Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Acta Haematol ; 126(4): 205-10, 2011.
Article in English | MEDLINE | ID: mdl-21934293

ABSTRACT

BRIT1 (BRCT-repeat inhibitor of hTERT expression), also known as microcephalin (MCPH1), is a crucial gene in the complex cellular machine that is devoted to DNA repair and acts as a regulator of both the intra-S and G2/M checkpoints. The most important role of BRIT1/MCPH1 in the regulation of cell cycle progression appears to be the G2/M checkpoint. The K562 and peripheral blood cells of chronic myeloid leukemia (CML) patients at diagnosis were found to downregulate BRIT1/MCPH1. However, we could not find any correlation between bcr/abl activity and the BRIT1/MCPH1 level. In order to study the genomic instability of CML cells, we evaluated the ability of these cells to arrest mitotic division after exposure to hydroxyurea, a known genotoxic agent. We showed that CML cells continue to proliferate without the activation of the G2/M cell cycle checkpoint arrest or of the apoptotic mechanism. This behavior may predispose the cells to accumulate genomic defects. In conclusion, we found that CML cells have a low BRIT1/MCPH1 level and show a defective G2/M arrest, confirming that these cells have a constitutive genomic instability.


Subject(s)
G2 Phase Cell Cycle Checkpoints , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Actins/antagonists & inhibitors , Adult , Aged , Cell Cycle Proteins , Cell Proliferation/drug effects , Cells, Cultured , Cytochalasin B/toxicity , Cytokinesis/drug effects , Cytoskeletal Proteins , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic , Genomic Instability , Humans , Hydroxyurea/antagonists & inhibitors , Hydroxyurea/toxicity , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Leukocytes/drug effects , Leukocytes/metabolism , Male , Middle Aged , Mutagens/toxicity , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , RNA, Messenger/metabolism
2.
Lung Cancer ; 71(1): 19-27, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20434789

ABSTRACT

The candidate tumor suppressor NORE1A is a nucleocytoplasmic shuttling protein, and although a fraction of the NORE1A in cells is localized to their centrosomes, the role of centrosomal NORE1A has not been elucidated. In this study we investigated the role of NORE1A in the numerical integrity of centrosomes and chromosome stability in lung cancer cells. Exposure of p53-deficient H1299 lung cancer cell line to hydroxyurea (HU) resulted in abnormal centrosome amplification (to 3 or more centrosomes per cell) as determined by immunofluorescence analysis with anti-γ-tubulin antibody, and forced expression of wild-type NORE1A partially suppressed the centrosome amplification. The nuclear export signal (NES) mutant (L377A/L384A) of NORE1A did not localize to centrosomes and did not suppress the centrosome amplification induced by HU. Fluorescence in situ hybridization analyses with probes specific for chromosomes 2 and 16 showed that wild-type NORE1A, but not NES-mutant NORE1A, suppressed chromosome instability in HU-exposed H1299 cells that was likely to have resulted from centrosome amplification. We next examined the status of NORE1A mRNA expression in non-small cell lung carcinoma (NSCLC) and detected down-regulation of NORE1A mRNA expression in 25 (49%) of 51 primary NSCLCs by quantitative real-time-polymerase chain reaction analysis. These results suggest that NORE1A has activity that suppresses the centrosome amplification induced by HU and that NORE1A mRNA down-regulation is one of the common gene abnormalities in NSCLCs, both of which imply a key preventive role of NORE1A against the carcinogenesis of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Centrosome/drug effects , Hydroxyurea/pharmacology , Lung Neoplasms/genetics , Monomeric GTP-Binding Proteins/genetics , Adaptor Proteins, Signal Transducing , Aged , Apoptosis Regulatory Proteins , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Centrosome/pathology , Down-Regulation/drug effects , Female , Genes, p53 , Humans , Hydroxyurea/antagonists & inhibitors , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Monomeric GTP-Binding Proteins/biosynthesis , Monomeric GTP-Binding Proteins/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transfection
3.
Teratology ; 49(4): 248-59, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8073363

ABSTRACT

Hydroxyurea (HU) is a potent mammalian teratogen. Within 2-4 hours after maternal injection, HU causes 1) a rapid episode of embryonic cell death and 2) profound inhibition of embryonic DNA synthesis. A variety of antioxidants delays the onset of embryonic cell death and reduces the incidence of birth defects. Antioxidants do not block the inhibition of DNA synthesis, indicating that early embryonic cell death is not caused by inhibited DNA synthesis. We have suggested that some HU molecules may react within the embryo to produce H2O2 and subsequent free radicals, including the very reactive hydroxyl free radical. The free radicals could cause the early cell death; antioxidants are believed to terminate the aberrant free radical reactions resulting in lessened developmental toxicity. To investigate whether hydroxyl free radicals cause the early episode of cell death, pregnant New Zealand white rabbits were injected subcutaneously on gestational day 12 with a teratogenic dose of HU (650 mg/kg) in the presence or absence of 550 mg/kg of D-mannitol (Man), a specific scavenger of hydroxyl free radicals. Osmotic control rabbits received HU plus 550 mg/kg of xylose (Xyl, a nonactive aldose). At term, the teratologic effects of HU were ameliorated by Man as evidenced by decreased incidences of the expected limb malformations. Xyl exerted no demonstrable effect on HU teratogenesis. Histological examination of limb buds at 3-8 hours after maternal injection, showed that Man delayed the onset of HU-induced cell death by as much as 4 hours. Xyl had no effect. That Man acts within the embryo was shown by performing intracoelomic injections on alternate implantation sites with Man, Xyl, or saline followed by subcutaneous injection of the pregnant doe with HU. Embryos were harvested 3-8 hours later. Limb buds from saline- and Xyl-injected embryos exhibited the typical pattern of widespread HU-induced cell death at 3-4 hours, whereas Man-injected embryos did not exhibit cell death until 5-8 hours. These results are consistent with those reported for antioxidant-mediated amelioration of HU-induced developmental toxicity and with the hypothesis that hydroxyl free radicals are the proximate reactive species in HU-induced early embryonic cell death.


Subject(s)
Free Radical Scavengers , Hydroxyurea/antagonists & inhibitors , Mannitol/pharmacology , Teratogens/toxicity , Animals , Embryo, Mammalian/pathology , Female , Hydroxyurea/toxicity , Mannitol/administration & dosage , Pregnancy , Rabbits , Uterus
4.
Braz J Med Biol Res ; 26(10): 1057-67, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8312837

ABSTRACT

1. Treatment with hydroxyurea (HU, 1 mg/g ip, 2 doses applied 7 h apart) eliminates the majority of cells undergoing mitosis (cycling cells) without affecting non-cycling cells. Oral tolerance, induced by a single gavage with 20 mg of ovalbumin, results in a drastic inhibition of anti-Ova antibody responses in young adult mice. Oral tolerance is actively maintained by the presence of specific suppressor T cells which may adoptively transfer the tolerance to naive syngeneic recipients. Under the clonal selection hypothesis, the induction of oral tolerance should be blocked by HU treatment applied soon after oral exposure to the antigen by the elimination of specific clones of lymphocytes activated by tolerogenic presentation of the antigen. 2. However, treatment with HU initiated 3, 6 or 24 h after oral exposure to ovalbumin had no effect on the induction of oral tolerance in B6D2F1 mice. However, treatment with HU 24 h before antigen exposure, totally blocked the induction of tolerance. Treatment with HU 72 h before ovalbumin had no effect. 3. In animals treated with HU 24 h before, the adoptive transfer of normal thymus, bone marrow or spleen cells partially restored the susceptibility to the induction of oral tolerance. 4. The results suggest that cycling cells, which may be totally regenerated within 72 h after treatment with HU, and are present in normal thymus, bone marrow and spleen, are crucially important for the induction of oral tolerance.


Subject(s)
Hydroxyurea/therapeutic use , Immune Tolerance/drug effects , Lymphocytes/drug effects , Ovalbumin/immunology , Administration, Oral , Animals , Antibodies/analysis , Enzyme-Linked Immunosorbent Assay , Hydroxyurea/antagonists & inhibitors , Immune Tolerance/immunology , Lymphocyte Activation/drug effects , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Ovalbumin/administration & dosage
5.
Braz. j. med. biol. res ; 26(10): 1057-67, Oct. 1993. ilus
Article in English | LILACS | ID: lil-148782

ABSTRACT

1. Treatment with hydroxyurea (HU, 1 mg/g ip, 2 doses applied 7 h apart) eliminates the majority of cells undergoing mitosis (cycling cells) without affecting non-cycling cells. Oral tolerance, induced by a single gavage with 20 mg of ovalbumin, results in a drastic inhibition of anti-Ova antibody responses in young adult mice. Oral tolerance is actively maintained by the presence of specific suppressor T cells which may adoptively transfer the tolerance to naive syngeneic recipients. Under the clonal selection hypothesis, the induction of oral tolerance should be blocked by HU treatment applied soon after oral exposure to the antigen by the elimination of specific clones of lymphocytes activated by tolerogenic presentation of the antigen. 2. However, treatment with HU initiated 3, 6 or 24 h after oral exposure to ovalbumin had no effect on the induction of oral tolerance in B6D2F1 mice. However, treatment with HU 24 h before antigen exposure, totally blocked the induction of tolerance. Treatment with HU 72 h before ovalbumin had no effect. 3. In animals treated with HU 24 h before, the adoptive transfer of normal thymus, bone marrow or spleen cells partially restored the susceptibility to the induction of oral tolerance. 4. The results suggest that cycling cells, which may be totally regenerated within 72 h after treatment with HU, and are present in normal thymus, bone marrow and spleen, are crucially important for the induction of oral tolerance


Subject(s)
Animals , Mice , Hydroxyurea/therapeutic use , Lymphocytes/drug effects , Ovalbumin/immunology , Immune Tolerance , Administration, Oral , Antibodies/analysis , Enzyme-Linked Immunosorbent Assay , Hydroxyurea/antagonists & inhibitors , Lymphocytes/immunology , Mice, Inbred DBA , Ovalbumin/administration & dosage , Immune Tolerance/immunology , Lymphocyte Activation
6.
Mutat Res ; 293(3): 215-23, 1993 Mar.
Article in English | MEDLINE | ID: mdl-7679471

ABSTRACT

Hydroxyurea, an inhibitor of ribonucleotide reductase, blocks the repair by normal human fibroblasts of ultraviolet light-damaged adenovirus 5. A mixture of all four DNA deoxynucleosides present with hydroxyurea reversed the inhibition, an effect consistent with the target of hydroxyurea being ribonucleotide reductase. Single deoxynucleosides were differentially effective in reversing the inhibition: deoxyguanosine reversed 100% of the HU block, deoxyadenosine 70%, deoxythymidine 55%, but deoxycytidine only 3%. The results are interpreted in the context of the known pattern of stimulation and inhibition of mammalian ribonucleotide reductase by dNTP effectors. We suggest that biological data may be important to assessing the availability of substrate for the ribonucleotide reductase molecules that provide dNTPs for DNA repair.


Subject(s)
Adenoviridae/genetics , DNA Repair/drug effects , Deoxyguanosine/pharmacology , Hydroxyurea/pharmacology , Adenoviridae/drug effects , Adenoviridae/growth & development , Cell Line , DNA, Viral/drug effects , Fibroblasts , Humans , Hydroxyurea/antagonists & inhibitors
7.
J Virol ; 65(5): 2290-8, 1991 May.
Article in English | MEDLINE | ID: mdl-2016760

ABSTRACT

Hydroxyurea, an inhibitor of ribonucleotide reductase, blocks replication of vaccinia virus. However, when medium containing hydroxyurea and dialyzed serum was supplemented with deoxyadenosine, the block to viral reproduction was circumvented, provided that an inhibitor of adenosine deaminase was also present. Deoxyguanosine, deoxycytidine, and deoxythymidine were ineffective alone and did not augment the deoxyadenosine effect. In fact, increasing concentrations of deoxyguanosine and deoxythymidine, but not deoxycytidine, eliminated the deoxyadenosine rescue, an effect that was reversed by the addition of low concentrations of deoxycytidine. These results suggested that the inhibition of viral replication by hydroxyurea was primarily due to a deficiency of dATP. Deoxyribonucleoside triphosphate pools in vaccinia virus-infected cells were measured at the height of viral DNA synthesis after a synchronous infection. With 0.5 mM hydroxyurea, the dATP pool was greater than 90% depleted, the dCTP and dGTP pools were 40 to 50% reduced, and the dTTP pool was increased. Assay of ribonucleotide reductase activity in intact virus-infected cells suggested that hydroxyurea may differentially affect reduction of the various substrates of the enzyme.


Subject(s)
Deoxyadenosines/pharmacology , Hydroxyurea/antagonists & inhibitors , Vaccinia virus/growth & development , Adenine/analogs & derivatives , Adenine/pharmacology , Adenosine Deaminase Inhibitors , Adenosine Triphosphate/metabolism , Antiviral Agents , Cells, Cultured , Deoxycytidine/pharmacology , Deoxyguanosine/pharmacology , Ribonucleotide Reductases/antagonists & inhibitors , Thymidine/pharmacology , Vaccinia virus/drug effects , Viral Plaque Assay , Virus Replication/drug effects
8.
Eur J Pharmacol ; 193(2): 173-7, 1991 Feb 07.
Article in English | MEDLINE | ID: mdl-1710991

ABSTRACT

The effects of 5-azacytidine and hydroxyurea on their independent ability to change adult hemoglobin proportions toward newborn proportions in adult rats were examined. The results revealed that both the chemotherapeutic agents were capable of switching certain hemoglobin components toward newborn values and required similar time-span to express their actions. However, the switching effect of these drugs was totally lost if aspirin was simultaneously administered into the rats, reflecting the need for concurrent prostaglandin synthesis.


Subject(s)
Aspirin/pharmacology , Azacitidine/antagonists & inhibitors , Fetal Hemoglobin/drug effects , Hydroxyurea/antagonists & inhibitors , Age Factors , Animals , Azacitidine/pharmacology , Fetal Hemoglobin/chemistry , Hydroxyurea/pharmacology , Male , Rats , Rats, Inbred Strains , Reference Values
9.
Reprod Toxicol ; 4(2): 145-52, 1990.
Article in English | MEDLINE | ID: mdl-2136029

ABSTRACT

Hydroxyurea (HU) is a swiftly acting cytotoxic teratogen and an inhibitor of DNA synthesis. Within 2 h of maternal treatment, HU causes necrosis in proliferating tissues of rabbit embryos on gestational day 12. Co-administration of the antioxidant propyl gallate (PG) delays the onset of necrosis until 6 h and ameliorates the teratogenic effects seen at term. Since HU also causes a rapid, profound decrease in uterine blood flow in pregnant rabbits, it is necessary to determine whether HU and PG interact within the pregnant female or within the embryo. In order to establish that the site of HU-PG interaction is embryonic, HU, PG, HU-PG, or vehicle was injected directly into implantation sites. When embryos were examined microscopically at 4 h, necrosis was observed only in the HU-treated embryos, indicating that the palliative interaction between HU and PG takes place within the embryo. To resolve whether the alleviation of HU-induced embryotoxicity was due to decreased HU levels within HU-PG embryos, HU concentrations were measured in embryos from HU-and HU-PG-treated maternal rabbits at 15 min to 8 h post injection. The HU levels of the two groups differed significantly only at 4 h. The rates of uptake during the linear phase (times from 15 min to 3 h) did not differ. When HU concentration was plotted versus time, measurements of the areas under the curve also did not differ. To determine whether PG alters the HU-induced inhibition of DNA synthesis, 3H-thymidine incorporation into embryonic DNA was assayed at 2 h after HU, HU-PG, or vehicle injections.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Abnormalities, Drug-Induced/prevention & control , Embryo, Mammalian/drug effects , Hydroxyurea/antagonists & inhibitors , Propyl Gallate/pharmacology , Animals , DNA/metabolism , Embryo, Mammalian/metabolism , Female , Hydroxyurea/pharmacokinetics , Hydroxyurea/toxicity , Injections , Necrosis/pathology , Pregnancy , Rabbits , Thymidine/metabolism , Uterus
10.
Reprod Toxicol ; 4(4): 267-75, 1990.
Article in English | MEDLINE | ID: mdl-2136047

ABSTRACT

Hydroxyurea (HU) is a potent teratogen that causes a characteristic, rapidly occurring episode of embryonic cell death 2 to 4 h after subcutaneously injecting 650 mg/kg of HU into pregnant New Zealand White rabbits on gestational day 12. Previous studies documented the ability of the phenolic antioxidant, propyl gallate, to delay the onset of embryonic cell death and to decrease the number and severity of defects seen at term. The present study investigated the ability of the structurally different antioxidants, ethoxyquin (ETX) and nordihydroguaiaretic acid (NDGA), to also ameliorate HU developmental toxicity. Injection of pregnant rabbits with either ETX or NDGA at 950 mg/kg, 15 to 30 min prior to HU injection, resulted in reduced developmental toxicity seen at term. The reduction was manifested by fewer malformed fetuses with increased body weights compared with fetuses from HU-only treated litters, greatly reduced incidences of specific malformations, and diminished severity of some HU-induced defects. Microscopic analysis of HU, HU-ETX, and HU-NDGA embryos was performed at 4 or 8 h after treatment. HU caused cell death in the limb bud mesenchyme, clearly evident at 4 h. In contrast, HU-NDGA embryos exhibited no signs of cell death until 8 h after treatment. Although most HU-ETX embryos exhibited little or no cell death at 4 h after treatment, in about 20% the level of cell death was indistinguishable from that in HU-treated embryos. The amelioration of HU developmental toxicity in the present study is consistent with the results of previous studies utilizing propyl gallate. The results suggest that the antioxidant properties of these substances interfere with the rapidly occurring toxic effects of HU and that this may account for amelioration of HU developmental toxicity.


Subject(s)
Abnormalities, Drug-Induced/prevention & control , Antioxidants/pharmacology , Ethoxyquin/pharmacology , Hydroxyurea/antagonists & inhibitors , Masoprocol/pharmacology , Abnormalities, Drug-Induced/pathology , Animals , Body Weight/drug effects , Embryo, Mammalian/drug effects , Female , Hydroxyurea/toxicity , Pregnancy , Rabbits
13.
Biochem Pharmacol ; 36(18): 2985-91, 1987 Sep 15.
Article in English | MEDLINE | ID: mdl-3498491

ABSTRACT

Inhibition of cell growth and DNA synthesis by hydroxyurea is thought to occur via an effect on the enzyme ribonucleotide reductase leading to a block of deoxyribonucleotide synthesis. Earlier attempts to bypass such a block by delivering deoxyribonucleosides to the medium of cultured cells have given equivocal results. Complications arise in such experiments from the specificity of the phosphorylating enzymes since 3 of the 4 deoxyribonucleosides are substrates for the same enzyme, with widely differing Km values, and from allosteric effects exerted by deoxyribonucleotides. We simplify this situation by using a mutant hamster V79 line that lacks the enzyme dCMP deaminase. The cells contain a 20-fold enlarged dCTP pool and require thymidine for optimal growth. Concentrations of hydroxyurea (50 or 100 microM) that in short-term experiments inhibited DNA synthesis depleted the dATP pool without seriously affecting pyrimidine deoxyribonucleotide pools. The dATP pool could be restored by addition of deoxyadenosine but this depleted the dGTP pool. This depletion could be counteracted by the simultaneous addition of deoxyguanosine but then critically depended on the relative concentrations of the two purine deoxyribonucleosides, with optimal results at 1 microM deoxyadenosine + 100 microM deoxyguanosine. Under those conditions the inhibition of DNA synthesis by hydroxyurea was partially reversed.


Subject(s)
DNA/biosynthesis , Hydroxyurea/antagonists & inhibitors , Nucleotides/metabolism , Purine Nucleosides/pharmacology , Animals , Cell Line , Cricetinae , Deoxyadenine Nucleotides/metabolism , Deoxyadenosines/pharmacology , Deoxycytosine Nucleotides/metabolism , Deoxyguanine Nucleotides/metabolism , Deoxyguanosine/pharmacology , Thymine Nucleotides/metabolism
14.
Teratology ; 30(1): 83-90, 1984 Aug.
Article in English | MEDLINE | ID: mdl-6484855

ABSTRACT

On day 10 of pregnancy NMRI mice received an intraperitoneal injection of either 500 mg/kg hydroxyurea (HU) or 500 mg/kg HU plus 500, 700, 800, or 900 mg/kg deoxycytidine monophosphate (dCMP). Three hours after application of the substances, the animals received 5 microCi/g 3H-thymidine i.p. One hour later the animals were sacrificed and the embryos were removed. Autoradiographs of the embryos served to determine the percentage of necroses, labelled necroses, mitoses, labelled mitoses, and labelled neuroepithelial cells of the spinal cord. Optimal inhibition of HU effects was achieved by application of 700 mg/kg dCMP. The cytotoxic effects of HU on neuroepithelial cells could be partially but not completely inhibited by simultaneous application of dCMP. The duration of HU-induced DNA synthesis inhibition was shortened after simultaneous dCMP application. A strong increase of the labelling index and shortening of the G2-phase duration of neuroepithelial cells after simultaneous application of HU and dCMP was striking as compared to the results obtained from untreated or HU-treated animals.


Subject(s)
Abnormalities, Drug-Induced , Deoxycytidine Monophosphate/pharmacology , Deoxycytosine Nucleotides/pharmacology , Hydroxyurea/toxicity , Spinal Cord/abnormalities , Animals , Drug Interactions , Female , Hydroxyurea/antagonists & inhibitors , Mice , Mitotic Index , Necrosis , Pregnancy , Spinal Cord/embryology , Spinal Cord/pathology
15.
Teratology ; 24(1): 19-35, 1981 Aug.
Article in English | MEDLINE | ID: mdl-7302871

ABSTRACT

Hydroxyurea (HU) is a potent teratogen which caused 100% embryotoxic effects in New Zealand white rabbits when injected sc on gestational day 12. These included a high percentage of resorptions and severe craniofacial, trunk and limb deformities of all survivors. Co-treatment of pregnant animals with the phenolic antioxidant propyl gallate (PG) resulted in amelioration of the embryotoxicity. Various amounts of PG (362-906 mg/kg) and HU (600-750 mg/kg) were eigher injected simultaneously or mixed together for periods of time up to 45 min. The extent of amelioration was dependent upon the amount of PG, although the highest dose of PG caused maternal toxicity. Simultaneous injections of HU and PG were not as efficacious as mixture of the two chemicals prior to injection. The length of time which the HU and PG mixture was allowed to stand prior to injection had no effect on the extent of amelioration. The protective action of PG resulted in significant linear reductions in both resorptions and specific malformations with increasing doses of PG. Histologic analysis of HU and HU-PG embryos disclosed that HU produced rapid cell death in the mesenchymal compartment of the embryo, particularly in the limb-buds, beginning at 2 h after treatment. Cell debris increased in amount until 8 h and remained extensive at 16 h. In contrast, HU-PG delayed the onset for cell death until 8 h. Nevertheless, at 16 h, the amount of cell debris in the limb-buds was appreciable. Thin layer chromatography (TLC) of HU-PG solution revealed no breakdown products or intermediate compounds suggesting that HU and PG do not react chemically. The presence of HU within both HU and HU-PG treated embryos was confirmed by TLC of embryonic sonicants. In contrast, TLC of embryonic extracts revealed PG only in HU-PG embryos. Light microscopy of other embryos from those same litters demonstrated extensive cell debris in the HU embryos but not in HU-PG embryos. It is suggested that the delay in onset of HU cytotoxicity is caused by the antioxidant properties of PG acting within the embryos, and that this may account for the amelioration of HU teratogenesis by PG.


Subject(s)
Gallic Acid/analogs & derivatives , Hydroxyurea/antagonists & inhibitors , Propyl Gallate/pharmacology , Teratogens/antagonists & inhibitors , Abnormalities, Drug-Induced/pathology , Animals , Bone and Bones/abnormalities , Bone and Bones/embryology , Female , Gestational Age , Hydroxyurea/pharmacology , Pregnancy , Rabbits , Time Factors
16.
Mutat Res ; 80(1): 121-31, 1981 Jan.
Article in English | MEDLINE | ID: mdl-6451801

ABSTRACT

Cycloheximide (CH) and puromycin (PM)strongly antagonize induction of sister-chromatid exchanges (SCEs) by exogenous agents regardless of the mechanism for initiating damage. 5-Bromodeoxyuridine (BUdR) substitution was used to monitor SCEs, but the background level of BUdR-induced SCEs was unaffected by the presence of protein inhibitors. Antagonism between DNA-damaging agents and protein inhibitors was strongest in euchromatic regions. Possible relationships between SCE formation and the mechanism of antagonism by protein inhibitors are discussed.


Subject(s)
Chromosome Aberrations , Crossing Over, Genetic/drug effects , Cycloheximide/pharmacology , Puromycin/pharmacology , Sister Chromatid Exchange/drug effects , Animals , Bromodeoxyuridine/antagonists & inhibitors , Cell Line , Cricetinae , Cricetulus , Ethyl Methanesulfonate/antagonists & inhibitors , Floxuridine/antagonists & inhibitors , Hydroxyurea/antagonists & inhibitors , Methyl Methanesulfonate/antagonists & inhibitors
18.
Cell Tissue Kinet ; 13(4): 415-24, 1980 Jul.
Article in English | MEDLINE | ID: mdl-7428015

ABSTRACT

Low dose (80 micrograms/kg) Actinomycin D (AD) produced a significant but transient inhibition of proliferation of the haemopoietic stem cells (CFUs) in chimaeras or in mice regenerating after sublethal irradiation. The same dose of AD had no effect on the resting CFUs population. During the period of proliferation inhibition, CFUs proved to be insensitive to the killing effect of [3H]thymidine in vitro and hydroxyurea (HU) in vivo. In Ehrlich ascites tumour (EAT) bearing mice enhanced CFUs turnover rate was found. Eighty micrograms/kg AD produced a selective effect in these mice: it protected the proliferating CFUs population without diminishing the effect of hydroxyurea on the tumour cells.


Subject(s)
Cell Division/drug effects , Dactinomycin/pharmacology , Hematopoietic Stem Cells/cytology , Hydroxyurea/antagonists & inhibitors , Animals , Autoradiography , Carcinoma, Ehrlich Tumor/physiopathology , Colony-Forming Units Assay , Dose-Response Relationship, Drug , Female , Hydroxyurea/pharmacology , Mice , Neoplasm Transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...