Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
BMC Pharmacol Toxicol ; 18(1): 53, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28662728

ABSTRACT

BACKGROUND: In order to obtain better clinical results in anticancer therapies, polychemotherapy or combination therapies are used. For this, the combinations are required to increase the efficacy and reduce the adverse reactions of the associated chemotherapies. The aim of this study was to evaluate the cytotoxic, apoptotic and (anti)proliferative potential of two sesquiterpene lactones isolated from Moquiniastrum polymorphum, 11,13-diidrozaluzanin C (1) and gochnatiolide C (2), and their associations with chemotherapeutic agents irinotecan, tamoxifen, cisplatin, 5-fluouracyl and doxorubicin in the tumoral lineage of MCF-7 breast adenocarcinoma. METHODS: The analyses were performed by MTT cytotoxicity assays, drug combination index (CI), apoptosis morphological assay and cell proliferation assay. Treatments were evaluated with short exposure times (4 h), followed or not by recovery in drug-free medium for 24 h. For the cell viability assay the statistical analysis was performed using software INSTAT, and the ANOVA/Tukey test was applied. Combination Indices (CI) was made using CompuSyn software and demonstrated through isoboles. The assays that evaluated cell death and proliferation used statistical analysis SAS 9.4 (Statistical Analysis System), and the procedure adopted was PROC NPAR1WAY. The Wilcoxon test at 5% level was applied for comparing statistical differences. RESULTS: The results demonstrated that the compounds decrease cell viability and increase their action when associated with irinotecan, tamoxifen and doxorubicin (CI < 1 and CI = 1). In periods of 4 h-exposure, the compounds cause cell death by apoptosis and after 24 h, they increase the mean number of cells in programmed cell death, especially when treated with 2. In addition, the association with doxorubicin increases the apoptotic potential induced by tested compounds. Both isolates had effect on the reduction of the number of mitoses, especially when 2 at its highest concentration is associated with doxorubicin. CONCLUSIONS: Finally, these compounds are presented as potential agents in chemotherapy combined with doxorubicin, since they trigger the mechanism of apoptosis, which, through the mechanism of action of sesquiterpene lactones, leads to a reduction in toxicity. In addition, the tested compounds have the ability to exert a synergistic action with doxorubicin, possibly by down-regulating the drug resistance mechanisms.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Lactones/pharmacology , Sesquiterpenes/pharmacology , Apoptosis/drug effects , Asteraceae , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Humans , MCF-7 Cells
2.
Genet Mol Biol ; 39(2): 279-89, 2016.
Article in English | MEDLINE | ID: mdl-27303909

ABSTRACT

Cardanol is an effective antioxidant and is a compound with antimutagenic and antitumoral activity. Here, we evaluated the genotoxic and mutagenic potential of saturated side chain cardanol and its effects in combination with cyclophosphamide in preventing DNA damage, apoptosis, and immunomodulation. Swiss mice were treated with cardanol (2.5, 5 and 10 mg/kg) alone or in combination with cyclophosphamide (100 mg/kg). The results showed that cardanol is an effective chemopreventive compound, with damage reduction percentages that ranged from 18.9 to 31.76% in the comet assay and from 45 to 97% in the micronucleus assay. Moreover, cardanol has the ability to reduce the frequency of apoptosis induced by cyclophosphamide. The compound did not show immunomodulatory activity. A final interpretation of the data showed that, despite its chemoprotective capacity, cardanol has a tendency to induce DNA damage. Hence, caution is needed if this compound is used as a chemopreventive agent. Also, this compound is likely not suitable as an adjuvant in chemotherapy treatments that use cyclophosphamide.

3.
Environ Toxicol Pharmacol ; 40(3): 715-21, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26410090

ABSTRACT

Combretastatin A-4 exhibits efficient anti-cancer potential in human tumors, including multidrug-resistant tumors. We evaluated the mutagenic, apoptotic and immunomodulatory potential of two diaryl sulfide analogs of combretastatin A-4, 1,2,3-trimethoxy-5-([4-methoxy-3-nitrophenyl]thio)benzene (analog 1) and 1,2,3-trimethoxy-5-([3-amino-4-methoxyphenyl]thio)benzene (analog 2), as well as their association with the anti-tumor agent cyclophosphamide, in Swiss mice. Such evaluation was achieved using the comet assay, peripheral blood micronucleus test, splenic phagocytosis assay, and apoptosis assay. Both analogs were found to be genotoxic, mutagenic and to induce apoptosis. They also increased splenic phagocytosis, although this increase was more pronounced for analog 2. When combined with cyclophosphamide, analog 1 enhanced the mutagenic and apoptotic effects of this anti-tumor agent. In contrast, analog 2 did not enhance the effects of cyclophosphamide and prevented apoptosis at lower doses. These data suggest that analog 1 could be an adjuvant chemotherapeutic agent and possibly improve the anti-neoplastic effect of cyclophosphamide. Additionally, this compound could be a candidate chemotherapeutic agent and/or an adjuvant for use in combined anti-cancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , DNA/drug effects , Sulfides/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Cell Survival/drug effects , Cyclophosphamide/pharmacology , Drug Synergism , Humans , Male , Mice , Micronucleus Tests , Phagocytosis/drug effects , Stilbenes/chemistry , Sulfides/pharmacology
4.
DNA Repair (Amst) ; 28: 107-15, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25758781

ABSTRACT

Loss of function of DNA repair (DNAR) genes is associated with genomic instability and cancer predisposition; it also makes cancer cells reliant on a reduced set of DNAR pathways to resist DNA-targeted therapy, which remains the core of the anticancer armamentarium. Because the landscape of DNAR defects across numerous types of cancers and its relation with drug activity have not been systematically examined, we took advantage of the unique drug and genomic databases of the US National Cancer Institute cancer cell lines (the NCI-60) to characterize 260 DNAR genes with respect to deleterious mutations and expression down-regulation; 169 genes exhibited a total of 549 function-affecting alterations, with 39 of them scoring as putative knockouts across 31 cell lines. Those mutations were compared to tumor samples from 12 studies of The Cancer Genome Atlas (TCGA) and The Cancer Cell Line Encyclopedia (CCLE). Based on this compendium of alterations, we determined which DNAR genomic alterations predicted drug response for 20,195 compounds present in the NCI-60 drug database. Among 242 DNA damaging agents, 202 showed associations with at least one DNAR genomic signature. In addition to SLFN11, the Fanconi anemia-scaffolding gene SLX4 (FANCP/BTBD12) stood out among the genes most significantly related with DNA synthesis and topoisomerase inhibitors. Depletion and complementation experiments validated the causal relationship between SLX4 defects and sensitivity to raltitrexed and cytarabine in addition to camptothecin. Therefore, we propose new rational uses for existing anticancer drugs based on a comprehensive analysis of DNAR genomic parameters.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Repair/genetics , Mutation , Cell Line, Tumor , Down-Regulation , Genes , Humans , National Cancer Institute (U.S.) , United States
5.
DNA Repair (Amst) ; 13: 1-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24355542

ABSTRACT

Tyrosyl-DNA-phosphodiesterase 1 (TDP1) repairs 3'-blocking DNA lesions by catalytically hydrolyzing the tyrosyl-DNA-phosphodiester bond of trapped topoisomerase I (Top1) cleavage complexes (Top1cc). It also removes 3'-blocking residues derived from oxidative damage or incorporation of chain terminating anticancer and antiviral nucleosides. Thus, TDP1 is regarded as a determinant of resistance to Top1 inhibitors and chain terminating nucleosides, and possibly of genomic stability. In the 60 cell lines of the NCI Developmental Therapeutic Anticancer Screen (the NCI-60), whose whole genome transcriptome and mutations have recently been characterized, we discovered two human lung cancer cell lines deficient for TDP1 (NCI_H522 and HOP_62). HOP_62 shows undetectable TDP1 mRNA and NCI_H522 bears a homozygous deleterious mutation of TDP1 at a highly conserved amino acid residue (K292E). Absence of TDP1 protein and lack of TDP1 catalytic activity were demonstrated in cell lysates from both cell lines. Lack of TDP1 expression in HOP_62 was shown to be due to TDP1 promoter hypermethylation. Our study provides insights into the possible inactivation of TDP1 in cancers and its relationship to cellular response to Top1-targeted drugs. It also reveals two TDP1 knockout lung cancer cell lines for further TDP1 functional analyses.


Subject(s)
DNA Topoisomerases, Type I/metabolism , Genetic Variation , Lung Neoplasms/genetics , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Amino Acid Sequence , Cell Line, Tumor , Conserved Sequence , DNA Methylation , DNA Topoisomerases, Type I/genetics , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , Humans , Mutation , National Cancer Institute (U.S.) , Promoter Regions, Genetic , United States
6.
Cancer Chemother Pharmacol ; 70(4): 491-502, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22851206

ABSTRACT

The development of new strategies for cancer therapeutics is indispensable for the improvement of standard protocols and the creation of other possibilities in cancer treatment. Yeast models have been employed to study numerous molecular aspects directly related to cancer development, as well as to determine the genetic contexts associated with anticancer drug sensitivity or resistance. The budding yeast Saccharomyces cerevisiae presents conserved cellular processes with high homology to humans, and it is a rapid, inexpensive and efficient compound screening tool. However, yeast models are still underused in cancer research and for screening of antineoplastic agents. Here, the employment of S. cerevisiae as a model system to anticancer research is discussed and exemplified. Focusing on the important determinants in genomic maintenance and cancer development, including DNA repair, cell cycle control and epigenetics, this review proposes the use of mutant yeast panels to mimic cancer phenotypes, screen and study tumor features and synthetic lethal interactions. Finally, the benefits and limitations of the yeast model are highlighted, as well as the strategies to overcome S. cerevisiae model limitations.


Subject(s)
Antineoplastic Agents/pharmacology , Saccharomyces cerevisiae/drug effects , Cell Cycle/drug effects , DNA Repair/drug effects , Drug Resistance , Epigenesis, Genetic , Humans , Mutation , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics
7.
Carcinogenesis ; 33(8): 1433-40, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22431722

ABSTRACT

Adenosine diphosphate (ADP)-ribosylation is an important posttranslational modification catalyzed by a variety of enzymes, including poly (ADP ribose) polymerases (PARPs), which use nicotinamide adenine dinucleotide (NAD(+)) as a substrate to synthesize and transfer ADP-ribose units to acceptor proteins. The PARP family members possess a variety of structural domains, span a wide range of functions and localize to various cellular compartments. Among the molecular actions attributed to PARPs, their role in the DNA damage response (DDR) has been widely documented. In particular, PARPs 1-3 are involved in several cellular processes that respond to DNA lesions, which include DNA damage recognition, signaling and repair as well as local transcriptional blockage, chromatin remodeling and cell death induction. However, how these enzymes are able to participate in such numerous and diverse mechanisms in response to DNA damage is not fully understood. Herein, the DDR functions of PARPs 1-3 and the emerging roles of poly (ADP ribose) polymers in DNA damage are reviewed. The development of PARP inhibitors, their applications and mechanisms of action are also discussed in the context of the DDR.


Subject(s)
DNA Damage , Poly(ADP-ribose) Polymerases/metabolism , Animals , Humans , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/chemistry , Protein Conformation
8.
Biochem J ; 437(1): 63-73, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21470188

