Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Dis Markers ; 2021: 5089872, 2021.
Article in English | MEDLINE | ID: mdl-34868394

ABSTRACT

OBJECTIVE: Ribonucleotide reductase M2 (RRM2) as an enzyme that catalyzes the deoxyreduction of nucleosides to deoxyribonucleoside triphosphate (dNTP) has been extensively studied, and it plays a crucial role in regulating cell proliferation. However, its role in ischemia-reperfusion injury (I/RI) is still unclear. METHODS: SD rats were used as the research object to detect the expression of RRM2 in the myocardium by constructing an I/RI model. At the same time, primary SD neonatal rat cardiomyocytes were extracted, and hypoxia/reoxygenation (H/R) treatment simulated the I/RI model. Using transfection technology to overexpress RRM2 in cardiomyocytes, quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) was used to detect the expression of RRM2, Cell Counting Kit-8 (CCK-8) assay was used to detect cell viability, and immunofluorescence staining was used to detect Ki67 and EdU-positive cells. Western blot (WB) technology was used to detect YAP and its phosphorylation expression. RESULTS: qRT-PCR results indicated that the expression of RRM2 was inhibited in the model group, and cardiomyocytes overexpressing RRM2 can obviously promote the proliferation of primary cardiomyocytes and improve the damage of cardiac structure and function caused by I/R. At the same time, RRM2 can promote the increase of YAP protein expression and the increase of Cyclin D1 mRNA expression. CONCLUSION: RRM2 expression was downregulated in myocardial tissue with I/R. After overexpression of RRM2, cardiomyocyte proliferation was upregulated and the Hippo-YAP signaling pathway was activated.


Subject(s)
Cell Proliferation/physiology , Hippo Signaling Pathway , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/cytology , Ribonucleoside Diphosphate Reductase/physiology , YAP-Signaling Proteins/metabolism , Animals , Humans , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
2.
Int J Mol Med ; 42(5): 2393-2402, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30106091

ABSTRACT

Rheumatoid arthritis (RA) is considered to be a systemic autoimmune disease that induces systemic complications and progressive disability. It affects a large number of people. RA fibroblast­like synoviocytes (RA­FLS) promote the progression of RA through the secretion of proinflammatory cytokines and increasing invasiveness into the extracellular matrix. Therefore, targeting RA­FLS represents a potential approach for the treatment of RA. Ribonucleotide reductase M2 (RRM2), a critical protein for DNA synthesis and repair, may promote the proliferation of cells and inhibit cellular apoptosis. In previous studies it has been confirmed that the suppression of RRM2 markedly suppressed the proliferation of liver cancer cells. In the present study, a cell permeable peptide­conjugated liposome­polycation­DNA (LPD) complex loaded with RRM2 small interfering RNA (siRNA) (CCP­LPDR) was developed, aiming to increase the levels of apoptosis and inhibit the proliferation of RA­FLS. CCP­LPDR is a small­sized molecule (~130 nm) with high encapsulation efficiency of siRNA (>90%) and high stability. Furthermore, it was verified that CCP­LPDR markedly suppressed RRM2 gene and protein expression by ~80%. Notably, CCP­LPDR efficiently targeted RA­FLS, resulting in a marked decrease in the proliferation and increase in the level of apoptosis in RA­FLS. In addition, the levels of proinflammatory cytokines tumor necrosis factor­α and interleukin­6 were markedly decreased in RA­FLS following CCP­LPDR treatment. Therefore, CCP­LPDR may efficiently deliver RRM2 to RA­FLS and represent a potential treatment for RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/therapy , Liposomes/chemistry , Peptides/chemistry , Protamines/chemistry , RNA, Small Interfering/physiology , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleoside Diphosphate Reductase/physiology , Synoviocytes/metabolism , Apoptosis/genetics , Apoptosis/physiology , Arthritis, Rheumatoid/genetics , Blotting, Western , Cell Line , Cell Proliferation/genetics , Cell Proliferation/physiology , Cell Survival/genetics , Cell Survival/physiology , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , RNA, Small Interfering/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
3.
Clin Sci (Lond) ; 132(13): 1417-1433, 2018 07 16.
Article in English | MEDLINE | ID: mdl-29853661

ABSTRACT

Although chemotherapeutic regimen containing gemcitabine is the first-line therapy for advanced lung squamous cell carcinoma (LSCC), gemcitabine resistance remains an important clinical problem. Some studies suggest that overexpressions of ribonucleotide reductase (RNR) subunit M2 (RRM2) may be involved in gemcitabine resistance. We used a novel RRM2 inhibitor, GW8510, as a gemcitabine sensitization agent to investigate the therapeutic utility in reversing gemcitabine resistance in LSCC. Results showed that the expressions of RRM2 were increased in gemcitabine intrinsic resistant LSCC cells upon gemcitabine treatment. GW8510 not only suppressed LSCC cell survival, but also sensitized gemcitabine-resistant cells to gemcitabine through autophagy induction mediated by RRM2 down-regulation along with decrease in dNTP levels. The combination of GW8510 and gemcitabine produced a synergistic effect on killing LSCC cells. The synergism of the two agents was impeded by addition of autophagy inhibitors chloroquine (CQ) or bafilomycin A1 (Baf A1), or knockdown of the autophagy gene, Bcl-2-interacting protein 1 (BECN1). Moreover, GW8510-caused LSCC cell sensitization to gemcitabine through autophagy induction was parallel with impairment of DNA double-strand break (DSB) repair and marked increase in cell apoptosis, revealing a cross-talk between autophagy and DNA damage repair, and an interplay between autophagy and apoptosis. Finally, gemcitabine sensitization mediated by autophagy induction through GW8510-caused RRM2 down-regulation was demonstrated in vivo in gemcitabine-resistant LSCC tumor xenograft, further indicating that the sensitization is dependent on autophagy activation. In conclusion, GW8510 can reverse gemcitabine resistance in LSCC cells through RRM2 downregulation-mediated autophagy induction, and GW850 may be a promising therapeutic agent against LSCC as it combined with gemcitabine.


Subject(s)
Autophagy/drug effects , Carcinoma, Squamous Cell/drug therapy , Deoxycytidine/analogs & derivatives , Indoles/pharmacology , Lung Neoplasms/drug therapy , Ribonucleoside Diphosphate Reductase/antagonists & inhibitors , Animals , Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Deoxycytidine/pharmacology , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Male , Mice, Inbred NOD , Ribonucleoside Diphosphate Reductase/physiology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Gemcitabine
4.
Curr Genet ; 64(3): 547-550, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29119271

ABSTRACT

Telomeres, the nucleoprotein complexes at the end of eukaryotic chromosomes, protect them from degradation and ensure the replicative capacity of cells. In most human tumors and in budding yeast, telomere length is maintained by the activity of telomerase, an enzyme that adds dNTPs according to an internal RNA template. The dNTPs are generated with the help of the ribonucleotide reductase (RNR) complex. We have recently generated strains lacking the large subunit of RNR, Rnr1, which were kept viable by the expression of RNR complexes containing the Rnr1 homolog, Rnr3. Interestingly, we found that these Rnr1-deficient strains have short telomeres that are stably maintained, but cannot become efficiently elongated by telomerase. Thus, a basic maintenance of short telomeres is possible under conditions, where Rnr1 activity is absent, but a sustained elongation of short telomeres fully depends on Rnr1 activity. We show that Rnr3 cannot compensate for this telomeric function of Rnr1 even when overall cellular dNTP values are restored. This suggests that Rnr1 plays a role in telomere elongation beyond increasing cellular dNTP levels. Furthermore, our data indicate that telomerase may act in two different modes, one that is capable of coping with the "end-replication problem" and is functional even in the absence of Rnr1 and another required for the sustained elongation of short telomeres, which fully depends on the presence of Rnr1. Supply of dNTPs for telomere elongation is provided by the Mec1ATR checkpoint, both during regular DNA replication and upon replication fork stalling. We discuss the implications of these results on telomere maintenance in yeast and cancer cells.


Subject(s)
Deoxyribonucleotides/physiology , Ribonucleoside Diphosphate Reductase/physiology , Ribonucleotide Reductases/physiology , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/genetics , Telomere Homeostasis/physiology , Humans , Neoplasms/genetics , Ribonucleotide Reductases/metabolism , Telomerase/metabolism
5.
Tumour Biol ; 37(3): 3515-26, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26453115

ABSTRACT

Ribonucleotide reductase (RR) has been reported to be associated with several types of cancer while the expression and role of RR in thyroid carcinoma (TC) has not been investigated. Here, we first examined the expression level of three RR subunit proteins (RRM1, RRM2, and RRM2B) in papillary thyroid carcinoma (PTC) and undifferentiated thyroid carcinoma (UTC) patient samples by immunohistochemistry. The results showed that RRM1 was higher expressed in 95.2 % cancer tissues compared with their adjacent normal tissues in 146 PTC samples. The expression level of RRM1 was positively correlated with T stage, lymph node metastasis (LNM), extrathyroidal invasion (ETI), and TNM stage in PTC patients. However, in 12 UTC samples, RRM1 expression was negatively expressed in six cases. To further determine the biological role of RRM1 in TC, ectopic expression or siRNA-mediated knockdown of RRM1 were carried out in the high-differentiated thyroid carcinoma cell line TPC-1 and the poor-differentiated thyroid carcinoma cell line SW579, respectively. In TPC-1 and SW579 cells, overexpression and siRNA knockdown of RRM1 demonstrated that RRM1 promoted DNA synthesis and proliferation in both cell lines as shown by EdU incorporation and cell viability assays. However, RRM1 enhanced cell migration and invasion in TPC-1 cells but inhibited that in SW579 cells as shown by wound healing and transwell assays. Moreover, we also found that RRM1 promoted PTEN expression and reduced Akt phosphorylation in a RR-activity-independent manner in the low-differentiated TC cells but not in the high-differentiated TC cells. In contrast, RRM2 expression was higher expressed in both PTC and UTC patient samples, consisting with its oncogenic role in other cancers. Therefore, we suggest that RRM1 promotes thyroid carcinoma proliferation as a component of RR but may play a different role in the invasion and metastasis of differently differentiated thyroid carcinomas through a non-RR pathway, which could be meaningful to precision treatment of thyroid carcinoma with RR inhibitors.


Subject(s)
Carcinoma/pathology , Thyroid Neoplasms/pathology , Tumor Suppressor Proteins/physiology , Adult , Aged , Carcinoma, Papillary , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , PTEN Phosphohydrolase/physiology , Ribonucleoside Diphosphate Reductase/physiology , Thyroid Cancer, Papillary
6.
World J Gastroenterol ; 18(34): 4704-13, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-23002339

ABSTRACT

AIM: To investigate the roles of the ribonucleotide reductase M2 (RRM2) subunit in colorectal cancer (CRC) and ultraviolet (UV)-induced DNA damage repair. METHODS: Immunohistochemical staining of tissue microarray was performed to detect the expression of RRM2. Seven CRC cell lines were cultured and three human colon cancer cell lines, i.e., HCT116, SW480 and SW620, were used. Reverse transcription polymerase chain reaction and Western blotting were performed to determine the mRNA and protein expression levels of RRM2, respectively. Cell proliferation assay, cell cycle analysis were performed. Cell apoptosis was evaluated by double staining with fluorescein isothiocyanate-conjugated Annexin V and propidium iodide (PI) using Annexin V/PI apoptosis kit. The motility and invasion of CRC cells were assessed by the Transwell chamber assay. Cells were irradiated with a 254 nm UV-C lamp to detect the UV sensitivity after RRM2 depletion. RESULTS: Immunohistochemical staining revealed elevated RRM2 levels in CRC tissues. RRM2 overexpression was positively correlated with invasion depth (P < 0.05), poorly differentiated type (P = 0.0051), and tumor node metastasis stage (P = 0.0015). The expression of RRM2 in HCT116 cells was downregulated after transfection, and HCT116 cell proliferation was obviously suppressed compared to control groups (P < 0.05). In the invasion test, the number of cells that passed through the chambers in the RRM2-siRNA group was 81 ± 3, which was lower than that in the negative control (289 ± 7) and blank control groups (301 ± 7.2). These differences were statistically significant (P < 0.01). Our data suggest that RRM2 overexpression may be associated with CRC progression. RRM2 silencing by siRNA may inhibit the hyperplasia and invasiveness of CRC cells, suggesting that RRM2 may play an important role in the infiltration and metastasis of CRC, which is a potential therapeutic strategy in CRC. In addition, RRM2 depletion increased UV sensitivity. CONCLUSION: These findings suggest that RRM2 may be a facilitating factor in colorectal tumorigenesis and UV-induced DNA damage repair.


Subject(s)
Colorectal Neoplasms/etiology , DNA Repair , Ribonucleoside Diphosphate Reductase/physiology , Ultraviolet Rays/adverse effects , Adult , Aged , Cell Cycle Proteins/physiology , Cell Line, Tumor , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Ribonucleoside Diphosphate Reductase/antagonists & inhibitors , Polo-Like Kinase 1
7.
Biochemistry ; 51(1): 205-13, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-22166006

ABSTRACT

Urokinase-type plasminogen activator (uPA) is expressed by lung epithelial cells and regulates fibrin turnover and epithelial cell viability. PMA, LPS, and TNF-alpha, as well as uPA itself, induce uPA expression in lung epithelial cells. PMA, LPS, and TNF-alpha induce uPA expression through increased synthesis as well as stabilization of uPA mRNA, while uPA increases its own expression solely through uPA mRNA stabilization. The mechanism by which lung epithelial cells regulate uPA expression at the level of mRNA stability is unclear. To elucidate this process, we sought to characterize protein-uPA mRNA interactions that regulate uPA expression. Regulation of uPA at the level of mRNA stability involves the interaction of a ~40 kDa cytoplasmic-nuclear shuttling protein with a 66 nt uPA mRNA 3'UTR sequence. We purified the uPA mRNA 3'UTR binding protein and identified it as ribonucleotide reductase M2 (RRM2). We expressed recombinant RRM2 and confirmed its interaction with a specific 66 nt uPA 3'UTR sequence. Immunoprecipitation of cell lysates with anti-RRM2 antibody and RT-PCR for uPA mRNA confirmed that RRM2 binds to uPA mRNA. Treatment of Beas2B cells with uPA or LPS attenuated RRM2-endogenous uPA mRNA interactions, while overexpression of RRM2 inhibited uPA protein and mRNA expression through destabilization of uPA mRNA. LPS exposure of lung epithelial cells translocates RRM2 from the cytoplasm to the nucleus in a time-dependent manner, leading to stabilization of uPA mRNA. This newly recognized pathway could influence uPA expression and a broad range of uPA-dependent functions in lung epithelial cells in the context of lung inflammation and repair.


Subject(s)
Lung/enzymology , RNA Processing, Post-Transcriptional/genetics , Respiratory Mucosa/enzymology , Ribonucleoside Diphosphate Reductase/physiology , Urokinase-Type Plasminogen Activator/biosynthesis , Animals , Cell Line , Cells, Cultured , Enzyme Stability/drug effects , Enzyme Stability/genetics , Humans , Lipopolysaccharides/pharmacology , Lung/cytology , Lung/drug effects , Mice , RNA Stability/drug effects , RNA Stability/genetics , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Ribonucleoside Diphosphate Reductase/biosynthesis , Ribonucleoside Diphosphate Reductase/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/genetics
8.
Anticancer Res ; 31(7): 2535-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21873171

ABSTRACT

BACKGROUND: We previously investigated the mRNA expression of colorectal cancer cell lines via a microarray analysis and found several genes that were significantly up-regulated by oncogenic KRAS under serum-starved conditions. Of these genes, we focused on ribonucleotide reductase M2 (RRM2), which was reported to be associated with DNA synthesis. MATERIALS AND METHODS: Cell proliferation and colony formation assays were performed using HCT116 cells transfected with lentiviral RRM2-shRNAs. RESULTS: Under serum-starved conditions, the expression level of RRM2 protein increased in HCT116 cells compared to HKe3 cells (HCT116 cells with a disruption in oncogenic KRAS), and the re-expression of KRAS in HKe3 cells induced the expression of RRM2. Both the cell proliferation under serum-depleted conditions and the anchorage-independent growth were impaired by the reduction of RRM2 protein expression. CONCLUSION: RRM2 represents a novel therapeutic target, thus highlighting the potential utility of RRM2 inhibitors in colorectal cancer with oncogenic KRAS.


Subject(s)
Adenocarcinoma/pathology , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/physiology , Oncogene Proteins/physiology , Proto-Oncogene Proteins/physiology , Ribonucleoside Diphosphate Reductase/biosynthesis , ras Proteins/physiology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Cell Adhesion , Cell Culture Techniques , Cell Division , Cell Line, Tumor/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Culture Media, Serum-Free , Genes, ras , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Oncogene Proteins/genetics , Point Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , RNA, Small Interfering/pharmacology , Ribonucleoside Diphosphate Reductase/antagonists & inhibitors , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleoside Diphosphate Reductase/physiology , Tumor Stem Cell Assay , Up-Regulation , ras Proteins/genetics
9.
J Bacteriol ; 189(9): 3496-501, 2007 May.
Article in English | MEDLINE | ID: mdl-17322311

ABSTRACT

The observed lengthening of the C period in the presence of a defective ribonucleoside diphosphate reductase has been assumed to be due solely to the low deoxyribonucleotide supply in the nrdA101 mutant strain. We show here that the nrdA101 mutation induces DNA double-strand breaks at the permissive temperature in a recB-deficient background, suggesting an increase in the number of stalled replication forks that could account for the slowing of replication fork progression observed in the nrdA101 strain in a Rec(+) context. These DNA double-strand breaks require the presence of the Holliday junction resolvase RuvABC, indicating that they have been generated from stalled replication forks that were processed by the specific reaction named "replication fork reversal." Viability results supported the occurrence of this process, as specific lethality was observed in the nrdA101 recB double mutant and was suppressed by the additional inactivation of ruvABC. None of these effects seem to be due to the limitation of the deoxyribonucleotide supply in the nrdA101 strain even at the permissive temperature, as we found the same level of DNA double-strand breaks in the nrdA(+) strain growing under limited (2-microg/ml) or under optimal (5-microg/ml) thymidine concentrations. We propose that the presence of an altered NDP reductase, as a component of the replication machinery, impairs the progression of the replication fork, contributing to the lengthening of the C period in the nrdA101 mutant at the permissive temperature.


Subject(s)
DNA Replication , Escherichia coli/enzymology , Escherichia coli/physiology , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleoside Diphosphate Reductase/physiology , Cell Division/genetics , Colony Count, Microbial , DNA Breaks, Double-Stranded , DNA Replication/genetics , DNA, Bacterial/metabolism , Deoxyribonucleotides/biosynthesis , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Exodeoxyribonuclease V/genetics , Holliday Junction Resolvases/physiology , Microbial Viability , Mutation
10.
Cancer Res ; 63(20): 6583-94, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14583450

ABSTRACT

Ribonucleotide reductase (RR) is responsible for the de novo conversion of the ribonucleoside diphosphates to deoxyribonucleoside diphosphates, which are essential for DNA synthesis and repair. RR consists of two subunits, hRRM1 and hRRM2. p53R2 is a new RR family member. Because the majority of human tumors possess mutant p53, it is important to know the molecular mechanism by which mutant p53 regulates RR and to what extent. In this study, we investigated the expression and function of p53R2 and hRRM2 after UV treatment in human prostate cancer PC3 cells, which possess mutant p53 with a truncated COOH-terminal, and in human oropharyngeal cancer KB cells, which possess wild-type p53. p53R2 (analyzed by Western blot and standardized relative to Coomassie Blue-stained band) was down-regulated in PC3 cells and up-regulated in KB cells after UV exposure. In contrast, hRRM2 was up-regulated by UV in both PC3 cells and KB cells. hRRM2 and p53R2 mRNA levels were assessed by Northern blot, and the results paralleled that of the Western blot. Coimmunoprecipitation assays using agarose-conjugated goat antihuman RRM1 antibody confirmed that the p53R2 binding to hRRM1 decreased in PC3 cells but increased in KB cells after UV treatment. hRRM2 binding to hRRM1 increased in both cell lines under the same conditions. These results suggest that PC3 cells are deficient in both transcription of p53R2 and binding to hRRM1 in response to UV irradiation. Confocal microscopy further confirmed that these findings were not due to translocation of hRRM2 and p53R2 from the cytoplasm to the nucleus. RR activity was measured following UV treatment and shown to increase in PC3 cells. It was unchanged in proportional of KB cells. The RR activity is consistent with the expression of hRRM2 seen in the Western blots. Thus, we hypothesize that hRRM2 complements p53R2 to form RR holoenzyme and maintain RR activity in PC3 cells after UV treatment. To further confirm this hypothesis, we examined the effect of RRM2 inhibitors on cells exposed to UV. In PC3 cells, hydroxyurea inhibited hRRM2 and resulted in increased sensitivity to UV irradiation. We also examined the effect of UV treatment on the colony-forming ability of cells transfected with hRRM2 as well as p53R2 sense or antisense expression vectors. Expression of antisense hRRM2 in PC3 cells led to decreased hRRM2 expression and resulted in greater sensitivity to UV than observed in wild-type PC3 cells. Taken together, we conclude that UV-induced activation of p53R2 transcription and binding of p53R2 to hRRM1 to form RR holoenzyme are impaired in the p53-mutant cell line PC3.


Subject(s)
Cell Cycle Proteins , DNA Repair , Ribonucleoside Diphosphate Reductase/physiology , Ribonucleotide Reductases/physiology , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , DNA Damage , Gene Expression Regulation, Neoplastic/physiology , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Hydroxyurea/pharmacology , KB Cells , Male , Mutation , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Binding/radiation effects , RNA, Antisense/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Ribonucleoside Diphosphate Reductase/biosynthesis , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleoside Diphosphate Reductase/metabolism , Ribonucleotide Reductases/biosynthesis , Ribonucleotide Reductases/genetics , Ribonucleotide Reductases/metabolism , Transfection , Tumor Suppressor Proteins/metabolism , Ultraviolet Rays
11.
Antisense Nucleic Acid Drug Dev ; 10(2): 111-6, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10805162

ABSTRACT

Ribonucleotide reductase (RR) is a rate-limiting enzyme in DNA synthesis and repair. The enzyme consists of two dissimilar subunits, M1 and M2. It is known that the M2 subunit plays a role in tumorgenicity and metastasis. In this study, we transfected human oropharyngeal KB cancer cells with human RR M1 and M2 antisense cDNA expressed by an inducible vector system. The transfectants were double-selected with hygromycin and G418. The clones, designated KB-M1AS, KB-M2AS and KB-CAT, represented transfectant clones that contained M1 antisense cDNA, M2 antisense cDNA, and a CAT reporter gene, respectively. In a colony-forming assay, colony formation for the KB-M2AS clone decreased approximately 50% when M2 antisense mRNA expression was induced by isopropylthiogalactose (IPTG). However, the KB-M1AS clone revealed no significant inhibition under IPTG induction. RR enzyme activity, as measured by 14CDP reduction assay, revealed a 30% decrease in the IPTG-induced KB-M2AS clone relative to non-IPTG-induced samples at 144 hours. As shown by Northern blot, expression of the M2 antisense mRNA showed peaks at 48 hours and 144 hours after induction by IPTG. M2 antisense mRNA expression induced by IPTG was 33-fold greater than the uninduced control at 144 hours. Western blot analysis showed that the M2 subunit protein level decreased in the KB-M2AS clone beginning at 72 hours after induction and continued to decrease to 50% of the uninduced control at 144 hours, then showed a slight recovery at 168 hours. In conclusion, M2 antisense mRNA expression by an inducible system can effectively decrease RR M2 protein expression, reduce enzyme activity, and inhibit growth. Furthermore, this approach can be employed in future antisense investigations.


Subject(s)
DNA, Antisense/biosynthesis , DNA, Complementary/biosynthesis , Growth Inhibitors/biosynthesis , Ribonucleoside Diphosphate Reductase/biosynthesis , Blotting, Western , Clone Cells , DNA, Antisense/physiology , DNA, Complementary/physiology , Enzyme Activation , Enzyme Induction , Growth Inhibitors/physiology , Humans , KB Cells , RNA, Messenger/biosynthesis , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleoside Diphosphate Reductase/metabolism , Ribonucleoside Diphosphate Reductase/physiology , Transfection , Tumor Stem Cell Assay
SELECTION OF CITATIONS
SEARCH DETAIL