Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 443
Filter
1.
Ann Oncol ; 35(7): 643-655, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38777726

ABSTRACT

BACKGROUND: POLE and POLD1 proofreading deficiency (POLE/D1pd) define a rare subtype of ultramutated metastatic colorectal cancer (mCRC; over 100 mut/Mb). Disease-specific data about the activity and efficacy of immune checkpoint inhibitors (ICIs) in POLE/D1pd mCRC are lacking and it is unknown whether outcomes may be different from mismatch repair-deficient (dMMR)/microsatellite instability-high (MSI-H) mCRCs treated with ICIs. PATIENTS AND METHODS: In this global study, we collected 27 patients with mCRC harboring POLE/D1 mutations leading to proofreading deficiency and treated with anti-programmed cell death-ligand 1 aloneĀ +/- anti-cytotoxic T-lymphocyte antigen-4 agents. We collected clinicopathological and genomic characteristics, response, and survival outcomes after ICIs of POLE/D1pd mCRC and compared them with a cohort of 610 dMMR/MSI-H mCRC patients treated with ICIs. Further genomic analyses were carried out in an independent cohort of 7241 CRCs to define POLE and POLD1pd molecular profiles and mutational signatures. RESULTS: POLE/D1pd was associated with younger age, male sex, fewer RAS/BRAF driver mutations, and predominance of right-sided colon cancers. Patients with POLE/D1pd mCRC showed a significantly higher overall response rate (ORR) compared to dMMR/MSI-H mCRC (89% versus 54%; PĀ = 0.01). After a median follow-up of 24.9 months (interquartile range: 11.3-43.0 months), patients with POLE/D1pd showed a significantly superior progression-free survival (PFS) compared to dMMR/MSI-H mCRC [hazard ratio (HR)Ā = 0.24, 95% confidence interval (CI) 0.08-0.74, PĀ = 0.01] and superior overall survival (OS) (HRĀ = 0.38, 95% CI 0.12-1.18, PĀ = 0.09). In multivariable analyses including the type of DNA repair defect, POLE/D1pd was associated with significantly improved PFS (HRĀ = 0.17, 95% CI 0.04-0.69, PĀ =Ā 0.013) and OS (HRĀ = 0.24, 95% CI 0.06-0.98, PĀ = 0.047). Molecular profiling showed that POLE/D1pd tumors have higher tumor mutational burden (TMB). Responses were observed in both subtypes and were associated with the intensity of POLE/D1pd signature. CONCLUSIONS: Patients with POLE/D1pd mCRC showed more favorable outcomes compared to dMMR/MSI-H mCRC to treatment with ICIs in terms of tumor response and survival.


Subject(s)
Colorectal Neoplasms , DNA Polymerase III , DNA Polymerase II , Immune Checkpoint Inhibitors , Mutation , Poly-ADP-Ribose Binding Proteins , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Mismatch Repair , DNA Polymerase II/antagonists & inhibitors , DNA Polymerase III/antagonists & inhibitors , Immune Checkpoint Inhibitors/therapeutic use , Microsatellite Instability , Poly-ADP-Ribose Binding Proteins/genetics
2.
BMC Cancer ; 15: 325, 2015 Apr 29.
Article in English | MEDLINE | ID: mdl-25924900

