Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 73
1.
Sci Rep ; 9(1): 13238, 2019 09 13.
Article En | MEDLINE | ID: mdl-31519977

Renal diseases impose considerable health and economic burdens on health systems worldwide, and there is a lack of efficient methods for the prevention and treatment due to their complexity and heterogeneity. Kidneys are organs with a high demand for energy produced by mitochondria, in which Rrm2b has critical functions as reported. The Rrm2b kidney-specific knockout mice we generated exhibited age-dependent exacerbated features, including mitochondrial dysfunction and increased oxidative stress; additionally, resulted in severe disruption of mitochondria-related metabolism. Rrm2b is vital not only to supply dNTPs for DNA replication and repair, but also to maintain structural integrity and metabolic homeostasis in mitochondria. Thence, Rrm2b deletion might induce chronic kidney defects in mice. This model can facilitate exploration of novel mechanisms and targeted therapies in the kidney diseases and has important translational and clinical implications.


Cell Cycle Proteins/physiology , Kidney Tubules/pathology , Mitochondria/pathology , Oxidative Stress , Ribonucleotide Reductases/physiology , Animals , DNA Replication , Female , Kidney Tubules/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Oxidation-Reduction
2.
Curr Genet ; 64(3): 547-550, 2018 Jun.
Article En | MEDLINE | ID: mdl-29119271

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.


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
3.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 41(11): 1155-1162, 2016 Nov 28.
Article Zh | MEDLINE | ID: mdl-27932760

OBJECTIVE: To explore the role of the special AT rich sequence binding protein-1 (SATB1) and ribonucleotide reductase M2 (RRM2) in enhancing malignant progression of non-Hodgkin lymphoma (NHL). 
 Methods: A total of 42 NHL and 42 chronic lymphadenitis patients were recruited. The protein expressions of SATB1 and RRM2 in cervical lymph nodes were determined by Western blot. After overexpression of SATB1, siSATB1 or siRRM2, the mRNA levels of SATB1 and RRM2 in cells were analyzed via RT-PCR, the cell proliferation was evaluated via MTT and EdU assays, while the migration and invasion of cells were assessed by transwell assays.
 Results: Compared with chronic lymphadenitis, the expressions of SATB1 and RRM2 in NHL patients were up-regulated. There was positive correlation between SATB1 and RRM2 in NHL patients. RRM2 mRNA level was up-regulated after transfection of SATB1 and down-regulated after transfection of siSATB1. Overexpression of SATB1 increased tumor cell proliferation, migration and invasion, while knockdown of RRM2 reversed those phenomena.
 Conclusion: SATB1 functions as an oncogene and promotes tumor cell proliferation, migration and invasion by up-regulation of RRM2 in NHL.


Cell Cycle Proteins/physiology , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Matrix Attachment Region Binding Proteins/physiology , Neoplasm Invasiveness/genetics , Ribonucleotide Reductases/physiology , Cell Cycle Proteins/genetics , Down-Regulation , Humans , Lymph Nodes/chemistry , Lymphoma, Non-Hodgkin , Matrix Attachment Region Binding Proteins/genetics , Oncogenes/genetics , Oncogenes/physiology , RNA, Messenger , RNA, Small Interfering , Ribonucleoside Diphosphate Reductase , Ribonucleotide Reductases/genetics , Signal Transduction , Transcription Factors , Transcriptional Activation , Transfection , Tumor Cells, Cultured , Up-Regulation
4.
Biochimie ; 123: 81-4, 2016 Apr.
Article En | MEDLINE | ID: mdl-26796884

p53R2 is a p53-inducible human ribonucleotide reductase subunit involved in critical cellular mechanisms, such as DNA repair, cell cycle arrest, and mitochondrial homeostasis. Molecular investigations and animal studies have revealed functional regulations of p53R2 and its disease relevance. The relationship between p53R2 expression and disease progression in different cancers has been evaluated, and researchers have discovered novel transcription factors and cellular mechanisms that control p53R2 in a p53-independent manner. In addition, p53R2-Mediated mechanisms that affect mitochondria, inflammation, and cancer have been addressed. The role of p53R2 in mitochondria diseases and in cancer is discussed. Finally, p53R2 is taken as a potential target for cancer treatment. This review summarizes the general background, novel regulatory findings, and medical prospect of p53R2.


Cell Cycle Proteins/physiology , Neoplasms/physiopathology , Ribonucleotide Reductases/physiology , Cell Cycle Proteins/genetics , Disease Progression , Gene Deletion , Humans , Mutation , Ribonucleotide Reductases/genetics
5.
J Pharmacol Sci ; 127(3): 319-25, 2015 Mar.
Article En | MEDLINE | ID: mdl-25837929

Gemcitabine is widely used for pancreatic, lung, and bladder cancer. However, drug resistance against gemcitabine is a large obstacle to effective chemotherapy. Nucleoside transporters, nucleoside and nucleotide metabolic enzymes, and efflux transporters have been reported to be involved in gemcitabine resistance. Although most of the resistant factors are supposed to be related to each other, it is unclear how one factor can affect the other one. In this study, we established gemcitabine-resistant pancreatic cancer cell lines. Gemcitabine resistance in these cells is caused by two major processes: a decrease in gemcitabine uptake and overexpression of ribonucleotide reductase large subunit (RRM1). Knockdown of RRM1, but not the overexpression of concentrative nucleoside transporter 1 (CNT1), could completely overcome the gemcitabine resistance. RRM1 knockdown in gemcitabine-resistant cells could increase the intracellular accumulation of gemcitabine by increasing the nucleoside transporter expression. Furthermore, a synergistic effect was observed between hydroxyurea, a ribonucleotide reductase (RR) inhibitor, and gemcitabine on the gemcitabine-resistant cells. Here we indicate that RR is one of the most promising targets to overcome gemcitabine resistance in gemcitabine-resistant cells with dual resistant factors.


Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/pharmacology , Pancreatic Neoplasms/pathology , Ribonucleotide Reductases/antagonists & inhibitors , Ribonucleotide Reductases/physiology , Deoxycytidine/metabolism , Deoxycytidine/pharmacology , Enzyme Inhibitors/metabolism , Humans , Pancreatic Neoplasms/metabolism , Tumor Cells, Cultured , Gemcitabine
6.
Article En | MEDLINE | ID: mdl-24809024

Ribonucleotide reductase (RNR) is a key enzyme that mediates the synthesis of deoxyribonucleotides, the DNA precursors, for DNA synthesis in every living cell. This enzyme converts ribonucleotides to deoxyribonucleotides, the building blocks for DNA replication, and repair. Clearly, RNR enzymes have contributed to the appearance of genetic material that exists today, being essential for the evolution of all organisms on Earth. The strict control of RNR activity and dNTP pool sizes is important, as pool imbalances increase mutation rates, replication anomalies, and genome instability. Thus, RNR activity should be finely regulated allosterically and at the transcriptional level. In this review we examine the distribution, the evolution, and the genetic regulation of bacterial RNRs. Moreover, this enzyme can be considered an ideal target for anti-proliferative compounds designed to inhibit cell replication in eukaryotic cells (cancer cells), parasites, viruses, and bacteria.


Bacteria/enzymology , Bacterial Physiological Phenomena , Ribonucleotide Reductases/physiology , Biological Evolution , Drug Discovery , Gene Expression Regulation, Bacterial , Humans , Ribonucleotide Reductases/antagonists & inhibitors , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/classification
7.
Semin Cell Dev Biol ; 30: 97-103, 2014 Jun.
Article En | MEDLINE | ID: mdl-24704278

Synthesis of deoxynucleoside triphosphates (dNTPs) is essential for both DNA replication and repair and a key step in this process is catalyzed by ribonucleotide reductases (RNRs), which reduce ribonucleotides (rNDPs) to their deoxy forms. Tight regulation of RNR is crucial for maintaining the correct levels of all four dNTPs, which is important for minimizing the mutation rate and avoiding genome instability. Although allosteric control of RNR was the first discovered mechanism involved in regulation of the enzyme, other controls have emerged in recent years. These include regulation of expression of RNR genes, proteolysis of RNR subunits, control of the cellular localization of the small RNR subunit, and regulation of RNR activity by small protein inhibitors. This review will focus on these additional mechanisms of control responsible for providing a balanced supply of dNTPs.


DNA Repair , DNA Replication , Ribonucleotide Reductases/physiology , Active Transport, Cell Nucleus , Animals , Cell Cycle , Deoxyribonucleotides/biosynthesis , Feedback, Physiological , Genomic Instability , Humans
8.
Life Sci ; 99(1-2): 14-7, 2014 Mar 18.
Article En | MEDLINE | ID: mdl-24486301

Although the deregulated expression of p53R2, a p53-inducible protein and homologue of the R2 subunit of ribonucleotide reductase, has been detected in several human cancers, p53R2 roles in cancer progression and malignancy still remains controversial. In this article, we present a viable hypothesis about the roles of p53R2 in cancer progression and therapy resistance based on the roles of cytoplasmic p21 and mutant p53. Since p53R2 can up-regulate p21 and p21, it in turn has a dual role in cell cycle. Hence, p53R2 can play a dual role in cell cycle progression. In addition, because p53 is the main regulator of p53R2, the mutant p53 may induce the expression of p53R2 in some cancer cells based on the "keep of function" phenomenon. Therefore, depending on the locations of p21 and the new abilities of mutant p53, p53R2 has dual role in cell cycle progression. Since the DNA damaging therapies induce p53R2 expression through the induction of p53, p53R2 can be the main therapy resistance mediator in cancers with cytoplasmic p21.


Cell Cycle Proteins/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Progression , Neoplasms/genetics , Ribonucleotide Reductases/physiology , Cell Cycle Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Humans , Neoplasms/physiopathology , Ribonucleotide Reductases/genetics
9.
Hepatology ; 59(4): 1459-70, 2014 Apr.
Article En | MEDLINE | ID: mdl-24214128

UNLABELLED: Ribonucleotide reductase (RR)M2B is an enzyme belonging to the ribonucleotide reductase enzyme family, which is essential for DNA synthesis and repair. RRM2B plays an important role in tumor progression and metastasis; however, little is known about the expression and underlying molecular mechanisms of RRM2B in hepatocellular carcinoma (HCC). In the present study, we report that down-regulation of RRM2B in HCC is negatively associated with intrahepatic metastasis, regardless of p53 status. Moreover, the ectopic overexpression of RRM2B decreased HCC cell migration and invasion in vitro, whereas silencing RRM2B expression resulted in increased migration and invasion in vitro and intrahepatic and lung metastasis in vivo. Additionally, knockdown of RRM2B by short hairpin RNA (shRNA) in HCC cells was associated with epithelial-mesenchymal transition (EMT), including the down-regulation of E-cadherin, and the concomitant up-regulation of N-cadherin and slug. A further experiment showed that RRM2B inhibited cell migration and spreading through regulation of the early growth response protein 1 (Egr-1)/phosphatase and tensin homolog (PTEN)/Akt1 pathway. Consistently, we also detected a significant correlation between RRM2B and E-cadherin protein expression in HCC tissues. Furthermore, Egr-1 also directly bound to the RRM2B promoter and repressed RRM2B transcription, thereby establishing a negative regulatory feedback loop. CONCLUSION: These findings indicate that RRM2B suppresses cell migration and spreading by way of modulation of the Egr-1/PTEN/Akt1 pathway.


Carcinoma, Hepatocellular/physiopathology , Cell Cycle Proteins/physiology , Cell Movement/physiology , Early Growth Response Protein 1/physiology , Liver Neoplasms/physiopathology , PTEN Phosphohydrolase/physiology , Proto-Oncogene Proteins c-akt/physiology , Ribonucleotide Reductases/physiology , Signal Transduction/physiology , Cadherins/physiology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Disease Progression , Down-Regulation/physiology , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , In Vitro Techniques , Liver Neoplasms/pathology , Male , Middle Aged , RNA, Small Interfering/pharmacology , Snail Family Transcription Factors , Transcription Factors/physiology
10.
Expert Opin Ther Targets ; 17(12): 1423-37, 2013 Dec.
Article En | MEDLINE | ID: mdl-24083455

INTRODUCTION: Ribonucleotide reductase (RR) is a unique enzyme, because it is responsible for reducing ribonucleotides to their corresponding deoxyribonucleotides, which are the building blocks required for DNA replication and repair. Dysregulated RR activity is associated with genomic instability, malignant transformation and cancer development. The use of RR inhibitors, either as a single agent or combined with other therapies, has proven to be a promising approach for treating solid tumors and hematological malignancies. AREAS COVERED: This review covers recent publications in the area of RR, which include: i) the structure, function and regulation of RR; ii) the roles of RR in cancer development; iii) the classification, mechanisms and clinical application of RR inhibitors for cancer therapy and iv) strategies for developing novel RR inhibitors in the future. EXPERT OPINION: Exploring the possible nonenzymatic roles of RR subunit proteins in carcinogenesis may lead to new rationales for developing novel anticancer drugs. Updated information about the structure and holoenzyme models of RR will help in identifying potential sites in the protein that could be targets for novel RR inhibitors. Determining RR activity and subunit levels in clinical samples will provide a rational platform for developing personalized cancer therapies that use RR inhibitors.


Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Ribonucleotide Reductases/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Humans , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/physiology
11.
Carcinogenesis ; 34(11): 2498-504, 2013 Nov.
Article En | MEDLINE | ID: mdl-23843040

Alternative splicing regulators have emerged as new players in cancer development, modulating the activities of many tumor suppressors and oncogenes and regulating the signaling pathways. However, little is known about the mechanisms by which these oncogenic splicing factors lead to cellular transformation. We have shown previously that the splicing factor serine and arginine splicing factor 1 (SRSF1; SF2/ASF) is a proto-oncogene, which is amplified in breast cancer and transforms immortal cells when overexpressed. In this study, we performed a structure-function analysis of SRSF1 and found that the RNA recognition motif 1 (RRM1) domain is required for its oncogenic activity. Deletion of RRM1 eliminated the splicing activity of SRSF1 on some of its endogenous targets. Moreover, we found that SRSF1 elevates the expression of B-Raf and activates the mitogen-activated protein kinase kinase (MEK) extracellular signal-regulated kinase (ERK) pathway and that RRM1 is required for this activation as well. B-Raf-MEK-ERK activation by SRSF1 contributes to transformation as pharmacological inhibition of MEK1 inhibits SRSF1-mediated transformation. In conclusion, RRM1 of SRSF1 is both required (and when tethered to the RS domain) also sufficient to activate the Raf-MEK-ERK pathway and to promote cellular transformation.


Cell Transformation, Neoplastic/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nuclear Proteins/physiology , RNA Splicing/genetics , RNA-Binding Proteins/physiology , Ribonucleotide Reductases/physiology , Amino Acid Motifs , Animals , Blotting, Western , Cell Adhesion , Cell Proliferation , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/genetics , HEK293 Cells , Humans , Liver/metabolism , Liver/pathology , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinases/genetics , Mutation , Protein Binding , Protein Structure, Tertiary/physiology , Proto-Oncogene Mas , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoside Diphosphate Reductase , Serine-Arginine Splicing Factors , Signal Transduction , Stem Cells/metabolism , Stem Cells/pathology , Tumor Suppressor Protein p53/physiology
12.
Am J Pathol ; 182(1): 142-51, 2013 Jan.
Article En | MEDLINE | ID: mdl-23245831

In normal human cells, oncogene-induced senescence (OIS) depends on induction of DNA damage response. Oxidative stress and hyperreplication of genomic DNA have been proposed as major causes of DNA damage in OIS cells. Here, we report that down-regulation of deoxyribonucleoside pools is another endogenous source of DNA damage in normal human fibroblasts (NHFs) undergoing HRAS(G12V)-induced senescence. NHF-HRAS(G12V) cells underexpressed thymidylate synthase (TS) and ribonucleotide reductase (RR), two enzymes required for the entire de novo deoxyribonucleotide biosynthesis, and possessed low dNTP levels. Chromatin at the promoters of the genes encoding TS and RR was enriched with retinoblastoma tumor suppressor protein and histone H3 tri-methylated at lysine 9. Importantly, ectopic coexpression of TS and RR or addition of deoxyribonucleosides substantially suppressed DNA damage, senescence-associated phenotypes, and proliferation arrest in two types of NHF-expressing HRAS(G12V). Reciprocally, short hairpin RNA-mediated suppression of TS and RR caused DNA damage and senescence in NHFs, although less efficiently than HRAS(G12V). However, overexpression of TS and RR in quiescent NHFs did not overcome proliferation arrest, suggesting that unlike quiescence, OIS requires depletion of dNTP pools and activated DNA replication. Our data identify a previously unknown role of deoxyribonucleotides in regulation of OIS.


Cellular Senescence/genetics , DNA Damage/genetics , Deoxyribonucleotides/metabolism , Oncogenes/physiology , Cell Proliferation , Cells, Cultured , Cellular Senescence/physiology , DNA Replication/genetics , Deoxyribonucleotides/genetics , Fibroblasts/metabolism , Fibroblasts/physiology , Humans , Proto-Oncogene Proteins p21(ras)/physiology , Ribonucleotide Reductases/biosynthesis , Ribonucleotide Reductases/physiology , Thymidylate Synthase/biosynthesis , Thymidylate Synthase/physiology
13.
Int J Gynecol Cancer ; 22(9): 1463-9, 2012 Nov.
Article En | MEDLINE | ID: mdl-23051959

OBJECTIVE: Ribonucleotide reductase (RNR) supplies deoxyribonucleotide diphosphates demanded by cells to repair radiation-induced DNA damage. Here, we investigate the impact of pretherapy RNR M1, M2, and M2b (p53R3) subunit level upon human cervical cancer radiochemosensitivity. MATERIALS/METHODS: Immunohistochemistry was performed on a tissue array comprised of 18 paired benign uterine cervix and stage IB2 cervical cancers to evaluate the relationship between cytosolic RNR M1, M2, and M2b staining intensity and radiochemotherapy cancer response. Patients underwent surgical hysterectomy (n = 8), or daily radiation (45 Gy), coadministered once-weekly cisplatin (40 mg/m), then low-dose rate brachytherapy (30 Gy) followed by adjuvant hysterectomy (n = 10). Radiochemotherapy response was determined by Response Evaluation Criteria In Solid Tumors version 1.0 criteria during brachytherapy. Cancer relapse rates and disease-free survival were calculated. RESULTS: M1, M2, and M2b antibody staining intensity was low (0-1+) in benign uterine cervical tissue. M1 and M2b immunoreactivity was 2+ or 3+ in most (13/18) cervical cancers. M2 immunoreactivity was 3+ in nearly all (16/18) cervical cancers. Cervical cancers overexpressing M1 and M2b had an increased hazard for incomplete radiochemotherapy response, relapse, and shortened disease-free survival. CONCLUSIONS: Ribonucleotide reductase subunit levels may predict human cervical cancer radiochemosensitivity and subsequent posttherapy cancer outcome. Further validation testing of RNR subunits as biomarkers for radiochemotherapy response is warranted.


Carcinoma, Squamous Cell/therapy , Chemoradiotherapy , Radiation Tolerance , Ribonucleotide Reductases/metabolism , Uterine Cervical Neoplasms/therapy , Adenocarcinoma/metabolism , Adenocarcinoma/therapy , Adult , Aged , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/physiology , Carcinoma, Squamous Cell/metabolism , Female , Humans , Hysterectomy , Middle Aged , Radiation Tolerance/physiology , Ribonucleotide Reductases/physiology , Tissue Array Analysis , Treatment Failure , Up-Regulation/physiology , Uterine Cervical Neoplasms/metabolism
14.
Gene ; 475(1): 30-8, 2011 Apr 01.
Article En | MEDLINE | ID: mdl-21194559

p53R2 is a newly identified small subunit of ribonucleotide reductase and plays a pivotal role in the supply of dNTPs for genomic DNA repair and mitochondrial DNA synthesis, but little is known about its functions in zebrafish. Herein, we obtained the cDNA of zebrafish p53R2 that shares 72.8% and 72.5% amino acid identities with human p53R2 and zebrafish R2, respectively. Residues crucial for enzymatic activity are highly conserved among p53R2 proteins from different species. p53R2 in zebrafish was maternally expressed, its transcripts were detected in developing embryos and all adult tissues examined. A 250-bp minimal promoter upstream of the translational initiation site was identified to drive basal expression of p53R2 in a p53-independent manner. Expression of p53R2 was induced by DNA-damaging reagents CPT or MMS, but suppressed by p53-knockdown in zebrafish embryos. Moreover, p53R2 was mainly distributed in the cytoplasm of cells under normal condition and upon DNA damage. Furthermore, overexpression of p53R2 attenuated apoptosis of embryonic cells caused by CPT or MMS treatment and protected developing embryos from death. Therefore, functions of p53R2 in zebrafish are closely associated with its activity in DNA repair and synthesis.


DNA Repair/genetics , DNA Replication/genetics , Ribonucleotide Reductases/physiology , Zebrafish Proteins/physiology , Zebrafish/metabolism , Amino Acid Sequence , Animals , Apoptosis/genetics , Base Sequence , DNA Damage , Embryo, Nonmammalian/enzymology , Molecular Sequence Data , Promoter Regions, Genetic , Ribonucleotide Reductases/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
15.
Free Radic Biol Med ; 49(11): 1617-28, 2010 Dec 01.
Article En | MEDLINE | ID: mdl-20851762

Ribonucleotide reductase (RNR) catalyzes the rate-limiting de novo synthesis of 2'-deoxyribonucleotides from the corresponding ribonucleotides and thereby provides balanced deoxyribonucleotide pools required for error-free DNA replication and repair. The essential role of RNR in DNA synthesis and the use of DNA as genetic material has made it an important target for the development of anticancer and antiviral agents. The most well known feature of the universal RNR reaction in all kingdoms of life is the involvement of protein free radicals. Redox-active cysteines, thiyl radicals, and thiol redox proteins of the thioredoxin superfamily play major roles in the catalytic mechanism. The involvement of cysteine residues in catalysis is common to all three classes of RNR. Taking account of the recent progress in this field of research, this review focuses on the use of thiols in the redox mechanism of RNR enzymes.


Ribonucleotide Reductases/metabolism , Ribonucleotide Reductases/physiology , Sulfhydryl Compounds/metabolism , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Catalysis/drug effects , Humans , Models, Biological , Ribonucleotide Reductases/chemistry , Structure-Activity Relationship , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/pharmacology , Sulfhydryl Reagents/chemistry , Sulfhydryl Reagents/pharmacology
16.
J Virol ; 84(20): 10937-42, 2010 Oct.
Article En | MEDLINE | ID: mdl-20668075

Viral enzymes that process small molecules provide potential chemotherapeutic targets. A key constraint-the replicative potential of spontaneous enzyme mutants-has been hard to define with human gammaherpesviruses because of their narrow species tropisms. Here, we disrupted the murid herpesvirus 4 (MuHV-4) ORF61, which encodes its ribonucleotide reductase (RNR) large subunit. Mutant viruses showed delayed in vitro lytic replication, failed to establish infection via the upper respiratory tract, and replicated to only a very limited extent in the lower respiratory tract without reaching lymphoid tissue. RNR could therefore provide a good target for gammaherpesvirus chemotherapy.


Rhadinovirus/enzymology , Rhadinovirus/pathogenicity , Ribonucleotide Reductases/physiology , Animals , Base Sequence , Cell Line , Cricetinae , DNA, Viral/genetics , Genes, Viral , Herpesviridae Infections/virology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutagenesis, Insertional , Respiratory System/virology , Rhadinovirus/genetics , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/genetics , Tumor Virus Infections/virology , Virulence/genetics , Virulence/physiology , Virus Replication/genetics , Virus Replication/physiology
17.
PLoS Pathog ; 6(7): e1000984, 2010 Jul 08.
Article En | MEDLINE | ID: mdl-20628573

Ribonucleotide reductases (RRs) are evolutionarily-conserved enzymes that catalyze the rate-limiting step during dNTP synthesis in mammals. RR consists of both large (R1) and small (R2) subunits, which are both required for catalysis by the R1(2)R2(2) heterotetrameric complex. Poxviruses also encode RR proteins, but while the Orthopoxviruses infecting humans [e.g. vaccinia (VACV), variola, cowpox, and monkeypox viruses] encode both R1 and R2 subunits, the vast majority of Chordopoxviruses encode only R2 subunits. Using plaque morphology, growth curve, and mouse model studies, we investigated the requirement of VACV R1 (I4) and R2 (F4) subunits for replication and pathogenesis using a panel of mutant viruses in which one or more viral RR genes had been inactivated. Surprisingly, VACV F4, but not I4, was required for efficient replication in culture and virulence in mice. The growth defects of VACV strains lacking F4 could be complemented by genes encoding other Chordopoxvirus R2 subunits, suggesting conservation of function between poxvirus R2 proteins. Expression of F4 proteins encoding a point mutation predicted to inactivate RR activity but still allow for interaction with R1 subunits, caused a dominant negative phenotype in growth experiments in the presence or absence of I4. Co-immunoprecipitation studies showed that F4 (as well as other Chordopoxvirus R2 subunits) form hybrid complexes with cellular R1 subunits. Mutant F4 proteins that are unable to interact with host R1 subunits failed to rescue the replication defect of strains lacking F4, suggesting that F4-host R1 complex formation is critical for VACV replication. Our results suggest that poxvirus R2 subunits form functional complexes with host R1 subunits to provide sufficient dNTPs for viral replication. Our results also suggest that R2-deficient poxviruses may be selective oncolytic agents and our bioinformatic analyses provide insights into how poxvirus nucleotide metabolism proteins may have influenced the base composition of these pathogens.


Ribonucleotide Reductases/physiology , Vaccinia virus/enzymology , Vaccinia virus/genetics , Virus Replication , Animals , DNA Replication , Mice , Mutation , Poxviridae , Protein Subunits/physiology , Vaccinia virus/pathogenicity , Virulence
18.
Biochemistry ; 48(49): 11612-21, 2009 Dec 15.
Article En | MEDLINE | ID: mdl-19899807

Ribonucleotide reductases (RNRs) catalyze the conversion of nucleoside 5'-diphosphates to the corresponding deoxynucleotides supplying the dNTPs required for DNA replication and DNA repair. Class I RNRs require two subunits, alpha and beta, for activity. Humans possess two beta subunits: one involved in S phase DNA replication (beta) and a second in mitochondrial DNA replication (beta' or p53R2) and potentially DNA repair. Gemcitabine (F(2)C) is used clinically as an anticancer agent, and its phosphorylated metabolites target many enzymes involved in nucleotide metabolism, including RNR. The present investigation with alpha (specific activity of 400 nmol min(-1) mg(-1)) and beta' (0.6 Y./beta'2 and a specific activity of 420 nmol min(-1) mg(-1)) establishes that F(2)CDP is a substoichiometric inactivator of RNR. Incubation of this alpha/beta' with [1'-(3)H]-F(2)CDP or [5-(3)H]-F(2)CDP and reisolation of the protein by Sephadex G-50 chromatography resulted in recovery 0.5 equiv of covalently bound sugar and 0.03 equiv of tightly associated cytosine to alpha2. SDS-PAGE analysis (loaded without boiling) of the inactivated RNR showed that 60% of alpha migrates as a 90 kDa protein and 40% as a 120 kDa protein. Incubation of [1'-(3)H]-F(2)CDP with active site mutants C444S/A, C218S/A, and E431Q/D-alpha and the C-terminal tail C787S/A and C790S/A mutants reveals that no sugar label is bound to the active site mutants of alpha and that, in the case of C218S-alpha, alpha migrates as a 90 kDa protein. Analysis of the inactivated wt-alpha/beta' RNR by size exclusion chromatography indicates a quaternary structure of alpha6beta'6. A mechanism of inactivation common with halpha/beta is presented.


Cell Cycle Proteins/physiology , Cytidine Diphosphate/analogs & derivatives , Enzyme Inhibitors/toxicity , Ribonucleotide Reductases/antagonists & inhibitors , Cell Cycle Proteins/isolation & purification , Chromatography, Gel , Cytidine Diphosphate/chemistry , Cytidine Diphosphate/toxicity , DNA Damage/genetics , DNA Repair/genetics , Enzyme Inhibitors/chemistry , Humans , Mutagenesis, Site-Directed , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Transport/genetics , Ribonucleotide Reductases/genetics , Ribonucleotide Reductases/isolation & purification , Ribonucleotide Reductases/metabolism , Ribonucleotide Reductases/physiology
19.
Int J Cancer ; 125(9): 2086-94, 2009 Nov 01.
Article En | MEDLINE | ID: mdl-19585502

Chlorophyllin (CHL) is a water-soluble derivative of chlorophyll that exhibits cancer chemopreventive properties, but which also has been studied for its possible cancer therapeutic effects. We report here that human colon cancer cells treated with CHL accumulate in S-phase of the cell cycle, and this is associated with reduced expression levels of p53, p21, and other G(1)/S checkpoint controls. At the same time, E2F1 and E2F4 transcription factors become elevated and exhibit increased DNA binding activity. In CHL-treated colon cancer cells, bromodeoxyuridine pulse-chase experiments provided evidence for the inhibition of DNA synthesis. Ribonucleotide reductase (RR), a pivotal enzyme for DNA synthesis and repair, was reduced at the mRNA and protein level after CHL treatment, and the enzymatic activity was inhibited in a concentration-dependent manner both in vitro and in vivo. Immunoblotting revealed that expression levels of RR subunits R1, R2, and p53R2 were reduced by CHL treatment in HCT116 (p53(+/+)) and HCT116 (p53(-/-)) cells, supporting a p53-independent mechanism. Prior studies have shown that reduced levels of RR small subunits can increase the sensitivity of colon cancer cells to clinically used DNA-damaging agents and RR inhibitors. We conclude that by inhibiting R1, R2, and p53R2, CHL has the potential to be effective in the clinical setting, when used alone or in combination with currently available cancer therapeutic agents.


Anticarcinogenic Agents/pharmacology , Chlorophyllides/pharmacology , Colonic Neoplasms/pathology , E2F4 Transcription Factor/physiology , Ribonucleotide Reductases/physiology , S Phase/drug effects , Cell Line, Tumor , Colonic Neoplasms/drug therapy , DNA/metabolism , E2F1 Transcription Factor/analysis , E2F1 Transcription Factor/metabolism , E2F4 Transcription Factor/analysis , Humans , Ribonucleotide Reductases/antagonists & inhibitors , Tumor Suppressor Protein p53/physiology
20.
J Virol ; 83(18): 9611-5, 2009 Sep.
Article En | MEDLINE | ID: mdl-19553308

Cytomegaloviruses (CMVs) code for immunoevasins, glycoproteins that are specifically dedicated to interfere with the presentation of antigenic peptides to CD8 T cells. Nonetheless, the biological outcome is not an immune evasion of the virus, since CD8 T cells can control CMV infection even when immunoevasins are expressed. Here, we compare the processing of a protective and a nonprotective epitope derived from the same viral protein, the antiapoptotic protein M45 in the murine model. The data provide evidence to conclude that protection against CMVs critically depends on antigenic peptides generated in an amount sufficient to exhaust the inhibitory capacity of immunoevasins.


Antigen Presentation , Cytomegalovirus Infections/immunology , Ribonucleotide Reductases/physiology , Viral Proteins/immunology , Animals , Apoptosis Regulatory Proteins , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus/pathogenicity , Epitopes/physiology , Humans , Mice , Peptide Fragments/immunology , Ribonucleotide Reductases/immunology , Viral Proteins/physiology
...