Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncol Rep ; 52(1)2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38847273

RESUMO

Pancreatic cancer (PC) is a malignant tumor possessing high mortality. The role of transcription factor Forkhead Box F2 (FOXF2) in PC remains unverified. The current study investigated the roles of FOXF2 in developing PC in vitro and in vivo. A xenograft tumor model was constructed with nude mice injected using FOXF2­overexpressing PC cells or FOXF2­silenced PC cells. High FOXF2 expression significantly enhanced the proliferation ability of PC cells in vitro and pancreatic tumor growth in vivo. The cell cycle analysis indicated that transition of G1­S phase was promoted by FOXF2. The cell cycle­associated proteins cyclin D1, CDK2, phosphorylated (p)­CDK2 and p­RB were upregulated in the FOXF2­overexpressing cells and downregulated in the cells with FOXF2 knockdown. Flow cytometric analysis and Hoechst staining showed that the percentage of apoptotic cells was significantly increased after FOXF2 was silenced. FOXF2 knockdown promoted expression of pro­apoptotic proteins (Bad, Bax and cleaved caspase­3) while suppressing the anti­apoptotic proteins (Bcl­2 and Bcl­xl) at the protein level. FOXF2 improved the migration and invasion of PC cells in vitro. Moreover, luciferase and chromatin immunoprecipitation assays revealed that FOXF2 binds to the MSI2 promoter, promoting its transcriptional expression. FOXF2 knockdown inhibited the MSI2 protein translation while enhancing the translation of NUMB protein, suppressing PC development in vivo. MSI2 silencing reversed the promotive effect mediated by FOXF2 on cell proliferation. These results demonstrated that FOXF2 is essential in PC progression, and the potential mechanism includes regulating MSI2 transcription.


Assuntos
Proliferação de Células , Progressão da Doença , Fatores de Transcrição Forkhead , Regulação Neoplásica da Expressão Gênica , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/metabolismo , Animais , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Camundongos , Proliferação de Células/genética , Linhagem Celular Tumoral , Apoptose/genética , Movimento Celular/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto , Masculino , Técnicas de Silenciamento de Genes , Feminino
2.
Cancer Cell Int ; 24(1): 146, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38654238

RESUMO

BACKGROUND: Colon cancer ranks third among global tumours and second in cancer-related mortality, prompting an urgent need to explore new therapeutic targets. C6orf15 is a novel gene that has been reported only in Sjogren's syndrome and systemic lupus erythematosus patients. We found a close correlation between increased C6orf15 expression and the occurrence of colon cancer. The aim of this study was to explore the potential of C6orf15 as a therapeutic target for colorectal cancer. METHOD: RNA-seq differential expression analysis of the TCGA database was performed using the R package 'limma.' The correlation between target genes and survival as well as tumour analysis was analysed using GEPIA. Western blot and PCR were used to assess C6orf15 expression in colorectal cancer tissue samples. Immunofluorescence and immunohistochemistry were used to assess C6orf15 subcellular localization and tissue expression. The role of C6orf15 in liver metastasis progression was investigated via a mouse spleen infection liver metastasis model. The association of C6orf15 with signalling pathways was assessed using the GSEA-Hallmark database. Immunohistochemistry (IHC), qPCR and western blotting were performed to assess the expression of related mRNAs or proteins. Biological characteristics were evaluated through cell migration assays, MTT assays, and Seahorse XF96 analysis to monitor fatty acid metabolism. RESULTS: C6orf15 was significantly associated with liver metastasis and survival in CRC patients as determined by the bioinformatic analysis and further verified by immunohistochemistry (IHC), qPCR and western blot results. The upregulation of C6orf15 expression in CRC cells can promote the nuclear translocation of ß-catenin and cause an increase in downstream transcription. This leads to changes in the epithelial-mesenchymal transition (EMT) and alterations in fatty acid metabolism, which together promote liver metastasis of CRC. CONCLUSION: Our study identified C6orf15 as a marker of liver metastasis in CRC. C6orf15 can activate the WNT/ß-catenin signalling pathway to promote EMT and fatty acid metabolism in CRC.

3.
Cancer Cell Int ; 24(1): 90, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38429738

RESUMO

BACKGROUND: Patients with colorectal cancer (CRC) with liver metastasis or drug resistance have a poor prognosis. Previous research has demonstrated that PPP2R1B inactivation results in the development of CRC. However, the role of PPP2R1B in CRC metastasis and drug resistance is unclear. METHODS: Venny 2.1 was used to determine the intersection between survival-related differentially expressed genes (DEGs) and liver metastasis-related DEGs according to RNA-seq data from The Cancer Genome Atlas (TCGA) and the GEO database (GSE179979). LC‒MS/MS and coimmunoprecipitation were performed to predict and verify the substrate protein of PPP2R1B. Gene Set Variation Analysis (GSVA) was subsequently utilized to assess pathway enrichment levels. The predictive performance of PPP2R1B was assessed by regression analysis, Kaplan-Meier (KM) survival analysis and drug sensitivity analysis. Immunohistochemistry (IHC), qRT-PCR and western blotting were performed to measure the expression levels of related mRNAs or proteins. Biological features were evaluated by wound healing, cell migration and invasion assays and CCK-8 assays. A mouse spleen infection liver metastasis model was generated to confirm the role of PPP2R1B in the progression of liver metastasis in vivo. RESULTS: According to bioinformatics analysis, PPP2R1B is significantly associated with liver metastasis and survival in CRC patients, and these findings were further verified via immunohistochemistry (IHC), qPCR and Western blotting. Pathway enrichment and LC‒MS/MS analysis revealed that PPP2R1B is negatively associated with the MAPK/ERK signalling pathway. Additionally, PD98059, a MAPK/ERK pathway inhibitor, inhibited EMT in vitro by reversing the changes in key proteins involved in EMT signalling (ZEB1, E-cadherin and Snail) and ERK/MAPK signalling (p-ERK) mediated by PPP2R1B. Oxaliplatin sensitivity prediction and validation revealed that PPP2R1B silencing decreased Oxaliplatin chemosensitivity, and these effects were reversed by PD98059 treatment. Moreover, PPP2R1B was coimmunoprecipitated with p-ERK in vitro. A negative correlation between PPP2R1B and p-ERK expression was also observed in clinical CRC samples, and the low PPP2R1B/high p-ERK coexpression pattern indicated a poor prognosis in CRC patients. In vivo, PPP2R1B silencing significantly promoted liver metastasis. CONCLUSIONS: This study revealed that PPP2R1B induces dephosphorylation of the p-ERK protein, inhibits liver metastasis and increases Oxaliplatin sensitivity in CRC patients and could be a potential candidate for therapeutic application in CRC.

4.
Onco Targets Ther ; 16: 1027-1042, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38107762

RESUMO

Purpose: Colon cancer is one of the leading causes of death worldwide, and screening of effective molecular markers for the diagnosis is prioritised for prevention and treatment. This study aimed to investigate the diagnostic and predictive potential of genes related to the lipid metabolism pathway, regulated by a protein called sterol-regulatory element-binding transcription Factor 2 (SREBF2), for colon cancer and patient outcomes. Methods: We used machine-learning algorithms to identify key genes associated with SREBF2 in colon cancer based on a public database. A nomogram was created to assess the diagnostic value of these genes and validated in the Cancer Genome Atlas. We also analysed the relationship between these genes and the immune microenvironment of colon tumours, as well as the correlation between gene expression and clinicopathological characteristics and prognosis in the China Medical University (CMU) clinical cohort. Results: Three genes, 7-dehydrocholesterol reductase (DHCR7), hydroxysteroid 11-beta dehydrogenase 2 (HSD11B2), and Ral guanine nucleotide dissociation stimulator-like 1 (RGL1), were identified as hub genes related to SREBF2 and colon cancer. Using the TCGA dataset, receiver operating characteristic curve analysis showed the area under the curve values of 0.943, 0.976, and 0.868 for DHCR7, HSD11B2, and RGL1, respectively. In the CMU cohort, SREBF2 and DHCR7 expression levels were correlated with TNM stage and tumour invasion depth (P < 0.05), and high DHCR7 expression was related to poor prognosis of colon cancer (P < 0.05). Furthermore, DHCR7 gene expression was positively correlated with the abundance of M0 and M1 macrophages and inversely correlated with the abundance of M2 macrophages, suggesting that the immune microenvironment may play a role in colon cancer surveillance. There was a correlation between SREBF2 and DHCR7 expression across cancers in the TCGA database. Conclusion: This study highlights the potential of DHCR7 as a diagnostic marker and therapeutic target for colon cancer.

5.
PeerJ ; 10: e13172, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35345586

RESUMO

Purpose: The Numb protein plays a vital role in tumor development. The main aim of this study was to identify ATP11A, which is associated with the biological behavior of pancreatic cancer, and elucidate its relationship with Numb and the underlying mechanism behind this relationship. Methods: First, data retrieved from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEX) databases was used to investigate the expression of ATP11A mRNA and its relationship with Numb mRNA in pancreatic cancer. Western blot assays on 31 pairs of pancreatic cancer tissues and paracancerous tissues, and immunohistochemical assays on 81 pancreatic cancer specimens were performed in order to verify the expression of ATP11A in pancreatic cancer at the protein level. Next, ATP11A was overexpressed or knocked down to observe its effects on the invasion and migration ability of pancreatic cancer cells and the changes of downstream proteins. Rescue assays were conducted to determine the mechanism through which ATP11A affects Numb, ZEB1, Snail2 and other proteins. Furthermore, immunoprecipitation assays were performed to explore the interaction between ATP11A and Numb. Finally, pancreatic cancer cells were stimulated with TGFB1 and ATP11A expression was examined to explore whether the effect of ATP11A on EMT was TGFB dependent. Results: At the mRNA level, the expression of ATP11A in pancreatic cancer tissues was significantly higher than in normal pancreatic tissues (P < 0.001). ATP11A expression was also highly correlated with Numb expression (R = 0.676). At the protein level, ATP11A expression in pancreatic cancer tissues was significantly higher than that in paracancerous tissues (P = 0.0009), and high ATP11A expression was also correlated with a worse prognosis. Moreover, our results showed that ATP11A can promote the invasion and migration of pancreatic cancer cells. Additionally, ATP11A could positively regulate the expression of Numb PRRL, Snail2 and ZEB1 proteins. The rescue experiment results showed that the enhancement effect of ATP11A on ZEB1/Snail2 was suppressed by the specific knockdown of Numb PRRL. In addition, the immunoprecipitation results showed that ATP11A could specifically bind to Numb PRRL. The expression of ATP11A was also upregulated after TGFB stimulation, suggesting that the effect of ATP11A on EMT is TGFB dependent. Conclusion: ATP11A is significantly upregulated in pancreatic cancer tissues, where it promotes the invasion and migration ability of pancreatic cancer cells. It is also associated with adverse prognosis in pancreatic cancer. Furthermore, ATP11A affects the epithelial-to-mesenchymal transition (EMT) of pancreatic cancer by regulating the TGFB dependent Numb PRRL-ZEB1/Snail2 pathway.


Assuntos
Neoplasias Pancreáticas , Humanos , Movimento Celular/genética , Neoplasias Pancreáticas/genética , Transição Epitelial-Mesenquimal/genética , RNA Mensageiro/genética , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Transportadores de Cassetes de Ligação de ATP , Neoplasias Pancreáticas
6.
Cell Death Dis ; 13(2): 173, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35197444

RESUMO

Isoform-specific functions of Numb in the development of cancers, especially in the initiation of epithelial-to-mesenchymal transition (EMT) remains controversial. We study the specific function of Numb-PRRL isoform in activated EMT of pancreatic ductal adenocarcinoma (PC), which is distinguished from our previous studies that only focused on the total Numb protein. Numb-PRRL isoform was specifically overexpressed and silenced in PC cells combining with TGF-ß1 and EGF stimulus. We systematically explored the potential effect of Numb-PRRL in the activated EMT of PC in vitro and in vivo. The total Numb protein was overexpressed in the normal pancreatic duct and well-differentiated PC by IHC. However, Numb-PRRS isoform but not Numb-PRRL showed dominant expression in PC tissues. Numb-PRRL overexpression promoted TGF-ß1-induced EMT in PANC-1 and Miapaca-2 cells. TGF-ß1-induced EMT-like cell morphology, cell invasion, and migration were enhanced in Numb-PRRL overexpressing groups following the increase of N-cadherin, Vimentin, Smad2/3, Snail1, Snail2, and cleaved-Notch1 and the decrease of E-cadherin. Numb-PRRL overexpression activated TGFß1-Smad2/3-Snail1 signaling was significantly reversed by the Notch1 inhibitor RO4929097. Conversely, Numb-PRRL silencing inhibited EGF-induced EMT in AsPC-1 and BxPC-3 cells following the activation of EGFR-ERK/MAPK signaling via phosphorylating EGFR at tyrosine 1045. In vivo, Numb-PRRL overexpression or silencing promoted or inhibited subcutaneous tumor size and distant liver metastases via regulating EMT and Snail signaling, respectively. Numb-PRRL promotes TGF-ß1- and EGF-induced EMT in PC by regulating TGF-ß1-Smad2/3-Snail and EGF-induced EGFR-ERK/MAPK signaling.


Assuntos
Transição Epitelial-Mesenquimal , Proteínas de Membrana , Proteínas do Tecido Nervoso , Neoplasias Pancreáticas , Fator de Crescimento Transformador beta1 , Linhagem Celular Tumoral , Movimento Celular , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB , Humanos , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Neoplasias Pancreáticas/patologia , Fator de Crescimento Transformador beta1/farmacologia , Neoplasias Pancreáticas
7.
Onco Targets Ther ; 14: 3503-3515, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34103941

RESUMO

BACKGROUND: Colorectal cancer is the fourth most common cancer and the second leading cause of cancer-related death in the USA. The aim of this study was to establish a tumor gene signature based on tumor stromal cell and autophagy for predicting the risk of recurrence in patients with colorectal cancer. METHODS: We used "Rtsne" and "xCell" R packages to estimate autophagy and stroma status, respectively. The discovery cohort used microarray gene expression data retrieved from the GSE39582 dataset. The Cox regression model and Least Absolute Shrinkage and Selection Operator (LASSO) were used to identify prognostic genes and to construct an autophagy-stroma-based gene signature. Moreover, external validation was conducted using GSE17538, GSE38832, TCGA database, and patient data obtained from the First Hospital of China Medical University (CMU). RESULTS: The LASSO model identified three genes (TNS1, TAGLN, and SFRP4) which were used to develop a risk stratification gene signature. The autophagy-stroma-based gene signature was identified as an independent prognostic factor by multivariate analysis (p = 0.0023). The results were validated in GSE17538 (p=0.0062), GSE38832 (p=0.028), TCGA (p=0.046) database, and patient data obtained from the First Hospital of China Medical University (CMU) (p=0.027). CONCLUSION: We have established and verified a feasible prognostic model of colorectal cancer based on autophagy and stromal cell characteristics of patients. The model can be used to evaluate recurrence risk of cancer patients, and the hub genes in the model provide potential targets for targeted colorectal cancer treatment.

8.
Onco Targets Ther ; 14: 2311-2325, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33833529

RESUMO

BACKGROUND: miR-944 belongs to the MicroRNAs family, as shown in our previous study, and is essential in the colorectal cancer (CRC) progression. It is negatively associated with invasion depth and lymph node status. Epithelial-mesenchymal transition (EMT) is essential in tumor invasion and metastasis. However, the relationship between miR-944 and EMT in CRC is unknown and should be further investigated. METHODS: Epithelial-mesenchymal transition (EMT) progression in CRC cell lines was detected with Cell morphology and Western blotting. CRC cell migration and invasion were examined using Transwell assays. Transcriptome and clinical data were obtained from The Cancer Genome Atlas (TCGA) database. The potential pathway of miR-944 and GATA6 were predicted using KEGG analysis. Colocalization was validated using immunofluorescence and Immunohistochemistry. Nuclear and Cytoplasmic Protein Extraction assays were conducted to determine the effects of miR-944 on Wnt/ß-catenin signaling. RESULTS: We found that miR­944 influences EGF-induced EMT malignant phenotype in vitro. KEGG analyses showed that miR-944 and GATA6 are associated with EMT related pathways, wnt signaling pathways. On the other hand, Western Blot analyses showed that miR-944 can regulate EMT and wnt-ß-catenin pathway-related protein, including ß-catenin, ZEB1, snail1 via GATA6 regulation. miR-944 also abrogates E-ca after EGF induction. Immunohistochemistry (IHC) and Immunofluorescence (IF) co-expression showed that GATA6 expression is positively associated with ß-catenin and ZEB1. GATA6 silencing can reverse EMT malignant phenotype and alterations of related protein induced by miR-944. Quantitative polymerase chain reaction analysis results showed that miR-944 is negatively associated with the UICC stage (P= 0.02), lymph nodes (p=0.04), and liver metastasis (p=0.03). Moreover, patients with high miR-944 expression have better survival (p=0.045). We finally combined miR-944 and GATA6 and found that miR-944/GATA6 ratio could be a novel prognostic biomarker in the TCGA dataset and it is an independent risk prognosis factor (p=0.045). CONCLUSION: Our results suggest that miR-944 suppresses the aggressive biological processes by directly repressing GATA6 expression and could be a potential candidate for therapeutic applications in CRC.

9.
Onco Targets Ther ; 13: 11045-11058, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33149617

RESUMO

OBJECTIVE: To investigate the effect of miR-590-3p on the malignant biological behavior of pancreatic cancer, and to explore the target genes and pathways directly affected by miR-590-3p, to provide new therapeutic ideas and targets for the study of the diagnosis and treatment of pancreatic cancer. METHODS: We used qRT-PCR to measure miR-590-3p expression quantities. We used cell cycle, CCK-8, clonal formation to verify the change of proliferation capacity of PC cells. We used transwell assay to detect the migration and invasion of PC cells. We used the bioinformatics tool TargetScan (http://www.targetscan.org) to identify the possible target genes of miR-590-3p. Immunohistochemistry revealed the clinicopathological significance of PPP2R2A, p27 and miR-590-3p in the expression of pancreatic cancer. Western blot was used to detect the expression changes of PPP2R2A, p27 and G1/S cell cycle pathway-related proteins CDK2, cyclinE2 and p21 after transfection of mimics and inhibitors of miR-590-3p. RESULTS: According to our study, hsa-miR-590-3p expression was significantly higher in PC tissues than that in paired normal pancreas, which was associated with PC tumor size (P=0.042) and preoperative CA19-9 level (P=0.046) of PC patients. Its overexpression promoted PC cell proliferation, invasion and migration following with the p27 and PPP2R2A protein downregulation in Capan-2, PANC-1 and BxPC-3 cells, and vice versa. Bioinformatics analysis and dual-luciferase reporter assay further confirmed that p27 and PPP2R2A were direct target genes of miR-590-3p. The negative relationship of miR-590-3p with p27 and PPP2R2A was also observed in PC tissues. CONCLUSION: MiR-590-3p promotes the proliferation, migration and invasion of pancreatic cancer cells. MiR-590-3p directly downregulated p27 and PPP2R2A and via the G1/S cell cycle pathway to promote the development of pancreatic cancer.

10.
J Exp Clin Cancer Res ; 39(1): 209, 2020 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-33028359

RESUMO

BACKGROUND: Our previous study showed that calreticulin (CRT) promoted EGF-induced epithelial-mesenchymal transition (EMT) in pancreatic cancer (PC) via Integrin/EGFR-ERK/MAPK signaling. We next investigated the novel signal pathway and molecular mechanism involving the oncogenic role of CRT in PC. METHODS: We investigated the potential role and mechanism of CRT in regulating intracellular free Ca2+ dependent acute and chronic endoplasmic reticulum stress (ERS)-induced EMT in PC in vitro and vivo. RESULTS: Thapsigargin (TG) induced acute ERS via increasing intracellular free Ca2+ in PC cells, which was reversed by CRT silencing. Additionally, CRT silencing inhibited TG-induced EMT in vitro by reversing TG-induced changes of the key proteins in EMT signaling (ZO-1, E-cadherin and Slug) and ERK/MAPK signaling (pERK). TG-promoted cell invasion and migration was also rescued by CRT silencing but enhanced by IRE1α silencing (one of the key stressors in unfolded protein response). Meanwhile, CRT was co-immunoprecipitated and co-localized with IRE1α in vitro and its silencing led to the chronic ERS via upregulating IRE1α independent of IRE1-XBP1 axis. Moreover, CRT silencing inhibited IRE1α silencing-promoted EMT, including inhibiting the activation of EMT and ERK/MAPK signaling and the promotion of cell mobility. In vivo, CRT silencing decreased subcutaneous tumor size and distant liver metastasis following with the increase of IRE1α expression. A negative relationship between CRT and IRE1α was also observed in clinical PC samples, which coordinately promoted the advanced clinical stages and poor prognosis of PC patients. CONCLUSIONS: CRT promotes EMT in PC via mediating intracellular free Ca2+ dependent TG-induced acute ERS and IRE1α-mediated chronic ERS via Slug and ERK/MAPK signaling.


Assuntos
Biomarcadores Tumorais/metabolismo , Cálcio/metabolismo , Calreticulina/metabolismo , Estresse do Retículo Endoplasmático , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Neoplasias Pancreáticas/patologia , Animais , Apoptose , Biomarcadores Tumorais/genética , Calreticulina/genética , Proliferação de Células , Endorribonucleases/genética , Endorribonucleases/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Prognóstico , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Taxa de Sobrevida , Células Tumorais Cultivadas , Proteína 1 de Ligação a X-Box/genética , Proteína 1 de Ligação a X-Box/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Exp Clin Cancer Res ; 39(1): 167, 2020 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-32838790

RESUMO

An amendment to this paper has been published and can be accessed via the original article.

12.
J Exp Clin Cancer Res ; 39(1): 16, 2020 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-31952541

RESUMO

BACKGROUND: Our previous study showed Musashi2 (MSI2) promoted chemotherapy resistance and pernicious biology of pancreatic cancer (PC) by down-regulating Numb and p53. We further explored the novel molecular mechanism involving its oncogenic role in PC development. METHODS: We investigated the potential role and mechanism of MSI2 in EGF-induced EMT in PC in vitro and vivo. RESULTS: EGF enhanced EGFR (epidermal growth factor receptor) phosphorylation, induced EMT and activated ZEB1-ERK/MAPK signaling in 2 PC cells. However, MSI2 silencing reversed EGF stimulated function, including inhibiting EGF-promoted EMT-like cell morphology and EGF-enhanced cell invasion and migration. Meanwhile, MSI2 silencing inhibited EGF-enhanced EGFR phosphorylation at tyrosine 1068 and reversed EGF-induced change of the key proteins in EMT and ZEB1-ERK/MAPK signaling (ZEB1, E-cad, ZO-1, ß-catenin, pERK and c-Myc). Additionally, MSI2 was co-stained and co-immunoprecipitated with ZEB1, pERK and c-Myc in PC cells by IF and co-IP, implying a close interaction between them. In vivo, MSI2 silencing inhibited pancreatic tumor size in situ and distant liver metastases. A close relationship of MSI2 with EMT and ZEB1-ERK/MAPK signaling were also observed in vivo and human PC samples, which coordinately promoted the poor prognosis of PC patients. CONCLUSIONS: MSI2 promotes EGF-induced EMT in PC via ZEB1-ERK/MAPK signaling.

13.
Biosci Rep ; 40(1)2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31898732

RESUMO

Serine-arginine protein kinase 2 (SRPK2) is aberrantly expressed in human malignancies including colorectal cancer (CRC). However, little is known about the molecular mechanisms, and the role of SRPK2 in chemosensitivity remains unexplored in CRC. We recently showed that SRPK2 promotes pancreatic cancer progression by down-regulating Numb and p53. Therefore, we investigated the cooperation between SRPK2, Numb and p53 in the cell migration, invasion and chemosensitivity of CRC in vitro. Here, we showed that SRPK2 expression was higher in CRC tumors than in nontumor tissues. SRPK2 expression was positively associated with clinicopathological characteristics of CRC patients, including tumor differentiation, T stage, N stage and UICC stage. Additionally, SRPK2 had no association with mutant p53 (mtp53) in SW480 and SW620 cells, but negatively regulated Numb and wild-type p53 (wtp53) in response to 5-fluorouracil or cisplatin treatment in HCT116 cells. Moreover, SRPK2, Numb and p53 coimmunoprecipitated into a triple complex with or without the treatment of 5-fluorouracil in HCT116 cells, and p53 knockdown reversed the up-regulation of wtp53 induced by SRPK2 silencing with chemical agent treatment. Furthermore, overexpression of SRPK2 increased cell migration and invasion and decreased chemosensitivity to 5-fluorouracil or cisplatin in HCT116 cells. Conversely, SRPK2 silencing decreased cell migration and invasion and increased chemosensitivity to 5-fluorouracil or cisplatin, yet these effects could be reversed by p53 knockdown under chemical agent treatment. These results thus reveal a novel role of SRPK2-Numb-p53 signaling in the progression of CRC and demonstrate that SRPK2 is a potential therapeutic target for CRC clinical therapy.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Cisplatino/farmacologia , Neoplasias Colorretais/patologia , Progressão da Doença , Feminino , Fluoruracila/farmacologia , Células HCT116 , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Estadiamento de Neoplasias/métodos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
14.
J Cell Mol Med ; 23(5): 3483-3494, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30873717

RESUMO

miR-944 is a microRNA that has been reported to play different important roles in the progression of cancer. Colorectal cancer (CRC) is a common cancer worldwide. A recent study has confirmed that miR-944 plays a tumour suppressive role in CRC. However, biological functions and the mechanism of miR-944 in CRC are poorly understood. Real-time reverse transcription polymerase chain reaction of 100 CRC tissues showed that miR-944 expression is frequently downregulated and is negatively associated with the T is the primary tumor, N is the lymph node, and M is the distant metastasis (TNM) stage (P = 0.009), depth of invasion (P = 0.001), and lymph node status (P = 0.002). Overexpression of mir-944 significantly impaired the functions of proliferation, migration and invasion in CRC cells, while these functions increased in knockdown experiments. GATA binding protein 6 (GATA6) knockdown can reverse the CRC cells functions induced by miR-944 inhibitor. Mechanistically, a Dual-Luciferase Reporter Assay showed that miR-944 is structurally combined with GATA6 and interacts with downstream proteins (CRT and p-AKT) in CRC cells. In conclusion, these findings indicated that miR-944 may be a tumour suppressor and could likely be used as a prognostic predictor and novel therapeutic target for CRC.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Expressão Ectópica do Gene , Fator de Transcrição GATA6/metabolismo , MicroRNAs/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação para Baixo/genética , Feminino , Fator de Transcrição GATA6/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Invasividade Neoplásica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...