ABSTRACT

BACKGROUND: Hepatocyte carcinoma (HCC) is one of the most common malignancies worldwide. Despite many achievements in diagnosis and treatment, HCC mortality remains high due to the malignant nature of the disease. Novel approaches, especially for targeted therapy, are being extensively explored. Gene therapy is ideal for such purpose for its specific expression of exogenous genes in HCC cells driven by tissue-specific promoter. However strategies based on correction of mutations or altered expressions of genes responsible for the development/progression of HCC have limitations because these aberrant molecules are not presented in all cancerous cells. In the current work, we adopted a novel strategy by targeting the DNA replication step which is essential for proliferation of every cancer cell. METHODS: A recombinant adenovirus with alpha fetoprotein (AFP) promoter-controlled expressions of artificial microRNAs targeting DNA polymerases α, ƎĀ“, ƎĀµ and recombinant active Caspase 3, namely Ad/AFP-Casp-AFP-amiR, was constructed. RESULTS: The artificial microRNAs could efficiently inhibit the expression of the target polymerases in AFP-positive HCC cells at both RNA and protein levels, and HCC cells treated with the recombinant virus Ad/AFP-Casp-AFP-amiR exhibited significant G0/1 phase arrest. The proliferation of HCC cells were significantly inhibited by Ad/AFP-Casp-AFP-amiR with increased apoptosis. On the contrary, the recombinant adenovirus Ad/AFP-Casp-AFP-amiR did not inhibit the expression of DNA polymerases α, ƎĀ“ or ƎĀµ in AFP-negative human normal liver cell HL7702, and showed no effect on the cell cycle progression, proliferation or apoptosis. CONCLUSIONS: Inhibition of DNA polymerases α, ƎĀ“ and ƎĀµ by AFP promoter-driven artificial microRNAs may lead to effective growth arrest of AFP-positive HCC cells, which may represent a novel strategy for gene therapy by targeting the genes that are essential for the growth/proliferation of cancer cells, avoiding the limitations set by any of the individually altered gene.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA Polymerase III/genetics , DNA Polymerase II/genetics , DNA Polymerase I/genetics , Liver Neoplasms/genetics , Adenoviridae/genetics , Animals , Apoptosis/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Line, Tumor , DNA Polymerase I/antagonists & inhibitors , DNA Polymerase II/antagonists & inhibitors , DNA Polymerase III/antagonists & inhibitors , Genetic Therapy , Humans , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Mice , MicroRNAs/genetics , Molecular Targeted Therapy , Promoter Regions, Genetic , Xenograft Model Antitumor Assays
3.
RNA ; 16(5): 904-12, 2010 May.
Article in English | MEDLINE | ID: mdl-20357345

ABSTRACT

Alternative splicing accounts for much of the complexity in higher eukaryotes. Thus, its regulation must allow for flexibility without hampering either its specificity or its fidelity. The mechanisms involved in alternative splicing regulation, especially those acting through coupling with transcription, have not been deeply studied in in vivo models. Much of our knowledge comes from in vitro approaches, where conditions can be precisely controlled at the expense of losing several levels of regulation present in intact cells. Here we studied the relative order of removal of the introns flanking a model alternative cassette exon. We show that there is a preferential removal of the intron downstream from the cassette exon before the upstream intron has been removed. Most importantly, both cis-acting mutations and trans-acting factors that regulate the model alternative splicing event differentially affect the relative order of removal. However, reduction of transcriptional elongation causing higher inclusion of the cassette exon does not change the order of intron removal, suggesting that the assumption, according to the "first come, first served" model, that slow elongation promotes preferential excision of the upstream intron has to be revised. We propose instead that slow elongation favors commitment to exon inclusion during spliceosome assembly. Our results reveal that measuring the order of intron removal may be a straightforward read-out to discriminate among different mechanisms of alternative splice site selection.


Subject(s)
Alternative Splicing , Introns , Base Sequence , Cell Line , DNA Polymerase II/antagonists & inhibitors , DNA Primers/genetics , Exons , Fibronectins/genetics , Humans , Kinetics , Models, Biological , Mutation , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splice Sites , Transfection
4.
Cancer Res ; 80(8): 1735-1747, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32161100

ABSTRACT

Checkpoint kinase 1 (CHK1) is a key mediator of the DNA damage response that regulates cell-cycle progression, DNA damage repair, and DNA replication. Small-molecule CHK1 inhibitors sensitize cancer cells to genotoxic agents and have shown single-agent preclinical activity in cancers with high levels of replication stress. However, the underlying genetic determinants of CHK1 inhibitor sensitivity remain unclear. We used the developmental clinical drug SRA737 in an unbiased large-scale siRNA screen to identify novel mediators of CHK1 inhibitor sensitivity and uncover potential combination therapies and biomarkers for patient selection. We identified subunits of the B-family of DNA polymerases (POLA1, POLE, and POLE2) whose silencing sensitized the human A549 non-small cell lung cancer (NSCLC) and SW620 colorectal cancer cell lines to SRA737. B-family polymerases were validated using multiple siRNAs in a panel of NSCLC and colorectal cancer cell lines. Replication stress, DNA damage, and apoptosis were increased in human cancer cells following depletion of the B-family DNA polymerases combined with SRA737 treatment. Moreover, pharmacologic blockade of B-family DNA polymerases using aphidicolin or CD437 combined with CHK1 inhibitors led to synergistic inhibition of cancer cell proliferation. Furthermore, low levels of POLA1, POLE, and POLE2 protein expression in NSCLC and colorectal cancer cells correlated with single-agent CHK1 inhibitor sensitivity and may constitute biomarkers of this phenotype. These findings provide a potential basis for combining CHK1 and B-family polymerase inhibitors in cancer therapy. SIGNIFICANCE: These findings demonstrate how the therapeutic benefit of CHK1 inhibitors may potentially be enhanced and could have implications for patient selection and future development of new combination therapies.


Subject(s)
Aphidicolin/pharmacology , Checkpoint Kinase 1/antagonists & inhibitors , Colorectal Neoplasms/drug therapy , Heterocyclic Compounds, 4 or More Rings/pharmacology , Lung Neoplasms/drug therapy , Retinoids/pharmacology , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Damage , DNA Polymerase I/antagonists & inhibitors , DNA Polymerase I/genetics , DNA Polymerase I/metabolism , DNA Polymerase II/antagonists & inhibitors , DNA Polymerase II/genetics , DNA Polymerase II/metabolism , DNA Polymerase beta , Drugs, Investigational/pharmacology , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Proteins/antagonists & inhibitors , Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors , Poly-ADP-Ribose Binding Proteins/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , RNA, Small Interfering/analysis , RNA, Small Interfering/genetics
5.
Mol Cell Biol ; 24(1): 217-27, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14673157

ABSTRACT

Relocation of euchromatic genes near the heterochromatin region often results in mosaic gene silencing. In Saccharomyces cerevisiae, cells with the genes inserted at telomeric heterochromatin-like regions show a phenotypic variegation known as the telomere-position effect, and the epigenetic states are stably passed on to following generations. Here we show that the epigenetic states of the telomere gene are not stably inherited in cells either bearing a mutation in a catalytic subunit (Pol2) of replicative DNA polymerase epsilon (Pol epsilon) or lacking one of the nonessential and histone fold motif-containing subunits of Pol epsilon, Dpb3 and Dpb4. We also report a novel and putative chromatin-remodeling complex, ISW2/yCHRAC, that contains Isw2, Itc1, Dpb3-like subunit (Dls1), and Dpb4. Using the single-cell method developed in this study, we demonstrate that without Pol epsilon and ISW2/yCHRAC, the epigenetic states of the telomere are frequently switched. Furthermore, we reveal that Pol epsilon and ISW2/yCHRAC function independently: Pol epsilon operates for the stable inheritance of a silent state, while ISW2/yCHRAC works for that of an expressed state. We therefore propose that inheritance of specific epigenetic states of a telomere requires at least two counteracting regulators.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Polymerase II/antagonists & inhibitors , Saccharomyces cerevisiae/genetics , Telomere/metabolism , Transcription Factors/metabolism , Chromatin/metabolism , DNA Replication/physiology , Saccharomyces cerevisiae/enzymology
6.
Mol Cell Biol ; 7(12): 4594-8, 1987 Dec.
Article in English | MEDLINE | ID: mdl-2830501

ABSTRACT

Antibodies against human c-myc protein have been reported to inhibit DNA polymerase activity and endogenous DNA synthesis in isolated nuclei, suggesting a role for c-myc in DNA replication. Using the same antibody preparations, we observed equivalent inhibition of simian virus 40 DNA replication and DNA polymerase alpha and delta activities in vitro, as well as inhibition of DNA synthesis in isolated nuclei. However, the c-myc antibodies could be completely separated from the DNA synthesis inhibition activity. c-myc antibodies prepared in other laboratories also did not interfere with initiation of simian virus 40 DNA replication, DNA synthesis at replication forks, or DNA polymerase alpha or delta activity. Therefore, the previously reported inhibition of DNA synthesis by some antibody preparations resulted from the presence of an unidentified inhibitor of DNA polymerases alpha and delta and not from the action of c-myc antibodies.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Antibodies/isolation & purification , DNA Replication , Proto-Oncogene Proteins/physiology , Animals , Cell Line , DNA Polymerase II/antagonists & inhibitors , DNA Polymerase III , DNA, Viral/biosynthesis , Humans , Immunologic Techniques , Immunosorbent Techniques , Nucleic Acid Synthesis Inhibitors , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-myc , Simian virus 40/genetics
7.
Mol Cell Biol ; 1(8): 680-6, 1981 Aug.
Article in English | MEDLINE | ID: mdl-9279381

ABSTRACT

Aphidicolin, a known inhibitor of eucaryotic deoxyribonucleic acid (DNA) polymerase alpha, efficiently inhibited amplification of ribosomal DNA during oogenesis in Xenopus laevis. DNA polymerase alpha, but not DNA polymerase gamma, as isolated from ovaries, was sensitive to aphidicolin. DNA polymerase beta was not detectable in Xenopus ovary extracts. Therefore, DNA polymerase alpha plays a major role in ribosomal ribonucleic acid gene amplification.


Subject(s)
DNA Polymerase II/metabolism , DNA, Ribosomal/metabolism , Gene Amplification/drug effects , Animals , Aphidicolin/pharmacology , DNA Polymerase II/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Female , Oogenesis/genetics , Xenopus laevis
8.
Mol Cell Biol ; 6(11): 3815-25, 1986 Nov.
Article in English | MEDLINE | ID: mdl-3025613

ABSTRACT

Aphidicolin, a specific inhibitor of DNA polymerase alpha, provided a novel method for distinguishing between initiation of DNA synthesis at the simian virus 40 (SV40) origin of replication (ori) and continuation of replication beyond ori. In the presence of sufficient aphidicolin to inhibit total DNA synthesis by 50%, initiation of DNA replication in SV40 chromosomes or ori-containing plasmids continued in vitro, whereas DNA synthesis in the bulk of SV40 replicative intermediate DNA (RI) that had initiated replication in vivo was rapidly inhibited. This resulted in accumulation of early RI in which most nascent DNA was localized within a 600- to 700-base-pair region centered at ori. Accumulation of early RI was observed only under conditions that permitted initiation of SV40 ori-dependent, T-antigen-dependent DNA replication and only when aphidicolin was added to the in vitro system. Increasing aphidicolin concentrations revealed that DNA synthesis in the ori region was not completely resistant to aphidicolin but simply less sensitive than DNA synthesis at forks that were farther away. Since DNA synthesized in the presence of aphidicolin was concentrated in the 300 base pairs on the early gene side of ori, we conclude that the initial direction of DNA synthesis was the same as that of early mRNA synthesis, consistent with the model proposed by Hay and DePamphilis (Cell 28:767-779, 1982). The data were also consistent with initiation of the first DNA chains in ori by CV-1 cell DNA primase-DNA polymerase alpha. Synthesis of pppA/G(pN)6-8(pdN)21-23 chains on a single-stranded DNA template by a purified preparation of this enzyme was completely resistant to aphidicolin, and further incorporation of deoxynucleotide monophosphates was inhibited. Therefore, in the presence of aphidicolin, this enzyme could initiate RNA-primed DNA synthesis at ori first in the early gene direction and then in the late gene direction, but could not continue DNA synthesis for an extended distance.


Subject(s)
DNA Replication/drug effects , Diterpenes/pharmacology , Simian virus 40/genetics , Animals , Aphidicolin , Cell Line , Cell Nucleus/metabolism , Chlorocebus aethiops , DNA Polymerase II/antagonists & inhibitors , DNA Restriction Enzymes , DNA, Viral/drug effects , Kidney , Plasmids
9.
Mol Cell Biol ; 15(5): 2374-82, 1995 May.
Article in English | MEDLINE | ID: mdl-7739521

ABSTRACT

Src and Yes protein-tyrosine kinase activities are elevated in malignant and premalignant tumors of the colon. To determine whether Src activity is elevated throughout the human colon carcinoma cell cycle as it is in polyomavirus middle T antigen- or F527 Src-transformed cells, and whether Yes activity, which is lower than that of Src in the carcinoma cells, is regulated differently, we measured their activities in cycling cells. We observed that the activities of both kinases were higher throughout all phases of the HT-29 colon carcinoma cell cycle than in corresponding phases of the fibroblast cycle. In addition, during mitosis of HT-29 cells, Src specific activity increased two- to threefold more, while Yes activity and abundance decreased threefold. The decreased steady-state protein levels of Yes during mitosis appeared to be due to both decreased synthesis and increased degradation of the protein. Inhibition of tyrosine but not serine/threonine phosphatases abolished the mitotic activation of Src. Mitotic Src was phosphorylated at novel serine and threonine sites and dephosphorylated at Tyr-527. Two cellular proteins (p160 and p180) were phosphorylated on tyrosine only during mitosis. Tyrosine phosphorylation of several other proteins decreased during mitosis. Thus, Src in HT-29 colon carcinoma cells, similar to Src complexed to polyomavirus middle T antigen or activated by mutation at Tyr-527, is highly active in all phases of the cell cycle. Moreover, Src activity further increases during mitosis, whereas Yes activity and abundance decrease. Thus, Src and Yes appear to be regulated differently during mitosis of HT-29 colon carcinoma cells.


Subject(s)
Colonic Neoplasms/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , src-Family Kinases , Aphidicolin/pharmacology , CSK Tyrosine-Protein Kinase , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA Polymerase II/antagonists & inhibitors , G2 Phase/genetics , G2 Phase/physiology , Humans , Mitosis/drug effects , Mitosis/genetics , Mitosis/physiology , Nocodazole/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphorylation , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-yes , Tumor Cells, Cultured
10.
Nucleic Acids Res ; 27(19): 3799-804, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10481018

ABSTRACT

The contribution of human DNA polymerase epsilon to nuclear DNA replication was studied. Antibody K18 that specifically inhibits DNA polymerase activity of human DNA polymerase epsilon in vitro significantly inhibits DNA synthesis both when microinjected into nuclei of exponentially growing human fibroblasts and in isolated HeLa cell nuclei. The capability of this neutralizing antibody to inhibit DNA synthesis in cells is comparable to that of monoclonal antibody SJK-132-20 against DNA polymerase alpha. Contrary to the antibody against DNA polymerase alpha, antibody K18 against DNA polymerase epsilon did not inhibit SV40 DNA replication in vitro. These results indicate that DNA polymerase epsilon plays a role in replicative DNA synthesis in proliferating human cells like DNA polymerase alpha, and that this role for DNA polymerase epsilon cannot be modeled by SV40 DNA replication.


Subject(s)
DNA Polymerase II/metabolism , DNA Replication , DNA, Viral/biosynthesis , Simian virus 40/genetics , Animals , Antibodies/immunology , Bromodeoxyuridine/metabolism , Catalytic Domain , Cattle , Cell Line , DNA Polymerase II/antagonists & inhibitors , DNA Polymerase II/immunology , Fibroblasts/cytology , HeLa Cells , Humans , Neutralization Tests , Rabbits , Simian virus 40/physiology , Virus Replication
11.
J Natl Cancer Inst ; 83(16): 1160-4, 1991 Aug 21.
Article in English | MEDLINE | ID: mdl-1886148

ABSTRACT

The toxicity profile and the pharmacokinetics of aphidicolin glycinate, a water-soluble analogue of aphidicolin, have been evaluated in two consecutive phase I clinical studies. In the first study, aphidicolin glycinate was given by 1-hour infusion for 5 consecutive days, every 3 weeks (daily x 5 study); in the second study, which was planned on the basis of the pharmacokinetic information obtained in the previous study, the drug was given by 24-hour continuous infusion. Treatment was repeated every 3 weeks. In the daily x 5 study, the daily dose was escalated from 12 mg/m2 to the maximum tolerated dose of 2250 mg/m2. Local toxicity was dose limiting. Elimination half-life was 2 +/- 0.2 hours (mean +/- SE) with aphidicolin being undetectable 6-8 hours after the end of the infusion. In the 24-hour continuous-infusion study, the dose was escalated from 435 mg/m2 to the maximum tolerated dose of 4500 mg/m2. Local toxicity was dose limiting, while other toxic effects were absent. The experimentally determined concentrations at the steady state were in agreement with those predicted on the basis of the available pharmacokinetic data. The targeted concentration at the steady state of 3 micrograms/mL was achieved at doses greater than or equal to 3000 mg/m2. Twenty-four-hour continuous infusion is the recommended schedule for clinical evaluations of aphidicolin glycinate as the synchronizing agent or in combination with cisplatin.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Aphidicolin/analogs & derivatives , DNA Polymerase II/antagonists & inhibitors , Diterpenes/pharmacokinetics , Adult , Aged , Antineoplastic Agents/administration & dosage , Diterpenes/administration & dosage , Drug Administration Schedule , Drug Evaluation , Female , Half-Life , Humans , Infusions, Intravenous , Male , Middle Aged
12.
J Natl Cancer Inst ; 81(7): 535-9, 1989 Apr 05.
Article in English | MEDLINE | ID: mdl-2493524

ABSTRACT

We have studied the effect of glutathione reduction by buthionine sulfoximine (BSO), a specific inhibitor of gamma -glutamyl cysteine synthetase, on DNA repair after cisplatin damage in an ovarian cancer cell line with in vitro induced resistance to cisplatin. In addition, we have examined the effect of aphidicolin, a specific inhibitor of DNA polymerase alpha, in combination with BSO on cisplatin-associated DNA repair. BSO treatment was found to partially inhibit DNA repair, and the addition of aphidicolin caused nearly a 100% inhibition in DNA repair activity. Treatment of cells with glutathione ester after BSO resulted in complete recovery of DNA repair activity or partial recovery if aphidicolin was present. The significance of these results to the chemosensitizing effects of BSO medicated glutathione reduction is discussed.


Subject(s)
Cisplatin/pharmacology , DNA Repair , Glutathione/physiology , Ovarian Neoplasms/genetics , Aphidicolin , Buthionine Sulfoximine , Centrifugation, Density Gradient , DNA Polymerase II/antagonists & inhibitors , DNA Repair/drug effects , Diterpenes/pharmacology , Drug Resistance , Female , Glutathione/metabolism , Humans , Methionine Sulfoximine/analogs & derivatives , Methionine Sulfoximine/pharmacology , Ovarian Neoplasms/metabolism , Tumor Cells, Cultured
13.
Cancer Res ; 47(14): 3725-8, 1987 Jul 15.
Article in English | MEDLINE | ID: mdl-3109732

ABSTRACT

Repair-proficient human cells can be sensitized to exposure to UV radiation at 254 nm by postirradiation incubation in the presence of the eukaryotic alpha polymerase inhibitor, aphidicolin. The degree of sensitization has been examined in cells cultured from humans suffering from various types of sun-sensitive syndromes. Xeroderma pigmentosum (XP) variant and Bloom's cell lines (both excision proficient) were strongly sensitized by aphidicolin. An excision repair proficient Cockayne's cell line and a deficient XPD line were both sensitized to a level similar to the sensitivity of excision deficient XPA cells. In contrast, three XPC cell lines which show intermediate UV-induced repair replication and UV sensitivity were sensitized little (in one case) or not at all (in two cases) to UV by postirradiation inhibition of the alpha polymerase. These results lead us to conclude that there are two independent pathways of biologically effective excision repair, the major one of which involves the alpha polymerase and a second, less efficient and slower pathway which is independent of the alpha polymerase and which is the only pathway operating in two of the three XPC strains tested. The rates of biologically effective excision repair were similar in normal, XP variant, and Cockayne's cell lines, but these rates were considerably higher than published rates of dimer excision measured under similar conditions.


Subject(s)
DNA Repair , Sunlight , Aphidicolin , Bloom Syndrome/genetics , Cells, Cultured , DNA Polymerase II/antagonists & inhibitors , Diterpenes/pharmacology , Humans , Ultraviolet Rays/adverse effects , Xeroderma Pigmentosum/genetics
14.
Cancer Res ; 42(6): 2260-4, 1982 Jun.
Article in English | MEDLINE | ID: mdl-7042080

ABSTRACT

9-beta-D-Arabinofuranosyl-2-fluoroadenine (2-F-ara-A), a derivative of 9-beta-D-arabinofuranosyladenine (ara-A) that is resistant to deamination, selectively inhibits DNA synthesis and has activity against mouse leukemia L1210 comparable to that of ara-A plus the adenosine deaminase inhibitor, 2'-deoxycoformycin. To determine if these two nucleosides have similar modes of action, comparisons were made of their effects and those of their triphosphates on enzymes known to be inhibited by ara-A or 9-beta-D-arabinofuranosyladenine 5'-triphosphate. 9-beta-D-Arabinofuranosyl-2-fluoroadenine 5'-triphosphate was more effective than 9-beta-D-arabinofuranosyladenine 5'-triphosphate in inhibiting the reduction of adenosine 5'-diphosphate and cytidine 5'-diphosphate by ribonucleotide reductase from HEp-2 cells or L1210 cells. DNA polymerase alpha from L1210 cells was equally sensitive to 9-beta-D-arabinofuranosyl-2-fluoroadenine 5'-triphosphate and 9-beta-D-arabinofuranosyladenine 5'-triphosphate, and DNA polymerase beta from L1210 cells was much less sensitive to both triphosphates. S-Adenosylhomocysteine hydrolase from L1210 cells was inactivated by 2-F-ara-A and ara-A, but higher concentrations of the fluoro derivative were required. These results are consistent with 2-F-ara-A and ara-A inhibition of DNA synthesis by inhibition of ribonucleotide reductase and DNA polymerase alpha.


Subject(s)
DNA Replication/drug effects , Hydrolases/antagonists & inhibitors , Leukemia L1210/enzymology , Nucleic Acid Synthesis Inhibitors , Ribonucleotide Reductases/antagonists & inhibitors , Vidarabine/analogs & derivatives , Vidarabine/pharmacology , Adenosylhomocysteinase , Animals , DNA Polymerase I/antagonists & inhibitors , DNA Polymerase II/antagonists & inhibitors , Mice
15.
Cancer Res ; 46(12 Pt 1): 6139-42, 1986 Dec.
Article in English | MEDLINE | ID: mdl-3490907

ABSTRACT

alpha 1-Antichymotrypsin (ACT), which is known as an efficient serum protease inhibitor and is detected in tumor cell nuclei, was found to inhibit the activity of DNA polymerase alpha purified from human stomach adenocarcinoma. The concentration of ACT required for 50% inhibition was 1.0 mg/ml and the manner of its inhibition showed the partially competitive relationship between ACT and DNA in the assay system. Furthermore the removal of ACT by anti-ACT antibody lost its antichymotryptic and anti-DNA polymerase activities in parallel. On the other hand, it did not inhibit the activity of human DNA polymerase beta. Other human serum proteins including serum albumin, alpha 1-acid glycoprotein, alpha 1-antitrypsin, and immunoglobulin G as well as other protease inhibitors such as leupeptin, pepstatin, phenylmethylsulfonyl fluoride, and chymostatin did not affect the activity of DNA polymerase alpha. Furthermore ACT heated at 60 degrees C did not inhibit DNA polymerase alpha, although it could still bind to DNA as well as native ACT. It was therefore concluded that the inhibitory action of ACT on DNA polymerase alpha was a direct phenomenon unrelated to its protease inhibitory or DNA binding activities.


Subject(s)
DNA Polymerase II/antagonists & inhibitors , Protease Inhibitors/pharmacology , alpha 1-Antichymotrypsin/pharmacology , Blood Proteins/pharmacology , DNA Polymerase II/metabolism , Hot Temperature , Humans , Neoplasms/analysis , alpha 1-Antichymotrypsin/analysis , alpha 1-Antichymotrypsin/metabolism
16.
Cancer Res ; 42(10): 4050-3, 1982 Oct.
Article in English | MEDLINE | ID: mdl-6809309

ABSTRACT

We have used alkaline elution analysis to determine whether strand breaks are detectable in L1210 DNA labeled during and prior to 1-beta-D-arabinofuranosylcytosine (ara-C) exposure. The results demonstrate that ara-C enhances elution of previously synthesized DNA as well as replicating DNA. ara-C induced dose-dependent strand breaks when DNA was labeled prior to drug exposure. In contrast, simultaneous exposure of cells to tritiated thymidine and drug resulted in maximal elution rates with 10(-6) M ara-C. These findings are compared to those obtained with aphidicolin, another inhibitor of DNA synthesis, which unlike ara-C is not incorporated into the DNA strand. These results suggest that both ara-C and aphidicolin increase DNA elution rates by impairing DNA synthesis. The different dose-dependent patterns during replicative DNA synthesis may result from the incorporation of ara-C in the DNA strand.


Subject(s)
Cytarabine/pharmacology , DNA Polymerase II/antagonists & inhibitors , DNA Replication/drug effects , Diterpenes/pharmacology , Leukemia L1210/metabolism , Nucleic Acid Synthesis Inhibitors , Animals , Aphidicolin , Kinetics , Mice
17.
Cancer Res ; 44(1): 319-23, 1984 Jan.
Article in English | MEDLINE | ID: mdl-6418378

ABSTRACT

An unusual isozyme of lactate dehydrogenase, originally detected in Kirsten sarcoma virus-infected cells and later shown to be induced in normal mammalian cells by anaerobic shock, has also been reported at elevated levels in several human carcinomas. This enzyme is subject to inhibition by guanosine triphosphate and by the dinucleosides 5',5"'-diadenosine tetraphosphate and 5',5"'-diguanosine tetraphosphate (4). Fluctuation of the activity of this enzyme in soluble extracts of synchronized HeLa cells suggests the enzyme may be linked to DNA synthesis. The lactate dehydrogenase K activity increased in early S phase and then decreased to nearly undetectable levels during the period of most active DNA synthesis. This was observed in cells synchronized by thymidine excess or by aphidicolin, an inhibitor of DNA polymerase alpha.


Subject(s)
Interphase , L-Lactate Dehydrogenase/metabolism , Aphidicolin , DNA Polymerase II/antagonists & inhibitors , DNA Replication , Diterpenes/toxicity , HeLa Cells/drug effects , HeLa Cells/enzymology , Humans , Isoenzymes , Kinetics , Neoplasms/enzymology
18.
Cancer Res ; 44(5): 1867-70, 1984 May.
Article in English | MEDLINE | ID: mdl-6424933

ABSTRACT

In vitro modification of M13 phage single-stranded DNA with 4-hydroxyaminoquinoline 1-oxide (4HAQO) resulted in four kinds of adducts: three guanine adducts, QGI, QGII, and QGIII; and one adenine adduct, QA, at ratios of 16.4 47.3, 13.7, and 22.6, respectively. The carcinogen-modified DNA, initiated with a sequence-defined oligodeoxynucleotide primer, was replicated in vitro with Escherichia coli DNA polymerase I (Klenow fragment) and calf thymus DNA polymerases alpha and beta. The reaction products were analyzed on a DNA-sequencing gel. DNA elongation by DNA polymerase I was arrested at putative guanine adducts on the template in three ways: at one base prior to guanine; at positions opposite to guanine; and at one base beyond guanine. Similar patterns of elongation arrest were also obtained with the mammalian DNA polymerases alpha and beta. In contrast to guanine adducts, the adenine adduct, QA, might lack the capacity to arrest DNA chain elongation by DNA polymerases.


Subject(s)
4-Hydroxyaminoquinoline-1-oxide/toxicity , Aminoquinolines/toxicity , DNA Polymerase II/antagonists & inhibitors , DNA Polymerase I/antagonists & inhibitors , Animals , Base Sequence , Cattle , Coliphages , DNA, Single-Stranded/metabolism , Escherichia coli/enzymology , Guanine , Kinetics , Templates, Genetic , Thymus Gland/enzymology
19.
Cancer Res ; 45(3): 1127-31, 1985 Mar.
Article in English | MEDLINE | ID: mdl-2578872

ABSTRACT

Base propenals arise from DNA by a Fe(II)-bleomycin-mediated reaction which leads to strand scission. These compounds undergo addition-elimination reactions with thiols and other nucleophilic groups under physiological conditions and form an addition product with glutathione. Thymine- and adenine-N1-propenals inhibit DNA synthesis in HeLa cells; both compounds are cytotoxic [50% inhibiting concentration (IC50) = 1 to 2 microM]. A structurally related nucleoside, thymidine-N3-propenal, designed as a metabolic pathway inhibitor, inhibits growth of HeLa, L1210 leukemia, Lewis lung carcinoma, B16 melanoma, and DLD-1 human colon carcinoma cells in culture (IC50 = 1 to 6 microM). A single injection of this compound, administered on the first day following transplant of L1210 leukemia cells, increased the mean survival time of mice by 50% (T/C = 154). Thymidine-N3-propenal selectively blocks DNA synthesis in HeLa cells and inhibits thymidine kinase (Ki = 5.1 microM) and DNA polymerase-alpha. We suggest that base propenals, rather than damaged DNA, account for some of the cytotoxic effects of bleomycin and that nucleoside propenals represent a novel class of site-directed inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Bleomycin/pharmacology , DNA, Neoplasm , Thymidine/analogs & derivatives , Thymine/analogs & derivatives , Bleomycin/metabolism , DNA Polymerase II/antagonists & inhibitors , DNA, Neoplasm/metabolism , Glutathione/metabolism , HeLa Cells , Humans , Neoplasm Proteins/biosynthesis , Nucleic Acids/biosynthesis , Thymidine/metabolism , Thymidine/pharmacology , Thymidine Kinase/antagonists & inhibitors , Thymine/pharmacology
20.
Cancer Res ; 42(9): 3810-3, 1982 Sep.
Article in English | MEDLINE | ID: mdl-6809314

ABSTRACT

Aphidicolin inhibits DNA replication and growth of all tested human and murine neoplastic cells including leukemic T- and B-lymphocytes and melanocarcinoma cells. The concentration of aphidicolin causing 50% inhibition of DNA synthesis in all of the tested neoplastic cell lines is similar to that necessary to inhibit DNA synthesis in HeLa cells by 50%. The mechanism of inhibition of DNA synthesis in neoplastic cells is again due to the inhibition of DNA polymerase alpha by aphidicolin. Aphidicolin at a concentration 100 times higher than that causing 50% inhibition of DNA synthesis and cell growth had no effect on total protein synthesis, on the secretion of immunoglobulins, or on the expression of HLA antigens which are involved in relevant phenomena of the immune response.


Subject(s)
Cell Division/drug effects , DNA Replication/drug effects , Diterpenes/pharmacology , HLA Antigens , Immunoglobulins/biosynthesis , Animals , Aphidicolin , Cell Line , DNA Polymerase II/antagonists & inhibitors , Humans , Leukemia , Lymphocytes , Melanoma , Mice , Multiple Myeloma , Protein Biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL