Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Ukr Biokhim Zh (1999) ; 79(5): 72-90, 2007.
Artigo em Russo | MEDLINE | ID: mdl-18357780

RESUMO

Data are presented concerning the basic metabolism sites, the reaction paths crossing in them and regulatory and toxical effect of formaldehyde and nitric oxide being mediated through them. In particular, they include: glutathione-formaldehyde-dependent dehydrogenase path of S-nitrosoglutathione reduction, semi-carbaside-sensitive amino-oxidase (SSAO) and NO-synthase systems; transformation of thioproline and metallothioneines, including nitrosation reactions. Possibilities of hexamethylenetetramine synthesis in the organism as well as its metabolism in conditions of formaldehyde hyperproduction and nitrosative stress are discussed. The role of metabolism sites, common for formaldehyde and nitrogen oxide, in the mechanisms of toxical effect of these compounds and development of pathologic states is considered.


Assuntos
Formaldeído , Óxido Nítrico , Animais , Formaldeído/metabolismo , Formaldeído/toxicidade , Humanos , Redes e Vias Metabólicas , Óxido Nítrico/metabolismo , Óxido Nítrico/toxicidade
2.
Ukr Biokhim Zh (1999) ; 77(1): 22-31, 2005.
Artigo em Russo | MEDLINE | ID: mdl-16335265

RESUMO

Possible exogenous sources of formaldehyde and nitric oxide have been considered; the environment pollution conditions under which these compounds and their precursors have mutual effect on the organism; endogenous sources of FA and NO which are intermediates of the metabolism and key enzymes of their transformation (semicarbazide-sensitive amine oxidase and NO-synthase) the role of the C1 metabolic cycle pathways and methyl cycles in the FA formation and accumulation have been considered as well, various paths of FA toxic action have been characterized.


Assuntos
Formaldeído/metabolismo , Formaldeído/toxicidade , Óxido Nítrico/metabolismo , Óxido Nítrico/toxicidade , Xenobióticos/metabolismo , Xenobióticos/toxicidade , Animais , Formaldeído/farmacocinética , Humanos , Inativação Metabólica , Óxido Nítrico/farmacocinética , Xenobióticos/farmacocinética
3.
Ukr Biokhim Zh (1999) ; 77(5): 5-23, 2005.
Artigo em Russo | MEDLINE | ID: mdl-16846066

RESUMO

Toxic properties of NO in organism are realized under its hyperproduction and inhibition of the system of anti-oxidant protection as a result of complex chemical transformations, the transient metals, oxygen, superoxide and other radicals being their main participant. Here direct paths (through formation of nitrosil complexes with the gem and nongem iron) of the toxic action of NO and the path mediated by active forms of nitrogen are found, which disturb various biomolecules and subcellular component through the reactions of S- and N-nitrozation, nitration, oxidation, desamination and other reaction, cause metabolic disbalance and death of cells by the type of apoptosis or necrosis. A possible mechanism of the death of cells caused by NO was considered on the example of thymocytes. According to this mechanism one of early stages of this death is a decrease of the cell fund of AP, intensification of catabolism of adenine nucleotides and transformation of xanthine oxidoreductase from D-form (xanthine dehydrogenase) of O-forms (xantine oxidase) which catalizes formation of cytotoxic molecules of superoxide and hydroperoxide. This cytotoxic mechanism which includes transformation of xanthine oxidase system, is probably, universal and does not depend essentially on the starting factor.


Assuntos
Apoptose/efeitos dos fármacos , Formaldeído/metabolismo , Óxido Nítrico/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Antioxidantes/metabolismo , Humanos , Ferro/metabolismo , Necrose , Doadores de Óxido Nítrico/farmacologia , Óxidos de Nitrogênio/metabolismo , Ácido Peroxinitroso/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Nitrito de Sódio/farmacologia
4.
Clin Exp Immunol ; 131(3): 415-21, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12605693

RESUMO

The genetic backgrounds of lupus-prone murine models are a valuable resource for studying the influence of environmental exposure on autoimmune diseases in sensitive populations. Epidemiological studies have shown associations between silica exposure and several autoimmune diseases, including scleroderma and systemic lupus erythematosus. To determine whether silica exposure can exacerbate systemic autoimmunity in genetically predisposed animals, New Zealand mixed mice were intranasally instilled twice with saline or saline suspensions of 1 mg silica or 500 micro g TiO2, a dose equivalent in surface area, and were evaluated with respect to health and immune status. Survival in silica exposed NZM mice was decreased compared to saline and TiO2 exposed mice. Proteinuria levels were elevated in silica exposed mice. Levels of circulating immune complexes, autoantibodies to nuclear antigen (ANA), histone, and double stranded DNA were measured every two weeks by ELISA. Circulating immune complexes showed a trend towards an increased acceleration in levels in the silica exposed mice compared to saline and TiO2 exposed mice. ANA levels were significantly higher in silica exposed animals compared to saline and TiO2 exposed animals (0.237 +/- 0.03 versus 0.140 +/- 0.029 and 0.125 +/- 0.03, P < 0.05) 16 weeks postexposure. Autoantibodies to histone were also significantly elevated after 16 weeks in silica exposed animals compared to saline and TiO2 exposed animals (0.227 +/- 0.03 versus 0.073 +/- 0.015 and 0.05 +/- 0.03, P < 0.05). In contrast, serum IgG levels were decreased in silica exposed NZM mice compared to the saline controls, however, IgM levels were unaffected. Lungs of the silica-exposed mice had increased inflammatory infiltrates as well as fibrotic lesions characterized by excess collagen deposition. Therefore, although NZM mice are susceptible to SLE, silica exposure significantly exacerbated the course of disease.


Assuntos
Doenças Autoimunes/etiologia , Predisposição Genética para Doença , Lúpus Eritematoso Sistêmico/etiologia , Dióxido de Silício/imunologia , Animais , Complexo Antígeno-Anticorpo/sangue , Autoanticorpos/biossíntese , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Feminino , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Glomérulos Renais/imunologia , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Masculino , Camundongos , Camundongos Endogâmicos NZB , Proteinúria/induzido quimicamente , Fibrose Pulmonar/etiologia , Fibrose Pulmonar/patologia , Silicose/patologia , Taxa de Sobrevida
5.
J Environ Pathol Toxicol Oncol ; 20 Suppl 1: 75-84, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11570676

RESUMO

Some inhaled particles are known to lead to inflammation and lung pathology, whereas others do not appear to have long-term effects. Potential mechanisms to account for these differences are only beginning to be understood. In this article we examine whether silica and PM1648 (a model urban particulate) caused selective deletion of the suppressor human alveolar macrophage (HAM) phenotype (RFD1+/7+), and whether this affected cytokine production in an antigen-presenting cell (APC) assay with autologous T lymphocytes. HAM were exposed to the bioactive particulates, silica and PM1648, for 24 hours, then isolated free of extracellular particulates and nonviable cells; HAM were then cultured with autologous lymphocytes in an 11-day APC assay. Silica exposure up-regulated a TH1 lymphocyte-derived cytokine, interferon gamma (IFN-gamma), and a TH2 lymphocyte-derived cytokine, interleukin-4 (IL-4). PM1648 exposure primarily upregulated IL-4. Neither particle exposure had a significant effect on interleukin-10 (IL-10) production. Control particulate exposures with titanium dioxide (TiO2) and wollastonite (Woll) caused no altered APC activity. Silica and PM1648 demonstrated selective toxicity to suppressor macrophages (RFD1+/7+). We propose that, because of the suppressor macrophage phenotype disabling, the activator macrophage (RFD1+/7-) operates free of the suppressor macrophage's influence, enhancing APC activity with increased lymphocyte-derived proinflammatory cytokine production.


Assuntos
Poluentes Ocupacionais do Ar/toxicidade , Células Apresentadoras de Antígenos/imunologia , Compostos de Cálcio/toxicidade , Pulmão/imunologia , Macrófagos Alveolares/imunologia , Silicatos/toxicidade , Dióxido de Silício/toxicidade , Titânio/toxicidade , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Cultivadas , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Interferon gama/metabolismo , Interleucina-4/metabolismo , Pulmão/efeitos dos fármacos , Macrófagos Alveolares/efeitos dos fármacos
6.
Toxicol Appl Pharmacol ; 174(1): 10-6, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11437644

RESUMO

Scavenger receptors (SR) are responsible for recognition of ligands as diverse as oxidized LDL (endogenous) to respirable particulates (exogenous). A number of recent studies have suggested that these SR ligands induce apoptosis of macrophages. However, the mechanism by which SR triggers apoptosis is not understood. This study used a murine alveolar macrophage cell line (MH-S) to investigate the role of the SR in caspase activation. The presence of SR on MH-S cells was confirmed by FACS analysis and was similar to the distribution found on murine alveolar macrophages. The activity of caspases 1, 3, and 6 was measured following a 6-h exposure to crystalline silica with and without blockers of the SR. Caspase activities were determined by hydrolysis of specific chromogenic substrates and formation of an active enzymatic form (Western for active caspase 3). Silica stimulated significant caspase activity, apoptosis, and necrosis of MH-S cells, which was attenuated by 2F8 (a blocking antibody) and polyinosinic acid (a nonspecific SR antagonist). The results indicate that the SR are necessary for caspase activation and subsequent apoptosis (as well as necrosis) caused by silica in macrophage cells.


Assuntos
Apoptose/efeitos dos fármacos , Macrófagos Alveolares/citologia , Proteínas de Membrana , Receptores Imunológicos/fisiologia , Receptores de Lipoproteínas , Dióxido de Silício/farmacologia , Animais , Caspase 3 , Caspases/metabolismo , Linhagem Celular , Membrana Celular/química , Fragmentação do DNA , Ativação Enzimática , Citometria de Fluxo , Macrófagos Alveolares/química , Macrófagos Alveolares/efeitos dos fármacos , Camundongos , Necrose , Receptores Imunológicos/análise , Receptores Depuradores , Receptores Depuradores Classe A , Receptores Depuradores Classe B
7.
J Exp Med ; 192(6): 857-69, 2000 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-10993916

RESUMO

Chronic inflammation leading to pulmonary fibrosis develops in response to environmental pollutants, radiotherapy, or certain cancer chemotherapeutic agents. We speculated that lung injury might be mediated by p53, a proapoptotic transcription factor widely implicated in the response of cells to DNA damage. Intratracheal administration of bleomycin led to caspase-mediated DNA fragmentation characteristic of apoptosis. The effects of bleomycin were associated with translocation of p53 from the cytosol to the nucleus only in alveolar macrophages that had been exposed to the drug in vivo, suggesting that the lung microenvironment regulated p53 activation. Experiments with a thiol antioxidant (N-acetylcysteine) in vivo and nitric oxide (NO) donors in vitro confirmed that reactive oxygen species were required for p53 activation. A specific role for NO was demonstrated in experiments with inducible nitric oxide synthase (iNOS)(-/)- macrophages, which failed to demonstrate nuclear p53 localization after in vivo bleomycin exposure. Strikingly, rates of bleomycin-induced apoptosis were at least twofold higher in p53(-/)- C57BL/6 mice compared with heterozygous or wild-type littermates. Similarly, levels of apoptosis were also twofold higher in the lungs of iNOS(-/)- mice than were observed in wild-type controls. Consistent with a role for apoptosis in chronic lung injury, levels of bleomycin-induced inflammation were substantially higher in iNOS(-/)- and p53(-/)- mice compared with wild-type controls. Together, our results demonstrate that iNOS and p53 mediate a novel apoptosis-suppressing pathway in the lung.


Assuntos
Apoptose/fisiologia , Bleomicina/toxicidade , Pulmão/patologia , Macrófagos Alveolares/fisiologia , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Acetilcisteína/farmacologia , Animais , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Fragmentação do DNA , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Pulmão/efeitos dos fármacos , Macrófagos Alveolares/citologia , Macrófagos Alveolares/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase/deficiência , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Espécies Reativas de Oxigênio/fisiologia , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
9.
J Rheumatol ; 27(2): 405-10, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10685806

RESUMO

OBJECTIVE: Autoantibodies to the U3 nucleolar ribonucleoprotein (RNP) fibrillarin occur in some patients with systemic sclerosis (SSc) or other connective tissue diseases and can be induced in certain mouse strains by injections of mercuric chloride, perhaps due to antigenic alteration of fibrillarin by mercury (Hg). Thus, potential occult exposure to Hg was explored in patients with SSc. METHODS: Urinary Hg levels were measured by cold vapor atomic absorption in 13 patients with antifibrillarin antibodies (11 with SSc), 39 SSc patients without antifibrillarin antibodies, and 32 healthy controls. RESULTS: Mean urinary Hg levels were significantly elevated in the antifibrillarin antibody positive patients compared to those in other patients with SSc and controls. After correction for urinary creatinine levels, mean urinary Hg levels remained significantly different than in the other 2 groups, although Hg levels in all were still within the normal or "unexposed" range. When patients and controls with low urinary creatinine levels were excluded from analysis, there were no significant differences in mean urinary Hg levels among the 3 groups. CONCLUSION: These findings suggest that further epidemiological and basic research studies of mercury are warranted in patients with SSc, especially those expressing antifibrillarin antibodies.


Assuntos
Autoanticorpos/imunologia , Proteínas Cromossômicas não Histona/imunologia , Mercúrio/urina , Escleroderma Sistêmico/imunologia , Escleroderma Sistêmico/urina , Animais , Humanos , Camundongos , Pessoa de Meia-Idade
10.
Toxicol Appl Pharmacol ; 162(2): 100-6, 2000 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-10637133

RESUMO

Macrophage scavenger receptors are known to bind endotoxins, oxidized low-density lipoproteins (Ox-LDL), and other proteins with clustered negative charges. Recent evidence indicates some particulates may also bind to the scavenger receptor and initiate apoptosis. In this study, chinese hamster ovary (CHO) cells stabily transfected with the murine class A type II scavenger receptor (SR-A II) were exposed to crystalline silica to examine the role of this receptor in apoptosis. In a 24-h culture, silica (250 microg/ml) induced significant cell injury (necrosis and apoptosis) in transfected cells (MSR II) but not in the control cells (KA-7). This effect was specific to silica, as a control particle titanium dioxide had no cytotoxic effects on the MSR II cells at equal particle mass concentrations. Furthermore, silica-induced apoptosis in the MSR II cells could be eliminated by preincubating the cells with SR-A II antagonists: polyinosinic acid or maleylated bovine serum albumin. This study further supports the hypothesis that the SR-A II is directly involved with silica toxicity and that certain scavenger receptor ligands may have an important role in regulating macrophage apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Receptores Imunológicos/fisiologia , Dióxido de Silício/toxicidade , Animais , Células CHO/efeitos dos fármacos , Células CHO/patologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Necrose , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/biossíntese , Receptores Imunológicos/genética , Receptores Depuradores , Receptores Depuradores Classe A , Titânio/toxicidade , Transfecção
11.
Inhal Toxicol ; 12(12): 1225-38, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11203434

RESUMO

Ozone is a ubiquitous air pollutant that can cause acute pulmonary inflammation and cell injury and may contribute to the exacerbation of chronic pulmonary diseases. The molecular mechanisms of ozone-induced cell injury, as well as protective mechanisms against ozone-injury, are not well understood. Since ozone is a reactive oxidant, and heme oxygenase-1 (HO-1) is an antioxidant enzyme induced by many oxidative stimuli, we hypothesized that HO-1 is one of the protective mechanisms against ozone-induced cell injury, as well as pulmonary inflammation. In the current study, C57Bl/6 mice were pretreated with a low level of endotoxin (lipopolysaccharide, LPS) (0.5 mg/kg) to induce HO-1, and 16 h later were exposed to 1 ppm ozone for 3 h. Endotoxin pretreatment caused a significant protection against ozone-induced pulmonary inflammation and cell injury in bronchoalveolar lavage (BAL) cells. The protection by endotoxin pretreatment against ozone-induced inflammation and necrosis in BAL cells was abolished by the cotreatment with a heme oxygenase inhibitor, tin protoporphyrin IX dichloride (SnPP), suggesting that HO-1 is responsible for the protection against ozone-induced pulmonary inflammation and BAL cell necrosis. Therefore, since HO-1 is induced following ozone exposure, HO-1 may contribute to the development of cellular adaptation to chronic ozone exposure.


Assuntos
Apoptose/efeitos dos fármacos , Heme Oxigenase (Desciclizante)/fisiologia , Inflamação/induzido quimicamente , Lipopolissacarídeos/farmacologia , Pulmão/efeitos dos fármacos , Ozônio/toxicidade , Animais , Líquido da Lavagem Broncoalveolar/citologia , Sobrevivência Celular/efeitos dos fármacos , Heme Oxigenase-1 , Pulmão/patologia , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL
12.
Am J Physiol ; 277(3): L550-7, 1999 09.
Artigo em Inglês | MEDLINE | ID: mdl-10484462

RESUMO

Acrolein is an environmental pollutant that is known to suppress respiratory host defense against infections; however, the mechanism of the decrease in host defense is not yet clear. We have previously reported that acrolein inhibited endotoxin-induced cytokine release and induced apoptosis in human alveolar macrophages, suggesting that the inhibition of cytokine release and/or cytotoxicity to alveolar macrophages may, in part, be responsible for acrolein-induced immunosuppression in the lung. Because nuclear factor-kappaB (NF-kappaB) is an important transcription factor for a number of cytokine genes and is also an important regulator of apoptosis, the effect of acrolein on NF-kappaB activity was examined by electrophoresis mobility shift assay. Acrolein caused a dose-dependent inhibition of endotoxin-induced NF-kappaB activation as well as an inhibition of basal level NF-kappaB activity. Because IkappaB is a principal regulator of NF-kappaB activity in the nucleus, changes in IkappaB were determined by Western blotting. Acrolein-inhibited IkappaB phosphorylation leads to an increase in cellular IkappaB levels preventing NF-kappaB nuclear translocation and is likely the mechanism of acrolein-induced inhibition of NF-kappaB activity. The role of basal level NF-kappaB in acrolein-induced apoptosis was also examined. An NF-kappaB inhibitor (MG-132) also induced apoptosis in human alveolar macrophages, suggesting that a certain basal level NF-kappaB activity may be required for macrophage cell survival. Taken together, our results suggest that the acrolein-inhibited endotoxin-induced NF-kappaB activation decreased the basal level NF-kappaB activity, which may be responsible for the inhibition of cytokine release and the induction of apoptosis in human alveolar macrophages.


Assuntos
Acroleína/farmacologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/metabolismo , NF-kappa B/metabolismo , Sobrevivência Celular/fisiologia , Células Cultivadas , Humanos , Proteínas I-kappa B/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos Alveolares/fisiologia , NF-kappa B/antagonistas & inibidores , Fosforilação/efeitos dos fármacos
13.
Am J Physiol ; 275(6): L1208-18, 1998 12.
Artigo em Inglês | MEDLINE | ID: mdl-9843859

RESUMO

Apoptosis is considered to be a protective mechanism that limits lung injury. However, apoptosis might contribute to the inflammatory burden present in the injured lung. The exposure of mice to bleomycin (BLM) is a well-established model for the study of lung injury. BLM exposure induces DNA damage and enhances tumor necrosis factor (TNF)-alpha expression in the lung. To evaluate the importance of alveolar macrophage (AM) apoptosis in the pathogenesis of lung injury, we exposed BLM-sensitive (C57BL/6) and BLM-resistant (BALB/c) mice to BLM (120 mg/kg) and studied the induction of apoptosis [by light-microscopy changes (2, 8, 12, 24, 48, and 72 h) and annexin V uptake by flow cytometry (24 h)], the secretion of TNF-alpha (measured by ELISA), and the expression of p53 (by immunoblotting) in AM retrieved from these mice. BLM, but not vehicle, induced apoptosis in AM from both murine strains. The numbers of apoptotic AM were significantly greater (P < 0.001) in C57BL/6 mice (52.9%) compared with BALB/c mice (40.8%) as demonstrated by annexin V uptake. BLM induction of apoptosis in AM was preceded by an increased secretion of TNF-alpha in C57BL/6 but not in BALB/c mice. Furthermore, double TNF-alpha receptor-deficient mice, developed on a C57BL/6 background, demonstrated significantly (P < 0.001) lower numbers of apoptotic AM compared with C57BL/6 and BALB/c mice. BLM also enhanced p53 expression in AM from both murine strains. However, p53-deficient mice developed BLM-induced lung injury, exhibited similar lung cell proliferation (measured as proliferating cell nuclear antigen immunostaining), and accumulated similar amounts of lung hydroxyproline (65 +/- 6.9 microgram/lung) as did C57BL/6 (62 +/- 6.5 microgram/lung) mice. Therefore, AM apoptosis is occurring during BLM-induced lung injury in a manner that correlates with murine strain sensitivity to BLM. Furthermore, TNF-alpha secretion rather than p53 expression contributes to the difference in murine strain response to BLM.tumor necrosis factor; strain susceptibility


Assuntos
Apoptose/fisiologia , Bleomicina/farmacologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose/efeitos dos fármacos , Feminino , Hidroxiprolina/metabolismo , Immunoblotting , Imuno-Histoquímica , Macrófagos Alveolares/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Antígeno Nuclear de Célula em Proliferação/metabolismo , Receptores do Fator de Necrose Tumoral/deficiência
14.
Exp Lung Res ; 24(6): 721-43, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9839161

RESUMO

Bleomycin (BLM) induction of lung fibrosis in mice is an established model to study the mechanism of pulmonary fibrosis. Cytokine secretion has been implicated as a fundamental component of the lung fibrotic process observed in response to BLM. Among the cytokines implicated in lung fibrosis, Tumor necrosis factor (TNF) alpha has been considered to play a fundamental role. In the present study, we characterized the cellular sources of TNF during BLM-induced lung injury and examined the importance of TNF receptors in this process. To characterize the expression of TNF, we utilized two strains of mice, one sensitive (C57BL/6) and one resistant (BALB/c) to BLM-induced lung injury. Mice received BLM (120 mg/kg total) or saline, as control, by multiple subcutaneous injections. BLM induced the development of inflammation in subpleural areas only in the lungs of BLM-sensitive mice. These subpleural areas were characterized by infiltration of CD68-positive macrophages and increased collagen deposition. BLM enhanced the expression of TNF mRNA in BLM-sensitive, but not in BLM-resistant, mice. In situ hybridization studies localized the expression of TNF in the areas of BLM-induced inflammation in 6% and 27% of macrophages at 14 and 21 days post BLM treatment. In addition to TNF, BLM exposure resulted in the upregulated expression of transforming growth factor (TGF)-beta 1, but not interleukin (IL)-1, mRNA in the lungs of both murine strains at 14 and 21 days. This upregulated expression of TGF-beta 1 mRNA was greater in the lungs of BLM-sensitive mice. In separate experiments, double TNF receptor knockout mice were exposed to BLM. These animals demonstrated an increased expression of TNF, but not TGF-beta 1, mRNA in response to BLM and did not exhibit histologic evidence of lung injury following BLM exposure. In summary, the upregulation of TNF mRNA in macrophages correlated with the appearance of inflammation following BLM exposure and was limited to the BLM-sensitive strain. Furthermore, in addition to the release of the TNF ligand, it appears that the presence of TNF receptors is necessary for the development of BLM-induced lung injury, and signaling through these receptors may contribute to the regulation of the TGF-beta 1 mRNA expression observed in response to bleomycin. These results provide further support for a role of macrophages and TNF in the induction of lung inflammation.


Assuntos
Fibrose Pulmonar/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Bleomicina/toxicidade , Primers do DNA/química , Feminino , Genótipo , Hidroxiprolina/metabolismo , Técnicas Imunoenzimáticas , Hibridização In Situ , Interleucina-1/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , RNA Mensageiro/biossíntese , Receptores do Fator de Necrose Tumoral/deficiência , Receptores do Fator de Necrose Tumoral/genética , Organismos Livres de Patógenos Específicos , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/genética
15.
Rev Environ Health ; 13(1-2): 99-108, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9718625

RESUMO

Acrolein is a ubiquitous environmental pollutant that is known to cause respiratory tract injury and suppression of pulmonary host defense against infections in animal models. The mechanisms of acrolein-induced suppression of pulmonary host defense are not well understood. It has been generally believed that epithelial injury is responsible for the acrolein-caused decrease in resistance to infection. Emerging evidence suggests, however, that the alveolar macrophage is also a key target for acrolein-induced suppression of pulmonary host defense. It is likely that the combination of epithelial cell injury and inhibition of macrophage function may be responsible for acrolein-induced suppression of pulmonary host defense. To better assess the health risk of exposure to environmental levels of acrolein, more population-based studies are needed to monitor the levels of acrolein exposure and the adverse health effects associated with such exposures.


Assuntos
Acroleína/toxicidade , Poluição do Ar/efeitos adversos , Doenças Respiratórias/induzido quimicamente , Acroleína/imunologia , Exposição Ambiental , Células Epiteliais/efeitos dos fármacos , Humanos , Tolerância Imunológica , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/imunologia , Saúde Pública , Doenças Respiratórias/imunologia
16.
Am J Physiol ; 274(1): L8-16, 1998 01.
Artigo em Inglês | MEDLINE | ID: mdl-9458795

RESUMO

Ozone is a photochemically generated pollutant that can cause acute pulmonary inflammation and induce cellular injury and may contribute to the development or exacerbation of chronic lung diseases. Despite much research, the mechanisms of ozone- and oxidant-induced cellular injury are still uncertain. Ozone and secondary free radicals have been reported to cause the formation of aldehydes in biological fluids. One of the most toxic aldehydes formed during oxidant-induced lipid peroxidation is 4-hydroxynonenal (HNE). HNE reacts primarily with Cys, Lys, and His amino acids, altering protein function and forming protein adducts. The purpose of this study was to determine whether HNE could account for the acute effects of ozone on lung cells. Human subjects were exposed to 0.4 parts/million ozone or air for 1 h with exercise (each subject served as his/her own control). Six hours after ozone exposure, cells obtained by airway lavage were examined for apoptotic cell injury, and cells from bronchoalveolar lavage were examined for apoptosis, presence of HNE adducts, and expression of stress proteins. Significant apoptosis was evident in airway lung cells after ozone exposure. Western analysis demonstrated an increase in a 32-kDa HNE protein adduct and a number of stress proteins, viz., 72-kDa heat shock protein and ferritin, in alveolar macrophages (AM) after ozone exposure. All of these effects could be replicated by in vitro exposure of AM to HNE. Consequently, the in vitro results and demonstration of HNE protein adducts after ozone exposure are consistent with a potential role for HNE in the cellular toxic effects of ozone.


Assuntos
Aldeídos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas de Bactérias , Líquido da Lavagem Broncoalveolar/citologia , Proteínas de Choque Térmico/biossíntese , Pulmão/patologia , Macrófagos Alveolares/patologia , Ozônio/toxicidade , Poluentes Atmosféricos , Aldeídos/metabolismo , Morte Celular , Sobrevivência Celular/efeitos dos fármacos , Chaperonina 60 , Chaperoninas/biossíntese , Reagentes de Ligações Cruzadas , Ferritinas/biossíntese , Proteínas de Choque Térmico HSP72 , Humanos , Pulmão/efeitos dos fármacos , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/metabolismo
17.
Environ Health Perspect ; 106(3): 127-32, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9441960

RESUMO

Epidemiological studies report a small but positive association between short-term increases in airborne particulate matter and small increases in morbidity and mortality from respiratory and cardiovascular disease in urban areas. However, the lack of a mechanistic explanation to link particle exposure and human health effects makes it difficult to validate the human health effects. The present study tested the hypothesis that urban particles could cause apoptosis of human alveolar macrophages(AM) and a shift of their phenotypes to a higher immune active state, which would provide a mechanism to explain an inflammatory response. Freshly isolated human AM were incubated for 24 hr with urban particles (#1648 and #1649), Mount Saint Helen's ash (MSH), and residual oil fly ash (ROFA). Cell viability was assessed by trypan blue exclusion and apoptosis was demonstrated by morphology, cell death ELISA, and DNA ladder formation. Additionally, AM were characterized according to RFD1(+) (immune stimulatory macrophages) and RFD1(+)7(+) (suppressor macrophages) phenotypes by flow cytometry. ROFA particles caused AM necrosis at concentrations as low as 10 microg/ml, urban particles had no effect except at 200 microg/ml, and MSH had no effect at 200 microg/ml. ROFA (25 microg/ml) and particles #1648 or #1649 (100 microg/ml) caused apoptosis of AM by all three criteria, but 200 microg/ml MSH had no effect. Finally, 25 microg/ml ROFA and 100 microg/ml particles #1648 or #1649 up regulated the expression of the RFD1(+) AM phenotype, while only ROFA decreased the RFD1(+)7(+) phenotype. Consequently, ROFA and urban particles can induce apoptosis of human AM and increase the ratio of AM phenotypes toward a higher immune active state (i.e., increased RFD1(+):RFD1(+)7(+) ratio). Ifurban particles cause similar changes in vivo, this could result in lung inflammation and possible increased pulmonary and cardiovascular disease.


Assuntos
Poluentes Atmosféricos/efeitos adversos , Apoptose/efeitos dos fármacos , Macrófagos Alveolares/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Fenótipo , Azul Tripano , Saúde da População Urbana
19.
Environ Health Perspect ; 105 Suppl 5: 1139-42, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9400713

RESUMO

The mechanism by which fibrogenic particulates induce inflammation that can progress to lung fibrosis is uncertain. The alveolar macrophage (AM) has been implicated in the inflammatory process because of its function and reported release of inflammatory mediators when isolated from fibrotic patients. It has been recently shown that fibrogenic, but not nonfibrogenic, particulates are highly potent in inducing apoptosis of human AM. In this study, we tested the hypothesis that fibrogenic particulates could shift the phenotypic ratio of human AM to a more inflammatory condition. The macrophage phenotypes were characterized by flow cytometry targeting the RFD1 and RFD7 epitopes. Results demonstrated that chrysotile and crocidolite asbestos, as well as crystalline silica, but not titanium dioxide or wollastonite, increased the RFD1+ phenotype (inducer or immune activator macrophages) and decreased the RFD1+ RFD7+ phenotype (suppressor macrophages). These results provide a mechanistic explanation that may link apoptosis (namely, suppressor macrophages) to a shift in the ratio of macrophage phenotypes that could initiate lung inflammation.


Assuntos
Amianto/toxicidade , Carcinógenos/toxicidade , Macrófagos Alveolares/efeitos dos fármacos , Dióxido de Silício/toxicidade , Apoptose/efeitos dos fármacos , Células Cultivadas , Epitopos/efeitos dos fármacos , Epitopos/genética , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Fenótipo
20.
Am J Physiol ; 273(4): L760-7, 1997 10.
Artigo em Inglês | MEDLINE | ID: mdl-9357850

RESUMO

Exposure to silica dust can result in lung inflammation that may progress to fibrosis for which there is no effective clinical treatment. The mechanisms involved in the development of pulmonary silicosis have not been well defined; however, most current evidence implicates a central role for alveolar macrophages in this process. We have previously demonstrated that fibrotic agents, such as asbestos and silica, induce apoptosis in human alveolar macrophages. The goal of this study was to identify molecular events in the silica-induced apoptotic process to better understand the mechanism by which fibrotic agents may be inducing apoptosis in human alveolar macrophages. To elucidate the possible mechanism by which silica causes apoptosis, we investigated the involvement of the interleukin-converting enzyme (ICE) family of proteases. Human alveolar macrophages were treated with silica in vitro and were examined for the involvement of ICE, Ich-1L, and cpp32beta in silica-induced apoptosis. Pretreatment of cells with 10 microM of the ICE inhibitor z-Val-Ala-Asp-fluoromethyl ketone and the cpp32beta inhibitor Asp-Glu-Val-Asp-fluoromethyl ketone completely blocked silica-induced apoptosis. Additionally, an increased formation of the active p20 fragments of ICE and Ich-1L as well as degradation of the inactive zymogen form of cpp32beta protein were observed in silica-treated human alveolar macrophages, indicating activation of these proteases. Furthermore, degradation of the nuclear protein poly(ADP-ribose) polymerase was observed within 2 h of silica treatment. These results suggest that silica-induced apoptosis involves activation of the ICE family of proteases and is the first step in elucidating the intracellular mechanism of particulate-induced apoptosis in human alveolar macrophages.


Assuntos
Apoptose/efeitos dos fármacos , Caspases , Cisteína Endopeptidases/metabolismo , Macrófagos Alveolares/efeitos dos fármacos , Dióxido de Silício/toxicidade , Adulto , Clorometilcetonas de Aminoácidos/farmacologia , Apoptose/fisiologia , Caspase 1 , Caspase 3 , Células Cultivadas , Inibidores de Cisteína Proteinase/farmacologia , Fragmentação do DNA , Precursores Enzimáticos/metabolismo , Feminino , Humanos , Macrófagos Alveolares/citologia , Macrófagos Alveolares/fisiologia , Masculino , Necrose , Poli(ADP-Ribose) Polimerases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...