Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Am J Physiol Heart Circ Physiol ; 321(5): H985-H1003, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34559580

RESUMO

Ventilation with gases containing enhanced fractions of oxygen is the cornerstone of therapy for patients with hypoxia and acute respiratory distress syndrome. Yet, hyperoxia treatment increases free reactive oxygen species (ROS)-induced lung injury, which is reported to disrupt autophagy/mitophagy. Altered extranuclear activity of the catalytic subunit of telomerase, telomerase reverse transcriptase (TERT), plays a protective role in ROS injury and autophagy in the systemic and coronary endothelium. We investigated interactions between autophagy/mitophagy and TERT that contribute to mitochondrial dysfunction and pulmonary injury in cultured rat lung microvascular endothelial cells (RLMVECs) exposed in vitro, and rat lungs exposed in vivo to hyperoxia for 48 h. Hyperoxia-induced mitochondrial damage in rat lungs [TOMM20, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT)], which was paralleled by increased markers of inflammation [myeloperoxidase (MPO), IL-1ß, TLR9], impaired autophagy signaling (Beclin-1, LC3B-II/1, and p62), and decreased the expression of TERT. Mitochondrial-specific autophagy (mitophagy) was not altered, as hyperoxia increased expression of Pink1 but not Parkin. Hyperoxia-induced mitochondrial damage (TOMM20) was more pronounced in rats that lack the catalytic subunit of TERT and resulted in a reduction in cellular proliferation rather than cell death in RLMVECs. Activation of TERT or autophagy individually offset mitochondrial damage (MTT). Combined activation/inhibition failed to alleviate hyperoxic-induced mitochondrial damage in vitro, whereas activation of autophagy in vivo decreased mitochondrial damage (MTT) in both wild type (WT) and rats lacking TERT. Functionally, activation of either TERT or autophagy preserved transendothelial membrane resistance. Altogether, these observations show that activation of autophagy/mitophagy and/or TERT mitigate loss of mitochondrial function and barrier integrity in hyperoxia.NEW & NOTEWORTHY In cultured pulmonary artery endothelial cells and in lungs exposed in vivo to hyperoxia, autophagy is activated, but clearance of autophagosomes is impaired in a manner that suggests cross talk between TERT and autophagy. Stimulation of autophagy prevents hyperoxia-induced decreases in mitochondrial metabolism and sustains monolayer resistance. Hyperoxia increases mitochondrial outer membrane (TOMM20) protein, decreases mitochondrial function, and reduces cellular proliferation without increasing cell death.


Assuntos
Células Endoteliais/enzimologia , Hiperóxia/complicações , Lesão Pulmonar/enzimologia , Pulmão/irrigação sanguínea , Microvasos/enzimologia , Mitocôndrias/enzimologia , Mitofagia , Telomerase/metabolismo , Animais , Proteínas Relacionadas à Autofagia/metabolismo , Permeabilidade Capilar , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/patologia , Feminino , Técnicas de Inativação de Genes , Mediadores da Inflamação/metabolismo , Lesão Pulmonar/etiologia , Lesão Pulmonar/genética , Lesão Pulmonar/patologia , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Microvasos/patologia , Mitocôndrias/genética , Mitocôndrias/patologia , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Ratos Sprague-Dawley , Ratos Transgênicos , Receptores de Superfície Celular/metabolismo , Telomerase/deficiência , Telomerase/genética , Receptor 4 Toll-Like/metabolismo , Receptor Toll-Like 9/metabolismo
2.
Respir Res ; 22(1): 258, 2021 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-34602075

RESUMO

BACKGROUND: Diabetes mellitus (DM) is a key contributing factor to poor survival in lung transplantation recipients. Mitochondrial dysfunction is recognized as a critical mediator in the pathogenesis of diabetic lung ischemia-reperfusion (IR) injury. The protective effects of adiponectin have been demonstrated in our previous study, but the underlying mechanism remains unclear. Here we demonstrated an important role of mitophagy in the protective effect of adiponectin during diabetic lung IR injury. METHODS: High-fat diet-fed streptozotocin-induced type 2 diabetic rats were exposed to adiponectin with or without administration of the SIRT1 inhibitor EX527 following lung transplantation. To determine the mechanisms underlying the action of adiponectin, rat pulmonary microvascular endothelial cells were transfected with SIRT1 small-interfering RNA or PINK1 small-interfering RNA and then subjected to in vitro diabetic lung IR injury. RESULTS: Mitophagy was impaired in diabetic lungs subjected to IR injury, which was accompanied by increased oxidative stress, inflammation, apoptosis, and mitochondrial dysfunction. Adiponectin induced mitophagy and attenuated subsequent diabetic lung IR injury by improving lung functional recovery, suppressing oxidative damage, diminishing inflammation, decreasing cell apoptosis, and preserving mitochondrial function. However, either administration of 3-methyladenine (3-MA), an autophagy antagonist or knockdown of PINK1 reduced the protective action of adiponectin. Furthermore, we demonstrated that APN affected PINK1 stabilization via the SIRT1 signaling pathway, and knockdown of SIRT1 suppressed PINK1 expression and compromised the protective effect of adiponectin. CONCLUSION: These data demonstrated that adiponectin attenuated reperfusion-induced oxidative stress, inflammation, apoptosis and mitochondrial dysfunction via activation of SIRT1- PINK1 signaling-mediated mitophagy in diabetic lung IR injury.


Assuntos
Adiponectina/farmacologia , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Lesão Pulmonar/prevenção & controle , Transplante de Pulmão/efeitos adversos , Pulmão/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitofagia/efeitos dos fármacos , Traumatismo por Reperfusão/prevenção & controle , Animais , Células Cultivadas , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/enzimologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Mediadores da Inflamação/metabolismo , Pulmão/enzimologia , Pulmão/patologia , Lesão Pulmonar/enzimologia , Lesão Pulmonar/etiologia , Lesão Pulmonar/patologia , Masculino , Mitocôndrias/enzimologia , Mitocôndrias/patologia , Estresse Oxidativo/efeitos dos fármacos , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Ratos Sprague-Dawley , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Sirtuína 1/genética , Sirtuína 1/metabolismo
3.
J Biochem Mol Toxicol ; 35(9): e22847, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34309930

RESUMO

In this study, we aimed to evaluate whether edaravone (EDA) has a protective role against valproic acid (VPA)-induced lung damage via its antioxidative activity. Male Sprague-Dawley rats were split into four groups. Control (n = 8) rats; rats given EDA (30 mg kg-1 day-1 ; n = 10); rats given only (VPA, 500 mg kg-1 day-1 ; n = 10); rats given VPA + EDA (in the same dose and time) for 7 days. EDA and VPA were applied intraperitoneally. After 8 days, lung tissues were immediately taken from the rats. In lung homogenates, reduced glutathione, total antioxidant status levels, and superoxide dismutase, glutathione peroxidase, sodium/potassium ATPase, paraoxonase1, and carbonic anhydrase activities significantly abated, whereas catalase, glutathione reductase, glutathione-S-transferase activities insignificantly decreased in the VPA-treated group. In contrast, lipid peroxidation, reactive oxygen species, and total oxidant status levels, glycoprotein and protein carbonyl contents, nitric oxide, hydroxyproline levels, and xanthine oxidase, lactate dehydrogenase, arginase, and prolidase activities significantly increased in the VPA-given group. Administration of EDA caused the reverse effects. As a consequence, EDA prevented oxidative stress-mediated lung injury via its robust antioxidant effects.


Assuntos
Edaravone/farmacologia , Sequestradores de Radicais Livres/farmacologia , Lesão Pulmonar/enzimologia , Pulmão/enzimologia , Ácido Valproico/efeitos adversos , Animais , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/tratamento farmacológico , Masculino , Oxirredutases/metabolismo , Ratos , Ratos Sprague-Dawley , ATPase Trocadora de Sódio-Potássio/metabolismo , Ácido Valproico/farmacologia
4.
Arch Toxicol ; 95(1): 103-116, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33033841

RESUMO

Pyrrolizidine alkaloids (PAs) are common phytotoxins with both hepatotoxicity and pneumotoxicity. Hepatic cytochrome P450 enzymes are known to bioactivate PAs into reactive metabolites, which can interact with proteins to form pyrrole-protein adducts and cause intrahepatic cytotoxicity. However, the metabolic and initiation biochemical mechanisms underlying PA-induced pneumotoxicity remain unclear. To investigate the in vivo metabolism basis for PA-induced lung injury, this study used mice with conditional deletion of the cytochrome P450 reductase (Cpr) gene and resultant tissue-selective ablation of microsomal P450 enzyme activities. After oral exposure to monocrotaline (MCT), a pneumotoxic PA widely used to establish animal lung injury models, liver-specific Cpr-null (LCN) mice, but not extrahepatic Cpr-low (xh-CL) mice, had significantly lower level of pyrrole-protein adducts in the serum, liver and lungs compared with wild-type (WT) mice. While MCT-exposed LCN mice had significantly higher blood concentration of intact MCT, compared to MCT-exposed WT or xh-CL mice. Consistent with the MCT in vivo bioactivation data, MCT-induced lung injury, represented by vasculature damage, in WT and xh-CL mice but not LCN mice. Furthermore, reactive metabolites of MCT were confirmed to exist in the blood efflux from the hepatic veins of MCT-exposed rats. Our results provide the first mode-of-action evidence that hepatic P450s are essential for the bioactivation of MCT, and blood circulating reactive metabolites of MCT to the lung causes pneumotoxicity. Collectively, this study presents the scientific basis for the application of MCT in animal lung injury models, and more importantly, warrants public awareness and further investigations of lung diseases associated with exposure to not only MCT but also different PAs.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Fígado/enzimologia , Lesão Pulmonar/induzido quimicamente , Pulmão/efeitos dos fármacos , Monocrotalina/toxicidade , NADPH-Ferri-Hemoproteína Redutase/metabolismo , Ativação Metabólica , Animais , Isoenzimas , Pulmão/metabolismo , Pulmão/patologia , Lesão Pulmonar/sangue , Lesão Pulmonar/enzimologia , Lesão Pulmonar/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monocrotalina/sangue , NADPH-Ferri-Hemoproteína Redutase/genética , Ligação Proteica , Ratos Sprague-Dawley , Toxicocinética
5.
Med Sci Monit ; 26: e919213, 2020 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-32034118

RESUMO

BACKGROUND This study aimed to investigate the effects of dexmedetomidine in a rat model of sepsis-induced lung injury and the role of the adenosine monophosphate-activated protein kinase (AMPK) gene and silent information regulator 1 (SIRT1) gene signaling pathway. MATERIAL AND METHODS Sixty 28-week-old healthy male Sprague-Dawley rats were randomly divided into three groups, the sham group, the model group, and the dexmedetomidine-treated group. The rat model of sepsis-induced lung injury was developed by surgical cecal ligation and puncture. Lung tissues examined histologically in the three study groups. Cell apoptosis was measured using the TUNEL assay, and the expression of inflammatory cytokines, tumor necrosis factor-alpha (TNF-alpha), interleukin-1ß (IL-1ß), and IL-10 were measured in rat lung tissue by enzyme-linked immunosorbent assay (ELISA). Apoptosis-associated proteins and AMPK/SIRT1 pathway-associated protein expression levels were detected using Western blot. RESULTS Dexmedetomidine significantly increased the survival rate and reduced the body temperature of rats in the model group with sepsis-induced lung injury, reduced lung injury, significantly reduced apoptosis in lung tissues, and reduced the expression levels of TNF-alpha, and IL-1ß, and increased the levels of IL-10. Dexmedetomidine significantly reduced the expression of caspase-3 in the rat lung tissue (P<0.01), and significantly increased the expression of Bcl-2/Bax and the phosphorylation levels of AMPK, SIRT1, nuclear factor-kappaB (NF-kappaB), and forkhead box class O 3a (FOXO3a). CONCLUSIONS In a rat model of sepsis-induced lung injury, dexmedetomidine reduced lung damage by activating the AMPK/SIRT1 signaling pathway and reduced the expression of inflammatory cytokines and cell apoptosis.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Dexmedetomidina/uso terapêutico , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/etiologia , Sepse/complicações , Transdução de Sinais , Sirtuína 1/metabolismo , Animais , Apoptose/efeitos dos fármacos , Temperatura Corporal/efeitos dos fármacos , Citocinas/metabolismo , Dexmedetomidina/farmacologia , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Pulmão/enzimologia , Pulmão/patologia , Lesão Pulmonar/enzimologia , Masculino , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Análise de Sobrevida
6.
Ecotoxicol Environ Saf ; 195: 110473, 2020 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-32199220

RESUMO

To investigate the effect of fermented black barley on cooking oil fume (COF)-induced lung injury, male ICR mice were randomized into five groups: normal control (NC), fermented black barley treatment (NF), COF exposure (O), COF + fermented black barley treatment (OF) and COF + Lactobacillus treatment (OL). The exposure of mice to COF was performed for 5 min per day and 4 days per week for a total of 9 weeks, and the mice in the OF, NF and OL groups were administered fermented black barley or Lactobacillus continuously for 9 weeks (1 mL/100 g). Our results showed that the gamma-aminobutyric acid (GABA), total phenolic, and flavonoid contents significantly increased after fermentation (P < 0.01). In addition, fermented black barley significantly increased SOD activity in the lung tissue, decreased the wet pulmonary coefficient, inhibited the reduction of microbial diversity and richness, and upregulated genes involved in cilium assembly and the cilium axoneme. These findings support the notion that fermented black barley can ameliorate COF-induced lung injury in mice.


Assuntos
Poluentes Atmosféricos/toxicidade , Microbioma Gastrointestinal , Hordeum , Lesão Pulmonar/terapia , Animais , Antioxidantes/química , Antioxidantes/metabolismo , Cílios/metabolismo , Culinária , Fermentação , Flavonoides/metabolismo , Hordeum/química , Hordeum/metabolismo , Lactobacillus , Pulmão/enzimologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/enzimologia , Lesão Pulmonar/microbiologia , Masculino , Camundongos Endogâmicos ICR , Fenóis/metabolismo , Superóxido Dismutase/metabolismo , Ácido gama-Aminobutírico/metabolismo
7.
Toxicol Appl Pharmacol ; 366: 25-34, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30641076

RESUMO

Mechanisms responsible for diesel exhaust particle (DEP)-induced toxicity in respiratory disorders are poorly understood, recent experimental and controlled exposure studies suggested that oxidative stress might be involved. To investigate the time-course effects DEP on nuclear factor erythroid 2-related factor 2 (Nrf2), a key regulator in cellular adaptive antioxidant response, mice were intratracheal instilled with 100 µg DEP/mouse and sacrificed after 30 min, 6 h, 12 h, 24 h, 48 h, and 72 h. We measured reactive oxygen species (ROS) as well as Nrf2 and antioxidant enzymes such as superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px) and phase II enzymes including heme oxygenase-1 (HO-1), NAD(P)H:quinone oxidoreductase-1 (NQO1), glutamate-cysteine ligase catalytic subunit (GCLC), glutamate-cysteine ligase modifier subunit (GCLM) in the lungs. Additionally, histopathological changes were examined. At 6 h, ROS peaked, most of the enzymes were activated, and the histology showed the lungs were damaged. At 12 h, ROS returned to normal level and CAT activity decreased, while protein expression of Nrf2, HO-1, NQO1, GCLC, and GCLM increased, and the lungs were recovering from damage. After 24 h, ROS started to decrease and Nrf2 showed a decreasing trend at both gene and protein levels, while the lung damage had been entirely restored. These results suggested that a single exposure to DEP induce transient oxidative stress in the lungs, with time-dependent effects on Nrf2 and antioxidant enzymes and phase II enzymes.


Assuntos
Antioxidantes/metabolismo , Enzimas/metabolismo , Lesão Pulmonar/enzimologia , Pulmão/enzimologia , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Material Particulado , Emissões de Veículos , Animais , Modelos Animais de Doenças , Enzimas/genética , Regulação Enzimológica da Expressão Gênica , Exposição por Inalação , Pulmão/patologia , Lesão Pulmonar/etiologia , Lesão Pulmonar/genética , Lesão Pulmonar/patologia , Masculino , Desintoxicação Metabólica Fase II , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/genética , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
8.
FASEB J ; 32(3): 1250-1264, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29122847

RESUMO

Idiopathic pulmonary fibrosis (IPF) is characterized by the deposition of excessive extracellular matrix and the destruction of lung parenchyma, resulting from an aberrant wound-healing response. Although IPF is often associated with an imbalance in protease activity, the mechanisms underlying the sustained repair mechanisms are not fully understood. Here, we addressed the role of the recently identified, membrane-anchored serine protease human airway trypsin-like protease (HAT). In the present study, we show that both HAT expression and activity were up-regulated in human IPF specimens. Next, adenoviral overexpression of HAT before bleomycin challenge attenuated lung injury as well as extracellular matrix deposition in the bleomycin-induced pulmonary fibrosis model. In vitro, HAT prevented specific fibrosis-associated responses in primary human pulmonary fibroblasts and induced the expression of mediators associated with the prostaglandin E2 pathway. Altogether, our findings suggested that HAT could have a protective role in IPF and other fibrotic lung disorders.-Menou, A., Flajolet, P., Duitmen, J., Justet, A., Moog, S., Jaillet, M., Tabèze, L., Solhonne, B., Garnier, M., Mal, H., Mordant, P., Castier, Y., Cazes, A., Sallenave, J.-M., Mailleux, A. A., Crestani, B. Human airway trypsin-like protease exerts potent, antifibrotic action in pulmonary fibrosis.


Assuntos
Lesão Pulmonar/prevenção & controle , Fibrose Pulmonar/prevenção & controle , Serina Endopeptidases/administração & dosagem , Animais , Antibióticos Antineoplásicos/toxicidade , Bleomicina/toxicidade , Estudos de Casos e Controles , Movimento Celular , Proliferação de Células , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Fibroblastos/patologia , Humanos , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Pulmão/patologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/enzimologia , Lesão Pulmonar/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/enzimologia , Fibrose Pulmonar/patologia , Serina Endopeptidases/metabolismo , Transdução de Sinais
9.
Drug Chem Toxicol ; 42(3): 257-263, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-29536762

RESUMO

CONTEXT: Sulfur mustard (SM) is a strong alkylating toxicant that targets different organs, particularly human lung tissue. Change in genes expression is one of the molecular mechanisms of SM toxicity in damaged tissue. OBJECTIVE: The purpose of this investigation is to characterize the expression of cyclooxygenase-2 (COX-2), 12-lipoxygenase (12-LO), inducible nitric oxide synthase 2 (iNOS2), and surfactant protein D (SFTPD) in lungs of patients who exposed to SM. METHODS: Lung biopsies were provided from SM-exposed patients (n = 6) and controls (n = 5). Total RNA were extracted from all specimens and then cDNA was synthesized for each sample. Changes in gene expression were measured using RT2 Profiler ™PCR Array. RESULTS: Pulmonary function tests revealed more obstructive and restrictive spirometric patterns among patients compared to the control group. Expression of COX-2 and 12-LO in the lung of patients was increased by 6.2555 (p = 0.004) and 6.2379-folds (p = 0.002), respectively. In contrast, expression of SF-D and iNOS genes was reduced by 8.5869-fold (p = 0.005) and 2.4466-folds (p = 0.011), respectively. CONCLUSIONS: Mustard lungs were associated with overexpression of COX-2 and 12-LO, which are responsible for inflammation, overproduction of free radicals and oxidative stress. Downregulation of iNOS2 and SF-D are probably the reason for lung disease and dysfunction among these patients. Therefore, the expression of these genes could be an important, routine part of the management of such patients.


Assuntos
Araquidonato 12-Lipoxigenase/genética , Substâncias para a Guerra Química/toxicidade , Ciclo-Oxigenase 2/genética , Expressão Gênica/efeitos dos fármacos , Lesão Pulmonar/induzido quimicamente , Gás de Mostarda/toxicidade , Óxido Nítrico Sintase Tipo II/genética , Proteína D Associada a Surfactante Pulmonar/genética , Estudos de Casos e Controles , Humanos , Irã (Geográfico) , Lesão Pulmonar/enzimologia , Lesão Pulmonar/genética , Testes de Função Respiratória
10.
Chin Med Sci J ; 34(4): 270-276, 2019 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-33906713

RESUMO

Objective To evaluate the antagonistic effects of N-acetylcysteine (NAC) on mitogen-activated protein kinases (MAPK) pathway activation, oxidative stress and inflammatory responses in rats with lung injury induced by fine particulate matter (PM2.5).Methods Forty eight male Wistar rats were randomly divided into six groups: blank control group (C1), water drip control group (C2), PM2.5 exposed group (P), low-dose NAC treated and PM2.5 exposed group (L), middle-dose NAC treated and PM2.5 exposed group (M), and high-dose NAC treated and PM2.5 exposed group (H). PM2.5 suspension (7.5 mg/kg) was administered tracheally once a week for four times. NAC of 125 mg/kg, 250 mg/kg and 500 mg/kg was delivered intragastrically to L, M and H group respectively by gavage (10 ml/kg) for six days before PM2.5 exposure. The histopathological changes and human mucin 5 subtype AC (MUC5AC) content in lung tissue of rats were evaluated. We investigated IL-6 in serum and bronchoalveolar lavage fluid (BALF) by Enzyme-linked immunosorbent assay (ELISA), MUC5AC in lung tissue homogenate by ELISA, glutathione peroxidase (GSH-PX) in serum and BALF by spectrophotometry, and the expression of p-ERK1/2, p-JNK1/2 and p-p38 proteins by Western blot. All the measurements were analyzed and compared statistically.Results Lung tissue of rats exposed to PM2.5 showed histological destruction and increased mucus secretion of bronchial epithelial cells. Rats receiving NAC treatment showed less histological destruction and mucus secretion. Of P, L, M and H group, MUC5AC in lung tissue, IL-6 in serum and BALF were higher than controls (C1 and C2) (all P<0.05), with the highest levels found in the P group and a decreasing trend with increase of NAC dose. The activity of GSH-PX in serum and BALF of PM2.5 exposed rats (P, L, M and H) was lower than that of controls (all P<0.05), with higher activities found in NAC treated rats (L, M, and H), and an increasing trend with increase of NAC dose. The expressions of p-ERK1/2, p-JNK1/2 and p-p38 proteins in PM2.5 exposed lung tissue (P, L, M and H) was higher than controls (all P<0.05), with decreased levels and dose dependent downregulation found in NAC treated rats.Conclusion NAC can antagonize major MAPK pathway activation, lung oxidative stress and inflammatory injury induced by PM2.5 in rats.


Assuntos
Acetilcisteína/farmacologia , Inflamação/patologia , Lesão Pulmonar/enzimologia , Lesão Pulmonar/patologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Estresse Oxidativo , Tamanho da Partícula , Material Particulado/toxicidade , Animais , Líquido da Lavagem Broncoalveolar , Ativação Enzimática/efeitos dos fármacos , Glutationa Peroxidase/sangue , Glutationa Peroxidase/metabolismo , Interleucina-6/sangue , Interleucina-6/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/patologia , Lesão Pulmonar/sangue , Masculino , Mucina-5AC/sangue , Mucina-5AC/metabolismo , Muco/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ratos Wistar
11.
Pulm Pharmacol Ther ; 50: 100-110, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29702255

RESUMO

AIM: This study assessed pulmonary outcomes generated by inhibiting key enzymes of sphingolipid metabolism pathways related to ceramide synthesis in a murine model of lung injury induced by lipopolysaccharide (LPS). METHODS: C57BL/6 male adult mice received LPS intratracheally and the expressions of acid sphingomyelinase (ASM), neutral sphingomyelinase (NSM), serine palmitoyl transferase (SPT) and dihydroceramide synthase (DS) were assessed at 2, 4, 6, 12 and 24 h after LPS instillation in lung homogenate (n = 30). The pharmacological inhibition of ASM, NSM, SPT and DS were assayed in other mice groups by three different doses of desipramine, GW4869, myriocin and fumonisin, respectively (n = 90). Their most effective doses were administered intraperitoneally 1 or 2 h before LPS to different animal groups (n = 120). Mice underwent determination of pulmonary mechanics, lung histopathological aspects and apoptosis. RESULTS: The expression levels of the enzymes reached their peak at 2-4 h after LPS administration. ASM inhibition attenuated alveolar collapse and GW4869 decreased lung elastance, proinflammatory cytokines' levels and was more effective to improve alveolar collapse than desipramine. On the other hand, SPT blockage aggravated lung lesion and no effects it was observed with fumonisin. Moreover, simultaneous administration of inhibitors (desipramine + GW4869, myriocin + fumonisin and all inhibitors together) resulted in no changes. CONCLUSION: Blockage of sphingomyelinases and the de novo pathways improved and aggravated lung injury, respectively, putatively suggesting specific targets to therapeutic strategies in LPS-induced lung injury.


Assuntos
Lipopolissacarídeos/farmacologia , Lesão Pulmonar/induzido quimicamente , Esfingolipídeos/metabolismo , Compostos de Anilina/farmacologia , Animais , Compostos de Benzilideno/farmacologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Pulmão/metabolismo , Pulmão/patologia , Lesão Pulmonar/enzimologia , Lesão Pulmonar/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxirredutases/antagonistas & inibidores , Oxirredutases/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Serina C-Palmitoiltransferase/antagonistas & inibidores , Serina C-Palmitoiltransferase/metabolismo , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Esfingomielina Fosfodiesterase/metabolismo
12.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 43(11): 1177-1181, 2018 Nov 28.
Artigo em Zh | MEDLINE | ID: mdl-30643060

RESUMO

OBJECTIVE: To establish rat model of lung ischemia/reperfusion (IR) in vivo, and to explore the effects of acidification pretreatment for respiratory acidosis on the expression of matrix metalloproteinase-9 (MMP-9) and the possible mechanisms.
 Methods: A total of 36 male Sprague-Dawley rats were divided into a sham group (S group), a IR group, and an experiment group (RA group) (n=12 in each group). The rat left lung hilum in the S group was dissociated, followed by perfusion without ischemia. After the left lung hilum in the IR group was blocked for 45 min, the rats were followed by reperfusion for 180 min. After left lung hilum in the RA group was dissociated, the respiratory parameters were adjusted so that pressure of end tidal carbon dioxide (PETCO2) reached 56-65 mmHg (1 mmHg=0.133 kPa) for 5 min, then the rats was subjected to IR. Lung tissue wet/dry (W/D) and lung permeability index (LPI) were calculated, while the lung histopathology was observed and the MMP-9 protein expression were measured.
 Results: Compared with the control group, the W/D and LPI in the IR group and the RA group increased after reperfusion (both P<0.05), and the levels of W/D and LPI in the group RA were lower than that in the IR group (P<0.05). LPI and pathology scores were significantly lower in the RA group than those in the IR group (both P<0.01). After IR, the expression of MMP9 in the lung tissues in the IR group and the RA group increased significantly (both P<0.01). The expression of MMP-9 protein in the RA group was significantly lower than that in the IR group (P<0.01).
 Conclusion: After lung IR injury, the expression of MMP-9 protein, vascular permeability and inflammatory exudation is increased. The acidification pretreatment for respiratory acidosis can inhibit the expression of MMP-9 protein and reduce inflammatory exudation after lung IR, showing a protective effect on lung IR injury.


Assuntos
Acidose Respiratória , Pulmão/enzimologia , Metaloproteinase 9 da Matriz , Traumatismo por Reperfusão/prevenção & controle , Acidose Respiratória/tratamento farmacológico , Acidose Respiratória/prevenção & controle , Animais , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Lesão Pulmonar/enzimologia , Masculino , Metaloproteinase 9 da Matriz/genética , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/tratamento farmacológico
13.
Am J Physiol Lung Cell Mol Physiol ; 313(1): L115-L125, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28385808

RESUMO

Supplemental oxygen (O2) increases the risk of lung injury in preterm infants, owing to an immature antioxidant system. Our objective was to determine whether impairing antioxidant defense by decreasing glutathione peroxidase 1 (GPx1) gene expression increases the injurious effects of hyperoxia (Hyp). GPx1+/+ and GPx1-/- C57Bl/6J mice were exposed to 21% O2 (Air) or 40% O2 (Hyp) from birth to postnatal day 7 (P7d); they were euthanized on P7d or maintained in air until adulthood [postnatal day 56 (P56d)] to assess short-term and long-term effects, respectively. We assessed lung architecture, three markers of pulmonary oxidative stress (P7d, P56d), macrophages in lung tissue (P7d), immune cells in bronchoalveolar lavage fluid (BALF; P56d), and GPx1-4 and catalase gene expression in lung tissue (P7d, P56d). On P7d, macrophages were decreased by lack of GPx1 expression and further decreased by hyperoxia. GPx1 expression was increased in GPx1+/+Hyp mice and decreased in both GPx1-/- groups. On P56d, heme oxygenase-1 was increased by hyperoxia when GPx1 was absent. There were significantly more immune cells from Hyp groups than from the GPx1+/+Air group and a greater proportion of lymphocytes in GPx1-/-Hyp mice. GPx1 expression was significantly decreased in GPx1-/- mice; GPx2-4 and catalase expression was increased in GPx1-/-Hyp mice compared with other groups. Tissue fraction was decreased in GPx1-/-Air mice; bronchiolar smooth muscle was decreased in GPx1-/- mice. GPx1 does not clearly exacerbate hyperoxia-induced increases in oxidative stress or lung injury but may alter pulmonary immune function. Increased expression of GPx2-4 and catalase in GPx1-/-Hyp mice suggests gene redundancy within the model.


Assuntos
Progressão da Doença , Regulação Enzimológica da Expressão Gênica , Glutationa Peroxidase/genética , Hiperóxia/enzimologia , Hiperóxia/genética , Lesão Pulmonar/enzimologia , Lesão Pulmonar/genética , Aldeídos/metabolismo , Animais , Animais Recém-Nascidos , Antioxidantes/metabolismo , Feminino , Glutationa Peroxidase/metabolismo , Heme Oxigenase-1/metabolismo , Pulmão/imunologia , Pulmão/patologia , Lesão Pulmonar/imunologia , Lesão Pulmonar/patologia , Masculino , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Tirosina/análogos & derivados , Tirosina/metabolismo , Glutationa Peroxidase GPX1
14.
Am J Physiol Lung Cell Mol Physiol ; 312(5): L722-L730, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28283477

RESUMO

Progressive pulmonary fibrosis is a devastating consequence of many acute and chronic insults to the lung. Lung injury leads to alveolar epithelial cell (AEC) death, destruction of the basement membrane, and activation of transforming growth factor-ß (TGF-ß). There is subsequent resolution of the injury and a coordinated and concurrent initiation of fibrosis. Both of these processes may involve activation of similar intracellular signaling pathways regulated in part by dynamic changes to the extracellular matrix. Matrix signaling can augment the profibrotic fibroblast response to TGF-ß. However, similar matrix/integrin signaling pathways may also be involved in the inhibition of ongoing TGF-ß-induced AEC apoptosis. Focal adhesion kinase (FAK) is an integrin-associated signaling molecule expressed by many cell types. We used mice with AEC-specific FAK deletion to isolate the epithelial aspect of integrin signaling in the bleomycin model of lung injury and fibrosis. Mice with AEC-specific deletion of FAK did not exhibit spontaneous lung injury but did have significantly greater terminal deoxynucleotidyl transferase dUTP-mediated nick-end labeling-positive cells (18.6 vs. 7.1) per ×200 field, greater bronchoalveolar lavage protein (3.2 vs. 1.8 mg/ml), and significantly greater death (77 vs. 19%) after bleomycin injury compared with littermate control mice. Within primary AECs, activated FAK directly associates with caspase-8 and inhibits activation of the caspase cascade resulting in less apoptosis in response to TGF-ß. Our studies support a model in which dynamic changes to the extracellular matrix after injury promote fibroblast activation and inhibition of epithelial cell apoptosis in response to TGF-ß through FAK activation potentially complicating attempts to nonspecifically target this pathway for antifibrotic therapy.


Assuntos
Células Epiteliais/enzimologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Lesão Pulmonar/enzimologia , Pulmão/patologia , Fibrose Pulmonar/enzimologia , Transdução de Sinais , Animais , Apoptose , Bleomicina , Caspase 8/metabolismo , Ativação Enzimática , Células Epiteliais/patologia , Deleção de Genes , Lesão Pulmonar/complicações , Lesão Pulmonar/patologia , Camundongos , Modelos Biológicos , Especificidade de Órgãos , Fibrose Pulmonar/complicações , Fibrose Pulmonar/patologia , Proteína C Associada a Surfactante Pulmonar/metabolismo
15.
Am J Physiol Lung Cell Mol Physiol ; 312(5): L586-L598, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28213471

RESUMO

Rodent pups exposed to hyperoxia develop lung changes similar to bronchopulmonary dysplasia (BPD) in extremely premature infants. Oxidative stress from hyperoxia can injure developing lungs through endoplasmic reticulum (ER) stress. Early caffeine treatment decreases the rate of BPD, but the mechanisms remain unclear. We hypothesized that caffeine attenuates hyperoxia-induced lung injury through its chemical chaperone property. Sprague-Dawley rat pups were raised either in 90 (hyperoxia) or 21% (normoxia) oxygen from postnatal day 1 (P1) to postnatal day 10 (P10) and then recovered in 21% oxygen until P21. Caffeine (20 mg/kg) or normal saline (control) was administered intraperitoneally daily starting from P2. Lungs were inflation-fixed for histology or snap-frozen for immunoblots. Blood caffeine levels were measured in treated pups at euthanasia and were found to be 18.4 ± 4.9 µg/ml. Hyperoxia impaired alveolar formation and increased ER stress markers and downstream effectors; caffeine treatment attenuated these changes at P10. Caffeine also attenuated the hyperoxia-induced activation of cyclooxygenase-2 and markers of apoptosis. In conclusion, hyperoxia-induced alveolar growth impairment is mediated, in part, by ER stress. Early caffeine treatment protects developing lungs from hyperoxia-induced injury by attenuating ER stress.


Assuntos
Cafeína/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Hiperóxia/complicações , Hiperóxia/patologia , Lesão Pulmonar/etiologia , Lesão Pulmonar/patologia , Animais , Apoptose/efeitos dos fármacos , Cafeína/sangue , Ciclo-Oxigenase 2/metabolismo , Metabolismo Energético/efeitos dos fármacos , Feminino , Proteínas de Choque Térmico/metabolismo , Hiperóxia/enzimologia , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Pulmão/patologia , Lesão Pulmonar/enzimologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Modelos Biológicos , Neovascularização Fisiológica/efeitos dos fármacos , Biogênese de Organelas , Estresse Oxidativo/efeitos dos fármacos , Peroxidase/metabolismo , Pneumonia/complicações , Pneumonia/patologia , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia , Ratos Sprague-Dawley , Resposta a Proteínas não Dobradas/efeitos dos fármacos
16.
J Surg Res ; 216: 9-17, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28807219

RESUMO

BACKGROUND: Lung ischemia-reperfusion injury (LIRI) is a pathologic process that is observed in several clinical conditions, and p38 mitogen-activated protein kinase (MAPK) is involved. Diabetes mellitus (DM) results in an increased incidence of ischemia-induced organ damage. The aims of this study were to examine the effects of DM on LIRI in a rat model of DM and to explore the possible mechanisms in relation to the p38 MAPK pathway. METHODS: Forty rats were randomly divided into the following five groups (n = 8 each): a control + sham group, a control + IR group (CIR), a DM + sham group, a DM + IR group (DIR), and a DM + IR + SB203580 group. The control and streptozotocin-induced diabetic rats underwent a sham operation or left hilum occlusion for 90 min followed by reperfusion for 4 h. SB203580 was used to inhibit the p38 MAPK pathway. The pulmonary oxygenation index, inflammatory cytokines in the serum, lung edema, histopathology, oxidant stress, apoptosis, and phosphorylated/total-p38 MAPK protein levels were measured. RESULTS: The DIR group displayed greater concentrations of tumor necrosis factor-α, interleukin-6, and intercellular adhesion molecule-1 and increases in the wet weight-to-dry weight ratio, lung injury scores, malondialdehyde levels, and cellular apoptosis, and these effects were accompanied by lower pulmonary oxygenation compared with the CIR group (P < 0.05). In the DIR group, the expression levels of p38 MAPK protein were significantly upregulated compared with those of the CIR group. Additionally, all of these alterations were attenuated in the DM + IR + SB203580 group compared with the DIR group. CONCLUSIONS: Diabetes exacerbates LIRI by activating the p38 MAPK pathway.


Assuntos
Diabetes Mellitus Experimental/complicações , Lesão Pulmonar/enzimologia , Traumatismo por Reperfusão/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Biomarcadores/metabolismo , Diabetes Mellitus Experimental/enzimologia , Pulmão/irrigação sanguínea , Pulmão/enzimologia , Pulmão/patologia , Lesão Pulmonar/complicações , Lesão Pulmonar/patologia , Masculino , Distribuição Aleatória , Ratos , Ratos Wistar , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/patologia
17.
Am J Respir Cell Mol Biol ; 55(6): 878-888, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27494234

RESUMO

Alveolar epithelial injury and increased alveolar permeability are hallmarks of acute respiratory distress syndrome. Apoptosis of lung epithelial cells via the Fas/Fas ligand (FasL) pathway plays a critical role in alveolar epithelial injury. Activation of hypoxia-inducible factor (HIF)-1 by inhibition of prolyl hydroxylase domain proteins (PHDs) is a possible therapeutic approach to attenuate apoptosis and organ injury. Here, we investigated whether treatment with dimethyloxalylglycine (DMOG), an inhibitor of PHDs, could attenuate Fas/FasL-dependent apoptosis in lung epithelial cells and lung injury. DMOG increased HIF-1α protein expression in vitro in MLE-12 cells, a murine alveolar epithelial cell line. Treatment of MLE-12 cells with DMOG significantly suppressed cell surface expression of Fas and attenuated FasL-induced caspase-3 activation and apoptotic cell death. Inhibition of the HIF-1 pathway by echinomycin or small interfering RNA transfection abolished these antiapoptotic effects of DMOG. Moreover, intraperitoneal injection of DMOG in mice increased HIF-1α expression and decreased Fas expression in lung tissues. DMOG treatment significantly attenuated caspase-3 activation, apoptotic cell death in lung tissue, and the increase in alveolar permeability in mice instilled intratracheally with FasL. In addition, inflammatory responses and histopathological changes were also significantly attenuated by DMOG treatment. In conclusion, inhibition of PHDs protects lung epithelial cells from Fas/FasL-dependent apoptosis through HIF-1 activation and attenuates lung injury in mice.


Assuntos
Apoptose/efeitos dos fármacos , Proteína Ligante Fas/farmacologia , Lesão Pulmonar/enzimologia , Lesão Pulmonar/patologia , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Aminoácidos Dicarboxílicos/farmacologia , Animais , Caspase 3/metabolismo , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Proteína de Domínio de Morte Associada a Fas/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos Endogâmicos C57BL , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Estabilidade Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
18.
Am J Respir Cell Mol Biol ; 55(3): 419-28, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27089175

RESUMO

Oxygen toxicity and antioxidant deficiencies contribute to the development of bronchopulmonary dysplasia. Aurothioglucose (ATG) and auranofin potently inhibit thioredoxin reductase-1 (TrxR1), and TrxR1 disruption activates nuclear factor E2-related factor 2 (Nrf2), a regulator of endogenous antioxidant responses. We have shown previously that ATG safely and effectively prevents lung injury in adult murine models, likely via Nrf2-dependent mechanisms. The current studies tested the hypothesis that ATG would attenuate hyperoxia-induced lung developmental deficits in newborn mice. Newborn C3H/HeN mice were treated with a single dose of ATG or saline within 12 hours of birth and were exposed to either room air or hyperoxia (85% O2). In hyperoxia, ATG potently inhibited TrxR1 activity in newborn murine lungs, attenuated decreases in body weight, increased the transcription of Nrf2-regulated genes nicotinamide adenine dinucleotide phosphate reduced quinone oxidoreductase-1 (NQO1) and heme oxygenase 1, and attenuated alterations in alveolar development. To determine the impact of TrxR1 inhibition on Nrf2 activation in vitro, murine alveolar epithelial-12 cells were treated with auranofin, which inhibited TrxR1 activity, enhanced Nrf2 nuclear levels, and increased NQO1 and heme oxygenase 1 transcription. Our novel data indicate that a single injection of the TrxR1 inhibitor ATG attenuates hyperoxia-induced alterations in alveolar development in newborn mice. Furthermore, our data support a model in which the effects of ATG treatment likely involve Nrf2 activation, which is consistent with our findings in other lung injury models. We conclude that TrxR1 represents a novel therapeutic target to prevent oxygen-mediated neonatal lung injury.


Assuntos
Hiperóxia/complicações , Hiperóxia/enzimologia , Lesão Pulmonar/complicações , Lesão Pulmonar/enzimologia , Fator 2 Relacionado a NF-E2/metabolismo , Tiorredoxina Dissulfeto Redutase/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Auranofina/farmacologia , Aurotioglucose/farmacologia , Peso Corporal/efeitos dos fármacos , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/metabolismo , Hiperóxia/patologia , Lesão Pulmonar/patologia , Camundongos , Camundongos Endogâmicos C3H , Morfogênese/efeitos dos fármacos , NAD(P)H Desidrogenase (Quinona)/metabolismo , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/crescimento & desenvolvimento , Alvéolos Pulmonares/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Tiorredoxina Dissulfeto Redutase/metabolismo
19.
Am J Respir Cell Mol Biol ; 55(1): 24-34, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26677751

RESUMO

Pneumococcal pneumonia is a leading cause of mortality worldwide. The inflammatory response to bacteria is necessary to control infection, but it may also contribute to tissue damage. Phosphodiesterase-4 inhibitors, such as rolipram (ROL), effectively reduce inflammation. Here, we examined the impact of ROL in a pneumococcal pneumonia murine model. Mice were infected intranasally with 10(5)-10(6) CFU of Streptococcus pneumoniae, treated with ROL in a prophylactic or therapeutic schedule in combination, or not, with the antibiotic ceftriaxone. Inflammation and bacteria counts were assessed, and ex vivo phagocytosis assays were performed. ROL treatment during S. pneumoniae infection decreased neutrophil recruitment into lungs and airways and reduced lung injury. Prophylactic ROL treatment also decreased cytokine levels in the airways. Although modulation of inflammation by ROL ameliorated pneumonia, bacteria burden was not reduced. On the other hand, antibiotic therapy reduced bacteria without reducing neutrophil infiltration, cytokine level, or lung injury. Combined ROL and ceftriaxone treatment decreased lethality rates and was more efficient in reducing inflammation, by increasing proresolving protein annexin A1 (AnxA1) expression, and bacterial burden by enhancing phagocytosis. Lack of AnxA1 increased inflammation and lethality induced by pneumococcal infection. These data show that immunomodulatory effects of phosphodiesterase-4 inhibitors are useful during severe pneumococcal pneumonia and suggest their potential benefit as adjunctive therapy during infectious diseases.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/enzimologia , Inibidores da Fosfodiesterase 4/uso terapêutico , Pneumonia Pneumocócica/complicações , Pneumonia Pneumocócica/tratamento farmacológico , Pneumonia Pneumocócica/enzimologia , Pneumonia/complicações , Animais , Anexina A1/metabolismo , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Ceftriaxona/farmacologia , Ceftriaxona/uso terapêutico , Pulmão/microbiologia , Pulmão/patologia , Lesão Pulmonar/complicações , Lesão Pulmonar/fisiopatologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Fagocitose/efeitos dos fármacos , Inibidores da Fosfodiesterase 4/farmacologia , Pneumonia/tratamento farmacológico , Pneumonia/patologia , Pneumonia/fisiopatologia , Pneumonia Pneumocócica/fisiopatologia , Testes de Função Respiratória , Rolipram/farmacologia , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/fisiologia
20.
Inflammopharmacology ; 24(4): 155-61, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27271689

RESUMO

OBJECTIVES: Paraquat (PQ) is a widely used herbicide. Exposure to PQ at toxic doses can result in fatal acute lung injury. Inhibition of the poly-(ADP-ribose) polymerase (PARP) enzyme alleviates inflammation and necrosis in various pathologies. Here we aimed to evaluate the effects of PARP inhibition on PQ-induced lung damage in a rat experimental model. METHODS: Female Sprague-Dawley rats (n = 24) were allocated into three groups: sham, PQ and PQ + 3-aminobenzamide (3-AB) that is a PARP inhibitor, groups. Experimental lung injury was induced by administration of 15 mg/kg PQ intraperitoneally in PQ and PQ + 3-AB groups. 3-AB (10 mg/kg twice per day) was administered to the PQ + 3-AB group for four consecutive days. The animals were killed on the fifth day following PQ administration. Lung tissue and blood samples were collected and stored until analysis. RESULTS: Serum lactate dehydrogenase (LDH) and neopterin levels, tissue oxidative stress parameters, transforming growth factor-beta1 (TGF-ß) levels and histological injury scores in the PQ + 3-AB group were significantly lower than in the PQ group (P < 0.05, PQ vs. PQ + 3-AB). Total antioxidant capacity in the PQ + 3-AB group was significantly higher than in the PQ group (P < 0.05, PQ + 3-AB vs. PQ). CONCLUSION: Our results suggested that the use of PARP inhibitors following PQ toxicity might be useful for minimizing lung injury due to paraquat toxicity.


Assuntos
Benzamidas/uso terapêutico , Lesão Pulmonar/prevenção & controle , Paraquat/toxicidade , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Poli(ADP-Ribose) Polimerases/metabolismo , Animais , Antioxidantes/metabolismo , Benzamidas/administração & dosagem , Biomarcadores/sangue , Modelos Animais de Doenças , Feminino , Injeções Intraperitoneais , Peroxidação de Lipídeos/efeitos dos fármacos , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/enzimologia , Lesão Pulmonar/patologia , Neopterina/sangue , Inibidores de Poli(ADP-Ribose) Polimerases/administração & dosagem , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa