Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Tumour Biol ; 35(3): 1899-906, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24155212

ABSTRACT

The aim of this study was to examine the prognostic value of BRCA1, RRM1, and RRM2 in patients with non-small cell lung cancer (NSCLC) who received adjuvant chemotherapy. A total of 418 patients who underwent curative pulmonary resection were obtained between January 2007 and November 2009. The relative cDNA quantification for BRCA1, RRM1, and RRM2 was conducted using a fluorescence-based, real-time detection method, and ß-actin was used as a reference gene. The low expression of RRM1 and RRM2 significantly increased the platinum-based chemotherapy response (For RRM1: odds ratio (OR) = 2.09, 95% confidence interval (CI) = 1.38-3.18; For RRM2: OR = 1.64, 95% CI = 1.09-2.48). The univariate analysis indicated that low expression of RRM1 attained a longer time to progression and overall survival time, with HR (95% CI) of 0.50 (0.33-0.77) and 0.60 (0.39-0.92), respectively. Similarly, low expression of RRM2 had a longer time to progression and overall survival, with HR (95% CI) of 0.57 (0.38-0.86) and 0.47 (0.31-0.71), respectively. In conclusion, low expression of RRM1 and RRM2 could be used to predict the treatment response to platinum-based chemotherapy and survival in NSCLC. The RRM1 and RRM2 could substantially contribute to the future design of individualized cancer treatment in NSCLC patients.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Ribonucleoside Diphosphate Reductase/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/mortality , Cisplatin/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Male , Middle Aged , Paclitaxel/administration & dosage , Prognosis , Proportional Hazards Models , Ribonucleoside Diphosphate Reductase/analysis , Tumor Suppressor Proteins/analysis , Vinblastine/administration & dosage , Vinblastine/analogs & derivatives , Vinorelbine , Gemcitabine
2.
Anticancer Agents Med Chem ; 21(16): 2204-2215, 2021 10 28.
Article in English | MEDLINE | ID: mdl-33397269

ABSTRACT

BACKGROUND: One of the main reasons for the poor survival rates of pancreatic cancer patients is the development of gemcitabine resistance, indicating that novel treatment strategies that have the ability to improve gemcitabine sensitivity are in need to combat this devastating disease. METHODS: TCGA PAAD data was used to determine the clinicopathological significance of high RRM2 (Ribonucleotide reductase subunit M2) expression for Pancreatic Ductal Adenocarcinoma (PDAC). The effects of GW8510 and gemcitabine on PANC-1 cell viability were determined using WST-8 assay. The potential synergistic interaction between GW8510 and gemcitabine was evaluated by the Combination Index (CI) analysis. The effects of GW8510 treatment on apoptosis, cell cycle, and cell migration, either in combination with gemcitabine or alone, were investigated. The effect of GW8510 on RRM2 protein levels was evaluated using ELISA assay. RESULTS: RRM2 is significantly over-expressed in PDAC compared to healthy pancreatic tissues (p <0.0001). RRM2 mRNA expression was found to be significantly correlated with the overall survival rate of patients (HR=2.17 [1.44-3.27], p=0.00016) and the pathological stages of the disease (p=0.0054). GW8510 significantly decreased the RRM2 protein levels compared to the control. Cell viability analysis showed that GW8510 has a similar effect to gemcitabine in inhibiting PANC-1 cell viability. GW8510 was found to synergize with gemcitabine to inhibit PANC-1 cell viability and migration. However, the effects of GW8510 on PANC-1 cells could not be explained by induction of apoptosis or cell cycle arrest. CONCLUSION: Targeting RRM2 using GW8510 may have the potential to increase gemcitabine sensitivity in pancreatic cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Deoxycytidine/analogs & derivatives , Indoles/pharmacology , Pancreatic Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Biomarkers, Tumor/analysis , Cell Proliferation/drug effects , Cell Survival/drug effects , Computational Biology , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Drug Screening Assays, Antitumor , Humans , Indoles/chemistry , Pancreatic Neoplasms/diagnosis , Ribonucleoside Diphosphate Reductase/analysis , Tumor Cells, Cultured , Gemcitabine
3.
Aging (Albany NY) ; 13(3): 3518-3535, 2020 12 19.
Article in English | MEDLINE | ID: mdl-33411689