ABSTRACT

Numerous anticancer agents and environmental mutagens target DNA. Although all such compounds interfere with the progression of the replication fork and inhibit DNA synthesis, there are marked differences in the DNA-damage response pathways they trigger, and the relative impact of the proximal or the distal signal transducers on cell survival is mainly lesion-specific. Accordingly, checkpoint kinase inhibitors in current clinical development show synergistic activity with some DNA-targeting agents, but not with others. In the present study, we characterize the DNA-damage response to the antitumour acronycine derivative S23906, which forms monofunctional adducts with guanine residues in the minor groove of DNA. S23906 exposure is accompanied by specific recruitment of RPA (replication protein A) at replication sites and rapid Chk1 activation. In contrast, neither MRN (Mre11-Rad50-Nbs1) nor ATM (ataxia-telangiectasia mutated), contributes to the initial response to S23906. Interestingly, genetic attenuation of ATR (ATM- and Ras3-related) activity inhibits not only the early phosphorylation of histone H2AX and Chk1, but also interferes with the late phosphorylation of Chk2. Moreover, loss of ATR function or pharmacological inhibition of the checkpoint kinases by AZD7762 is accompanied by abrogation of the S-phase arrest and increased sensitivity towards S23906. These findings identify ATR as a central co-ordinator of the DNA-damage response to S23906, and provide a mechanistic rationale for combinations of S23906 and similar agents with checkpoint abrogators.


Subject(s)
Acronine/analogs & derivatives , Antineoplastic Agents, Alkylating/pharmacology , Cell Cycle Proteins/physiology , DNA Damage , Mutation , Protein Serine-Threonine Kinases/physiology , Acid Anhydride Hydrolases , Acronine/pharmacology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Checkpoint Kinase 1 , Checkpoint Kinase 2 , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , HeLa Cells , Histones/metabolism , Humans , MRE11 Homologue Protein , Microscopy, Fluorescence , Nuclear Proteins/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Replication Protein A/metabolism , Thiophenes/pharmacology , Urea/analogs & derivatives , Urea/pharmacology
9.
Biochem Pharmacol ; 79(2): 147-53, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19712668

ABSTRACT

5-Fluorouracil (5-FU) is an antitumor antimetabolite that can be converted into fluoronucleotides and FdUMP. Fluoronucleotides are incorporated into DNA and RNA, while FdUMP results in nucleotide pool imbalance. Saccharomyces cerevisiae is unable to convert 5-FU into FdUMP, making yeast a unique model system to study the cellular effects of 5-FU and FdUMP independently. A panel of repair-deficient yeast strains was used to identify the DNA repair pathways needed for repair of lesions generated by 5-FU or FdUMP. This included yeast deficient in base excision repair (BER), nucleotide excision repair (NER), translesion synthesis (TLS), mismatch repair (MMR), post-replication repair (PRR), homologous recombination (HR) and non-homologous end-joining (NHEJ). The results revealed an important role of BER, since BER-mutants (ntg1, ntg2, apn1, apn2) showed pronounced sensitivity to both 5-FU and FdUMP. MMR mutants also showed high sensitivity to both compounds. In contrast, deficiencies in NER, NHEJ and TLS repair had only minor influence on the sensitivity to FU and FdUMP. Interestingly, deficiencies in HR (rad52) and PPR (rad6, rad18) were associated with increased sensitivity to 5-FU, but not to FdUMP. Taken together, our study reveals an important contribution of DNA repair pathways on the sensitivity to 5-FU and its active metabolite FdUMP. Importantly, the repair mechanisms differed for the 2 antimetabolites since lesions induced by 5-FU were repaired by BER, MMR, HR and PRR, while only BER and MMR were required for repair of FdUMP-induced lesions.


Subject(s)
Antimetabolites, Antineoplastic/adverse effects , DNA Repair , Fluorouracil/adverse effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development
10.
J Appl Toxicol ; 29(4): 308-16, 2009 May.
Article in English | MEDLINE | ID: mdl-19115314

ABSTRACT

5-Fluorouracil (5-FU) is an antineoplasic drug widely used to treat cancer. Its cytotoxic effect has been principally ascribed to the misincorporation of fluoronucleotides into DNA and RNA during their synthesis, and the inhibition of thymidylate synthase (TS) by FdUMP (one of the 5-FU active metabolites), which leads to nucleotide pool imbalance. In the present study, we compared the ability of 5-FU and FdUMP to induce apoptosis and to influence the cell cycle progression in human colon SW620 adenocarcinoma cells in regards to their genotoxic and clastogenic activities. Our study demonstrates that 5-FU induces SSB, DSB and apoptosis earlier than FdUMP. Interestingly, while both drugs are able to induce apoptosis, their effect on the cell cycle progression differed. Indeed, 5-FU induces an arrest in G1/S while FdUMP causes an arrest in G2/M. Independently of the temporal difference in strand breaks and apoptosis induction, as well as the differential cell cycle modulation, both drugs presented similar clastogenic effects. The different pattern of cell cycle arrest suggests that the two drugs induce different types of primary DNA lesions that could lead to the activation of different checkpoints and recruit different DNA repair pathways.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , DNA, Neoplasm/drug effects , Fluorodeoxyuridylate/toxicity , Fluorouracil/toxicity , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacokinetics , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Comet Assay , DNA Damage , Fluorodeoxyuridylate/chemistry , Fluorodeoxyuridylate/pharmacokinetics , Fluorouracil/chemistry , Fluorouracil/pharmacokinetics , Histones/genetics , Humans , Micronucleus Tests , Mutagens/toxicity
11.
Environ Toxicol Pharmacol ; 28(1): 120-4, 2009 Jul.
Article in English | MEDLINE | ID: mdl-21783991

ABSTRACT

This research has evaluated the effects of enteral supplementation of glutamine in clastogens and genotoxic damages caused by the acute administration of cisplatin. For this, it was utilized Swiss mice distributed in eight experimental groups: control, cisplatin, glutamine, in three different doses and the combination of these with cisplatin. The results show that the glutamine was present in neither genotoxic nor mutagenic activity. When in association with glutamine and cisplatin, in simultaneous treatment, it was verified the frequency decreased of micronuclei and comets. The damage reduction percentages to the micronucleus ranged from 95.4 to 91.8% after 24h of administration of these compounds and 76.7 to 56.8% after 48h. In the same time the damage reduction percentages to the comet test ranged from 117.0 to 115.0%. The results suggest that glutamine is capable of preventing genotoxic and mutagenic damage according to the experimental design proposed.

12.
Toxicol In Vitro ; 21(1): 41-52, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17055696

ABSTRACT

A large number of functional foods, including those that contain beta-glucan, have been shown to prevent the development of cancer and other chronic diseases. The aim of the present study was to elucidate its mechanism of action, as well as to understand its effects as an antigenotoxic, anticlastogenic agent, and to determine its capacity to preserve cell viability. The investigation was carried out in the CHO-k1 and CHO-xrs5 cell lines. The cytokinesis-blocked micronucleus assay indicated that the different doses of beta-glucan examined (5, 10, 20 and 40 microg/ml) did not show clastogenic effects. In the CHO-k1 cell line, a chemopreventive effect could be observed in all the protocols tested: pre-treatment (% reduction of 35.0-57.3), simultaneous treatment (simple--5 reduction of 19.7-55.6 and with pre-incubation--of 42.7-56.4) and post-treatment (% reduction of 17.9-37.6). This finding indicates mechanisms of action involving desmutagenesis and bioantimutagenesis, albeit the latter having a lesser role. However, in the repair-deficient CHO-xrs5 cells, beta-glucan did not show a protective effect with post-treatment (% reduction of 2.96), thus supporting the involvement of bioantimutagenesis. The comet assay in CHO-k1 cells demonstrated that beta-glucan has neither a genotoxic nor an antigenotoxic effect. Cell viability tests indicated that beta-glucan preserves cell viability in both cell lines, preventing apoptotic events. These findings suggest that beta-glucan, when present in foods, could provide them with nutraceutical characteristics and act as a dietary supplement, or that beta-glucan could be used in new drug development.


Subject(s)
DNA Damage/drug effects , DNA Repair/genetics , Saccharomyces cerevisiae/chemistry , beta-Glucans/pharmacology , Acridine Orange , Animals , Antigens/toxicity , Antimutagenic Agents , Apoptosis/drug effects , CHO Cells , Cell Survival/drug effects , Comet Assay , Cricetinae , Cricetulus , Ethidium , Fluorescent Dyes , Micronucleus Tests , Necrosis , beta-Glucans/isolation & purification
13.
Toxicol In Vitro ; 20(7): 1225-33, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16716562

ABSTRACT

Due to the need to identify new antimutagenic agents and to determine their mechanism of action, the present study examined the mechanism of action of the beta-glucan with regard to antimutagenicity using the micronucleus assay in CHO-k1 and HTC cell lines. The mutagenicity experiments were performed with three different concentrations of beta-glucan (5, 10, and 20 microg/mL), in wich only the highest dose showed mutagenic activity. In the antimutagenicity experiments, the same concentrations of beta-glucan were combined with a mutagenic agent, methylmethane sulfonate, or 2-aminoanthracene, using four different treatment protocols: pre-treatment, simultaneous treatment (simple and with pre-incubation), and post-treatment. The results indicate that the CHO-k1 cell line treated with MMS presented a chemopreventive activity for all the doses of beta-glucan in the different treatment protocols, except for the lowest dose in post-treatment. When HTC cell line treated with MMS is analysed, a chemopreventive activity can be verified for the highest dose in both pre- and post-treatment. For the simple simultaneous treatment, the three doses demonstrated efficacy, while for the simultaneous treatment with pre-incubation only the intermediate concentration was effective. In HTC treated with 2AA both the lowest dose in the pre-treatment protocol and the post-treatment protocol did not show efficacy in preventing DNA damage. The evaluation of the different protocols and the damage decrease percentages observed suggest that beta-glucan has both desmutagenic and bioantimutagenic activity. It is necessary, however, to note that efficacy and mechanism of action are subject to variation when compared the two cell lines, since in HTC, representing a drug-metabolizing system, this substance can show a diminished chemopreventive capacity.


Subject(s)
Antimutagenic Agents/pharmacology , Hordeum/chemistry , Micronucleus Tests/methods , beta-Glucans/pharmacology , Animals , Anthracenes/toxicity , Antimutagenic Agents/chemistry , Antimutagenic Agents/isolation & purification , CHO Cells , Cell Line , Cricetinae , Cricetulus , DNA Damage/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Methyl Methanesulfonate/toxicity , Micronuclei, Chromosome-Defective/chemically induced , Micronuclei, Chromosome-Defective/drug effects , Mutagens/toxicity , beta-Glucans/chemistry , beta-Glucans/isolation & purification
14.
Semina cienc. biol. saude ; 26(2): 131-140, out.-dez. 2005. tab
Article in Portuguese | LILACS | ID: lil-429356

ABSTRACT

O Agaricus blazei Murril ss. Heinemann (ABM), cogumelo comestível nativo do Brasil, tem sido utilizado na medicina popular no tratamento de inúmeras doenças, incluindo o câncer. O objetivo do presente estudo foi avaliar os efeitos do extrato ABM (0,4 por cento) na clastogenicidade induzida pela exposição à radiação ultravioleta (UV), em células CHO-k1, pelo teste de aberração cromossômica. As células foram tratadas em diferentes condições (tratamento contínuo, pré-tratamento e pós-tratamento), associadas à indução de danos no DNA pela UV. A análise dos dados demonstrou que a UV e o ABM apresentaram atividade clastogênica. Nos protocolos de pré e pós-tratamento não foram evidenciados efeitos anticlastogênicos. No entanto, o protocolo de tratamento contínuo demonstrou efeito protetor com redução de danos de 86,1 por cento. Os resultados não permitem inferir com clareza o tipo de mecanismo de ação do extrato de ABM, o qual poderia agir tanto por desmutagênese, quanto por bioantimutagênese. no entanto, é evidente o seu efeito na diminuição de danos causados por radiação não-ionizante, apesar de, em concentração muito elevada, apresentar atividade clastogênica.


Subject(s)
Radiation, Nonionizing , Sunscreening Agents , Ultraviolet Rays , Agaricus , Light , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...