testis specific gene Opa interacting protein 5 (OIP5) is reactivated in many human cancers, but its functions in glioblastoma remain unclear. Here, we assessed the significance of OIP5 in the tumorigenesis and metastasis of glioblastoma for the first time. Methods: An immunohistochemistry assay was performed to detect OIP5 expression changes in glioblastoma patients. Overall survival analysis was performed to evaluate the prognostic significance of OIP5. Growth curve, colony formation, and transwell assays were used to analyze cell proliferation and metastasis. Tumorigenicity potential was investigated in orthotopic tumor models, and immunoprecipitation, chromatin immunoprecipitation, and luciferase assays were employed to explore the mechanisms underlying the activation of OIP5 expression by E2F transcription factor 1 (E2F1) to stabilize and maintain E2F1 signaling. Results: OIP5 was found to be upregulated in glioblastoma patients and to impair patient survival, and the increased expression of OIP5 was positively correlated with tumor stage. Compared with short hairpin green fluorescent protein cells, cells in which OIP5 was knocked down exhibited significantly reduced proliferation, metastasis, colony formation, and tumorigenicity abilities, whereas OIP5 recovery enhanced these abilities. OIP5 was highly correlated with cell cycle progression but had no obvious effects on apoptosis. Notably, we demonstrated a feedback loop in which E2F1 activates the expression of OIP5 to stabilize and maintain E2F1 signaling and promote the E2F1-regulated gene expression that is required for aggressive tumor biology. Conclusions: Collectively, our findings demonstrate that OIP5 promotes glioblastoma progression and metastasis, suggesting that OIP5 is a potential target for anticancer therapy.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinogênese/patologia , Proteínas Cromossômicas não Histona/metabolismo , Fator de Transcrição E2F1/química , Fator de Transcrição E2F1/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/secundário , Animais , Apoptose , Biomarcadores Tumorais/genética , Carcinogênese/metabolismo , Estudos de Casos e Controles , Ciclo Celular , Proteínas de Ciclo Celular , Movimento Celular , Proliferação de Células , Proteínas Cromossômicas não Histona/genética , Fator de Transcrição E2F1/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica , Prognóstico , Estabilidade Proteica , Taxa de Sobrevida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Cell Death Dis ; 8(10): e3133, 2017 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-29072681

RESUMO

Demethylzeylasteral is one of the extracts of Tripterygium wilfordii Hook F, which plays important roles in multiple biological processes such as inflammation inhibition, as well as immunosuppression. However, anti-cancer function and the underlying mechanisms of demethylzeylasteral in melanoma cells remain unclear. In this study, we demonstrate that demethylzeylasteral has an anti-tumor property in melanoma cells. Demethylzeylasteral not only inhibits cell proliferation through cell cycle arrest at S phase, but also induces cell apoptosis in melanoma cells. MCL1 is an anti-apoptotic protein in BCL2 family, and amplifies frequently in multiple human cancers. MCL1 is also known as a potential contributor for the resistance of BCL2 inhibitors, as well as various chemotherapeutic drugs. MCL1 is, therefore, regarded as a potential target for cancer therapy. Here, for the first time, we unveil that demethylzeylasteral suppresses the expression of MCL1. Interestingly, MCL1 interacts with S phase-related protein CDK2, and thereby inhibits it's ubiquitin-dependent degradation. Together, demethylzeylasteral is a promising anti-tumor compound in melanoma cells. Demethylzeylasteral is also a potential inhibitor of MCL1.


Assuntos
Melanoma/tratamento farmacológico , Proteína de Sequência 1 de Leucemia de Células Mieloides/antagonistas & inibidores , Triterpenos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Células HEK293 , Humanos , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Nus , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Distribuição Aleatória , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA