Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Publication year range
1.
Br J Pharmacol ; 178(11): 2351-2369, 2021 06.
Article in English | MEDLINE | ID: mdl-33645631

ABSTRACT

BACKGROUND AND PURPOSE: It is well known that microsatellite instability-high (MSI-H) is associated with 5-fluorouracil (5-FU) resistance in colorectal cancer. MSI-H is the phenotype of DNA mismatch repair deficiency (MMR-D), mainly occurring due to hypermethylation of MLH1 promoter CpG island. However, the mechanisms of MMR-D/MSI-H are unclear. We aim to investigate the pathway of MMR-D/MSI-H involved in 5-FU resistance. EXPERIMENTAL APPROACH: Human colorectal cancer specimens were diagnosed for MSI-H by immunohistochemistry and western blotting. Proteome microarray interactome assay was performed to screen nuclear proteins interacting with ATG5. Nuclear ATG5 and ATG5-Mis18α overexpression were analysed in ATG5high colorectal cancer bearing mice. The methylation assay determined the hypermethylation of hMLH1 promoter CpG island in freshly isolated human colorectal cancer tissue samples and HT29atg5 and SW480atg5 cancer cells. KEY RESULTS: In ATG5high colorectal cancer patients, 5-FU-based therapy resulted in nuclear translocation of ATG5, leading to MSI-H. Colorectal cancer in Atg5 Tg mice demonstrated 5-FU resistance, compared to Atg5+/- and WT mice. Proteome microarray assay identified Mis18α, a protein localized on the centromere and a source for methylation of the underlying chromatin, which responded to the translocated nuclear ATG5 leading to ATG5-Mis18α conjugate overexpression. This resulted in MLH1 deficiency due to hypermethylation of hMLH1 promoter CpG island, while the deletion of nuclear Mis18α failed to induce ATG5-Mis18α complex and MMR-D/MSI-H. CONCLUSIONS AND IMPLICATIONS: Nuclear ATG5 resulted in MMR-D/MSI-H through its interaction with Mis18α in ATG5high colorectal cancer cells. We suggest that ATG5-Mis18α or Mis18α may be a therapeutic target for treating colorectal cancer.


Subject(s)
Adaptor Proteins, Signal Transducing , Autophagy-Related Protein 5 , Colorectal Neoplasms , Microsatellite Instability , Adaptor Proteins, Signal Transducing/genetics , Animals , Brain Neoplasms , Chromosomal Proteins, Non-Histone , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , DNA , DNA Methylation , Humans , Mice , MutL Protein Homolog 1/genetics , Neoplastic Syndromes, Hereditary
2.
Int J Clin Exp Pathol ; 12(8): 2909-2919, 2019.
Article in English | MEDLINE | ID: mdl-31934127

ABSTRACT

PH domain leucine-rich repeat protein phosphatase 2 (PHLPP2) belongs to the phosphokinase family, that has been reported to play an important role in several cancers. However, the expression of PHLPP2 and its correlation with clinicopathologic characteristics in colorectal cancer (CRC) have yet to be determined. The aim of this study is to investigate the expression of PHLPP2 and explore its role in CRC. The expression of PHLPP2, PTEN, PI3KCA, and PI3KCB in 130 cases of CRC and normal tissues was assessed by immunohistochemistry. In addition, the expression of PHLPP2, PTEN, PI3KCA, and PI3KCB in 32 pairs of CRC tissues and their corresponding normal tissues was determined by RT-PCR and western blotting, respectively. PHLPP2 expression in CRC was significantly lower than that of normal tissues. However, PHLPP2 mRNA shows no significant difference between CRC and normal tissue. PTEN expression in left colorectal cancer (LCC) was absent, while PI3KCA and PI3KCB in right colorectal cancer (RCC) were significantly higher than those in LCC. PHLPP2 was negatively correlated with p-Akt1 in CRC. The expression of p-Akt1 in PHLPP2 (+)/PTEN (+) in CRC tissues was significantly lower than that in other groups. PHLPP2 expression was correlated with differentiation, invasion, and lymph node metastasis. Kaplan-Meier analysis and multivariate analysis reveal that PHLPP2 is closely related to prognosis; more importantly, it is an independent prognostic factor for CRC. In conclusion, PHLPP2 may play a major role in the development, metastasis, and prognosis of CRC.

3.
Oncol Rep ; 41(2): 895-907, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30431131

ABSTRACT

Long non­coding RNAs (lncRNAs) have been consistently demonstrated to be involved in oral squamous cell carcinoma (OSCC) as either tumor oncogenes or tumor suppressors. However, the underlying mechanisms of OSCC tumorigenesis and development have not yet been fully elucidated. The expression profiles of mRNAs and lncRNAs in OSCC were analyzed by a microarray assay. To verify the results of the microarray, 10 differentially expressed lncRNAs were randomly selected and measured by quantitative RT­PCR (qRT­PCR). Gene Ontology (GO) and metabolic pathway analyses were performed to analyze gene function and identify enriched pathways. Subsequently, two independent algorithms were used to predict the target genes of the lncRNAs. We identified 2,294 lncRNAs and 1,938 mRNAs that were differentially expressed in all three OSCC tissues by a microarray assay. Through the construction of co­expression networks of differentially expressed genes, 4 critical lncRNAs nodes were identified as potential key factors in the pathogenesis of OSCC. Expression of the 4 critical lncRNA nodes was not associated with age, sex, smoking or tumor location (P>0.05) but was positively correlated with clinical stage, lymphatic metastasis, distant metastasis and survival status (P<0.05). Kaplan­Meier analysis demonstrated that low expression levels of these 4 critical lncRNA nodes contributed to poor median progression­free survival (PFS) and overall survival (OS) (P<0.05). GO and pathway analyses indicated that the functions and enriched pathways of many dysregulated genes are associated with cancer. Potential target genes of dysregulated lncRNAs were enriched in 43 metabolic pathways, with cancer pathways being the primary enrichment pathways. In summary, we analyzed the profile of lncRNAs in OSCC and identified the functions and enriched metabolic pathways of both dysregulated mRNAs and the target genes of dysregulated lncRNAs, providing new insights into molecular markers and therapeutic targets for OSCC.


Subject(s)
Gene Expression Regulation, Neoplastic , Mouth Neoplasms/genetics , RNA, Long Noncoding/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Female , Follow-Up Studies , Gene Expression Profiling , Gene Ontology , Gene Regulatory Networks , Genome-Wide Association Study , Humans , Lymphatic Metastasis , Male , Metabolic Networks and Pathways/genetics , Middle Aged , Mouth Neoplasms/mortality , Mouth Neoplasms/pathology , Neoplasm Staging , Progression-Free Survival , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Squamous Cell Carcinoma of Head and Neck/mortality , Squamous Cell Carcinoma of Head and Neck/pathology
4.
Med Sci Monit ; 22: 1761-5, 2016 May 25.
Article in English | MEDLINE | ID: mdl-27222034

ABSTRACT

BACKGROUND Early metastasis of osteosarcoma (OS) is highly lethal and responds poorly to drug and radiation therapies. MicroRNAs (miRNAs) are a class of small noncoding RNAs that modulate gene expression at the post-transcriptional level. However, the detailed functions of specific miRNAs are not entirely understood. The aim of the present study was to investigate the role of miR-184 as a mediator of drug resistance in human osteosarcoma. MATERIAL AND METHODS qRT-PCR was used to analyze the expression level of miR-184 in OS cell line U-2 OS and MG-63 treated with doxorubicin. MiR-184 agomir or miR-184 antagomir was transferred into cells to regulated miR-184. The target of miR-184 was predicted by TargetScan and confirmed by luciferase reporter assay. Bcl-2-like protein 1 (BCL2L1) expression was detected by Western blot. Cell apoptosis was determined by Annexin V staining and analysis by flow cytometry. RESULTS Doxorubicin induced time-dependent expression of miR-184 in OS cell line U-2 OS and MG-63. Luciferase reporter assay identified BCL2L1 as the direct target gene of miR-184. Furthermore, doxorubicin reduced BCL2L1 expression, which was reversed by miR-184 overexpression and further decreased by miR-184 inhibition in OS cells. In addition, miR-184 agomir reduced doxorubicin-induced cell apoptosis, whereas miR-184 antagomir enhanced apoptosis in OS cells, suggesting that up-regulation of miR-184 contributes to chemoresistance of the OS cell line. CONCLUSIONS Our data show that miR-184 was up-regulated in OS patients treated with doxorubicin therapy and leads to poor response to drug therapy by targeting BCL2L1.


Subject(s)
Bone Neoplasms/drug therapy , Doxorubicin/pharmacology , MicroRNAs/metabolism , Osteosarcoma/drug therapy , bcl-X Protein/metabolism , 3' Untranslated Regions , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Humans , MicroRNAs/genetics , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , bcl-X Protein/genetics
5.
Yi Chuan ; 37(4): 367-373, 2015 Apr.
Article in Chinese | MEDLINE | ID: mdl-25881702

ABSTRACT

Human NKX2.5 (NK2 homeobox 5) premature stop codon (PTC) mutations cause congenital heart diseases such as atrial septal defect and atrioventricular block. At present, eight NKX2.5 PTC mutations were reported as E109X, Q149X, Q170X, Q187X, Q198X, Y256X, Y259X and C264X. To observe the ability of tRNA suppressors to read through NKX2.5 PTC mutations and produce functional full-length proteins, eight NKX2.5 PTC mutations were cloned into pcDNA3.1(-) vectors and four fragments (wild-type NKX2.5, E109X, Q149X and C264X) were cloned in pEGFP-N1 vectors to acquire NKX2.5-EGFP fusing plasmids. After transfection of NKX2.5-EGFP with or without corresponding tRNA suppressor into HeLa cells, the quantity of EGFP was measured to confirm the readthrough ability of the PTCs. NKX2.5 full-length and truncated protein expression levels were examined by Western blotting and the readthrough efficiency of tRNA suppressors on the PTCs was calculated respectively. The activity of NKX2.5 full-length and truncated protein was confirmed on NKX2.5 target gene-Cx43 mRNA level measured by Real-time PCR. Three tRNA suppressors were used: tRNA am, tRNA oc and tRNA op. tRNA am could suppress UAG-containing PTCs Q149X, Q170X, Q187X, Q198X and the readthrough efficiency for the latter three was above 50%. tRNA op could suppress UGA-containing PTC C264X with ~50% readthrough efficiency. tRNA oc failed to read through NKX2.5 PTC mutations. The relative Cx43 mRNA level in all readthrough samples was increased to 7%-41.7%. In conclusion, tRNA am and tRNA op could suppress NKX2.5 PTCs and induce functional protein expression. However, the effects of tRNA suppressors on cellular function are not clear yet, warranting further researches.


Subject(s)
Codon, Terminator/genetics , Gene Expression Regulation , Homeodomain Proteins/genetics , Peptide Chain Termination, Translational , RNA, Transfer/genetics , Transcription Factors/genetics , Codon, Terminator/metabolism , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/metabolism , Humans , RNA, Transfer/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...