Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem J ; 477(10): 1939-1950, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32391557

ABSTRACT

eIF3a is a putative subunit of the eukaryotic translation initiation factor 3 complex. Accumulating evidence suggests that eIF3a may have a translational regulatory function by suppressing translation of a subset of mRNAs while accelerating that of other mRNAs. Albeit the suppression of mRNA translation may derive from eIF3a binding to the 5'-UTRs of target mRNAs, how eIF3a may accelerate mRNA translation remains unknown. In this study, we show that eIF3a up-regulates translation of Chk1 but not Chk2 mRNA by interacting with HuR, which binds directly to the 3'-UTR of Chk1 mRNA. The interaction between eIF3a and HuR occurs at the 10-amino-acid repeat domain of eIF3a and the RNA recognition motif domain of HuR. This interaction may effectively circularize Chk1 mRNA to form an end-to-end complex that has recently been suggested to accelerate mRNA translation. Together with previous findings, we conclude that eIF3a may regulate mRNA translation by directly binding to the 5'-UTR to suppress or by interacting with RNA-binding proteins at 3'-UTRs to accelerate mRNA translation.


Subject(s)
Checkpoint Kinase 1/biosynthesis , ELAV-Like Protein 1 , Eukaryotic Initiation Factor-3 , Protein Biosynthesis/physiology , Cell Line , ELAV-Like Protein 1/chemistry , ELAV-Like Protein 1/metabolism , Eukaryotic Initiation Factor-3/chemistry , Eukaryotic Initiation Factor-3/metabolism , Gene Expression Regulation , Humans , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA-Binding Proteins
2.
Int J Biol Sci ; 16(8): 1388-1402, 2020.
Article in English | MEDLINE | ID: mdl-32210727

ABSTRACT

Objective: The therapeutic effects of the checkpoint kinase 1 (CHK1)-targeted inhibition in tumor therapy have been confirmed, but how to choose an effective application method in breast cancer with heterogeneous molecular characteristics has remained unclear. Methods: We evaluated the status of CHK1 in breast cancer using the cancer genome atlas database. Chemosensitivity and single-agent antitumor activity of CHK1 inhibition were measured by drug sensitivity assay, cell proliferation assay, cell cycle and apoptosis analysis in breast cancer with different ER/PR status. And based on the conjoint transcriptome atlas analyses, the corresponding mechanism were explored. Results: In ER-/PR-/HER2- breast cancer, CHK1 inhibition enhanced adriamycin (ADR) chemosensitivity which was mediated by the mitotic checkpoint complex (MCC)-anaphase-promoting complex/cyclosome (APC/C)-cyclin B1 axis, Msh homeobox 2 (MSX2) and Bcl-2-like protein 11 (BIM). However, in ER+/PR+/HER2- breast cancer, because of the significant suppression for centromere protein F (CENPF)-mediated transcriptional activation of CHK1 induced by ADR itself, CHK1 inhibition fails to sensitize ADR toxicity. Interestingly, CHK1 inhibition showed the single-agent antitumor activity in ER+/PR+/HER2- breast cancer which was mediated by the cyclin dependent kinase inhibitor 1A (p21), kinesin family member 11 (Eg5) and cell surface death receptor (Fas). Conclusions: CHK1's variable role determines the application of CHK1 inhibition in breast cancer with ER/PR heterogeneity.


Subject(s)
Breast Neoplasms/metabolism , Checkpoint Kinase 1/biosynthesis , Estrogen Receptor alpha/biosynthesis , Receptors, Progesterone/biosynthesis , Apoptosis , Breast Neoplasms/genetics , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Survival , Computational Biology , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor/methods , Female , Genome, Human , Humans , Kaplan-Meier Estimate , MCF-7 Cells , RNA Interference , Treatment Outcome
3.
Cancer Lett ; 433: 232-241, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30003927

ABSTRACT

Radiation therapy (XRT) is a standard treatment for prostate cancer (PCa). Although dose escalation increases local control, toxicity hampers further escalation. Broader improvement will be possible by the addition of adjuvant therapies, which can synergize with radiation and thus improve efficacy. We have identified a natural compound (Nexrutine, Nx) that inhibits the survival and growth of PCa cells in combination with radiation. Combination studies demonstrated strong interaction between Nx and radiation both in vitro in multiple PCa cell lines and in the Transgenic adenocarcinoma of mouse prostate (TRAMP) model. Nx potentiated growth inhibitory effects of IR by down regulating ribosomal protein S6K (RPS6KB1), CyclinD1, Chk1 and HIF-1 α and prolonging G2/M checkpoint block. RPS6KB1 is upregulated in prostate cancers and its expression is correlated with tumor grade. Knockdown of RPS6KB1 in PCa cells increased their sensitivity toward radiation-induced survival inhibition. Overall, we provide scientific evidence (i) in support of Nx as an adjuvant in PCa patients receiving XRT (ii) suggesting that RPS6KB1 is an important player in Nx-mediated combinatorial benefits and emphasizes that RPS6KB1 is a novel target for PCa treatment. These data underscore the need to test the agent in additional preclinical models to validate these observations.


Subject(s)
Antineoplastic Agents/pharmacology , Plant Extracts/pharmacology , Prostatic Neoplasms/radiotherapy , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Checkpoint Kinase 1/biosynthesis , Cyclin D1/biosynthesis , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Male , Mice , PC-3 Cells , Ribosomal Protein S6 Kinases, 70-kDa/biosynthesis
4.
Gynecol Oncol ; 150(1): 136-142, 2018 07.
Article in English | MEDLINE | ID: mdl-29804637

ABSTRACT

OBJECTIVE: To analyze the expression and clinical role of CHK1 and CHK2 in metastatic high-grade serous carcinoma (HGSC). METHODS: HGSC effusions (n = 335; 280 peritoneal, 55 pleural) were analyzed for protein expression of total CHK1 and its phosphorylated forms p-ser317 and p-ser296, as well as total CHK2 and its phosphorylated form p-thr68 using immunohistochemistry. Expression was analyzed for association with clinicopathologic parameters, including chemotherapy response, and survival. RESULTS: Carcinoma cells stained positive, predominantly at the nuclei, in the majority of cases (range 83-100% for the five antibodies), while expression in reactive mesothelial cells and tumor-associated macrophages was more variable. Total CHK1 (p = 0.037), p-CHK1ser317 (p = 0.001), p-CHK1ser296 (p = 0.002) and p-CHK2thr68 (p < 0.001) expression was significantly higher in post-chemotherapy disease recurrence compared to pre-chemotherapy effusions obtained at diagnosis. CHK1, p-CHK1ser296, p-CHK2thr68 and p-CHK1ser317 nuclear expression was positively related to expression of the checkpoint regulator WEE1, previously studied in this cohort (p = 0.003, p = 0.013, p = 0.001 and p = 0.01, respectively). Higher total CHK1 (p = 0.007), p-CHK1ser317 (p = 0.004), CHK2 (p = 0.01) and p-CHK2thr68 (p = 0.048) expression was significantly related to shorter overall survival in univariate analysis, and CHK1ser317 was an independent prognostic marker in multivariate analysis (p = 0.025). Higher p-CHK1ser317 (p = 0.03) and CHK2 (p = 0.034) expression was additionally associated with poor progression-free survival. CONCLUSIONS: CHK1 and CHK2 and their activated forms are frequently expressed in HGSC effusions, with higher expression following exposure to chemotherapy, and their expression is related to survival.


Subject(s)
Checkpoint Kinase 1/metabolism , Checkpoint Kinase 2/biosynthesis , Checkpoint Kinase 2/metabolism , Cystadenocarcinoma, Serous/enzymology , Adult , Aged , Aged, 80 and over , Checkpoint Kinase 1/biosynthesis , Checkpoint Kinase 1/genetics , Checkpoint Kinase 2/genetics , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Enzyme Activation , Female , Humans , Middle Aged , Neoplasm Grading , Survival Analysis , Young Adult
5.
Oncotarget ; 7(33): 53881-53894, 2016 Aug 16.
Article in English | MEDLINE | ID: mdl-27449089

ABSTRACT

Checkpoint kinase 1 (CHK1) is an integral component of the cell cycle as well as the DNA Damage Response (DDR) pathway. Previous work has demonstrated the effectiveness of inhibiting CHK1 with small-molecule inhibitors, but the role of CHK1 mediated DDR in medulloblastoma is unknown. CHK1, both at the mRNA and protein level, is highly expressed in medulloblastoma and elevated CHK1 expression in Group3 medulloblastoma is an adverse prognostic marker. CHK1 inhibition with the small-molecule drug AZD7762, results in decreased cell growth, increased DNA damage and cell apoptosis. Furthermore, AZD7762 acts in synergy with cisplatin in reducing cell proliferation in medulloblastoma. Similar phenotypic changes were observed with another CHK1 inhibitor, PF477736, as well as genetic knockdown using siRNA against CHK1. Treatments with small-molecule inhibitors of CHK1 profoundly modulated the expression of both upstream and downstream target proteins within the CHK1 signaling pathways. This suggests the presence of a feedback loop in activating CHK1. Overall, our results demonstrate that small-molecule inhibition of CHK1 in combination with, cisplatin, is more advantageous than either treatment alone, especially for Group 3 medulloblastoma, and therefore this combined therapeutic approach serves as an avenue for further investigation.


Subject(s)
Biomarkers, Tumor/analysis , Cerebellar Neoplasms/pathology , Checkpoint Kinase 1/biosynthesis , Medulloblastoma/pathology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Benzodiazepinones/pharmacology , Biomarkers, Tumor/metabolism , Cell Survival/drug effects , Cerebellar Neoplasms/enzymology , Cerebellar Neoplasms/mortality , Cisplatin/pharmacology , Disease-Free Survival , Genes, myc , Humans , Kaplan-Meier Estimate , Medulloblastoma/enzymology , Medulloblastoma/mortality , Prognosis , Pyrazoles/pharmacology , Thiophenes/pharmacology , Urea/analogs & derivatives , Urea/pharmacology
6.
Oncotarget ; 7(29): 45776-45788, 2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27329729

ABSTRACT

The search for preoperative biomarkers for thyroid malignancies, in particular for follicular thyroid carcinoma (FTC) diagnostics, is of utmost clinical importance. We thus aimed at screening for potential biomarker candidates for FTC. To evaluate dynamic alterations in molecular patterns as a function of thyroid malignancy progression, a comparative analysis was conducted in clinically distinct subgroups of FTC and poorly differentiated thyroid carcinoma (PDTC) nodules. NanoString analysis of FFPE samples was performed in 22 follicular adenomas, 56 FTC and 25 PDTC nodules, including oncocytic and non-oncocytic subgroups. The expression levels of CHEK1, c-KIT, SLC26A4, TG and TPO were significantly altered in all types of thyroid carcinomas. Based on collective changes of these biomarkers which correlating among each other, a predictive score has been established, allowing for discrimination between benign and FTC samples with high sensitivity and specificity. Additional transcripts related to thyroid function, cell cycle, circadian clock, and apoptosis regulation were altered in the more aggressive oncocytic subgroups only, with expression levels correlating with disease progression. Distinct molecular patterns were observed for oncocytic and non-oncocytic FTCs and PDTCs. A predictive score correlation coefficient based on collective alterations of identified here biomarkers might help to improve the preoperative diagnosis of FTC nodules.


Subject(s)
Adenocarcinoma, Follicular/metabolism , Biomarkers, Tumor/analysis , Thyroid Neoplasms/metabolism , Transcriptome , Autoantigens/analysis , Autoantigens/biosynthesis , Checkpoint Kinase 1/analysis , Checkpoint Kinase 1/biosynthesis , Gene Expression Profiling , Humans , Iodide Peroxidase/analysis , Iodide Peroxidase/biosynthesis , Iron-Binding Proteins/analysis , Iron-Binding Proteins/biosynthesis , Membrane Transport Proteins/analysis , Membrane Transport Proteins/biosynthesis , Microfilament Proteins/analysis , Microfilament Proteins/biosynthesis , Muscle Proteins/analysis , Muscle Proteins/biosynthesis , Proto-Oncogene Proteins c-kit/analysis , Proto-Oncogene Proteins c-kit/biosynthesis , Sulfate Transporters
7.
Sci Rep ; 6: 21519, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26867682

ABSTRACT

Radiation has a limited but relevant role in the adjuvant therapy of gastric cancer (GC) patients. Since Chk1 plays a critical function in cellular response to genotoxic agents, we aimed to analyze the role of Chk1 in GC as a biomarker for radiotherapy resistance. We analyzed Chk1 expression in AGS and MKN45 human GC cell lines by RT-QPCR and WB and in a small cohort of human patient's samples. We demonstrated that Chk1 overexpression specifically increases resistance to radiation in GC cells. Accordingly, abrogation of Chk1 activity with UCN-01 and its expression with shChk1 increased sensitivity to bleomycin and radiation. Furthermore, when we assessed Chk1 expression in human samples, we found a correlation between nuclear Chk1 accumulation and a decrease in progression free survival. Moreover, using a luciferase assay we found that Chk1's expression is controlled by p53 and RB/E2F1 at the transcriptional level. Additionally, we present preliminary data suggesting a posttranscriptional regulation mechanism, involving miR-195 and miR-503, which are inversely correlated with expression of Chk1 in radioresistant cells. In conclusion, Chk1/microRNA axis is involved in resistance to radiation in GC, and suggests Chk1 as a potential tool for optimal stratification of patients susceptible to receive adjuvant radiotherapy after surgery.


Subject(s)
Checkpoint Kinase 1/biosynthesis , Chemoradiotherapy , E2F1 Transcription Factor/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Retinoblastoma Binding Proteins/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/therapy , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Bleomycin/pharmacology , Cell Line, Tumor , Checkpoint Kinase 1/genetics , E2F1 Transcription Factor/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/radiation effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Retinoblastoma Binding Proteins/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...