Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Mutat Res ; 480-481: 9-22, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11506795

ABSTRACT

Multiple points of intervention are the target for dietary and pharmacological interventions aimed at preventing cancer and other diseases in which mutations in somatic cells play a pathogenetic role. For instance, our studies showed that DNA adducts can be consistently detected in arterial smooth muscle cells from human atherosclerotic lesions. Their levels were significantly correlated with the occurrence of atherogenic risk factors known from traditional epidemiology and were strikingly enhanced in atherosclerotic patients lacking the GSTM1 genotype. Cancer chemoprevention has a dual goal, i.e. prevention of occurrence of the disease (primary prevention) and early detection and reversion of tumors at a premalignant stage (secondary prevention). At a later stage, attempts can be made to prevent local recurrences as well as invasion and metastasis of malignant cells (tertiary prevention). For a rational use of chemopreventive agents it is essential not only to evaluate their efficacy and safety but also to understand the mechanisms involved. Sometimes it is difficult to discriminate whether modulation of a given end-point is actually a specific mechanism or rather the epiphenomenon of other events. For instance, we recently found that apoptosis is considerably stimulated in the respiratory tract of smoke-exposed rats; whereas certain chemopreventive agents work by further enhancing smoke-related apoptosis, other agents appear to downregulate apoptosis simply because they inhibit the genotoxic events signaling this process. We propose here a detailed, updated classification of the points of intervention exploitable in the prevention of mutation and cancer. The general outline includes a variety of extracellular and cellular mechanisms modulating the genotoxic response and tumor initiation as well as tumor promotion, progression, angiogenesis, invasion, and metastasis. This classification is not intended to provide a rigid scheme, since several intervention points are reiterated several times over different phases of the process. Moreover, some mechanisms are strictly interconnected or partially overlapping. Interestingly, a number of chemopreventive agents work through multiple mechanisms, which warrants a higher efficacy and a broader spectrum of action. It is also convenient to combine chemopreventive agents working through complementary mechanisms. In recent preclinical studies, we observed that combination of N-acetylcysteine with either oltipraz or ascorbic acid produces additive or more than additive protective effects towards early biomarkers and/or experimentally-induced tumors.


Subject(s)
Chemoprevention , Mutation , Neoplasms/genetics , Neoplasms/prevention & control , Animals , Antioxidants/pharmacology , Biomarkers, Tumor , Diet , Disease Progression , Drug Therapy, Combination , Humans , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/prevention & control , Retinoids/pharmacology , Risk Factors
2.
Carcinogenesis ; 22(7): 999-1013, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11408342

ABSTRACT

Although smoking cessation is the primary goal for the control of cancer and other smoking-related diseases, chemoprevention provides a complementary approach applicable to high risk individuals such as current smokers and ex-smokers. The thiol N-acetylcysteine (NAC) works per se in the extracellular environment, and is a precursor of intracellular cysteine and glutathione (GSH). Almost 40 years of experience in the prophylaxis and therapy of a variety of clinical conditions, mostly involving GSH depletion and alterations of the redox status, have established the safety of this drug, even at very high doses and for long-term treatments. A number of studies performed since 1984 have indicated that NAC has the potential to prevent cancer and other mutation-related diseases. N-Acetylcysteine has an impressive array of mechanisms and protective effects towards DNA damage and carcinogenesis, which are related to its nucleophilicity, antioxidant activity, modulation of metabolism, effects in mitochondria, decrease of the biologically effective dose of carcinogens, modulation of DNA repair, inhibition of genotoxicity and cell transformation, modulation of gene expression and signal transduction pathways, regulation of cell survival and apoptosis, anti-inflammatory activity, anti-angiogenetic activity, immunological effects, inhibition of progression to malignancy, influence on cell cycle progression, inhibition of pre-neoplastic and neoplastic lesions, inhibition of invasion and metastasis, and protection towards adverse effects of other chemopreventive agents or chemotherapeutical agents. These mechanisms are herein reviewed and commented on with special reference to smoking-related end-points, as evaluated in in vitro test systems, experimental animals and clinical trials. It is important that all protective effects of NAC were observed under a range of conditions produced by a variety of treatments or imbalances of homeostasis. However, our recent data show that, at least in mouse lung, under physiological conditions NAC does not alter per se the expression of multiple genes detected by cDNA array technology. On the whole, there is overwhelming evidence that NAC has the ability to modulate a variety of DNA damage- and cancer-related end-points.


Subject(s)
Acetylcysteine/pharmacology , Anticarcinogenic Agents/pharmacology , Antimutagenic Agents/pharmacology , Smoking/genetics , Animals , Humans , Smoking/adverse effects
3.
Cancer Res ; 61(6): 2472-9, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11289117

