Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
1.
Cells ; 11(20)2022 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-36291178

RESUMO

The binding of SARS-CoV-2 spikes to the cell receptor angiotensin-converting enzyme 2 (ACE2) is a crucial target both in the prevention and in the therapy of COVID-19. We explored the involvement of oxidoreductive mechanisms by investigating the effects of oxidants and antioxidants on virus uptake by ACE2-expressing cells of human origin (ACE2-HEK293). The cell uptake of pseudoviruses carrying the envelope of either Delta or Omicron variants of SARS-CoV-2 was evaluated by means of a cytofluorimetric approach. The thiol N-acetyl-L-cysteine (NAC) inhibited the uptake of both variants in a reproducible and dose-dependent fashion. Ascorbic acid showed modest effects. In contrast, neither hydrogen peroxide (H2O2) nor a system-generating reactive oxygen species (ROS), which play an important role in the intracellular alterations produced by SARS-CoV-2, were able to affect the ability of either Delta or Omicron SARS-CoV-2 pseudoviruses to be internalized into ACE2-expressing cells. In addition, neither H2O2 nor the ROS generating system interfered with the ability of NAC to inhibit that mechanism. Moreover, based on previous studies, a preventive pharmacological approach with NAC would have the advantage of decreasing the risk of developing COVID-19, irrespective of its variants, and at the same time other respiratory viral infections and associated comorbidities.


Assuntos
Enzima de Conversão de Angiotensina 2 , Tratamento Farmacológico da COVID-19 , Humanos , SARS-CoV-2 , Acetilcisteína/farmacologia , Peróxido de Hidrogênio/farmacologia , Espécies Reativas de Oxigênio , Antioxidantes/farmacologia , Células HEK293 , Peptidil Dipeptidase A/metabolismo , Ácido Ascórbico/farmacologia , Oxidantes/farmacologia , Compostos de Sulfidrila/farmacologia
3.
Food Chem Toxicol ; 155: 112383, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34224802

RESUMO

In spite of the overwhelming epidemiological evidence for cigarette smoke (CS) carcinogenicity, less attention has been paid to the effects of CS as a complex mixture. As assessed in a series of experiments in murine models, the whole-body exposure to mainstream CS induced significant increases of micronucleated cells in the respiratory tract, bone marrow and peripheral blood of adult mice as well as in the liver and peripheral blood of foetuses whose mothers had been exposed throughout pregnancy. Urethane was potently clastogenic in the same cells when injected intraperitoneally. The daily administration of extra-virgin olive oil by gavage produced evident and consistent protective effects in all monitored experimental systems. In contrast, sunflower oil exhibited some adverse effects. Curcumin did not produce any significant effect in the bone marrow of both CS-exposed adults and foetuses but it elicited a dose-dependent protective effect traceable in blood erythrocytes. However, the higher curcumin dose further increased the frequency of micronucleated pulmonary alveolar macrophages. The apparent protective effects produced by lycopene and by a carotenoid mix were overwhelmed by those produced by olive oil, and lycopene even exhibited a worsening effect on the frequency of micronucleated erythroblasts in the bone marrow of urethane-treated adult mice.


Assuntos
Mutagênicos/toxicidade , Substâncias Protetoras/farmacologia , Poluição por Fumaça de Tabaco/efeitos adversos , Uretana/toxicidade , Animais , Curcumina/farmacologia , Eritrócitos/efeitos dos fármacos , Feminino , Feto/efeitos dos fármacos , Licopeno/farmacologia , Macrófagos Alveolares/efeitos dos fármacos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Azeite de Oliva/farmacologia , Gravidez
4.
J Med Virol ; 93(3): 1613-1619, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32902903

RESUMO

March 21, 2020 was the ridgeline between the growth of the coronavirus disease 2019 (COVID-19) epidemic wave in Italy and the start of its decline. We analyzed the epidemic patterns from March 1 to June 30. There was a progressive drop of cases from March (104,710) to April (94,888), May (25,705) and June (8110). Likewise, after a slight increase of deaths in April (14,804) compared to March (12,396), a considerable decline occurred in May (5170) and June (1464). Doubling times of cumulative cases grew from 2 to 6 days until March 20 to 2 weeks up to April 5, and thereafter no further doubling occurred until June 30. There was a striking North-South gradient of both cases and deaths. At the end of June, the nine Northern Italian regions or provinces, five central regions, and seven southern regions had contributed to the 81.1%, 12.4%, and 6.5% of the 240,578 national cases, respectively. Lombardy, the most populous region, was by far the most heavily affected one, accounting for the 39.0% of the national cases occurring over the analyzed 4-month period. However, in relative terms, it was preceded by Aosta Valley, the least populous region, less than 1% of the population of both regions having been affected by cases of COVID-19. The curves showing the ratio of daily cumulative cases and deaths to those of the previous day tended to flatten with time by approaching the zero growth but without reaching it, which documents a persisting circulation of severe acute respiratory syndrome coronavirus 2 in the Italian territory.


Assuntos
COVID-19/epidemiologia , Epidemias/prevenção & controle , Humanos , Itália/epidemiologia , SARS-CoV-2/patogenicidade
5.
FASEB J ; 34(10): 13185-13193, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32780893

RESUMO

COVID-19 may cause pneumonia, acute respiratory distress syndrome, cardiovascular alterations, and multiple organ failure, which have been ascribed to a cytokine storm, a systemic inflammatory response, and an attack by the immune system. Moreover, an oxidative stress imbalance has been demonstrated to occur in COVID-19 patients. N- Acetyl-L-cysteine (NAC) is a precursor of reduced glutathione (GSH). Due to its tolerability, this pleiotropic drug has been proposed not only as a mucolytic agent, but also as a preventive/therapeutic agent in a variety of disorders involving GSH depletion and oxidative stress. At very high doses, NAC is also used as an antidote against paracetamol intoxication. Thiols block the angiotensin-converting enzyme 2 thereby hampering penetration of SARS-CoV-2 into cells. Based on a broad range of antioxidant and anti-inflammatory mechanisms, which are herein reviewed, the oral administration of NAC is likely to attenuate the risk of developing COVID-19, as it was previously demonstrated for influenza and influenza-like illnesses. Moreover, high-dose intravenous NAC may be expected to play an adjuvant role in the treatment of severe COVID-19 cases and in the control of its lethal complications, also including pulmonary and cardiovascular adverse events.


Assuntos
Acetilcisteína/uso terapêutico , Antioxidantes/uso terapêutico , Tratamento Farmacológico da COVID-19 , COVID-19 , SARS-CoV-2/metabolismo , Internalização do Vírus/efeitos dos fármacos , Enzima de Conversão de Angiotensina 2/antagonistas & inibidores , Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/metabolismo , COVID-19/patologia , Quimioterapia Adjuvante , Humanos , Estresse Oxidativo/efeitos dos fármacos
6.
Sci Rep ; 10(1): 11290, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32647312

RESUMO

Smoking-related lung tumors are characterized by profound epigenetic changes including scrambled patterns of DNA methylation, deregulated histone acetylation, altered gene expression levels, distorted microRNA profiles, and a global loss of cytosine hydroxymethylation marks. Here, we employed an enhanced version of bisulfite sequencing (RRBS/oxRRBS) followed by next generation sequencing to separately map DNA epigenetic marks 5-methyl-dC and 5-hydroxymethyl-dC in genomic DNA isolated from lungs of A/J mice exposed whole-body to environmental cigarette smoke for 10 weeks. Exposure to cigarette smoke significantly affected the patterns of cytosine methylation and hydroxymethylation in the lungs. Differentially hydroxymethylated regions were associated with inflammatory response/disease, organismal injury, and respiratory diseases and were involved in regulation of cellular development, function, growth, and proliferation. To identify epigenetic changes in the lung associated with exposure to tobacco carcinogens and inflammation, A/J mice were intranasally treated with the tobacco carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), the inflammatory agent lipopolysaccharide (LPS), or both. NNK alone caused minimal epigenetic alterations, while exposure either to LPS or NNK/LPS in combination led to increased levels of global cytosine methylation and formylation, reduced cytosine hydroxymethylation, decreased histone acetylation, and altered expression levels of multiple genes. Our results suggest that inflammatory processes are responsible for epigenetic changes contributing to lung cancer development.


Assuntos
Epigênese Genética , Exposição por Inalação , Neoplasias Pulmonares/genética , Pulmão/efeitos dos fármacos , Fumaça/efeitos adversos , Animais , Carcinógenos/metabolismo , Proliferação de Células , Cromatografia Líquida de Alta Pressão , Ilhas de CpG , Citosina/química , DNA/metabolismo , Metilação de DNA , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Histonas/química , Histonas/metabolismo , Inflamação , Camundongos , Camundongos Endogâmicos , Nitrosaminas/metabolismo , Fumar , Sulfitos/farmacologia , Nicotiana , Produtos do Tabaco
7.
J Med Virol ; 92(10): 1956-1961, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32314804

RESUMO

The coronavirus disease 2019 epidemic started in Italy by the end of January 2020 and, after 1 month, it affected 1049 persons. Based on the Italian Ministry of Health data, we reconstructed the daily course of virus-positive cases and deaths over March 2020 for the whole of Italy, 19 regions and 2 provinces. From 29 February to 31 March, there was a 100.9-fold increase in the cumulative number of cases and a 428.6-fold increase in the number of deaths in Italy. When plotted on a semilogarithmic scale, the curves tended to diverge from linearity with 23%, 16%, and 7% average daily increases during the three decades of March. Similarly, the number of deaths decreased from an average daily growth of 19% over the second decade to 10% over the third decade. The correlation coefficients relating the days to cases or deaths over each one of the three decades approached unity. As inferred from the equations of the regression lines relative to the three decades, the doubling times of cases were 3.4, 5.1, and 9.6 days, respectively. The doubling times of deaths over the second and third decades were 4.9 and 7.0 days, respectively. There was a broad geographic variability, with a striking gradient from the North, where 40.8% of cases and 57.9% of deaths occurred in Lombardy, to the South. On the whole, over March there was a trend to epidemic growth decline but the time for the end of the epidemic will depend on a variety of factors and, at present, it is unpredictable.


Assuntos
COVID-19/epidemiologia , COVID-19/mortalidade , Pandemias , Humanos , Itália/epidemiologia
8.
Carcinogenesis ; 41(1): 91-99, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-31562745

RESUMO

Chronic inflammation plays a crucial role in the carcinogenesis process and, in particular, in smoking-related carcinogenesis. Therefore, anti-inflammatory agents provide an interesting perspective in the prevention of smoking-associated cancers. Among nonsteroidal anti-inflammatory drugs (NSAIDs), licofelone is a triple inhibitor of both cyclooxygenases (COX-1 and COX-2) and of 5-lipooxygenase (5-LOX) that has shown some encouraging results in cancer prevention models. We previously showed that the dietary administration of licofelone, starting after weanling, to Swiss H mice exposed for 4 months to mainstream cigarette smoke since birth attenuated preneoplastic lesions of inflammatory nature in both lung and urinary tract, and had some effects on the yield of lung tumors at 7.5 months of age. The present study aimed at evaluating the early modulation by licofelone of pulmonary DNA and RNA alterations either in smoke-free or smoke-exposed H mice after 10 weeks of exposure. Licofelone protected the mice from the smoke-induced loss of body weight and significantly attenuated smoke-induced nucleotide alterations by decreasing the levels of bulky DNA adducts and 8-hydroxy-2'-deoxyguanosine in mouse lung. Moreover, the drug counteracted dysregulation by smoke of several pulmonary microRNAs involved in stress response, inflammation, apoptosis, and oncogene suppression. However, even in smoke-free mice administration of the drug had significant effects on a broad panel of microRNAs and, as assessed in a subset of mice used in a parallel cancer chemoprevention study, licofelone even enhanced the smoke-induced systemic genotoxic damage after 4 months of exposure. Therefore, caution should be paid when administering licofelone to smokers for long periods.


Assuntos
Anticarcinógenos/administração & dosagem , Dano ao DNA/efeitos dos fármacos , Inflamação/tratamento farmacológico , Neoplasias Pulmonares/prevenção & controle , Pirróis/administração & dosagem , Poluição por Fumaça de Tabaco/efeitos adversos , Animais , Anticarcinógenos/toxicidade , Araquidonato 5-Lipoxigenase/metabolismo , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Adutos de DNA/imunologia , Adutos de DNA/metabolismo , Modelos Animais de Doenças , Esquema de Medicação , Feminino , Humanos , Inflamação/etiologia , Inflamação/imunologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Camundongos , MicroRNAs/imunologia , MicroRNAs/metabolismo , Pirróis/toxicidade , Fatores de Tempo , Testes de Toxicidade Subcrônica
9.
Oncotarget ; 9(72): 33656-33681, 2018 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-30263093

RESUMO

Celecoxib, a nonsteroidal anti-inflammatory drug that selectively targets cyclooxygenase-2, is a promising cancer chemopreventive agent. However, safety concerns have been raised in clinical trials evaluating its ability to prevent colorectal adenomas. The rationale for the herein reported studies was to analyze genomic and epigenetic end-points aimed at investigating both the chemopreventive properties of celecoxib towards cigarette smoke-associated molecular alterations and its possible adverse effects. We carried out three consecutive studies in mice treated with either smoke and/or celecoxib. Study 1 investigated early DNA alterations (DNA adducts, oxidative DNA damage, and systemic genotoxic damage) and epigenetic alterations (expression of 1,135 microRNAs) in lung and blood of Swiss H mice; Study 2 evaluated the formation of DNA adducts in lung, liver, and heart; and Study 3 evaluated the expression of microRNAs in 10 organs and 3 body fluids of ICR (CD-1) mice. Surprisingly, the oral administration of celecoxib to smoke-free mice resulted in the formation of DNA adducts in both lung and heart and in dysregulation of microRNAs in mouse organs and body fluids. On the other hand, celecoxib attenuated smoke-related DNA damage and dysregulation of microRNA expression. In conclusion, celecoxib showed pleiotropic properties and multiple mechanisms by counteracting the molecular damage produced by smoke in a variety of organs and body fluids. However, administration of celecoxib to non-smoking mice resulted in evident molecular alterations, also including DNA and RNA alterations in the heart, which may bear relevance in the pathogenesis of the cardiovascular adverse effects of this drug.

10.
Theranostics ; 8(8): 2147-2160, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29721069

RESUMO

Purpose: MicroRNAs are small non-coding RNAs that regulate gene expression, thereby playing a role in a variety of physiological and pathophysiological states. Exposure to cigarette smoke extensively downregulates microRNA expression in pulmonary cells of mice, rats, and humans. Cellular microRNAs are released into body fluids, but a poor parallelism was previously observed between lung microRNAs and circulating microRNAs. The purpose of the present study was to validate the application of this epigenetic biomarker by using less invasive collection procedures. Experimental design: Using microarray analyses, we measured 1135 microRNAs in 10 organs and 3 body fluids of mice that were either unexposed or exposed to mainstream cigarette smoke for up to 8 weeks. The results obtained with selected miRNAs were validated by qPCR. Results: The lung was the main target affected by smoke (190 dysregulated miRNAs), followed by skeletal muscle (180), liver (138), blood serum (109), kidney (96), spleen (89), stomach (36), heart (33), bronchoalveolar lavage fluid (32), urine (27), urinary bladder (12), colon (5), and brain (0). Skeletal muscle, kidney, and lung were the most important sources of smoke-altered microRNAs in blood serum, urine, and bronchoalveolar lavage fluid, respectively. Conclusions: microRNA expression analysis was able to identify target organs after just 8 weeks of exposure to smoke, well before the occurrence of any detectable histopathological alteration. The present translational study validates the use of body fluid microRNAs as biomarkers applicable to human biomonitoring for mechanistic studies, diagnostic purposes, preventive medicine, and therapeutic strategies.


Assuntos
Líquidos Corporais/metabolismo , MicroRNAs/metabolismo , Especificidade de Órgãos , Fumar/efeitos adversos , Animais , Peso Corporal , Análise por Conglomerados , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos Endogâmicos ICR , MicroRNAs/genética , Análise de Componente Principal , RNA/isolamento & purificação , Reprodutibilidade dos Testes , Fatores de Tempo
11.
Carcinogenesis ; 39(8): 1037-1044, 2018 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-29788174

RESUMO

Epidemiological studies show that there is limited evidence that tobacco smoking causes breast cancer in humans. In rodents, many tobacco smoke chemicals cause mammary gland tumors. This study evaluated the mammary gland differentiation in mice exposed to environmental cigarette smoke (ECS), using 3R4F Kentucky reference cigarettes, starting after birth and continuing daily for 10 weeks (total particulate exposure 95 mg/m3; CO 610 ppm). We also analyzed the effects of oral administration of non-steroidal anti-inflammatory drugs (NSAIDs), aspirin (1600 mg/kg) or naproxen (320 mg/kg), on mammary gland differentiation, either in unexposed or ECS-exposed mice. The ECS exposure caused delay of mammary glands development. We speculate that this delay may result from aryl hydrocarbon receptor (AHR) signaling activation, which has an antiestrogenic effect and crosstalk to the estrogen metabolism pathway. Similarly, naproxen impaired gland differentiation in unexposed and ECS-exposed mice, while aspirin hindered its development only in unexposed mice. The lack of differentiation induced by the NSAIDs could be explained by their antiestrogenic effect through inhibition of aldo-keto reductases. In ECS-exposed animals, aspirin induced intense lobular formation, which could indicate that aspirin is counteracting the AHR signaling induced by ECS. Based on the differentiation induced by aspirin in ECS-exposed animals, we postulate that these mice would be less susceptible to mammary carcinogenesis. Our results suggest that exposure to smoke at an early age impairs the development of the mammary gland, thus resulting in a longer period of susceptibility and increased risk of breast cancer. However, addition of aspirin can abrogate this effect.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Aspirina/administração & dosagem , Glândulas Mamárias Animais/efeitos dos fármacos , Neoplasias Mamárias Experimentais/prevenção & controle , Poluição por Fumaça de Tabaco/efeitos adversos , Administração Oral , Animais , Carcinogênese/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Suscetibilidade a Doenças/etiologia , Feminino , Masculino , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/etiologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Naproxeno/administração & dosagem , Receptores de Hidrocarboneto Arílico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fumaça/efeitos adversos , Nicotiana/efeitos adversos
12.
Carcinogenesis ; 39(4): 580-587, 2018 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-29370344

RESUMO

In spite of the outstanding role of tobacco smoking in human carcinogenesis, it is difficult to reproduce its effects in experimental animals. Based on the knowledge that a variety of mechanisms account for a higher susceptibility to carcinogens early in life, we have developed a murine model in which mainstream cigarette smoke becomes convincingly carcinogenic. The standard model involves exposure to smoke for 4 months, starting after birth, followed by an additional 3-4 months in filtered air. We evaluated herein the time- and dose-dependent response, at 7.5 months of life, of Swiss H mice that had been exposed to smoke for either 1, 2 or 4 months after birth. A one-month exposure, corresponding to a period of intense alveolarization, was sufficient to induce most inflammatory, degenerative and preneoplastic pulmonary lesions, including emphysema and alveolar epithelial hyperplasia, blood vessel proliferation and hemangiomas, reflecting an early proangiogenic role of smoking, and microadenomas bearing ki-67-positive proliferating cells as well as urinary bladder epithelial hyperplasia. Two months of exposure were needed to induce pulmonary adenomas and urinary bladder papillomas in males only, which highlights a protective role of estrogens in urinary bladder carcinogenesis. Four months, which in humans would correspond to the postnatal period, puberty, adolescence and early adulthood, were needed to induce other lesions, including tubular epithelial hyperplasia of kidney, bronchial epithelial hyperplasia and especially pulmonary malignant tumors. These findings highlight the concept that preneoplastic and neoplastic lesions occurring in adulthood can be induced by exposure to smoke early in life.


Assuntos
Carcinogênese/induzido quimicamente , Modelos Animais de Doenças , Neoplasias/etiologia , Nicotiana/efeitos adversos , Fumaça/efeitos adversos , Animais , Animais Recém-Nascidos , Feminino , Masculino , Camundongos , Lesões Pré-Cancerosas/etiologia , Fumar/efeitos adversos , Poluição por Fumaça de Tabaco/efeitos adversos
13.
Oncotarget ; 8(49): 85716-85748, 2017 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-29156752

RESUMO

We recently showed that nonsteroidal anti-inflammatory drugs (NSAIDs) are able to inhibit the lung tumors induced by cigarette smoke, either mainstream (MCS) or environmental (ECS), in female mice. We used subsets of mice to analyze the expression of 1135 microRNAs in both lung and blood serum, as related to the whole-body exposure to smoke and/or oral administration of either aspirin or naproxen. In a first study, we evaluated early microRNA alterations in A/J mice exposed to ECS for 10 weeks, starting at birth, and/or treated with NSAIDs for 6 weeks, starting after weaning. At that time, when no histopathological change were apparent, ECS caused a considerable downregulation of pulmonary microRNAs affecting both adaptive mechanisms and disease-related pathways. Aspirin and naproxen modulated, with intergender differences, the expression of microRNAs having a variety of functions, also including regulation of cyclooxygenases and inflammation. In a second study, we evaluated late microRNA alterations in Swiss H mice exposed to MCS during the first 4 months of life and treated with NSAIDs after weaning until 7.5 months of life, when tumors were detected in mouse lung. Modulation of pulmonary microRNAs by the two NSAIDs was correlated with their ability to prevent preneoplastic lesions (microadenomas) and adenomas in the lung. In both studies, exposure to smoke and/or treatment with NSAIDs also modulated microRNA profiles in the blood serum. However, their levels were poorly correlated with those of pulmonary microRNAs, presumably because circulating microRNAs reflect the contributions from multiple organs and not only from lung.

14.
PLoS One ; 11(12): e0167239, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27907070

RESUMO

Cigarette smoke (CS) and ethanol (EtOH) are known to synergize in the causation of cancers of the upper aerodigestive tract and of the liver. Little is known about possible interactions between these agents in other organs. These premises prompted us to evaluate the clastogenic effects resulting from the inhalation for 3 weeks of mainstream CS and oral administration of EtOH, which were tested either individually or in combination in cells of adult BDF1 mice and their fetuses. CS exerted clastogenic effects in haematopoietic cells of adult male mice by increasing the frequency of micronucleated erythroid cells both in bone marrow and in peripheral blood as well as the frequency of micronucleated and polynucleated pulmonary alveolar macrophages. Likewise, exposure to CS of pregnant mice resulted in a clastogenic damage in maternal bone marrow cells and in the liver and peripheral blood of their fetuses. Under all experimental conditions, EtOH was consistently devoid of clastogenic effects when given alone. In adult mice, EtOH exhibited a mild stimulating effect on the clastogenicity of CS in haematopoietic cells, while an opposite effect was observed in the respiratory tract, where EtOH attenuated the cytogenetic alterations induced by CS in pulmonary alveolar macrophages. At variance with the mild synergism observed in haematopoietic cells of adult mice, EtOH inhibited the clastogenicity of CS in the liver and peripheral blood cells of transplacentally exposed fetuses. Therefore, the effects of EtOH in CS-exposed mice show different trends depending both on the life stage and on the cells analyzed.


Assuntos
Etanol/efeitos adversos , Feto/efeitos dos fármacos , Exposição Materna/efeitos adversos , Mutagênese/efeitos dos fármacos , Fumar/efeitos adversos , Animais , Células Sanguíneas/efeitos dos fármacos , Células Sanguíneas/metabolismo , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Aberrações Cromossômicas/induzido quimicamente , Feminino , Fígado/efeitos dos fármacos , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/metabolismo , Masculino , Camundongos , Gravidez , Fatores de Tempo
15.
Toxicology ; 373: 54-62, 2016 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-27840117

RESUMO

Both ethanol and cigarette smoke are classified as human carcinogens. They can synergize, especially in tissues of the upper aerodigestive tract that are targeted by both agents. The main objective of the present study was to evaluate the individual and combined effects of ethanol and smoke in the respiratory tract, either following transplacental exposure and/or postnatal exposure. We designed two consecutive studies in mouse models by exposing Swiss H mice to oral ethanol and/or inhaled mainstream cigarette smoke for up to 4 months, at various prenatal and postnatal life stages. Clastogenic effects and histopathological alterations were evaluated after 4 and 8 months, respectively. Ethanol was per se devoid of clastogenic effects in mouse peripheral blood erythrocytes. However, especially in mice exposed both transplacentally throughout pregnancy and in the postnatal life, ethanol administration was associated not only with liver damage but also with pro-angiogenetic effects in the lung by stimulating the proliferation of blood vessels. In addition, these mice developed pulmonary emphysema, alveolar epithelial hyperplasias, microadenomas, and benign tumors. On the other hand, ethanol interfered in the lung carcinogenesis process resulting from the concomitant exposure of mice to smoke. In fact, ethanol significantly attenuated some smoke-related preneoplastic and neoplastic lesions in the respiratory tract, such as alveolar epithelial hyperplasia, microadenomas, and even malignant tumors. In addition, ethanol attenuated cigarette smoke clastogenicity. In conclusion, preclinical studies provide evidence that, in spite of its pulmonary toxicity, ethanol may mitigate some noxious effects of cigarette smoke in the respiratory tract.


Assuntos
Carcinogênese/efeitos dos fármacos , Depressores do Sistema Nervoso Central/toxicidade , Etanol/toxicidade , Neoplasias Pulmonares/induzido quimicamente , Nicotiana , Fumaça/efeitos adversos , Poluição por Fumaça de Tabaco/efeitos adversos , Animais , Peso Corporal/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/patologia , Interações Medicamentosas , Feminino , Pneumopatias/induzido quimicamente , Pneumopatias/patologia , Masculino , Camundongos , Mutagênicos/toxicidade , Neovascularização Patológica/induzido quimicamente , Neovascularização Patológica/patologia , Gravidez , Análise de Sobrevida
16.
Oncotarget ; 7(51): 84758-84774, 2016 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-27713172

RESUMO

Cigarette smoke (CS) is known to dysregulate microRNA expression profiles in the lungs of mice, rats, and humans, thereby modulating several pathways involved in lung carcinogenesis and other CS-related diseases. We designed a study aimed at evaluating (a) the expression of 1135 microRNAs in the lung of Swiss H mice exposed to mainstream CS during the first 4 months of life and thereafter kept in filtered air for an additional 3.5 months, (b) the relationship between lung microRNA profiles and histopathological alterations in the lung, (c) intergender differences in microRNA expression, and (d) the comparison with microRNA profiles in blood serum. CS caused multiple histopathological alterations in the lung, which were almost absent in sham-exposed mice. An extensive microRNA dysregulation was detected in the lung of CS-exposed mice. Modulation of microRNA profiles was specifically related to the histopathological picture, no effect being detected in lung fragments with non-neoplastic lung diseases (emphysema or alveolar epithelial hyperplasia), whereas a close association occurred with the presence and multiplicity of preneoplastic lesions (microadenomas) and benign lung tumors (adenomas). Three microRNAs regulating estrogen and HER2-dependent mechanisms were modulated in the lung of adenoma-bearing female mice. Blood microRNAs were also modulated in mice affected by early neoplastic lesions. However, there was a poor association between lung microRNAs and circulating microRNAs, which can be ascribed to an impaired release of mature microRNAs from the damaged lung. Studies in progress are evaluating the feasibility of analyzing blood microRNAs as a molecular tool for lung cancer secondary prevention.


Assuntos
Adenoma/diagnóstico , Biomarcadores Tumorais/genética , Neoplasias Pulmonares/diagnóstico , Pulmão/fisiologia , MicroRNAs/genética , Adenoma/genética , Animais , Carcinogênese/genética , Fumar Cigarros/efeitos adversos , Estrogênios/metabolismo , Feminino , Humanos , Pulmão/patologia , Neoplasias Pulmonares/genética , Masculino , Camundongos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo
17.
Toxicol Appl Pharmacol ; 306: 113-9, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27404458

RESUMO

Evaluation of the reducing capacity of human gastric fluid from healthy individuals, under fasted and fed conditions, is critical for assessing the cancer hazard posed by ingested hexavalent chromium [Cr(VI)] and for developing quantitative physiologically-based pharmacokinetic models used in risk assessment. In the present study, the patterns of Cr(VI) reduction were evaluated in 16 paired pre- and post-meal gastric fluid samples collected from 8 healthy volunteers. Human gastric fluid was effective both in reducing Cr(VI), as measured by using the s-diphenylcarbazide colorimetric method, and in attenuating mutagenicity in the Ames test. The mean (±SE) Cr(VI)-reducing ability of post-meal samples (20.4±2.6µgCr(VI)/mL gastric fluid) was significantly higher than that of pre-meal samples (10.2±2.3µgCr(VI)/mL gastric fluid). When using the mutagenicity assay, the decrease of mutagenicity produced by pre-meal and post-meal samples corresponded to reduction of 13.3±1.9 and 25.6±2.8µgCr(VI)/mL gastric fluid, respectively. These data are comparable to parallel results conducted by using speciated isotope dilution mass spectrometry. Cr(VI) reduction was rapid, with >70% of total reduction occurring within 1min and 98% of reduction is achieved within 30min with post-meal gastric fluid at pH2.0. pH dependence was observed with decreasing Cr(VI) reducing capacity at higher pH. Attenuation of the mutagenic response is consistent with the lack of DNA damage observed in the gastrointestinal tract of rodents following administration of ≤180ppm Cr(VI) for up to 90days in drinking water. Quantifying Cr(VI) reduction kinetics in the human gastrointestinal tract is necessary for assessing the potential hazards posed by Cr(VI) in drinking water.


Assuntos
Cromo/química , Suco Gástrico/química , Poluentes Químicos da Água/química , Adulto , Cromo/toxicidade , Jejum , Histidina/genética , Humanos , Concentração de Íons de Hidrogênio , Mutagênese , Testes de Mutagenicidade , Oxirredução , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/genética , Poluentes Químicos da Água/toxicidade
18.
Toxicol Appl Pharmacol ; 306: 120-33, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27396814

RESUMO

To extend previous models of hexavalent chromium [Cr(VI)] reduction by gastric fluid (GF), ex vivo experiments were conducted to address data gaps and limitations identified with respect to (1) GF dilution in the model; (2) reduction of Cr(VI) in fed human GF samples; (3) the number of Cr(VI) reduction pools present in human GF under fed, fasted, and proton pump inhibitor (PPI)-use conditions; and (4) an appropriate form for the pH-dependence of Cr(VI) reduction rate constants. Rates and capacities of Cr(VI) reduction were characterized in gastric contents from fed and fasted volunteers, and from fasted pre-operative patients treated with PPIs. Reduction capacities were first estimated over a 4-h reduction period. Once reduction capacity was established, a dual-spike approach was used in speciated isotope dilution mass spectrometry analyses to characterize the concentration-dependence of the 2nd order reduction rate constants. These data, when combined with previously collected data, were well described by a three-pool model (pool 1 = fast reaction with low capacity; pool 2 = slow reaction with higher capacity; pool 3 = very slow reaction with higher capacity) using pH-dependent rate constants characterized by a piecewise, log-linear relationship. These data indicate that human gastric samples, like those collected from rats and mice, contain multiple pools of reducing agents, and low concentrations of Cr(VI) (<0.7 mg/L) are reduced more rapidly than high concentrations. The data and revised modeling results herein provide improved characterization of Cr(VI) gastric reduction kinetics, critical for Cr(VI) pharmacokinetic modeling and human health risk assessment.


Assuntos
Cromo/química , Suco Gástrico/química , Modelos Biológicos , Poluentes Químicos da Água/química , Jejum , Humanos , Oxirredução
19.
Trends Pharmacol Sci ; 37(2): 120-142, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26726119

RESUMO

Many drugs in common use possess pleiotropic properties that make them capable of interfering with carcinogenesis mechanisms. We discuss here the ability of pharmacological agents to mitigate the pulmonary carcinogenicity of mainstream cigarette smoke. The evaluated agents include anti-inflammatory drugs (budesonide, celecoxib, aspirin, naproxen, licofelone), antidiabetic drugs (metformin, pioglitazone), antineoplastic agents (lapatinib, bexarotene, vorinostat), and other drugs and supplements (phenethyl isothiocyanate, myo-inositol, N-acetylcysteine, ascorbic acid, berry extracts). These drugs have been evaluated in mouse models mimicking interventions either in current smokers or in ex-smokers, or in prenatal chemoprevention. They display a broad spectrum of activities by attenuating either smoke-induced preneoplastic lesions or benign tumors and/or malignant tumors. Together with epidemiological data, these findings provide useful information to predict the potential effects of pharmacological agents in smokers.


Assuntos
Anticarcinógenos/farmacologia , Carcinogênese/efeitos dos fármacos , Neoplasias Pulmonares/epidemiologia , Neoplasias Pulmonares/prevenção & controle , Fumar/epidemiologia , Animais , Anticarcinógenos/administração & dosagem , Modelos Animais de Doenças , Humanos , Abandono do Hábito de Fumar , Prevenção do Hábito de Fumar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...