Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Proc Natl Acad Sci U S A ; 114(31): 8241-8246, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28716944

ABSTRACT

Human ribonucleotide reductase (hRR) is crucial for DNA replication and maintenance of a balanced dNTP pool, and is an established cancer target. Nucleoside analogs such as gemcitabine diphosphate and clofarabine nucleotides target the large subunit (hRRM1) of hRR. These drugs have a poor therapeutic index due to toxicity caused by additional effects, including DNA chain termination. The discovery of nonnucleoside, reversible, small-molecule inhibitors with greater specificity against hRRM1 is a key step in the development of more effective treatments for cancer. Here, we report the identification and characterization of a unique nonnucleoside small-molecule hRR inhibitor, naphthyl salicylic acyl hydrazone (NSAH), using virtual screening, binding affinity, inhibition, and cell toxicity assays. NSAH binds to hRRM1 with an apparent dissociation constant of 37 µM, and steady-state kinetics reveal a competitive mode of inhibition. A 2.66-Å resolution crystal structure of NSAH in complex with hRRM1 demonstrates that NSAH functions by binding at the catalytic site (C-site) where it makes both common and unique contacts with the enzyme compared with NDP substrates. Importantly, the IC50 for NSAH is within twofold of gemcitabine for growth inhibition of multiple cancer cell lines, while demonstrating little cytotoxicity against normal mobilized peripheral blood progenitor cells. NSAH depresses dGTP and dATP levels in the dNTP pool causing S-phase arrest, providing evidence for RR inhibition in cells. This report of a nonnucleoside reversible inhibitor binding at the catalytic site of hRRM1 provides a starting point for the design of a unique class of hRR inhibitors.


Subject(s)
Hydrazones/pharmacology , Naphthalenes/pharmacology , Ribonucleotide Reductases/antagonists & inhibitors , Salicylates/pharmacology , Catalytic Domain , Cell Cycle/drug effects , Crystallography, X-Ray , Deoxyadenine Nucleotides/metabolism , Drug Screening Assays, Antitumor/methods , Humans , Hydrazones/chemistry , Naphthalenes/chemistry , Ribonucleoside Diphosphate Reductase , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/metabolism , Salicylates/chemistry , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism
2.
Methods Mol Biol ; 1534: 165-173, 2017.
Article in English | MEDLINE | ID: mdl-27812878

ABSTRACT

DNA damage response has been characterized as an important mediator of senescence phenotypes induced by activated oncogenes in normal human cells. Depletion of intracellular deoxyribonucleotide pools has been recently recognized as one of the major causes for DNA damage in these cells. Cells undergoing oncogene-induced senescence display decreased expression of several rate-limiting enzymes involved in the biosynthesis of deoxyribonucleotides, including thymidylate synthase (TS) and ribonucleotide reductase (RR). Individual depletion of these enzymes leads to premature senescence. Reciprocally, ectopic expression of TS and RR or addition of deoxyribonucleosides resulted in suppression of senescence phenotypes in normal or tumor cells caused by overexpression of activated HRAS or depletion of C-MYC, respectively. Therefore, in the current chapter, we will describe a methodology for the quantitative measurement of nucleotide pools in senescent cells.


Subject(s)
Cellular Senescence , Nucleotides/metabolism , Oncogenes , Cell Line , Cellular Senescence/genetics , Chromatography, High Pressure Liquid , DNA Damage , Deoxyribonucleotides/chemistry , Deoxyribonucleotides/metabolism , Fibroblasts/metabolism , Gene Expression , Genes, myc , Humans , Nucleotides/chemistry , Oncogenes/genetics , Ribonucleotide Reductases/metabolism , Thymidylate Synthase/metabolism
3.
Radiat Res ; 186(5): 466-477, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27740890

ABSTRACT

Gemcitabine (dFdCyd) shows broad antitumor activity in solid tumors in chemotherapeutic regimens or when combined with ionizing radiation (radiosensitization). While it is known that mismatches in DNA are necessary for dFdCyd radiosensitization, the critical event resulting in radiosensitization has not been identified. Here we hypothesized that late DNA damage (≥24 h after drug washout/irradiation) is a causal event in radiosensitization by dFdCyd, and that homologous recombination repair (HRR) is required for this late DNA damage. Using γ-H2AX as a measurement of DNA damage in MCF-7 breast cancer cells, we demonstrate that 10 or 80 nM dFdCyd alone produced significantly more late DNA damage compared to that observed within 4 h after treatment. The combination of dFdCyd treatment followed by irradiation did not produce a consistent increase in DNA damage in the first 4 h after treatment, however, there was a synergistic increase 24-48 h later relative to treatment with dFdCyd or radiation alone. RNAi suppression of the essential HRR protein, XRCC3, significantly decreased both radiosensitization and late DNA damage. Furthermore, inhibition of HRR with the Rad51 inhibitor B02 prevented radiosensitization when added after, but not during, treatment with dFdCyd and radiation. To our knowledge, this is the first published study to show that radiosensitization with dFdCyd results from a synergistic increase in DNA damage at 24-48 h after drug and radiation treatment, and that this damage and radiosensitization require HRR. These results suggest that tumors that overexpress HRR will be more vulnerable to chemoradiotherapy, and treatments that increase HRR and/or mismatches in DNA will enhance dFdCyd radiosensitization.


Subject(s)
DNA Damage , Deoxycytidine/analogs & derivatives , Radiation-Sensitizing Agents/pharmacology , Recombinational DNA Repair/drug effects , Recombinational DNA Repair/radiation effects , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA-Binding Proteins/metabolism , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Gamma Rays , Histones/metabolism , Humans , MCF-7 Cells , Rad51 Recombinase/antagonists & inhibitors , Time Factors , Gemcitabine
4.
EMBO Mol Med ; 7(9): 1138-52, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26197802

ABSTRACT

Chromosomal instability in early cancer stages is caused by replication stress. One mechanism by which oncogene expression induces replication stress is to drive cell proliferation with insufficient nucleotide levels. Cancer development is driven by alterations in both genetic and environmental factors. Here, we investigated whether replication stress can be modulated by both genetic and non-genetic factors and whether the extent of replication stress affects the probability of neoplastic transformation. To do so, we studied the effect of folate, a micronutrient that is essential for nucleotide biosynthesis, on oncogene-induced tumorigenicity. We show that folate deficiency by itself leads to replication stress in a concentration-dependent manner. Folate deficiency significantly enhances oncogene-induced replication stress, leading to increased DNA damage and tumorigenicity in vitro. Importantly, oncogene-expressing cells, when grown under folate deficiency, exhibit a significantly increased frequency of tumor development in mice. These findings suggest that replication stress is a quantitative trait affected by both genetic and non-genetic factors and that the extent of replication stress plays an important role in cancer development.


Subject(s)
Carcinogenesis/drug effects , DNA Replication/drug effects , Folic Acid/metabolism , Genomic Instability/drug effects , Oncogenes/drug effects , Animals , Mice
5.
Radiat Res ; 183(1): 114-23, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25564718

ABSTRACT

Gemcitabine (difluorodeoxycytidine; dFdCyd) is a potent radiosensitizer, noted for its ability to enhance cytotoxicity with radiation at noncytotoxic concentrations in vitro and subchemotherapeutic doses in patients. Radiosensitization in human tumor cells requires dFdCyd-mediated accumulation of cells in S phase with inhibition of ribonucleotide reductase, resulting in ≥80% deoxyadenosine triphosphate (dATP) depletion and errors of replication in DNA. Less is known of the role of specific DNA replication and repair pathways in the radiosensitization mechanism. Here the role of homologous recombination (HR) in relationship to the metabolic and cell cycle effects of dFdCyd was investigated using a matched pair of CHO cell lines that are either proficient (AA8 cells) or deficient (irs1SF cells) in HR based on expression of the HR protein XRCC3. The results demonstrated that the characteristics of radiosensitization in the rodent AA8 cells differed significantly from those in human tumor cells. In the AA8 cells, radiosensitization was achieved only under short (≤4 h) cytotoxic incubations, and S-phase accumulation did not appear to be required for radiosensitization. In contrast, human tumor cell lines were radiosensitized using noncytotoxic concentrations of dFdCyd and required early S-phase accumulation. Studies of the metabolic effects of dFdCyd demonstrated low dFdCyd concentrations did not deplete dATP by ≥80% in AA8 and irs1SF cells. However, at higher concentrations of dFdCyd, failure to radiosensitize the HR-deficient irs1SF cells could not be explained by a lack of dATP depletion or lack of S-phase accumulation. Thus, these parameters did not correspond to dFdCyd radiosensitization in the CHO cells. To evaluate directly the role of HR in radiosensitization, XRCC3 expression was suppressed in the AA8 cells with a lentiviral-delivered shRNA. Partial XRCC3 suppression significantly decreased radiosensitization [radiation enhancement ratio (RER) = 1.6 ± 0.15], compared to nontransduced (RER = 2.7 ± 0.27; P = 0.012), and a substantial decrease compared to nonspecific shRNA-transduced (RER = 2.5 ± 0.42; P = 0.056) AA8 cells. Although the results support a role for HR in radiosensitization with dFdCyd in CHO cells, the differences in the underlying metabolic and cell cycle characteristics suggest that dFdCyd radiosensitization in the nontumor-derived CHO cells is mechanistically distinct from that in human tumor cells.


Subject(s)
Deoxycytidine/analogs & derivatives , Radiation-Sensitizing Agents/metabolism , Radiation-Sensitizing Agents/pharmacology , Recombinational DNA Repair/drug effects , Recombinational DNA Repair/radiation effects , Animals , CHO Cells , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Cricetinae , Cricetulus , DNA-Binding Proteins/metabolism , Deoxycytidine/metabolism , Deoxycytidine/pharmacology , Humans , Phosphates/metabolism , Gemcitabine
6.
DNA Repair (Amst) ; 12(12): 1114-21, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24231389

ABSTRACT

The nucleoside analog ganciclovir (GCV) elicits cytotoxicity in tumor cells via a novel mechanism in which drug incorporation into DNA produces minimal disruption of replication, but numerous DNA double strand breaks occur during the second S-phase after drug exposure. We propose that homologous recombination (HR), a major repair pathway for DNA double strand breaks, can prevent GCV-induced DNA damage, and that inhibition of HR will enhance cytotoxicity with GCV. Survival after GCV treatment in cells expressing a herpes simplex virus thymidine kinase was strongly dependent on HR (>14-fold decrease in IC50 in HR-deficient vs. HR-proficient CHO cells). In a homologous recombination reporter assay, the histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA; vorinostat), decreased HR repair events up to 85%. SAHA plus GCV produced synergistic cytotoxicity in U251tk human glioblastoma cells. Elucidation of the synergistic mechanism demonstrated that SAHA produced a concentration-dependent decrease in the HR proteins Rad51 and CtIP. GCV alone produced numerous Rad51 foci, demonstrating activation of HR. However, the addition of SAHA blocked GCV-induced Rad51 foci formation completely and increased γH2AX, a marker of DNA double strand breaks. SAHA plus GCV also produced synergistic cytotoxicity in HR-proficient CHO cells, but the combination was antagonistic or additive in HR-deficient CHO cells. Collectively, these data demonstrate that HR promotes survival with GCV and compromise of HR by SAHA results in synergistic cytotoxicity, revealing a new mechanism for enhancing anticancer activity with GCV.


Subject(s)
Antineoplastic Agents/pharmacology , Ganciclovir/pharmacology , Homologous Recombination/drug effects , Hydroxamic Acids/pharmacology , Animals , Apoptosis/drug effects , CHO Cells , Carrier Proteins/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cricetulus , Endodeoxyribonucleases , HeLa Cells , Humans , Nuclear Proteins/metabolism , Rad51 Recombinase/metabolism , Vorinostat
7.
Cell Rep ; 5(2): 493-507, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-24139804

ABSTRACT

Melanoma is one of the most aggressive types of human cancers, and the mechanisms underlying melanoma invasive phenotype are not completely understood. Here, we report that expression of guanosine monophosphate reductase (GMPR), an enzyme involved in de novo biosynthesis of purine nucleotides, was downregulated in the invasive stages of human melanoma. Loss- and gain-of-function experiments revealed that GMPR downregulates the amounts of several GTP-bound (active) Rho-GTPases and suppresses the ability of melanoma cells to form invadopodia, degrade extracellular matrix, invade in vitro, and grow as tumor xenografts in vivo. Mechanistically, we demonstrated that GMPR partially depletes intracellular GTP pools. Pharmacological inhibition of de novo GTP biosynthesis suppressed whereas addition of exogenous guanosine increased invasion of melanoma cells as well as cells from other cancer types. Our data identify GMPR as a melanoma invasion suppressor and establish a link between guanosine metabolism and Rho-GTPase-dependent melanoma cell invasion.


Subject(s)
GMP Reductase/metabolism , Melanoma/enzymology , Purine Nucleosides/biosynthesis , Animals , Cell Line, Tumor , Cell Movement , Extracellular Matrix/metabolism , GMP Reductase/antagonists & inhibitors , GMP Reductase/genetics , Guanosine Triphosphate/metabolism , HCT116 Cells , Humans , IMP Dehydrogenase/metabolism , Melanoma/metabolism , Melanoma/pathology , Mice , Phenotype , RNA Interference , RNA, Small Interfering/metabolism , Transplantation, Heterologous , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism
8.
Adv Healthc Mater ; 2(10): 1337-50, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23554387

ABSTRACT

Poly(amidoamine) (PAMAM) dendrimers are branched water-soluble polymers defined by consecutive generation numbers (Gn) indicating a parallel increase in size, molecular weight, and number of surface groups available for conjugation of bioactive agents. In this article, we compare the biodistribution of N-acetylgalactosamine (NAcGal)-targeted [(14) C]1 -G5-(NH2 )5 -(Ac)108 -(NAcGal)14 particles to non-targeted [(14) C]1 -G5-(NH2 )127 and PEGylated [(14) C]1 -G5-(NH2 )44 -(Ac)73 -(PEG)10 particles in a mouse hepatic cancer model. Results show that both NAcGal-targeted and non-targeted particles are rapidly cleared from the systemic circulation with high distribution to the liver. However, NAcGal-targeted particles exhibited 2.5-fold higher accumulation in tumor tissue compared to non-targeted ones. In comparison, PEGylated particles showed a 16-fold increase in plasma residence time and a 5-fold reduction in liver accumulation. These results motivated us to engineer new PEGylated G5 particles with PEG chains anchored to the G5 surface via acid-labile cis-aconityl linkages where the free PEG tips are functionalized with NAcGal or SP94 peptide to investigate their potential as targeting ligands for hepatic cancer cells as a function of sugar conformation (α versus ß), ligand concentration (100-4000 nM), and incubation time (2 and 24 hours) compared to fluorescently (Fl)-labeled and non-targeted G5-(Fl)6 -(NH2 )122 and G5-(Fl)6 -(Ac)107 -(cPEG)15 particles. Results show G5-(Fl)6 -(Ac)107 -(cPEG[NAcGalß ])14 particles achieve faster uptake and higher intracellular concentrations in HepG2 cancer cells compared to other G5 particles while escaping the non-specific adsorption of serum protein and phagocytosis by Kupffer cells, which make these particles the ideal carrier for selective drug delivery into hepatic cancer cells.


Subject(s)
Acetylgalactosamine/chemistry , Acetylglucosamine/analogs & derivatives , Dendrimers/chemistry , Peptides/chemistry , Polyethylene Glycols/chemistry , Acetylglucosamine/chemistry , Acetylglucosamine/pharmacokinetics , Acetylglucosamine/therapeutic use , Animals , Antigens, CD7/chemistry , Antigens, CD7/metabolism , Cells, Cultured , Dendrimers/pharmacokinetics , Dendrimers/therapeutic use , Fluorescent Dyes/chemistry , Half-Life , Hep G2 Cells , Humans , Kupffer Cells/cytology , Kupffer Cells/drug effects , Ligands , Liver/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Nude , Particle Size , Peptides/metabolism , Phagocytosis , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/therapeutic use , Rats , Time Factors , Tissue Distribution , Transplantation, Heterologous
9.
Am J Pathol ; 182(1): 142-51, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23245831

ABSTRACT

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.


Subject(s)
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
10.
Aging (Albany NY) ; 4(12): 917-22, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23249808

ABSTRACT

The down-regulation of dominant oncogenes, including C-MYC, in tumor cells often leads to the induction of senescence via mechanisms that are not completely identified. In the current study, we demonstrate that MYC-depleted melanoma cells undergo extensive DNA damage that is caused by the underexpression of thymidylate synthase (TS) and ribonucleotide reductase (RR) and subsequent depletion of deoxyribonucleoside triphosphate pools. Simultaneous genetic inhibition of TS and RR in melanoma cells induced DNA damage and senescence phenotypes very similar to the ones caused by MYC-depletion. Reciprocally, overexpression of TS and RR in melanoma cells or addition of deoxyribo-nucleosides to culture media substantially inhibited DNA damage and senescence-associated phenotypes caused by C-MYC depletion. Our data demonstrate the essential role of TS and RR in C-MYC-dependent suppression of senescence in melanoma cells.


Subject(s)
Cellular Senescence/drug effects , DNA Damage/drug effects , Deoxyribonucleosides/pharmacology , Melanoma/enzymology , Proto-Oncogene Proteins c-myc/metabolism , Ribonucleotide Reductases/metabolism , Skin Neoplasms/enzymology , Thymidylate Synthase/metabolism , Cell Line, Tumor , Down-Regulation , Gene Expression Regulation, Neoplastic , Genotype , Humans , Melanoma/genetics , Melanoma/pathology , Phenotype , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , Ribonucleoside Diphosphate Reductase/metabolism , Ribonucleotide Reductases/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Thymidylate Synthase/genetics , Time Factors , Transfection , Tumor Suppressor Proteins/metabolism
11.
Int J Radiat Oncol Biol Phys ; 84(5): e613-20, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22867891

ABSTRACT

PURPOSE: To determine the effect of short hairpin ribonucleic acid (shRNA)-mediated suppression of thymidylate synthase (TS) on cytotoxicity and radiosensitization and the mechanism by which these events occur. METHODS AND MATERIALS: shRNA suppression of TS was compared with 5-fluoro-2'-deoxyuridine (FdUrd) inactivation of TS with or without ionizing radiation in HCT116 and HT29 colon cancer cells. Cytotoxicity and radiosensitization were measured by clonogenic assay. Cell cycle effects were measured by flow cytometry. The effects of FdUrd or shRNA suppression of TS on dNTP deoxynucleotide triphosphate imbalances and consequent nucleotide misincorporations into deoxyribonucleic acid (DNA) were analyzed by high-pressure liquid chromatography and as pSP189 plasmid mutations, respectively. RESULTS: TS shRNA produced profound (≥ 90%) and prolonged (≥ 8 days) suppression of TS in HCT116 and HT29 cells, whereas FdUrd increased TS expression. TS shRNA also produced more specific and prolonged effects on dNTPs deoxynucleotide triphosphates compared with FdUrd. TS shRNA suppression allowed accumulation of cells in S-phase, although its effects were not as long-lasting as those of FdUrd. Both treatments resulted in phosphorylation of Chk1. TS shRNA alone was less cytotoxic than FdUrd but was equally effective as FdUrd in eliciting radiosensitization (radiation enhancement ratio: TS shRNA, 1.5-1.7; FdUrd, 1.4-1.6). TS shRNA and FdUrd produced a similar increase in the number and type of pSP189 mutations. CONCLUSIONS: TS shRNA produced less cytotoxicity than FdUrd but was equally effective at radiosensitizing tumor cells. Thus, the inhibitory effect of FdUrd on TS alone is sufficient to elicit radiosensitization with FdUrd, but it only partially explains FdUrd-mediated cytotoxicity and cell cycle inhibition. The increase in DNA mismatches after TS shRNA or FdUrd supports a causal and sufficient role for the depletion of dTTP thymidine triphosphate and consequent DNA mismatches underlying radiosensitization. Importantly, shRNA suppression of TS avoids FP-mediated TS elevation and its negative prognostic role. These studies support the further exploration of TS suppression as a novel radiosensitizing strategy.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , DNA Mismatch Repair , Floxuridine/pharmacology , RNA, Small Interfering/pharmacology , Radiation Tolerance/genetics , Thymidylate Synthase/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Checkpoint Kinase 1 , Cytidine Triphosphate/metabolism , Enzyme Activation/drug effects , Guanosine Triphosphate/metabolism , HT29 Cells , Humans , Phosphorylation , Protein Kinases/metabolism , Tumor Stem Cell Assay/methods
12.
Cell ; 145(3): 435-46, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21529715

ABSTRACT

Chromosomal instability in early cancer stages is caused by stress on DNA replication. The molecular basis for replication perturbation in this context is currently unknown. We studied the replication dynamics in cells in which a regulator of S phase entry and cell proliferation, the Rb-E2F pathway, is aberrantly activated. Aberrant activation of this pathway by HPV-16 E6/E7 or cyclin E oncogenes significantly decreased the cellular nucleotide levels in the newly transformed cells. Exogenously supplied nucleosides rescued the replication stress and DNA damage and dramatically decreased oncogene-induced transformation. Increased transcription of nucleotide biosynthesis genes, mediated by expressing the transcription factor c-myc, increased the nucleotide pool and also rescued the replication-induced DNA damage. Our results suggest a model for early oncogenesis in which uncoordinated activation of factors regulating cell proliferation leads to insufficient nucleotides that fail to support normal replication and genome stability.


Subject(s)
Genomic Instability , Neoplasms/genetics , Nucleotides/biosynthesis , Cyclin E/metabolism , DNA Replication , E2F Transcription Factors/metabolism , Humans , Loss of Heterozygosity , Neoplasms/metabolism , Neoplasms/pathology , Nucleotides/metabolism , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/metabolism , Retinoblastoma Protein/metabolism , S Phase
13.
Biomaterials ; 32(17): 4118-29, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21429574

ABSTRACT

There is an urgent need for novel polymeric carriers that can selectively deliver a large dose of chemotherapeutic agents into hepatic cancer cells to achieve high therapeutic activity with minimal systemic side effects. PAMAM dendrimers are characterized by a unique branching architecture and a large number of chemical surface groups suitable for coupling of chemotherapeutic agents. In this article, we report the coupling of N-acetylgalactosamine (NAcGal) to generation 5 (G5) of poly(amidoamine) (PAMAM-NH2) dendrimers via peptide and thiourea linkages to prepare NAcGal-targeted carriers used for targeted delivery of chemotherapeutic agents into hepatic cancer cells. We describe the uptake of NAcGal-targeted and non-targeted G5 dendrimers into hepatic cancer cells (HepG2) as a function of G5 concentration and incubation time. We examine the contribution of the asialoglycoprotein receptor (ASGPR) to the internalization of NAcGal-targeted dendrimers into hepatic cancer cells through a competitive inhibition assay. Our results show that uptake of NAcGal-targeted G5 dendrimers into hepatic cancer cells occurs via ASGPR-mediated endocytosis. Internalization of these targeted carriers increased with the increase in G5 concentration and incubation time following Michaelis-Menten kinetics characteristic of receptor-mediated endocytosis. These results collectively indicate that G5-NAcGal conjugates function as targeted carriers for selective delivery of chemotherapeutic agents into hepatic cancer cells.


Subject(s)
Acetylgalactosamine/pharmacokinetics , Antineoplastic Agents/pharmacology , Dendrimers/chemical synthesis , Dendrimers/pharmacokinetics , Drug Carriers/chemical synthesis , Polyamines/pharmacology , Biological Transport , Drug Delivery Systems/methods , Endocytosis , Hep G2 Cells , Humans , Polyamines/chemistry , Polymers
14.
Mutat Res ; 684(1-2): 1-10, 2010 Feb 03.
Article in English | MEDLINE | ID: mdl-20004674

ABSTRACT

Nucleoside analogs are efficacious cancer chemotherapeutics due to their incorporation into tumor cell DNA. However, they exhibit vastly different antitumor efficacies, suggesting that incorporation produces divergent effects on DNA replication. Here we have evaluated the consequences of incorporation on DNA replication and its fidelity for three structurally related deoxyguanosine analogs: ganciclovir (GCV), currently in clinical trials in a suicide gene therapy approach for cancer, D-carbocyclic 2'-deoxyguanosine (CdG) and penciclovir (PCV). GCV and CdG elicited similar cytotoxicity at low concentrations, whereas PCV was 10-100-fold less cytotoxic in human tumor cells. DNA replication fidelity was evaluated using a supF plasmid-based mutation assay. Only GCV induced a dose-dependent increase in mutation frequency, predominantly GC-->TA transversions, which contributed to cytotoxicity and implicated the ether oxygen in mutagenicity. Activation of mismatch repair with hydroxyurea decreased mutations but failed to repair the GC-->TA transversions. GCV slowed S-phase progression and CdG also induced a G2/M block, but both drugs allowed completion of one cell cycle after drug treatment followed by cell death in the second cell cycle. In contrast, PCV induced a lengthy early S-phase block due to profound suppression of DNA synthesis, with cell death in the first cell cycle after drug treatment. These data suggest that GCV and CdG elicit superior cytotoxicity due to their effects in template DNA, whereas strong inhibition of nascent strand synthesis by PCV may protect against cytotoxicity. Nucleoside analogs based on the carbohydrate structures of GCV and CdG is a promising area for antitumor drug development.


Subject(s)
Antineoplastic Agents/pharmacology , Carbohydrates/chemistry , Cell Cycle/drug effects , Cell Death/drug effects , DNA Replication/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/chemistry , Ganciclovir/pharmacology , Acyclovir/analogs & derivatives , Acyclovir/pharmacology , Base Sequence , Cell Line, Tumor , DNA Mismatch Repair , Deoxyguanosine/agonists , Deoxyguanosine/pharmacology , Genes, Transgenic, Suicide , Guanine , HCT116 Cells , Humans , Molecular Sequence Data , Mutation
16.
Cell Cycle ; 7(15): 2392-400, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18677108

ABSTRACT

To identify C-MYC targets rate-limiting for proliferation of malignant melanoma, we stably inhibited C-MYC in several human metastatic melanoma lines via lentivirus-based shRNAs approximately to the levels detected in normal melanocytes. C-MYC depletion did not significantly affect levels of E2F1 protein reported to regulate expression of many S-phase specific genes, but resulted in the repression of several genes encoding enzymes rate-limiting for dNTP metabolism. These included thymidylate synthase (TS), inosine monophosphate dehydrogenase 2 (IMPDH2) and phosphoribosyl pyrophosphate synthetase 2 (PRPS2). C-MYC depletion also resulted in reduction in the amounts of deoxyribonucleoside triphosphates (dNTPs) and inhibition of proliferation. shRNA-mediated suppression of TS, IMPDH2 or PRPS2 resulted in the decrease of dNTP pools and retardation of the cell cycle progression of melanoma cells in a manner similar to that of C-MYC-depletion in those cells. Reciprocally, concurrent overexpression of cDNAs for TS, IMPDH2 and PRPS2 delayed proliferative arrest caused by inhibition of C-MYC in melanoma cells. Overexpression of C-MYC in normal melanocytes enhanced expression of the above enzymes and increased individual dNTP pools. Analysis of in vivo C-MYC interactions with TS, IMPDH2 and PRPS2 genes confirmed that they are direct C-MYC targets. Moreover, all three proteins express at higher levels in cells from several metastatic melanoma lines compared to normal melanocytes. Our data establish a novel functional link between C-MYC and dNTP metabolism and identify its role in proliferation of tumor cells.


Subject(s)
Cell Proliferation , Melanoma/metabolism , Melanoma/pathology , Nucleotides/biosynthesis , Proto-Oncogene Proteins c-myc/physiology , Cell Proliferation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , IMP Dehydrogenase/genetics , IMP Dehydrogenase/metabolism , IMP Dehydrogenase/physiology , Melanocytes/metabolism , Melanoma/genetics , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering/pharmacology , Ribose-Phosphate Pyrophosphokinase/genetics , Ribose-Phosphate Pyrophosphokinase/metabolism , Ribose-Phosphate Pyrophosphokinase/physiology , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Thymidylate Synthase/physiology , Transfection , Tumor Cells, Cultured
17.
Mol Pharmacol ; 74(3): 863-71, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18535288

ABSTRACT

The antitumor drug 5-fluoro-2'-deoxyuridine (FdUrd) also sensitizes tumor cells to ionizing radiation in vitro and in vivo. Although radiosensitization with FdUrd requires dTTP depletion and S-phase arrest, the exact mechanism by which these events produce radiosensitization remains unknown. We hypothesized that the depletion of dTTP produces DNA mismatches that, if not repaired before irradiation, would result in radiosensitization. We evaluated this hypothesis in mismatch repair (MMR)-deficient HCT116 0-1 cells that lack the expression of the required MMR protein MLH1 (inactive MLH1), and in MMR-proficient (wild-type MLH1) HCT116 1-2 cells. Although HCT116 0-1 cells were less sensitive to FdUrd (IC(50) = 3.5 microM) versus HCT116 1-2 cells (IC(50) = 0.75 microM), when irradiation followed FdUrd (IC(50)) the MLH1-inactivated cells exhibited greater radiosensitization compared with MMR-wild-type cells [radiation enhancement ratio (RER) = 1.8 +/- 0.28 versus 1.1 +/- 0.1, respectively] and an increase (> or =8-fold) in nucleotide misincorporations. In SW620 cells and HCT116 1-2 MLH1-wild-type cells, FdUrd (IC(50)) did not produce radiosensitization nor did it increase the mutation frequency, but after short hairpin RNA-directed suppression of MLH1 this concentration produced excellent radiosensitization (RER = 1.6 +/- 0.10 and 1.5 +/- 0.06, respectively) and an increase in nucleotide misincorporations (8-fold and 6-fold, respectively). Incubation with higher concentrations of FdUrd (IC(90)) after suppression of MLH1 produced a further increase in ionizing radiation sensitivity in both SW620 and HCT116 1-2 cells (RER = 1.8 +/- 0.03 and 1.7 +/- 0.13, respectively) and nucleotide misincorporations (>10-fold in both cell lines). These results demonstrate an important role for MLH1 and implicate mismatches in radiosensitization by FdUrd.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Base Pair Mismatch/drug effects , Floxuridine/pharmacology , Nuclear Proteins/deficiency , Radiation Tolerance/drug effects , Base Pair Mismatch/radiation effects , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Death/drug effects , Cell Death/radiation effects , HCT116 Cells , Humans , MutL Protein Homolog 1 , Mutation/genetics , Nucleotides/metabolism , Plasmids/genetics , RNA, Small Interfering/metabolism , Radiation Tolerance/radiation effects , Radiation, Ionizing
18.
J Clin Oncol ; 25(26): 4043-50, 2007 Sep 10.
Article in English | MEDLINE | ID: mdl-17827452

ABSTRACT

Radiosensitization with antimetabolites has improved clinical outcome for patients with solid malignancies, especially cancers of the GI tract, cervix, and head and neck. Fluorouracil (FU) and hydroxyurea have been widely used clinically during the last four decades, and promising results have been observed more recently with gemcitabine. Although the antimetabolites all target DNA replication, they differ with respect to the mechanisms by which they produce radiosensitization. The antimetabolite radiosensitizers may inhibit thymidylate synthase (TS) or ribonucleotide reductase, and the nucleoside/nucleobase analogs can be incorporated into DNA. Radiosensitization can result from chemotherapy-induced increase in DNA double-strand breaks or inhibition of their repair. Studies of repair pathways involved in radiosensitization with antimetabolites implicate base excision repair with the TS inhibitors, homologous recombination with gemcitabine, and mismatch repair with FU and gemcitabine. Gemcitabine can also stimulate epidermal growth factor receptor (EGFR) phosphorylation; inhibiting this effect with EGFR inhibitors can potentiate cytotoxicity and radiosensitization. Additional work is necessary to determine more precisely the processes by which antimetabolites act as radiation sensitizers and to define the optimal sequencing of these agents with EGFR inhibitors to provide better guidance for clinical protocols combining these drugs with radiotherapy.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Neoplasms/drug therapy , Neoplasms/radiotherapy , Radiation-Sensitizing Agents/therapeutic use , Animals , Combined Modality Therapy , Humans
19.
Mol Cancer Ther ; 6(6): 1858-68, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17575114

ABSTRACT

Radiation sensitization by 2',2'-difluoro-2'-deoxycytidine (dFdCyd) has correlated with dATP depletion [dFdCDP-mediated inhibition of ribonucleotide reductase (RR)] and S-phase accumulation. We hypothesized that radiosensitization by dFdCyd is due to nucleotide misincorporations in the presence of deoxynucleotide triphosphate pool imbalances, which, if not repaired, augments cell death following irradiation. The ability of dFdCyd to produce misincorporations was measured as pSP189 plasmid mutations in hMLH1-deficient [mismatch repair (MMR) deficient] and hMLH1-expressing (MMR proficient) HCT116 cells. Only MMR-deficient cells showed a significant increase in nucleotide misincorporations (2- to 3-fold increase; P or=5-fold increase; P < 0.05), thus further implicating the inhibition of RR as the mechanism underlying radiosensitization by dFdCyd. These data showed that the presence and persistence of mismatched nucleotides is integral to radiosensitization by dFdCyd and suggest a role for hMLH1 deficiency in eliciting the radiosensitizing effect.


Subject(s)
Base Pair Mismatch , Deoxycytidine/analogs & derivatives , Radiation-Sensitizing Agents/pharmacology , Cell Line, Tumor , Deoxycytidine/pharmacology , Humans , Mutation , RNA, Small Interfering , Gemcitabine
20.
Cancer Res ; 66(7): 3845-51, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16585212

ABSTRACT

Suicide gene therapy with the herpes simplex virus thymidine kinase (HSV-TK) cDNA and ganciclovir can elicit cytotoxicity to transgene-expressing and nonexpressing bystander cells via transfer of ganciclovir phosphates through gap junctions. HeLa cells do not exhibit bystander cytotoxicity, although we showed recently that they transfer low levels of ganciclovir phosphates to bystander cells. Here, we attempted to induce bystander cytotoxicity using hydroxyurea, an inhibitor of ribonucleotide reductase, to decrease the endogenous dGTP pool, which should lessen competition with ganciclovir triphosphate for DNA incorporation. Addition of hydroxyurea to cocultures of HSV-TK-expressing and bystander cells synergistically increased ganciclovir-mediated cytotoxicity to both cell populations while producing primarily an additive effect in cultures of 100% HSV-TK-expressing cells. Whereas HSV-TK-expressing cells in coculture were approximately 50-fold less sensitive to ganciclovir compared with cultures of 100% HSV-TK-expressing cells, addition of hydroxyurea restored ganciclovir sensitivity. Quantification of deoxynucleoside triphosphate pools showed that hydroxyurea decreased dGTP pools without significantly affecting ganciclovir triphosphate levels. Although hydroxyurea significantly increased the ganciclovir triphosphate:dGTP value for 12 to 24 hours in HSV-TK-expressing and bystander cells from coculture (1.4- to 4.9-fold), this value was increased for <12 hours (2.5-fold) in 100% HSV-TK-expressing cells. These data suggest that the prolonged increase in the ganciclovir triphosphate:dGTP value in cells in coculture resulted in synergistic cytotoxicity. Compared with enhancement of bystander cytotoxicity through modulation of gap junction intercellular communication, this strategy is superior because it increased cytotoxicity to both HSV-TK-expressing and bystander cells in coculture. This approach may improve clinical efficacy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Ganciclovir/pharmacology , Hydroxyurea/pharmacology , Simplexvirus/genetics , Thymidine Kinase/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Coculture Techniques , Deoxyguanine Nucleotides/metabolism , Drug Synergism , Ganciclovir/adverse effects , Ganciclovir/analogs & derivatives , Ganciclovir/metabolism , Ganciclovir/pharmacokinetics , Genetic Therapy/methods , HeLa Cells , Humans , Hydroxyurea/adverse effects , Nucleotides/metabolism , Simplexvirus/enzymology , Thymidine Kinase/biosynthesis , Thymidine Kinase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...