ABSTRACT

Chemoprevention opens new perspectives in the prevention of cancer and other chronic degenerative diseases associated with tobacco smoking, exploitable in current smokers and, even more, in exsmokers and passive smokers. Evaluation of biomarkers in animal models is an essential step for the preclinical assessment of efficacy and safety of potential chemopreventive agents. Groups of Sprague Dawley rats were exposed whole body to a mixture of mainstream and sidestream cigarette smoke for 28 consecutive days. Five chemopreventive agents were given either with drinking water (N-acetyl-L-cysteine, 1 g/kg body weight/day) or with the diet (1,2-dithiole-3-thione, 400 mg; Oltipraz, 400 mg; phenethyl isothiocyanate, 500 mg; and 5,6-benzoflavone, 500 mg/kg diet). The monitored biomarkers included: DNA adducts in bronchoalveolar lavage cells, tracheal epithelium, lung and heart; oxidative damage to pulmonary DNA; hemoglobin adducts of 4-aminobiphenyl and benzo(a)pyrene-7,8-diol-9,10-epoxide; micronucleated and polynucleated alveolar macrophages and micronucleated polychromatic erythrocytes in bone marrow. Exposure of rats to smoke resulted in dramatic alterations of all investigated parameters. N-Acetyl-L-cysteine, phenylethyl isothiocyanate, and 5,6-benzoflavone exerted a significant protective effect on all alterations. 1,2-Dithiole-3-thione was a less effective inhibitor and exhibited both a systemic toxicity and genotoxicity in alveolar macrophages, whereas its substituted analogue Oltipraz showed limited protective effects in this model. Interestingly, combination of N-acetyl-L-cysteine with Oltipraz was the most potent treatment, resulting in an additive or more than additive inhibition of smoke-related DNA adducts in the lung and hemoglobin adducts. These results provide evidence for the differential ability of test agents to modulate smoke-related biomarkers in the respiratory tract and other body compartments and highlight the potential advantages in combining chemopreventive agents working with distinctive mechanisms.


Subject(s)
Anticarcinogenic Agents/pharmacology , Nicotiana/adverse effects , Plants, Toxic , Smoke/adverse effects , Smoking/metabolism , Acetylcysteine/pharmacology , Animals , Biomarkers/analysis , DNA Adducts/antagonists & inhibitors , DNA Adducts/metabolism , DNA Damage , Eating/drug effects , Hemoglobins/metabolism , Inhalation Exposure , Lung/metabolism , Male , Micronuclei, Chromosome-Defective , Oxidation-Reduction , Pyrazines/pharmacology , Rats , Rats, Sprague-Dawley , Smoking/blood , Thiones , Thiophenes , Tobacco Smoke Pollution , Weight Gain/drug effects
4.
Carcinogenesis ; 22(3): 375-80, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238175

ABSTRACT

Preclinical studies may elucidate the meaning of biomarkers applicable to epidemiologic studies and to clinical trials for cancer prevention. No study has explored so far the effect of cigarette smoke on apoptosis in vivo. We evaluated modulation of apoptosis in cells of the respiratory tract of smoke-exposed Sprague-Dawley rats both by morphological analysis and TUNEL method. In a first study, exposure of rats to mainstream cigarette smoke for either 18 or 100 consecutive days produced a significant and time-dependent increase in the proportion of apoptotic cells in the bronchial and bronchiolar epithelium. Oral N:-acetylcysteine did not affect the background frequency of apoptosis but significantly and sharply decreased smoke-induced apoptosis. In a second study, exposure of rats to a mixture of sidestream and mainstream smoke for 28 consecutive days resulted in a >10-fold increase in the frequency of pulmonary alveolar macrophages undergoing apoptosis. Dietary administration of either 5,6-benzoflavone, 1,2-dithiole-3-thione or oltipraz did not affect the frequency of smoke-induced apoptosis, whereas phenethyl isothiocyanate produced a further significant enhancement. Again, N-acetylcysteine and its combination with oltipraz significantly decreased smoke-induced apoptosis. In both studies exposure to smoke resulted in a sharp increase of cells positive for proliferating cell nuclear antigen (PCNA), which was unaffected by the examined chemopreventive agents. These findings highlight the concept that modulation of apoptosis has diversified meanings. Different meanings (as explained in the following lines). First, the apoptotic process is triggered as a defense system against genotoxic agents, such as the components of cigarette smoke. The further induction produced by phenethyl isothiocyanate, favoring removal of damaged cells, represents an example of a detoxification mechanism. Inhibition of smoke-induced apoptosis by N:-acetylcysteine should be interpreted as an epiphenomenon of antigenotoxic mechanisms, as shown in parallel studies evaluating modulation of DNA alterations in the respiratory tract of the same animals. Thus, it is important to discriminate between whether the opposite modulation of apoptosis is per se a protective mechanism or the beneficial outcome of other mechanisms inhibiting genotoxicity.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Respiratory System/drug effects , Smoke/adverse effects , Animals , In Situ Nick-End Labeling , Macrophages, Alveolar/cytology , Macrophages, Alveolar/drug effects , Male , Plants, Toxic , Rats , Rats, Sprague-Dawley , Respiratory System/cytology , Nicotiana
5.
Int J Oncol ; 18(3): 607-15, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179494

ABSTRACT

In spite of the major role played by cigarette smoking in the epidemiology of lung cancer, it is very difficult to reproduce the carcinogenicity of this complex mixture in animal models. We implemented a series of pilot experiments in three mouse strains, exposed either to environmental cigarette smoke (ECS) or mainstream cigarette smoke (MCS) or its condensate (MCSC). The whole-body exposure of Aroclor-treated A/J mice to ECS resulted in a rapid and potent induction of micronuclei in peripheral blood erythrocytes. After 6 months of exposure, 6 h a day, followed by 4 months of recovery in filtered air, both lung tumor incidence and multiplicity were significantly increased as compared to sham-exposed mice (77.8% vs. 22.2%, and 1.11+/-0.26 vs. 0.22+/-0.15, means +/- SE). Multiple i.p. injections of butylated hydroxytoluene did not significantly enhance the tumor yield. Another experiment confirmed the responsiveness of A/J mice exposed to ECS for 5 months, followed by 4 months of recovery in air (75.0% vs. 25.0%, and 1.05+/-0.17 vs. 0.25+/-0.10). In contrast, the increase in lung tumor yield after exposure to ECS for 2 months, followed by recovery in air for 7 months, was not significant, and the continuous exposure to ECS for 9 months was totally ineffective. These data, in agreement with previous results of others, show that exposure of A/J mice to ECS for 5-6 months, followed by recovery in air for 4 months, is successful in inducing a weak but significant and reproducible increase in lung tumor yield. Furthermore, the simultaneous exposure to the light emitted by halogen quartz bulbs for 9 months and to ECS for 5 months, followed by 4 months in air, was again weakly tumorigenic (incidence of 55.0% and multiplicity of 0.75+/-0.19), whereas exposure to both ECS and light for 9 months was devoid of effect. The whole-body exposure of A/J mice to MCS, 1 h a day for 5 months, or weekly i.p. injections of MCSC for 5 months, followed in both cases by 4 months of recovery in air, failed to enhance the lung tumor yield. The whole-body exposure of SKH-1 hairless mice to ECS for 6 months, followed by exposure to halogen light for 8 months, resulted in the formation of multiple skin tumors but failed to produce lung tumors. The whole-body exposure of C57BL/6 mice to ECS for 6 months failed to induce any lung tumor but caused alopecia, gray hair, and hair bulb cell apoptosis, which were prevented by the oral administration of N-acetylcysteine.


Subject(s)
Adenoma/etiology , Lung Neoplasms/etiology , Smoking/adverse effects , Acetylcysteine/pharmacology , Adenoma/genetics , Adenoma/pathology , Animals , Body Weight , Butylated Hydroxytoluene/toxicity , Cytogenetic Analysis , DNA Damage/drug effects , DNA, Neoplasm/drug effects , Female , Incidence , Injections, Intraperitoneal , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Hairless , Mice, Inbred C57BL , Pilot Projects
6.
Int J Cancer ; 88(5): 702-7, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11072237

ABSTRACT

Both ascorbic acid (AsA, vitamin C) and N-acetylcysteine (NAC), a precursor and analogue of glutathione, possess a broad array of biological properties underlying their protective role in a variety of pathophysiological conditions. However, under certain circumstances, AsA behaves as a pro-oxidant rather than an anti-oxidant and produces adverse effects. This prompted us to evaluate whether NAC could interact with AsA in preventing mutation and cancer. AsA significantly increased spontaneous revertants in the Salmonella typhimurium strains TA102 and TA104, which are sensitive to oxidative mutagens. In contrast, NAC lowered the spontaneous background in TA104 and neutralized the negative effects of AsA. Moreover, NAC and AsA showed additive effects in reducing chromium(VI) and in reverting its mutagenicity. A single i.p. injection of urethane (1 g/kg body weight) to 120 A/J mice resulted, after 4 months, in the formation of a total of 1,532 lung tumors, 425 in the 30 mice treated with the carcinogen only, 404 in those treated with urethane plus AsA, 365 in those treated with urethane plus NAC and 338 in those treated with urethane plus the combination of AsA and NAC (both given daily with drinking water at the dose of 1 g/kg body weight). Compared to positive controls, tumor multiplicity was poorly affected by AsA, whereas it was significantly decreased by NAC and even more so by its combination with AsA. The overall volumes of lung tumors in the 4 groups were 107.5, 89.3, 61.3 and 49.7 mm(3), respectively. Tumor sizes were slightly but significantly decreased in mice treated with AsA and more so in those treated with NAC and NAC plus AsA, their combination being significantly more effective than each individually. All protective effects elicited by combining the 2 drugs were additive. Therefore, NAC prevents the adverse effects of AsA on spontaneous mutagenicity; at the same time, this thiol behaves in an additive fashion with AsA, inhibiting the mutagenicity of chromium(VI) and the lung tumorigenicity of urethane in mice. These findings suggest that NAC and AsA could conveniently be combined in cancer chemoprevention and other pharmacological interventions.


Subject(s)
Acetylcysteine/metabolism , Ascorbic Acid/metabolism , Cell Transformation, Neoplastic/metabolism , Lung Neoplasms/metabolism , Mutagenesis/physiology , Animals , Carcinogenicity Tests , Chromium/metabolism , Drug Stability , Female , Lung Neoplasms/chemically induced , Mice , Urethane
7.
Carcinogenesis ; 21(9): 1677-82, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10964099

ABSTRACT

A combination of tobacco smoking with certain agents has been shown to exert synergistic carcinogenic effects. On the other hand, antagonism betweeen smoke and other pulmonary carcinogens has also been documented by both epidemiological and experimental data. In spite of a very large number of studies carried out for decades in workers exposed to hexavalent chromium, the influence of smoking habits on lung carcinogenesis induced by this metal has not been clarified. For this reason, we performed two studies evaluating clastogenic effects in rodents. In the first one, BDF(1) mice were exposed whole-body to mainstream cigarette smoke for 5 days and, on the last day, they received an i.p. injection of potassium dichromate. In the second study, Sprague-Dawley rats were exposed whole-body to environmental cigarette smoke for 18 consecutive days and for the same period of time they received daily intra-tracheal instillations of sodium dichromate. Individually, the two hexavalent chromium salts and cigarette smoke, either mainstream or environmental, enhanced the frequency of micronuclei in bone marrow polychromatic erythrocytes of both mice and rats. Moreover, individual exposure to either environmental cigarette smoke or sodium dichromate enhanced the frequency of micronuclei and multiple nuclei in pulmonary alveolar macrophages of rats. In both studies, combined exposure to cigarette smoke and hexavalent chromium produced less than additive clastogenic effects. These results are consistent with our previous data, showing that hexavalent chromium and either benzo[a]pyrene or cigarette smoke condensate behave antagonistically in in vitro mutagenicity test systems and that the chromium reducing capacity of human pulmonary alveolar macrophages and peripheral lung parenchyma is enhanced in smokers. Taken together, in the absence of any epidemiological evidence, these findings rule out any occurrence of synergism between cigarette smoke and hexavalent chromium, at least in certain stages of the carcinogenesis process.


Subject(s)
Cocarcinogenesis , Mutagens/toxicity , Potassium Dichromate/toxicity , Smoking/adverse effects , Animals , Bone Marrow Cells/drug effects , Bronchoalveolar Lavage , Dose-Response Relationship, Drug , Erythrocytes/drug effects , Injections, Intraperitoneal , Instillation, Drug , Macrophages, Alveolar/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Micronuclei, Chromosome-Defective/drug effects , Rats , Rats, Sprague-Dawley , Tobacco Smoke Pollution/adverse effects , Trachea
8.
Carcinogenesis ; 20(8): 1491-7, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10426797

ABSTRACT

In spite of the major role played by smoking tobacco in the epidemiology of chronic degenerative diseases, it is difficult to mimic the genotoxic and carcinogenic effects of this complex mixture in animal models. We undertook an experimental study evaluating the time-course induction, persistence and modulation of cytogenetic alterations induced in BDF(1) mice exposed whole-body to mainstream cigarette smoke. The animals were divided into five groups, including: (i) 72 sham-exposed mice; (ii) 72 mice exposed to smoke for up to 3 weeks; (iii) 72 mice treated daily with the thiol N-acetylcysteine (NAC, 0.5 g/kg body weight) with drinking water; (iv) 72 mice exposed to smoke and treated daily with NAC, starting 5 days before exposure to smoke; and (v) 48 mice exposed to smoke and treated daily with NAC, starting 1 day after discontinuation of exposure to smoke. After 1, 2, 3, 4, 5, 6, 7, 10 and 14 weeks since the start of exposure to cigarette smoke, eight mice per group were killed, and cytogenetic parameters were evaluated. Exposure to smoke induced a high frequency of micronucleated and binucleated (BN) pulmonary alveolar macrophages, which persisted for at least 14 weeks. The frequency of micronuclei increased early in bone marrow polychromatic erythrocytes, but declined to background levels upon discontinuation of exposure to smoke. By comparison, their induction in circulating normochromatic erythrocytes (NCE) was slightly delayed, less intense but still significant, and persisting for an additional 3 weeks. Administration of NAC, throughout duration of the experiment, strongly inhibited the smoke-induced formation of micronuclei in alveolar macrophages and had some transiently significant effect on the induction of BN macrophages. NAC did not significantly decrease the smoke-induced formation of micronuclei in bone marrow cells, whereas it attenuated the formation of micronuclei in peripheral blood NCE. When given after discontinuation of exposure to cigarette smoke, NAC did not affect the cytogenetic alterations but normalized the altered bronchoalveolar lavage cellularity. The present data provide a detailed analysis of time-related cytogenetic alterations in smoke-exposed mice, both in the respiratory tract and at a systemic level, and show the effects of NAC on these parameters and on the pulmonary inflammatory response.


Subject(s)
Chromosome Aberrations , Micronuclei, Chromosome-Defective , Tobacco Smoke Pollution/adverse effects , Acetylcysteine/administration & dosage , Animals , Erythrocytes , Female , Free Radical Scavengers/administration & dosage , Macrophages, Alveolar , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Micronucleus Tests
9.
Int J Cancer ; 77(2): 302-5, 1998 Jul 17.
Article in English | MEDLINE | ID: mdl-9650568

ABSTRACT

A major goal in pre-clinical cancer chemoprevention research is to assess the predictive value of intermediate biomarker modulation towards tumor prevention. With this aim, BALB/c mice were treated with 10 daily i.p. injections of urethane (ethyl carbamate), each of 400 mg/kg body weight. Groups of mice received with drinking water either a drug containing the thiol N-acetylcysteine (NAC), at 0.1 or 0.5 g/kg body weight, or its excipient, starting 27 days before the first injection of the carcinogen until the end of the experiment. Out of the 30 mice, 10 per group were identified and individually monitored for 8 sequential times in order to assess the course of micronucleated normochromatic erythrocytes in peripheral blood. This systemic genotoxicity biomarker increased during the 10-day period of treatment with urethane, reached a peak 2 to 6 days after the last injection, and was still significantly higher than the baseline after 10 additional days. Clastogenicity was significantly inhibited by NAC, with a dose-related effect, but not by the drug excipient. As evaluated 4 months after the first injection of urethane, most mice developed lung tumors, whose multiplicity was not affected by the drug excipient but was significantly decreased in the presence of NAC. Correlation between the frequency of micronucleated normochromatic erythrocytes at peak levels and lung-tumor multiplicity was highly significant when evaluated in the context of all 40 mice undergoing cytogenetic analyses (r = 0.561, p = 0.0002). It was similarly high, but did not reach the significance threshold, within each treatment group, due to the lower number of animals and some deviations from the regression line. Therefore, the prediction of lung-tumor yield based on the intensity of the early genotoxicity biomarker is justified when formulated within a sufficiently large group of animals, but is not absolute at individual level.


Subject(s)
Acetylcysteine/pharmacology , Anticarcinogenic Agents/pharmacology , Antimutagenic Agents/pharmacology , Lung Neoplasms/chemically induced , Micronuclei, Chromosome-Defective , Urethane/antagonists & inhibitors , Acetylcysteine/administration & dosage , Administration, Oral , Animals , Body Weight , Carcinogens , Lung Neoplasms/prevention & control , Male , Mice , Mice, Inbred BALB C , Time Factors
10.
FASEB J ; 12(9): 753-8, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9619454

ABSTRACT

In spite of the epidemiological evidence supporting a synergism between alcohol consumption and cigarette smoking in the pathogenesis of cancers of the aerodigestive tract, there is a paucity of experimental studies evaluating the effects of these agents under well-controlled conditions and exploring the mechanisms involved. We exposed groups of female BD6 rats, aged 8 months, to ethanol (5% in drinking water for 8 consecutive months) and/or whole-body to mainstream cigarette smoke (1 h/day, 5 days/week for 8 months). DNA was purified from different organs and analyzed for the presence of DNA-protein crosslinks and 32P-postlabeled DNA adducts after butanol enrichment. No significant increase of DNA-protein crosslinks, compared to untreated controls, was induced by any treatment in liver, lung, or heart. 'Spontaneous' nucleotidic modifications were detected by 32P-postlabeling in organs of untreated rats, with the highest levels occurring in the heart. Ingestion of ethanol did not affect DNA adduct levels in any of the organs examined: esophagus, liver, lung, and heart. Exposure to cigarette smoke induced formation of DNA adducts in the lung and heart, but not in the esophagus or liver. The combined ingestion of ethanol resulted in a significant formation of smoke-related DNA adducts in the esophagus and in their further, dramatic increase in the heart. It thus appears that ethanol consumption increases the bioavailability of DNA binding smoke components in the upper digestive tract and favors their systemic distribution. The mechanisms responsible for the interaction between ethanol and smoke and for the selective localization of DNA alterations in different organs are discussed. Formation of DNA adducts in the organs examined may be relevant in the pathogenesis of lung and esophageal cancers as well as in the pathogenesis of other types of chronic degenerative diseases, such as chronic obstructive pulmonary diseases and cardiomyopathies.


Subject(s)
Alcohol Drinking/adverse effects , DNA Adducts/analysis , DNA Damage , Nicotiana/adverse effects , Plants, Toxic , Smoke/adverse effects , Animals , Drug Interactions , Esophagus/pathology , Female , Liver/pathology , Lung/pathology , Myocardium/pathology , Rats
11.
Cancer Lett ; 94(1): 91-5, 1995 Jul 20.
Article in English | MEDLINE | ID: mdl-7621451

ABSTRACT

Exposure of male Balb/C mice to mainstream cigarette smoke for 4 months, starting 10 or 30 days before the administration of ethyl carbamate (0.3% in drinking water for 3 weeks), resulted in an up to 57.6% (P < 0.05) decrease of lung adenoma multiplicity. However, the number of ethyl carbamate-induced lung tumors was not significantly affected by exposure to cigarette smoke when ethyl carbamate was injected i.p. in single doses of 0.5 or 1.0 g/kg, irrespective of the different treatment schedules used, i.e. (a) 10 days before and 4 days after the ethyl carbamate injection; (b) throughout the experiment starting 10 days before the ethyl carbamate injection, and (c) until the end of the experiment, starting 30 days after the ethyl carbamate injection. Disulfiram (500 mg/kg), given by gavage 24 h and 1 h before the ethyl carbamate injection, decreased by 88.5% (P < 0.001) the multiplicity of lung adenomas but had no effect on tumorigenesis when administered after the carcinogen injection. Proadifen (SKF-525 A, 50 mg/kg) injected i.p. 24 h and 1 h before and 24 h and 48 h after the injection with ethyl carbamate tended to decrease the multiplicity of lung adenomas, but not to a significant extent. Furthermore, disulfiram given 24 h and 1 h before the i.p. administration of ethyl carbamate completely prevented its clastogenicity in mouse bone marrow. On the other hand, cigarette smoke, which was per se a weak clastogen in bone marrow erythroblasts, synergistically potentiated the clastogenic response to ethyl carbamate in a more than additive fashion.


Subject(s)
Disulfiram/pharmacology , Lung Neoplasms/etiology , Nicotiana , Plants, Toxic , Smoke/adverse effects , Urethane , Animals , Bone Marrow/drug effects , Cytochrome P-450 Enzyme System/metabolism , Female , Lung Neoplasms/chemically induced , Male , Mice , Mice, Inbred BALB C , Micronucleus Tests , Urethane/administration & dosage , Water Supply
12.
J Cell Biochem Suppl ; 22: 33-41, 1995.
Article in English | MEDLINE | ID: mdl-8538208

ABSTRACT

The thiol N-acetylcysteine (NAC), now under clinical trial for cancer chemoprevention both in Europe (project Euroscan) and in the US (National Cancer Institute), has been shown during the past decade to exert protective effects in a variety of experimental test systems. NAC inhibited spontaneous mutagenicity and that induced by a number of chemical compounds and complex mixtures. Moreover, NAC significantly decreased the incidence of neoplastic and preneoplastic lesions induced by several chemical carcinogens in rodents (mice, rats, hamsters), e.g., in lung, trachea, colon, liver, mammary gland, Zymbal gland, bladder and skin. Our studies provided evidence that multiple mechanisms contribute to NAC antimutagenicity and anticarcinogenicity. They include extracellular mechanisms, such as detoxification of reactive compounds due to the nucleophilic and antioxidant properties of NAC, inhibition of nitrosation products, and enhancement of thiol concentration in intestinal bacteria; trapping and enhanced detoxification of carcinogens in long-lived non-target cells, such as erythrocytes and bronchoalveolar lavage cells; mechanisms working in the cytoplasm of target cells, such as replenishment of GSH stores, modulation of metabolism of mutagens/carcinogens, blocking of electrophiles, and scavenging of reactive oxygen species; and nuclear effects, such as inhibition of DNA adduction by metabolites of carcinogens, inhibition of "spontaneous" mutations, attenuation of carcinogen-induced DNA damage, and protection of nuclear enzymes, such as poly(ADP-ribose) polymerase.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Acetylcysteine/therapeutic use , Anticarcinogenic Agents/therapeutic use , Animals , Biomarkers/chemistry , Cricetinae , Cytoplasm/drug effects , Humans , Inactivation, Metabolic , Mice , Mutagenicity Tests , Neoplasm Invasiveness , Neoplasm Metastasis , Rats
13.
J Cell Biochem ; 56(4): 449-54, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7890804

ABSTRACT

A series of 16 experiments, using a total of 2,000 BD6 rats, was designed in order to assess the ability of 8 individual agents or their combinations to modulate the liver and oesophageal carcinogenesis induced by multiple doses of diethylnitrosamine (DEN). Of the antioxidants tested, sodium selenite, ascorbic acid, and butylated hydroxytoluene generally exhibited protective effects on both types of tumors. In contrast, retinoic acid behaved as a promoter of DEN hepatocarcinogenesis, but this effect could be eliminated by its combination with either selenite or butylated hydroxytoluene. Caffeine and theophylline, when individually assayed, were devoid of significant protective effects, and the latter methylxanthine stimulated oesophageal tumorigenesis when administered after exposure to the carcinogen. Caffeine tended to decrease the multiplicity of liver tumors and potentiated the inhibitory effect of selenite in the liver. Irrespective of combination with caffeine, treatment with phenobarbital before each DEN injection tended to reduce the multiplicity of both liver and oesophageal tumors. On the other hand, the metabolic inhibitor diethyldithiocarbamate, given after each DEN injection, dramatically enhanced the incidence and multiplicity of oesophageal tumors. Thus, on the whole, modulation of DEN carcinogenesis varied depending on test agents, their combinations, dosages, treatment schedules, and target organ.


Subject(s)
Esophageal Neoplasms/prevention & control , Neoplasms, Experimental/prevention & control , Animals , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Caffeine/pharmacology , Diethylnitrosamine , Disease Models, Animal , Female , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/prevention & control , Neoplasms, Experimental/chemically induced , Phenobarbital/pharmacology , Rats , Rats, Inbred Strains , Survival , Theophylline/pharmacology , Tretinoin/pharmacology
14.
Mutat Res ; 317(2): 89-109, 1994 Apr.
Article in English | MEDLINE | ID: mdl-7511795

ABSTRACT

Two antimutagenicity databases were prepared by applying a co-treatment procedure to the Salmonella reversion assay. Ninety compounds belonging to various chemical classes were quantitatively tested for antimutagenicity towards the direct-acting mutagen 4-nitroquinoline 1-oxide (4NQO) in strain TA100 of S. typhimurium and 63 of them were additionally tested for antimutagenicity towards unfractionated mainstream cigarette smoke (CS) in strain TA98, in the presence of S9 mix. Twelve compounds (13.3%) inhibited 4NQO mutagenicity by at least 50%, with a MID50 (dose inhibiting 50% of mutagenicity) varying over a 1226-fold range. Twenty-six compounds (41.3%) inhibited CS mutagenicity, with a MID50 varying over a 520-fold range. Three compounds only, i.e., bilirubin, curcumin and myricetin, were capable of inhibiting the mutagenicities of both 4NQO and CS. However, myricetin and the other flavonoid rutin were at the same time mutagenic by inducing frameshift mutations following metabolic activation. There was a rather rigorous selectivity of antimutagenicity data depending on the chemical class of inhibitors and it was possible to discriminate protective effects within several pairs or series of structurally related compounds. For instance, all eight thiols and aminothiols inhibited 4NQO mutagenicity, which contrasted with the inactivity of the remaining 17 sulfur compounds tested, all of them lacking a free sulfhydryl group. The mutagenicity of CS was consistently inhibited by the majority of phenols (eight out of 10 tested) and by all two isothiocyanates, two dithiocarbamates, three indole derivatives, three tetrapyrrole compounds and three flavonoids tested. Although the results obtained cannot be extrapolated to other mutagens or test systems, they may provide a useful source of information for research in the area of antimutagenesis and for the development of chemopreventive agents.


Subject(s)
4-Nitroquinoline-1-oxide/toxicity , Antimutagenic Agents/pharmacology , Information Systems , Nicotiana , Plants, Toxic , Smoke/adverse effects , Mutagenicity Tests , Structure-Activity Relationship
15.
Cancer Res ; 54(7 Suppl): 1994s-1998s, 1994 Apr 01.
Article in English | MEDLINE | ID: mdl-8137327

ABSTRACT

Molecular dosimetry techniques were exploited in order to assess the efficacy of experimental chemoprevention assays and to evaluate the involvement of DNA alterations, not only in cancer but also in other chronic degenerative diseases. In agreement with other protective effects previously observed in the same animal models, the thiol N-acetylcysteine (NAC) totally prevented or significantly reduced the formation of carcinogen-DNA adducts in three experimental systems in rats. Thus, as assessed by 32P postlabeling, supplement of the diet with NAC decreased both deoxyguanosine-C8-aminofluorene adducts (butanol enrichment) and deoxyguanosine-N2-acetylaminofluorene adducts (nuclease P1 enrichment) formed in rat liver following dietary administration of 2-acetylaminofluorene for 3 weeks. DNA adducts were detected by synchronous fluorescence spectrophotometry in rat liver, lung, heart, and testis following a daily i.t. instillation of benzo(a)pyrene for 3 consecutive days. The whole-body exposure of rats to mainstream cigarette smoke for 40 consecutive days resulted in the appearance of DNA adducts in heart, lung, and aorta, whereas no adduct was detected by synchronous fluorescence spectrophotometry in liver, brain, and testis. Multiple DNA adducts in the aorta were also measured by 32P postlabeling. Administration of NAC by gavage inhibited the formation of DNA adducts in all organs of rats treated with benzo(a)pyrene or exposed to cigarette smoke. It is of interest that a single chemopreventive agent can display a broad-spectrum protective ability. The selective localization of DNA adducts in different organs depends on pharmacokinetics, metabolic capacity, DNA repair efficiency, and cell proliferation rate. Whereas inhibition by NAC of DNA adducts in testis can be correlated with its demonstrated ability to prevent dominant lethal mutations, we raise the hypothesis that DNA adducts in lung, heart, and aorta may be pathogenetically associated with lung cancer, cardiomyopathies, and arteriosclerosis, respectively. In order to explore the involvement of molecular and biochemical alterations in human arteriosclerosis, we started an extensive collaborative project and report here preliminary data showing the presence of DNA adducts in aorta smooth muscle cells obtained from arteriosclerotic patients.


Subject(s)
Acetylcysteine/pharmacology , Anticarcinogenic Agents/pharmacology , Arteriosclerosis/etiology , Carcinogens/metabolism , Carcinogens/toxicity , DNA/metabolism , 2-Acetylaminofluorene/toxicity , Animals , Aorta, Abdominal/pathology , Arteriosclerosis/pathology , Arteriosclerosis/prevention & control , Cricetinae , DNA/drug effects , Diet , Humans , Male , Mice , Rats , Rats, Sprague-Dawley , Rats, Wistar , Smoke/adverse effects , Smoking/adverse effects
16.
Cancer Lett ; 72(3): 183-9, 1993 Aug 31.
Article in English | MEDLINE | ID: mdl-8402590

ABSTRACT

A series of experiments was carried out to assess cytotoxic and cytogenetic effects in bone marrow polychromatic erythrocytes (PCE) and pulmonary alveolar macrophages (PAM) resulting from individual or combined exposure of male BD6 rats to ethanol, cigarette smoke and Aroclor 1254. Addition of 5% ethanol to drinking water did not affect the micronucleus frequency but consistently enhanced the proportion of polynucleated PAM. Moreover, the higher concentration used (10%) was cytotoxic in the bone marrow. Whole-body exposure to cigarette smoke elevated the micronucleus frequency in both PCE (4.0-4.4-fold) and PAM (2.0-3.6-fold) and enhanced the frequency of polynucleated PAM. After 3 weeks of combined exposure, ethanol produced contrasting effects in smoke-exposed rats, i.e. an increase of micronuclei in PCE and a decrease in PAM. An i.p. injection of Aroclor 1254 was per se devoid of any influence on the monitored parameters but tended to attenuate the cytotoxic and cytogenetic changes produced by cigarette smoke or ethanol in both types of cell.


Subject(s)
Erythroblasts/pathology , Ethanol/toxicity , Macrophages, Alveolar/pathology , Mutagens/toxicity , Smoke , Animals , Aroclors/toxicity , Erythroblasts/drug effects , Macrophages, Alveolar/drug effects , Male , Micronucleus Tests , Rats , Rats, Inbred Strains
17.
Environ Health Perspect ; 99: 249-52, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8319635

ABSTRACT

In addition to possessing an extraordinary sweeping activity, pulmonary alveolar macrophages (PAM) are equipped with the biochemical mechanisms involved in the metabolism of carcinogens, which were found to be inducible in humans by cigarette smoke. Moreover, several defense processes were stimulated in rat PAM after in vivo administration of the anticarcinogen N-acetylcysteine (NAC). Benzo[a]pyrene diol epoxide (BPDE)-DNA adducts, as revealed by synchronous fluorescence spectrophotometry, were selectively detected in PAM of smokers and persisted up to 6 months. The amount of adducts was significantly correlated with the number of currently smoked cigarettes but not with the cigarettes smoked in a lifetime (pack-years). Nevertheless, deviations from the regression line pointed out the role of interindividual variability factors in adduct formation. Probably due to the low mitotic rate of PAM in the respiratory lumen, the frequency of micronuclei was not enhanced in smokers. However, parallel assays in rats showed that micronuclei can be enhanced after massive intratracheal administration of benzo[a]pyrene or whole-body exposure to high amounts of mainstream cigarette smoke, which also induced BPDE-DNA adducts in lung cells and other organs, including the heart. All these adverse effects were markedly inhibited by the oral administration of NAC, which provides the premise and rationale for a future study on the protective effects of oral NAC in heavy smokers.


Subject(s)
DNA Adducts , Macrophages, Alveolar/drug effects , Neoplasms/prevention & control , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/analysis , Acetylcysteine/pharmacology , Adolescent , Adult , Aged , Animals , Benzo(a)pyrene/toxicity , DNA/analysis , Female , Humans , Macrophages, Alveolar/chemistry , Macrophages, Alveolar/ultrastructure , Male , Micronuclei, Chromosome-Defective/drug effects , Micronuclei, Chromosome-Defective/ultrastructure , Middle Aged , Rats , Rats, Sprague-Dawley , Smoking/pathology
18.
Carcinogenesis ; 13(11): 2187-90, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1423892

ABSTRACT

The formation of smoke-related DNA adducts and their chemoprevention were investigated in tissues of male Sprague-Dawley rats, by testing a total of 132 DNA samples by synchronous fluorescence spectrophotometry (SFS), which mainly detects benzo[a]pyrene diolepoxide (BPDE)-DNA adducts. Groups of six animals each were exposed whole-body to mainstream cigarette smoke, once daily, for up to 40 consecutive days. No adduct was revealed in liver DNA, whereas smoke-related DNA adducts were detectable in the lung from the 8th day of exposure and continued to increase until the 40th day. Adducts to heart DNA, which were monitored after 28 and 40 days of exposure, attained even higher levels than those detected in the lungs of the same animals. A high correlation existed between the amounts of smoke-related DNA adducts measured in these two organs. The daily administration by gavage of the thiol N-acetyl-L-cysteine (NAC), an effective mutation and cancer chemopreventive agent, which had been previously shown to inhibit the formation of SFS-positive DNA adducts in rats receiving intratracheal instillations of benzo[a]pyrene, significantly prevented occurrence of the same adducts in both heart and lungs of smoke-exposed rats. No fluorescence signal was observed in liver, lung, or heart DNA of sham-exposed animals. The findings of this molecular dosimetry study complement the results of parallel histopathologic, cytogenetic, biochemical and metabolic analyses of tissues and cells from the same rats, providing evidence for a variety of significant alterations produced by exposure to cigarette smoke and for the specific protective role of NAC.


Subject(s)
7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/toxicity , Acetylcysteine/pharmacology , DNA/drug effects , Heart/drug effects , Lung/drug effects , Neoplasms, Experimental/prevention & control , Animals , Carcinogens/toxicity , Male , Plants, Toxic , Rats , Rats, Sprague-Dawley , Smoke , Nicotiana
19.
Mutagenesis ; 7(4): 295-301, 1992 Jul.
Article in English | MEDLINE | ID: mdl-1518414

ABSTRACT

Male Sprague-Dawley rats were exposed whole-body to the mainstream smoke produced by a commercial filter cigarette for 8 consecutive days, accounting for a cumulative exposure to the smoke of 75 cigarettes. Liver and lung S12 fractions were used in the Salmonella mutagenicity test in order to assess either the decrease of potency of a direct-acting mutagen (sodium dichromate) or the metabolic activation of promutagens, including cigarette smoke itself and its condensate, benzo[a]pyrene and its 7,8-diol, the aromatic amine 2-aminofluorene, and the heterocyclic amine 3-amino-1-methyl-5H-pyrido(4,3)indole. Moreover, individual biochemical parameters were measured in the liver and lung of the same rats and, in the case of cytochrome P-450-dependent monooxygenases, also in the heart of untreated or Aroclor-treated rats. The monitored biochemical parameters included aryl hydrocarbon (benzo[a]pyrene) hydroxylase and ethoxyresorufin deethylase in microsomal fractions, epoxide (benzo[a]pyrene-4,5-oxide) hydrolase in both microsomal and cytosolic fractions, glutathione (GSH) and GSH S-transferase in the cytosol. Exposure to cigarette smoke resulted in a number of significant metabolic changes, as compared to sham-exposed rats. The most pronounced alterations consisted in a 2.6-fold induction of aryl hydrocarbon hydroxylase in the lung and 8-fold induction of ethoxyresorufin deethylase in the liver, and in a marked stimulation of the liver metabolic activation of all promutagens. The last effect was inhibited by the oral administration of the chemopreventive agent N-acetylcysteine. On the whole, there was a poor correlation between the monitored biochemical and mutagenicity endpoints.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Acetylcysteine/administration & dosage , Liver/drug effects , Lung/drug effects , Nicotiana , Plants, Toxic , Smoke/adverse effects , Administration, Oral , Animals , Liver/metabolism , Lung/metabolism , Male , Mutagenicity Tests/methods , Mutagens/metabolism , Rats , Rats, Inbred Strains , Salmonella typhimurium/genetics
20.
Mutat Res ; 268(1): 77-82, 1992 Jul.
Article in English | MEDLINE | ID: mdl-1378189

ABSTRACT

The genotoxic effects of mitomycin C (MMC) and farmorubicin (FR) in a free form and included in polybutylcyanoacrylate nanoparticles (PBCN) were studied employing the Salmonella/microsome mutagenicity assay and the micronucleus test in mouse bone marrow as well as in mouse fetal liver. The data obtained clearly indicated that MMC (0.25-2.00 micrograms/plate) was a strong mutagen in S. typhimurium TA102, while the same concentrations of this compound in PBCN were ineffective in inducing his+ revertant mutations in bacterial cells. A similar total suppression of mutagenic activity of FR (1.0-20.0 micrograms/plate) was registered in S. typhimurium TA98 when the drug was included in PBCN. Furthermore, the incorporation of MMC (2.0 or 4.0 mg/kg, i.p.) into PBCN strongly diminished or even abolished its clastogenic activity in the bone marrow of virgin and pregnant mice as well as in mouse fetal liver, respectively. In addition, a lack of genotoxic effect of PBCN only was also established. The toxic activity of MMC in mouse bone marrow was significantly reduced or completely abolished after its inclusion in PBCN. A conclusion might be drawn that the genotoxic activity of some antitumor drugs might be markedly diminished or even abolished after their incorporation in PBCN.


Subject(s)
Enbucrilate , Epirubicin/toxicity , Mitomycin/toxicity , Mutagens/toxicity , Animals , Bone Marrow/drug effects , Drug Carriers , Female , Liver/drug effects , Liver/embryology , Maternal-Fetal Exchange , Mice , Micronuclei, Chromosome-Defective/drug effects , Micronucleus Tests , Mutagenicity Tests , Pregnancy , Salmonella typhimurium/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...