ABSTRACT

Ribonucleotide reductase subunit M2 may play a role as a potential prognostic biomarker in several cancers. In this study, we evaluated whether RRM2 gene expression is associated with the prognosis of patients with lung adenocarcinoma (LUAD) using publicly available data from The Cancer Genome Atlas (TCGA). Wilcoxon signed-rank test and logistic regression were performed to evaluate the association between RRM2 expression and clinical features in patients with LUAD. Kaplan-Meier and Cox regression methods were used to examine the effect of RRM2 expression level in the overall survival, and a nomogram was performed to illustrate the correlation between the RRM2 gene expression and the risk of LUAD. TCGA data set was used for gene set enrichment analysis (GSEA). We also performed a further experiment in vitro to assess the effect of RRM2 expression on the proliferation and invasive abilities of LUAD cells and its key signaling pathway proteins. Our results revealed that the expression level of RRM2 in patients with LUAD was much higher than that in normal tissues (p = 3.99e-32). High expression of RRM2 was significantly associated with tumor stage (IV vs. I: OR = 3.02, p = 0.012) and TNM classification (T2 vs. T1: OR = 1.88, p = 0.001; N2 vs. N0: OR = 2.69, p < 0.001). Kaplan-Meier survival analysis showed that high expression of RRM2 was associated with a worse prognosis of LUAD compared low expression of RRM2 (p = 7.86e-04). Multivariate analysis showed that high RRM2 expression was an independent factor affecting overall survival (HR = 1.29, p < 0.001). The association between RRM2 gene expression and the risk of LUAD was presented in a nomogram. GSEA revealed that the cell cycle, p53 signaling pathway, DNA replication, small cell lung cancer, apoptosis, and pathways in cancer were differentially enriched in patients with high expression of RRM2. RRM2 over-expression promoted the proliferation and invasive abilities of LUAD cells. RRM2 over-expression increased the activation of Bcl-2 and E-cadherin signaling pathways, and reduced the activation of p53 signaling pathway. In summary, high RRM2 expression is an independent predictive factor of poor prognosis in patients with lung adenocarcinoma.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Ribonucleoside Diphosphate Reductase , Adenocarcinoma of Lung/diagnosis , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/mortality , Adenocarcinoma of Lung/pathology , Aged , Cell Cycle/genetics , Cell Line , Female , Humans , Lung/metabolism , Lung/pathology , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Prognosis , Ribonucleoside Diphosphate Reductase/analysis , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleoside Diphosphate Reductase/metabolism , Signal Transduction/genetics
4.
Mol Cell Biol ; 38(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29263158

ABSTRACT

A tightly controlled cellular deoxyribonucleotide (deoxynucleoside triphosphate [dNTP]) pool is critical for maintenance of genome integrity. One mode of dNTP pool regulation is through subcellular localization of ribonucleotide reductase (RNR), the enzyme that catalyzes the rate-limiting step of dNTP biosynthesis. In Saccharomyces cerevisiae, the RNR small subunit, Rnr2-Rnr4, is localized to the nucleus, whereas the large subunit, Rnr1, is cytoplasmic. As cells enter S phase or encounter DNA damage, Rnr2-Rnr4 relocalizes to the cytoplasm to form an active holoenzyme complex with Rnr1. Although the DNA damage-induced relocalization requires the checkpoint kinases Mec1-Rad53-Dun1, the S-phase-specific redistribution does not. Here, we report that the S-phase cyclin-cyclin-dependent kinase (CDK) complex Clb6-Cdc28 controls Rnr2-Rnr4 relocalization in S phase. Rnr2 contains a consensus CDK site and exhibits Clb6-dependent phosphorylation in S phase. Deletion of CLB6 or removal of the CDK site results in an increased association of Rnr2 with its nuclear anchor Wtm1, nuclear retention of Rnr2-Rnr4, and an enhanced sensitivity to the RNR inhibitor hydroxyurea. Thus, we propose that Rnr2-Rnr4 redistribution in S phase is triggered by Clb6-Cdc28-mediated phosphorylation of Rnr2, which disrupts the Rnr2-Wtm1 interaction and promotes the release of Rnr2-Rnr4 from the nucleus.


Subject(s)
CDC28 Protein Kinase, S cerevisiae/metabolism , Cyclin B/metabolism , Ribonucleoside Diphosphate Reductase/metabolism , Ribonucleotide Reductases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , CDC28 Protein Kinase, S cerevisiae/analysis , Cyclin B/analysis , Phosphorylation , Protein Transport , Ribonucleoside Diphosphate Reductase/analysis , Ribonucleotide Reductases/analysis , S Phase , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/analysis
5.
Oncogene ; 23(21): 3790-801, 2004 May 06.
Article in English | MEDLINE | ID: mdl-15094776

ABSTRACT

Translation initiation in eukaryotes is a rate-limiting step in protein synthesis. It is a complicated process that involves many eukaryotic initiation factors (eIFs). Altering the expression level or the function of eIFs may influence the synthesis of some proteins and consequently cause abnormal cell growth and malignant transformation. P170, the largest putative subunit of eIF3, has been found elevated in human breast, cervical, esophageal, and lung cancers, suggesting that p170 may have a potential role in malignant transformation and/or cell growth control. Our recent studies suggested that p170 is likely a translational regulator and it may mediate the effect of mimosine on the translation of a subset mRNAs. Mimosine, a plant nonprotein amino acid, inhibits mammalian DNA synthesis, an essential event of cell growth. The rate-limiting step in DNA synthesis is the conversion of the ribonucleotides to their corresponding deoxyribonucleotides catalysed by ribonucleotide reductase of which the activity is regulated by the level of its M2 subunit. It has been reported that inhibiting the activity of M2 also inhibits cell growth. To understand the relationship between protein and DNA synthesis and between p170 and cell growth control, we investigated in this study whether p170 regulates the synthesis of M2 and, thus, cell growth. We found that altering the expression level of p170 changes the synthesis rate of both M2 and DNA. Decreasing p170 expression in human lung cancer cell line H1299 and breast cancer cell line MCF7 significantly reversed their malignant growth phenotype. However, the overall [35S]methionine incorporation following dramatic decrease in p170 expression was only approximately 25% less than the control cells. These observations, together with our previous findings, suggest that p170 may regulate the translation of a subset mRNAs and its elevated expression level may be important for cancer cell growth and for maintaining their malignant phenotype.


Subject(s)
Eukaryotic Initiation Factor-3/physiology , Ribonucleoside Diphosphate Reductase/biosynthesis , 5' Untranslated Regions/metabolism , Animals , Cell Division , Cell Line, Tumor , DNA/biosynthesis , DNA, Antisense/pharmacology , Eukaryotic Initiation Factor-3/antagonists & inhibitors , Humans , Mice , Mimosine/pharmacology , NIH 3T3 Cells , Protein Biosynthesis , Protein Subunits , Ribonucleoside Diphosphate Reductase/analysis , Transfection
6.
Pharmacol Ther ; 29(1): 111-27, 1985.
Article in English | MEDLINE | ID: mdl-3914643

ABSTRACT

It would be expected that drugs directed at the rate-limiting step in a key metabolic pathway in tumor cell proliferation would provide a useful basis for therapy of neoplasms. Ribonucleotide reductase catalyzes the rate-limiting step in the de novo synthesis of dNTP's for DNA synthesis. Further, ribonucleotide reductase is composed of two non-identical protein subunits (non-heme iron and effector-binding subunits) which can be specifically and independently inhibited. As a result, combinations of drugs specifically directed at each of the subunits of ribonucleotide reductase have been shown to cause synergistic inhibition of L1210 cell growth in culture and synergistic cell kill. This approach offers a novel basis for the design of combination chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Ribonucleoside Diphosphate Reductase/analysis , Ribonucleotide Reductases/analysis , Animals , Cytarabine/pharmacology , Humans , Kinetics , Nucleic Acid Synthesis Inhibitors , Pyrimidines/pharmacology , Ribonucleoside Diphosphate Reductase/antagonists & inhibitors , Ribonucleoside Diphosphate Reductase/metabolism
7.
Int J Clin Exp Pathol ; 5(4): 347-55, 2012.
Article in English | MEDLINE | ID: mdl-22670179

ABSTRACT

BACKGROUND: Ribonucleotide reductase M2 (RRM2) was associated with pancreatic tumor progression and resistance to gemcitabine. This study aimed to determine if RRM2 protein expression was prognostic in patients with resectable pancreatic adenocarcinoma and predictive of adjuvant gemcitabine benefit. METHODS: 117 patients underwent tumor resection for pancreatic adenocarcinoma from 10/1999 to 12/2007. We constructed tissue microarrays from paraffin-embedded tumors and determined RRM2 protein expression using immunohistochemistry and grouped as negative or positive. We estimated overall survival (OS) and progression-free survival (PFS) using the Kaplan-Meier method and examined the prognostic and predictive value of RRM2 expression using Cox proportional hazards model. RESULTS: RRM2 expression showed no prognostic value in the entire group regarding OS (median OS 30.9 months in RRM2-positive versus 13.7 months in RRM2-negative, P = 0.26) and PFS (median OS 20.6 months in RRM2-positive versus 11.8 months in RRM2-negative, P = 0.46). RRM2 expression did not predict adjuvant gemcitabine benefit in the subgroup of 44 patients who received gemcitabine therapy (median OS 31.2 versus 15.2 months, P = 0.62; median PFS 11.3 versus 14.0 months, P = 0.35). Cox proportional hazards regression showed no prognostic effect of RRM2 expression on OS and PFS in the subgroup of 44 patients. However, the number of positive lymph nodes and perineural invasion were prognostic factors for OS (HR 1.2, P = 0.005) and for PFS (HR 5.5, P = 0.007), respectively. CONCLUSION: RRM2 protein expression in pancreatic adenocarcinoma is neither prognostic nor predictive of adjuvant gemcitabine benefit in patients with resectable pancreatic adenocarcinoma.


Subject(s)
Carcinoma, Pancreatic Ductal/surgery , Pancreatectomy , Pancreatic Neoplasms/surgery , Ribonucleoside Diphosphate Reductase/analysis , Adult , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/therapeutic use , Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/secondary , Chemotherapy, Adjuvant , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Disease-Free Survival , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Ohio , Pancreatectomy/adverse effects , Pancreatectomy/mortality , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Paraffin Embedding , Predictive Value of Tests , Proportional Hazards Models , Retrospective Studies , Risk Assessment , Risk Factors , Time Factors , Tissue Array Analysis , Treatment Outcome , Gemcitabine
9.
J Bacteriol ; 130(2): 957-9, 1977 May.
Article in English | MEDLINE | ID: mdl-400802

ABSTRACT

Ribonucleoside diphosphate reductase subunits B1 and B2 and ether-permeabilized cell activities of Escherichia coli increase in parallel during thymine deprivation. Thioredoxin and thioredoxin reductase activities are not affected by thymine deprivation.


Subject(s)
Escherichia coli/enzymology , Ribonucleoside Diphosphate Reductase/biosynthesis , Ribonucleotide Reductases/biosynthesis , Ribonucleoside Diphosphate Reductase/analysis , Thymine Nucleotides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL