Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.158
Filtrar
1.
Res Vet Sci ; 172: 105254, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38582048

RESUMO

Two randomized crossover trials evaluated the effects of nicardipine constant rate infusion (CRI) on 1) the anesthetic potency of sevoflurane and 2) the ability to attenuate dexmedetomidine-induced cardiovascular depression in anesthetized dogs. First, six healthy Beagle dogs weighing 11.7 ± 0.9 kg were allocated to one of three treatments that administered a CRI of carrier (saline) or dexmedetomidine 0.5 or 3.0 µg/kg/h following a loading dose. The minimum alveolar concentration (MAC) of sevoflurane was determined utilizing electric stimuli before and after the loading dose of nicardipine (20 µg/kg intravenously for 10 min), followed by CRI at 40 µg/kg/h with 60 min of equilibration. Subsequently, cardiovascular and blood gas variables were evaluated in another trial under sevoflurane anesthesia at the individual 1.5 MAC. After baseline measurements, the dogs were assigned to two treatments (dexmedetomidine CRI at 0.5 or 3.0 µg/kg/h following a loading dose) with sevoflurane doses adjusted to 1.5 times of MAC equivalent, and the measurements were repeated every 15 min for 120 min. After 60 min, nicardipine CRI at 40 µg/kg/h with a loading dose was added to the dexmedetomidine CRI. Dexmedetomidine infusions significantly decreased the sevoflurane MAC but nicardipine did not significantly alter the MAC either with or without dexmedetomidine CRI in dogs. Dexmedetomidine dose-dependently decreased the cardiac index and increased the systemic vascular resistance index; these effects were fully counteracted by concomitant nicardipine CRI. Nicardipine CRI can be useful for controlling the cardiovascular depression elicited by dexmedetomidine in anesthetized dogs without affecting the anesthetic potency of sevoflurane.


Assuntos
Anestésicos Inalatórios , Dexmedetomidina , Nicardipino , Sevoflurano , Animais , Dexmedetomidina/farmacologia , Dexmedetomidina/administração & dosagem , Cães , Sevoflurano/farmacologia , Sevoflurano/administração & dosagem , Nicardipino/farmacologia , Nicardipino/administração & dosagem , Anestésicos Inalatórios/farmacologia , Anestésicos Inalatórios/administração & dosagem , Masculino , Estudos Cross-Over , Feminino , Alvéolos Pulmonares/efeitos dos fármacos , Infusões Intravenosas/veterinária , Frequência Cardíaca/efeitos dos fármacos , Hipnóticos e Sedativos/farmacologia , Hipnóticos e Sedativos/administração & dosagem , Pressão Sanguínea/efeitos dos fármacos
2.
Exp Cell Res ; 438(1): 114030, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38583855

RESUMO

Acute respiratory distress syndrome (ARDS) is a serious lung condition that often leads to hospitalization in intensive care units and a high mortality rate. Sevoflurane is a volatile anesthetic with growing interest for sedation in ventilated patients with ARDS. It has been shown to have potential lung-protective effects, such as reduced inflammation and lung edema, or improved arterial oxygenation. In this study, we investigated the effects of sevoflurane on lung injury in cultured human carcinoma-derived lung alveolar epithelial (A549) cells. We found that sevoflurane was associated with improved wound healing after exposure to inflammatory cytokines, with preserved cell proliferation but no effect on cell migration properties. Sevoflurane exposure was also associated with enhanced cell viability and active autophagy in A549 cells exposed to cytokines. These findings suggest that sevoflurane may have beneficial effects on lung epithelial injury by promoting alveolar epithelial wound healing and by influencing the survival and proliferation of A549 epithelial cells in vitro. Further research is needed to confirm these findings and to investigate the key cellular mechanisms explaining sevoflurane's potential effects on lung epithelial injury.


Assuntos
Proliferação de Células , Sobrevivência Celular , Síndrome do Desconforto Respiratório , Sevoflurano , Cicatrização , Sevoflurano/farmacologia , Humanos , Síndrome do Desconforto Respiratório/tratamento farmacológico , Síndrome do Desconforto Respiratório/patologia , Cicatrização/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células A549 , Proliferação de Células/efeitos dos fármacos , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Movimento Celular/efeitos dos fármacos , Anestésicos Inalatórios/farmacologia , Citocinas/metabolismo , Autofagia/efeitos dos fármacos , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia
3.
Biomed Pharmacother ; 174: 116447, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38518606

RESUMO

Sepsis-induced acute respiratory distress syndrome (ARDS) causes significant fatalities worldwide and lacks pharmacological intervention. Alveolar fluid clearance (AFC) plays a pivotal role in the remission of ARDS and is markedly impaired in the pathogenesis of ARDS. Here, we demonstrated that erythropoietin could effectively ameliorate lung injury manifestations and lethality, restore lung function and promote AFC in a rat model of lipopolysaccharide (LPS)-induced ARDS. Moreover, it was proven that EPO-induced restoration of AFC occurs through triggering the total protein expression of ENaC and Na,K-ATPase channels, enhancing their protein abundance in the membrane, and suppressing their ubiquitination for degeneration. Mechanistically, the data indicated the possible involvement of EPOR/JAK2/STAT3/SGK1/Nedd4-2 signaling in this process, and the pharmacological inhibition of the pathway markedly eliminated the stimulating effects of EPO on ENaC and Na,K-ATPase, and subsequently reversed the augmentation of AFC by EPO. Consistently, in vitro studies of alveolar epithelial cells paralleled with that EPO upregulated the expression of ENaC and Na,K-ATPase, and patch-clamp studies further demonstrated that EPO substantially strengthened sodium ion currents. Collectively, EPO could effectively promote AFC by improving ENaC and Na,K-ATPase protein expression and abundance in the membrane, dependent on inhibition of ENaC and Na,K-ATPase ubiquitination, and resulting in diminishing LPS-associated lung injuries.


Assuntos
Canais Epiteliais de Sódio , Eritropoetina , Ratos Sprague-Dawley , Síndrome do Desconforto Respiratório , Sepse , ATPase Trocadora de Sódio-Potássio , Ubiquitinação , Animais , Canais Epiteliais de Sódio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Eritropoetina/farmacologia , Sepse/complicações , Sepse/tratamento farmacológico , Sepse/metabolismo , Ubiquitinação/efeitos dos fármacos , Síndrome do Desconforto Respiratório/tratamento farmacológico , Síndrome do Desconforto Respiratório/metabolismo , Masculino , Ratos , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Lipopolissacarídeos , Transdução de Sinais/efeitos dos fármacos , Modelos Animais de Doenças
4.
Toxicol Appl Pharmacol ; 485: 116886, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38452946

RESUMO

Despite extensive preclinical testing, cancer therapeutics can result in unanticipated toxicity to non-tumor tissue in patients. These toxicities may pass undetected in preclinical experiments due to modeling limitations involving poor biomimicry of 2-dimensional in vitro cell cultures and due to lack of interspecies translatability in in vivo studies. Instead, primary cells can be grown into miniature 3-dimensional structures that recapitulate morphological and functional aspects of native tissue, termed "organoids." Here, human bronchioalveolar organoids grown from primary alveolar epithelial cells were employed to model lung epithelium and investigate off-target toxicities associated with antibody-drug conjugates (ADCs). ADCs with three different linker-payload combinations (mafodotin, vedotin, and deruxtecan) were tested in bronchioalveolar organoids generated from human, rat, and nonhuman primate lung cells. Organoids demonstrated antibody uptake and changes in viability in response to ADC exposure that model in vivo drug sensitivity. RNA sequencing identified inflammatory activation in bronchioalveolar cells in response to deruxtecan. Future studies will explore specific cell populations involved in interstitial lung disease and incorporate immune cells to the culture.


Assuntos
Imunoconjugados , Organoides , Organoides/efeitos dos fármacos , Organoides/patologia , Animais , Imunoconjugados/toxicidade , Humanos , Ratos , Avaliação Pré-Clínica de Medicamentos/métodos , Macaca fascicularis , Células Cultivadas , Testes de Toxicidade/métodos , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/patologia
5.
Food Chem Toxicol ; 161: 112829, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35093429

RESUMO

Particulate matter (PM) generally consists of aggregated particles containing trace metals and polycyclic aromatic hydrocarbons (PAHs). Cytochrome P450 (CYP) 1A1, one of the extensively investigated biomarkers, is highly inducible when PAHs activate the aryl hydrocarbon receptor (AhR). The present study focused on developing a LC-MS/MS-based assay to evaluate CYP1A1 induction potential following PM exposure. This assay adapted a CYP1A1 selective reaction of granisetron 7-hydroxylation in response to an AhR inducer, 6-formylindolo[3,2-b]carbazole (FICZ), in HepaRG and A549 cell lines. Exposure to FICZ (10 nM) increased the levels of granisetron 7-hydroxylation significantly, whereas no elevation of ethoxyresorufin-O-deethylation (EROD) activity was found in HepaRG cells. In A549 cells, granisetron 7-hydroxylation showed a better dose-response from 0 to 10000 nM FICZ treatment than EROD. EROD Additionally, the application of the assay with diesel PM exposure showed a concentration-dependent induction of CYP1A1 in HepaRG, A549, and human nasal epithelial cells. The granisetron assay has better selectivity for CYP1A1 than the conventional EROD assay, which is overlapped reaction with CYP1A2 and CYP1B1, with high correlations between AhR activation and CYP1A1 mRNA levels. Accompanying the great application potential to different organs and cell culture systems, future studies will implement the granisetron assay for the respiratory toxicity evaluation.


Assuntos
Cromatografia Líquida , Citocromo P-450 CYP1A1/metabolismo , Gasolina/análise , Granisetron/farmacologia , Espectrometria de Massas , Material Particulado/toxicidade , Linhagem Celular , Citocromo P-450 CYP1A1/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Humanos , Hidroxilação , Material Particulado/química , Alvéolos Pulmonares/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
6.
Biomed Pharmacother ; 143: 112209, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34649343

RESUMO

BACKGROUND: Alveolar hypercoagulation and fibrinolytic inhibition are important characteristics during acute respiratory distress syndrome (ARDS), and NF-κB p65 signaling pathway is involved to regulate these pathophysiologies. We hypothesize that targeting NF-κB signal pathway could ameliorate alveolar hypercoagulation and fibrinolyitc inhibition, thus attenuating lung injury in ARDS. PURPOSE: We explore the efficacy and the potential mechanism of andrographolide sulfonate (Andro-S) on alveolar hypercoagulation and fibrinolytic inhibition in LPS-induced ARDS in mice. METHODS: ARDS was made by lipopolysaccharide (LPS) inhalation in C57BLmice. Andrographolide sulfonate (2.5, 5 and 10 mg/kg) was intraperitoneally given to the mice (once a day for three consecutive days) before LPS administration. NEMO binding domain peptide (NBD), an inhibitor of NF-κB, was used as the positive control and it replaced Andro-S in mice of NBD group. Mice in normal control received saline instead of LPS. Lung tissues and bronchoalveolar lavage fluid (BALF) were collected for analysis of alveolar coagulation, fibrinolytic inhibition as well as of pulmonary inflammatory response after 8 h of LPS inhalation. NF-κB signal pathway in lung tissue was simultaneously determined. RESULTS: Andro-S dose-dependently inhibited tissue factor (TF) and plasminogen activator inhibitor (PAI)-1 expressions either in mRNA or in protein in lung tissue of ARDS mice, and it also decreased the concentrations of TF, PAI-1, thrombin-antithrombin complex (TAT), procollagen peptide type Ⅲ (PⅢP) while promoting the production of activated protein C (APC) in BALF. Meanwhile, Andro-S effectively inhibited inflammatory response (interleukin 1ß and myeloperoxidase) induced by LPS. LPS stimulation dramatically activated NF-κB signal pathway, indicated by increased expressions of phosphorylation of p65 (p-p65), p-IKKα/ß and p-IκBα and the higher p65-DNA binding activity, which were all dose-dependently reversed by Andro-S. Andro-S and NBD presented similar efficacies. CONCLUSIONS: Andro-S treatment improves alveolar hypercoagulation and fibrinolytic inhibition and attenuates pulmonary inflammation in LPS-induced ARDS in mice partly through NF-κB pathway inactivation. The drug is expected to be an effective choice for ARDS.


Assuntos
Anti-Inflamatórios/farmacologia , Diterpenos/farmacologia , Fibrinólise/efeitos dos fármacos , Fibrinolíticos/farmacologia , NF-kappa B/metabolismo , Alvéolos Pulmonares/efeitos dos fármacos , Síndrome do Desconforto Respiratório/tratamento farmacológico , Trombofilia/tratamento farmacológico , Animais , Fatores de Coagulação Sanguínea/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos , Masculino , Camundongos Endogâmicos C57BL , Fosforilação , Alvéolos Pulmonares/metabolismo , Síndrome do Desconforto Respiratório/sangue , Síndrome do Desconforto Respiratório/induzido quimicamente , Transdução de Sinais , Trombofilia/sangue , Trombofilia/induzido quimicamente
7.
Toxicol Lett ; 352: 61-69, 2021 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-34624459

RESUMO

Mitomycin treatment induces pulmonary toxicity, and alveolar epithelial cell senescence is crucial in the pathogenesis of the latter. However, the mechanism by which mitomycin induces alveolar epithelial cell senescence has yet to be elucidated. In this work, different doses (37.5-300 nM) of mitomycin induced the senescence of human alveolar type II-like epithelial cells and enhanced the phosphorylation of GSK3ß (S9). The GSK3ß (S9A) mutant reversed the senescence of mitomycin-treated alveolar epithelial cells. Pharmacological inhibition and gene deletion of Akt1, a kinase that regulates the phosphorylation of GSK3ß (S9), suppressed mitomycin-induced alveolar epithelial cell senescence. The knockdown of p53, a downstream effector of GSK3ß and an important regulator of cell senescence, repressed mitomycin-induced alveolar epithelial cell senescence. Treatment with baicalein weakened the phosphorylation of GSK3ß (S9) and alleviated the senescence of alveolar epithelial cells brought about by mitomycin treatment. GSK3ß (S9) phosphorylation appears to be the first signal involved in the mitomycin-induced senescence of alveolar epithelial cells and may present a potential target for attenuating mitomycin-induced pulmonary toxicity.


Assuntos
Alquilantes/toxicidade , Regulação para Baixo/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/metabolismo , Mitomicina/toxicidade , Alvéolos Pulmonares/efeitos dos fármacos , Células A549 , Senescência Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Flavanonas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/genética , Humanos , Imidazóis/farmacologia , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Alvéolos Pulmonares/citologia , Piridinas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
J Pharmacol Exp Ther ; 379(2): 156-165, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34465632

RESUMO

Acute respiratory distress syndrome (ARDS), a common and fatal clinical condition, is characterized by the destruction of epithelium and augmented permeability of the alveolar-capillary barrier. Resolvin conjugates in tissue regeneration 1 (RCTR1) is an endogenous lipid mediator derived from docosahexaenoic acid , exerting proresolution effects in the process of inflammation. In our research, we evaluated the role of RCTR1 in alveolar fluid clearance (AFC) in lipopolysaccharide-induced ARDS/acute lung injury (ALI) rat model. Rats were injected with RCTR1 (5 µg/kg) via caudal veins 8 hours after lipopolysaccharide (LPS) (14 mg/kg) treatment, and then AFC was estimated after 1 hour of ventilation. Primary type II alveolar epithelial cells were incubated with LPS (1 ug/ml) with or without RCTR1 (10 nM) for 8 hours. Our results showed that RCTR1 significantly enhanced the survival rate, promoted the AFC, and alleviated LPS-induced ARDS/ALI in vivo. Furthermore, RCTR1 remarkably elevated the protein expression of sodium channels and Na, K-ATPase and the activity of Na, K-ATPase in vivo and in vitro. Additionally, RCTR1 also decreased neural precursor cell expressed developmentally downregulated 4-2 (Nedd4-2) level via upregulating Ser473-phosphorylated-Akt expression. Besides this, inhibitors of receptor for lipoxin A4 (ALX), cAMP, and phosphatidylinositol 3-kinase (PI3K) (BOC-2, KH-7, and LY294002) notably inhibited the effects of RCTR1 on AFC. In summary, RCTR1 enhances the protein levels of sodium channels and Na, K-ATPase and the Na, K-ATPase activity to improve AFC in ALI through ALX/cAMP/PI3K/Nedd4-2 pathway, suggesting that RCTR1 may become a therapeutic drug for ARDS/ALI. SIGNIFICANCE STATEMENT: RCTR1, an endogenous lipid mediator, enhanced the rate of AFC to accelerate the resolution of inflammation in the LPS-induced murine lung injury model. RCTR1 upregulates the expression of epithelial sodium channels (ENaCs) and Na, K-ATPase in vivo and in vitro to accelerate the AFC. The efficacy of RCTR1 on the ENaC and Na, K-ATPase level was in an ALX/cAMP/PI3K/Nedd4-2-dependent manner.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Ácidos Docosa-Hexaenoicos/farmacologia , Agonistas do Canal de Sódio Epitelial/farmacologia , Canais Epiteliais de Sódio/metabolismo , Alvéolos Pulmonares/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Animais , Ácidos Docosa-Hexaenoicos/análogos & derivados , Ácidos Docosa-Hexaenoicos/uso terapêutico , Lipopolissacarídeos/toxicidade , Masculino , Alvéolos Pulmonares/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
9.
Comput Math Methods Med ; 2021: 7710129, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34471421

RESUMO

OBJECTIVE: We aimed to explore the level of PS, cell viability, inflammatory factors, and apoptosis in neonatal respiratory distress syndrome (ARDS). Besides, we explored the potential relationship between ACE2, SIRT1/eNOS pathway, and hypoxia-induced AT II cell damage. METHODS: The hUC-MSC-derived AT II cells were verified by IF and ICC, whereas qRT-PCR was used for PS and AT II cell marker (CK-8 and KGF). The AT II cell damage model was established by hypoxia exposure. The enhanced expression of ACE2 was tested after transfection with pcDNA3.1-ACE2 by western blot. The effects of hypoxia and ACE2 on AT II cells were evaluated by MTT, western blot, ELISA, and flow cytometry. The involvement of the SIRT1/eNOS pathway in AT II cell's protective functions against NRDS was verified with the addition of SIRT1 inhibitor EX527. RESULTS: Based on the successful differentiation of AT II cells from hUC-MSCs and the buildup of AT II cell damage model, the overexpressed ACE2 impeded the hypoxia-induced cellular damage of AT II cells. It also counteracted the inhibitory effects of hypoxia on the secretion of PS. Overexpression of ACE2 rescued the cell viability and suppressed the secretion of inflammatory cytokines and the apoptosis of AT II cells triggered by hypoxia. And ACE2 activated the SIRT1/eNOS pathway to play its cell-protective and anti-inflammatory roles. CONCLUSION: Our findings provided information that ACE2 prevented AT II cells from inflammatory damage through activating the SIRT1/eNOS pathway, which suggested that ACE2 might become a novel protective agent applied in the protection and treatment of NRDS.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Alvéolos Pulmonares/lesões , Alvéolos Pulmonares/metabolismo , Surfactantes Pulmonares/metabolismo , Sirtuína 1/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Apoptose , Carbazóis/farmacologia , Diferenciação Celular , Hipóxia Celular , Sobrevivência Celular , Células Cultivadas , Biologia Computacional , Feminino , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Redes e Vias Metabólicas/efeitos dos fármacos , Alvéolos Pulmonares/efeitos dos fármacos , Síndrome do Desconforto Respiratório do Recém-Nascido/etiologia , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/prevenção & controle , Sirtuína 1/antagonistas & inibidores , Regulação para Cima
10.
Int Immunopharmacol ; 100: 108077, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34464887

RESUMO

BACKGROUND: Disruption of alveolar endothelial barrier caused by inflammation drives the progression of septic acute lung injury (ALI). Pravastatin, an inhibitor of HMG Co-A reductase, has potent anti-inflammatory effects. In the present study, we aim to explore the beneficial role of pravastatin in sepsis-induced ALI and its related mechanisms. METHODS: A septic ALI model was established by cecal ligation and puncture (CLP) in mice. The pulmonary microvascular endothelial cells (PMVECs) were challenged with lipopolysaccharide (LPS). The pathological changes in lung tissues were examined by HE staining. The pulmonary microvascular permeability was determined by lung wet-to-dry (W/D) weight ratio and Evans blue staining. The total protein concentration in bronchoalveolar lavage fluid (BALF) was detected by BCA assay. The levels of TNF-α, IL-1ß, and IL-6 were assessed by qRT-PCR and ELISA. Apoptosis was determined by flow cytometry and TUNEL. Western blotting was performed for detection of target protein levels. The expression of VE-Cadherin in lung tissues was evaluated by immunohistochemical staining. RESULTS: Pravastatin improved survival rate, attenuated lung pathological changes and reduced pulmonary microvascular permeability in septic mice. In addition, pravastatin restrained sepsis-induced inflammatory response and apoptosis in the lung tissues and PMVECs. Moreover, pravastatin up-regulated the levels of junction proteins ZO-1, JAM-C, and VE-Cadherin. Finally, pravastatin suppressed inflammation, apoptosis and enhanced the expression of junction proteins via regulating Cav-1/eNOS signaling pathway in LPS-exposed PMVECs. CONCLUSION: Pravastatin ameliorates sepsis-induced ALI through improving alveolar endothelial barrier disruption via modulating Cav-1/eNOS pathway, which may be an effective candidate for treating septic ALI.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Células Endoteliais/efeitos dos fármacos , Pravastatina/farmacologia , Sepse/tratamento farmacológico , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Líquido da Lavagem Broncoalveolar/imunologia , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/imunologia , Caveolina 1/metabolismo , Modelos Animais de Doenças , Células Endoteliais/imunologia , Células Endoteliais/patologia , Humanos , Masculino , Camundongos , Óxido Nítrico Sintase Tipo III/metabolismo , Pravastatina/uso terapêutico , Alvéolos Pulmonares/irrigação sanguínea , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/patologia , Sepse/complicações , Sepse/imunologia , Sepse/patologia
11.
Aging (Albany NY) ; 13(13): 16922-16937, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34238764

RESUMO

Idiopathic pulmonary fibrosis (IPF) is an age-related disorder that carries a universally poor prognosis and is thought to arise from repetitive micro injuries to the alveolar epithelium. To date, a major factor limiting our understanding of IPF is a deficiency of disease models, particularly in vitro models that can recapitulate the full complement of molecular attributes in the human condition. In this study, we aimed to develop a model that more closely resembles the aberrant IPF lung epithelium. By exposing mouse alveolar epithelial cells to repeated, low doses of bleomycin, instead of usual one-time exposures, we uncovered changes strikingly similar to those in the IPF lung epithelium. This included the acquisition of multiple phenotypic and functional characteristics of senescent cells and the adoption of previously described changes in mitochondrial homeostasis, including alterations in redox balance, energy production and activity of the mitochondrial unfolded protein response. We also uncovered dramatic changes in cellular metabolism and detected a profound loss of proteostasis, as characterized by the accumulation of cytoplasmic protein aggregates, dysregulated expression of chaperone proteins and decreased activity of the ubiquitin proteasome system. In summary, we describe an in vitro model that closely resembles the aberrant lung epithelium in IPF. We propose that this simple yet powerful tool could help uncover new biological mechanisms and assist in developing new pharmacological tools to treat the disease.


Assuntos
Fibrose Pulmonar Idiopática/patologia , Pulmão/crescimento & desenvolvimento , Pulmão/patologia , Mucosa Respiratória/crescimento & desenvolvimento , Mucosa Respiratória/patologia , Animais , Antibióticos Antineoplásicos/toxicidade , Bleomicina/toxicidade , Linhagem Celular , Senescência Celular , Modelos Animais de Doenças , Metabolismo Energético , Homeostase , Humanos , Camundongos , Mitocôndrias/metabolismo , Oxirredução , Complexo de Endopeptidases do Proteassoma , Proteínas/metabolismo , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia , Resposta a Proteínas não Dobradas
12.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166204, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34175430

RESUMO

Ferroptosis is a newly discovered type of regulated cell death, characterized by the iron-dependent accumulation of lipid reactive oxygen species, which has been implicated in numerous human diseases. However, its role in pulmonary fibrosis, a fatal lung disease with unknown etiology, is largely unknown. Here, we investigated the role of ferroptosis in pulmonary fibrosis. We found a large amount of iron deposition in the lung tissue of patients with pulmonary fibrosis. We observed ferroptosis in alveolar type II (ATII) cells, fibrotic lung tissues of BLM-induced pulmonary fibrosis mice. BLM-induced increase in iron level was accompanied by pathological changes, collagen deposition, and ferroptosis in ATII cells, indicating iron deposition-induced ferroptosis, which promoted the development of pulmonary fibrosis. Moreover, deferoxamine (DFO) completely prevented the pro-fibrosis effects of BLM by reducing iron deposition and ferroptosis in ATII cells. Genes associated with intracellular iron metabolism and homeostasis, such as transferrin receptor 1, divalent metal transporter 1, and ferroportin-1, and showed abnormal expression levels in animal tissues and lung epithelial MLE-12 cells, which responded to BLM stimulation. Overall, we demonstrated that BLM-induced iron deposition in MLE-12 cells is prone to both mitochondrial dysfunction and ferroptosis and that DFO reverses this phenotype. In the future, understanding the role of ferroptosis may shed new light on the etiology of pulmonary fibrosis. Ferroptosis inhibitors or genetic engineering of ferroptosis-related genes might offer potential targets to treat pulmonary fibrosis.


Assuntos
Ferroptose/efeitos dos fármacos , Ferro/metabolismo , Pulmão/metabolismo , Fibrose Pulmonar/metabolismo , Animais , Bleomicina/toxicidade , Proteínas de Transporte de Cátions/genética , Desferroxamina/farmacologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Ferro/efeitos adversos , Pulmão/efeitos dos fármacos , Pulmão/patologia , Camundongos , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/patologia , Espécies Reativas de Oxigênio/metabolismo , Receptores da Transferrina/genética
13.
Int J Mol Sci ; 22(10)2021 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-34069552

RESUMO

There is little in vitro data available on long-term effects of TiO2 exposure. Such data are important for improving the understanding of underlying mechanisms of adverse health effects of TiO2. Here, we exposed pulmonary epithelial cells to two doses (0.96 and 1.92 µg/cm2) of TiO2 for 13 weeks and effects on cell cycle and cell death mechanisms, i.e., apoptosis and autophagy were determined after 4, 8 and 13 weeks of exposure. Changes in telomere length, cellular protein levels and lipid classes were also analyzed at 13 weeks of exposure. We observed that the TiO2 exposure increased the fraction of cells in G1-phase and reduced the fraction of cells in G2-phase, which was accompanied by an increase in the fraction of late apoptotic/necrotic cells. This corresponded with an induced expression of key apoptotic proteins i.e., BAD and BAX, and an accumulation of several lipid classes involved in cellular stress and apoptosis. These findings were further supported by quantitative proteome profiling data showing an increase in proteins involved in cell stress and genomic maintenance pathways following TiO2 exposure. Altogether, we suggest that cell stress response and cell death pathways may be important molecular events in long-term health effects of TiO2.


Assuntos
Células Epiteliais Alveolares/metabolismo , Titânio/efeitos adversos , Células Epiteliais Alveolares/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Divisão Celular , Linhagem Celular , Células Epiteliais/metabolismo , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Humanos , Pulmão/metabolismo , Nanopartículas Metálicas/efeitos adversos , Nanopartículas/efeitos adversos , Estresse Oxidativo/efeitos dos fármacos , Proteômica/métodos , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Titânio/metabolismo , Transcriptoma/genética
14.
Innate Immun ; 27(4): 294-312, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34000873

RESUMO

In China, baicalin is the main active component of Scutellaria baicalensis, which has been used in the treatment of inflammation-related diseases, such as inflammation-induced acute lung injury. However, its specific mechanism remains unclear. This study examined the protective effect of baicalin on LPS-induced inflammation injury of alveolar epithelial cell line A549 and explored its protective mechanism. Compared with the LPS-induced group, the proliferation inhibition rates of alveolar type II epithelial cell line A549 intervened by different concentrations of baicalin decreased significantly, as did the levels of inflammatory factors IL-6, IL-1ß, prostaglandin 2 and TNF-α in the supernatant. The expression levels of inflammatory proteins inducible NO synthase (iNOS), NF-κB65, phosphorylated ERK (p-ERK1/2), and phosphorylated c-Jun N-terminal kinase (p-JNK1) significantly decreased, as did the protein expression of follistatin-like protein 1 (FSTL1). In contrast, expression of miR-200b-3p significantly increased in a dose-dependent manner. These results suggested that baicalin could significantly inhibit the expression of inflammation-related proteins and improve LPS-induced inflammatory injury in alveolar type II epithelial cells. The mechanism may be related to the inhibition of ERK/JNK inflammatory pathway activation by increasing the expression of miR-200b-3p. Thus, FSTL1 is the regulatory target of miR-200b-3p.


Assuntos
Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Flavonoides/uso terapêutico , Proteínas Relacionadas à Folistatina/efeitos dos fármacos , Lipopolissacarídeos , MicroRNAs/biossíntese , Alvéolos Pulmonares/lesões , Transdução de Sinais/efeitos dos fármacos , Células A549 , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Inflamação/induzido quimicamente , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , MicroRNAs/efeitos dos fármacos , MicroRNAs/genética , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia
15.
Biomolecules ; 11(5)2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33946440

RESUMO

Cell's microenvironment has been shown to exert influence on cell behavior. In particular, matrix-cell interactions strongly impact cell morphology and function. The purpose of this study was to analyze the influence of different culture substrate materials on phenotype and functional properties of lung epithelial adenocarcinoma (A549) cells. A549 cells were seeded onto two different biocompatible, commercially available substrates: a polyester coverslip (Thermanox™ Coverslips), that was used as cell culture plate control, and a polydimethylsiloxane membrane (PDMS, Elastosil® Film) investigated in this study as alternative material for A549 cells culture. The two substrates influenced cell morphology and the actin cytoskeleton organization. Further, the Yes-associated protein (YAP) and its transcriptional coactivator PDZ-binding motif (TAZ) were translocated to the nucleus in A549 cells cultured on polyester substrate, yet it remained mostly cytosolic in cells on PDMS substrate. By SEM analysis, we observed that cells grown on Elastosil® Film maintained an alveolar Type II cell morphology. Immunofluorescence staining for surfactant-C revealing a high expression of surfactant-C in cells cultured on Elastosil® Film, but not in cells cultured on Thermanox™ Coverslips. A549 cells grown onto Elastosil® Film exhibited morphology and functionality that suggest retainment of alveolar epithelial Type II phenotype, while A549 cells grown onto conventional plastic substrates acquired an alveolar Type I phenotype.


Assuntos
Células Epiteliais Alveolares/citologia , Células Epiteliais Alveolares/efeitos dos fármacos , Dimetilpolisiloxanos/farmacologia , Poliésteres/farmacologia , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/efeitos dos fármacos , Células A549 , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Técnicas de Cultura de Células/métodos , Linhagem Celular Tumoral , Meios de Cultura , Humanos , Lipopeptídeos/biossíntese , Pulmão/citologia , Pulmão/efeitos dos fármacos , Microscopia Eletroquímica de Varredura , Peptídeos Cíclicos/biossíntese , Fatores de Transcrição/biossíntese , Proteínas de Sinalização YAP
16.
Allergol Immunopathol (Madr) ; 49(3): 91-99, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33938193

RESUMO

INTRODUCTION AND OBJECTIVES: There are a few reports in the literature about the successful use of sugammadex in the treatment of hypersensitivity reactions caused by rocuronium; however, the pathophysiological mechanism is still unknown. This study aims to investigate the changes caused by rocuronium in the lung and the effect of sugammadex on these changes with biochemical, light microscopic and immunohistochemical parameters on a rat model. MATERIALS AND METHODS: For the study, 28-male Sprague-Dawley rats were randomly divided, seven of each, into four groups. Group C (control) received only 0. 9 % NaCl without any drug. Group R received rocuronium alone 1mg/kg. Group S received sugammadex alone 96 mg/kg. Group RS received rocuronium 1mg/kg and sugammadex 96 mg/kg. After 24 h later, the animals were sacrificed and their tissues were removed. Biochemical (IgE/CRP), light microscopic and immunohistochemical findings were recorded. RESULTS: Immunoglobulin E and CRP levels, peribronchial, alveolar septal lymphocytic infiltration, thickening of the alveolar membranes and bleeding sites in Group R were significantly higher than all the other groups. In Group RS, while these parameters were significantly lower than that of Group R and Group S, it was significantly higher than that of Group C. Total mast cells and tryptase-positive mast cells counts were significantly higher in Group R than in all other groups. In Group RS, these parameters were statistically lower than that of Group R and Group S, but higher than that of Group C. CONCLUSIONS: This study shows that allergic inflammatory changes due to rocuronium in the lungs of rats are reduced with sugammadex. These results support cases of anaphylaxis due to rocuronium which improved with sugammadex.


Assuntos
Hipersensibilidade/complicações , Inflamação/prevenção & controle , Pulmão/efeitos dos fármacos , Fármacos Neuromusculares não Despolarizantes/efeitos adversos , Rocurônio/efeitos adversos , Sugammadex/farmacologia , Anafilaxia/induzido quimicamente , Anafilaxia/prevenção & controle , Animais , Proteína C-Reativa/análise , Modelos Animais de Doenças , Hemorragia/induzido quimicamente , Imunoglobulina E/análise , Inflamação/induzido quimicamente , Inflamação/imunologia , Linfócitos , Masculino , Mastócitos/citologia , Mastócitos/enzimologia , Fármacos Neuromusculares não Despolarizantes/antagonistas & inibidores , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Rocurônio/antagonistas & inibidores , Triptases/análise
17.
Int J Mol Sci ; 22(6)2021 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-33805743

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a lethal age-related lung disease whose pathogenesis involves an aberrant response of alveolar epithelial cells (AEC). Activated epithelial cells secrete mediators that participate in the activation of fibroblasts and the excessive deposition of extracellular matrix proteins. Previous studies indicate that matrix metalloproteinase 14 (MMP14) is increased in the lung epithelium in patients with IPF, however, the role of this membrane-type matrix metalloproteinase has not been elucidated. In this study, the role of Mmp14 was explored in experimental lung fibrosis induced with bleomycin in a conditional mouse model of lung epithelial MMP14-specific genetic deletion. Our results show that epithelial Mmp14 deficiency in mice increases the severity and extension of fibrotic injury and affects the resolution of the lesions. Gain-and loss-of-function experiments with human epithelial cell line A549 demonstrated that cells with a deficiency of MMP14 exhibited increased senescence-associated markers. Moreover, conditioned medium from these cells increased fibroblast expression of fibrotic molecules. These findings suggest a new anti-fibrotic mechanism of MMP14 associated with anti-senescent activity, and consequently, its absence results in impaired lung repair. Increased MMP14 in IPF may represent an anti-fibrotic mechanism that is overwhelmed by the strong profibrotic microenvironment that characterizes this disease.


Assuntos
Células Epiteliais/patologia , Fibrose Pulmonar Idiopática/genética , Metaloproteinase 14 da Matriz/genética , Alvéolos Pulmonares/metabolismo , Células A549 , Actinas/genética , Actinas/metabolismo , Animais , Bleomicina/administração & dosagem , Senescência Celular/genética , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Regulação da Expressão Gênica , Humanos , Hipoxantina Fosforribosiltransferase/genética , Hipoxantina Fosforribosiltransferase/metabolismo , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Metaloproteinase 14 da Matriz/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Cultura Primária de Células , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia , Succinato Desidrogenase/genética , Succinato Desidrogenase/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
18.
Int J Mol Sci ; 22(5)2021 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-33800016

RESUMO

Carbon-based nanomaterials are nowadays attracting lots of attention, in particular in the biomedical field, where they find a wide spectrum of applications, including, just to name a few, the drug delivery to specific tumor cells and the improvement of non-invasive imaging methods. Nanoparticles inhaled during breathing accumulate in the lung alveoli, where they interact and are covered with lung surfactants. We recently demonstrated that an apparently non-toxic concentration of engineered carbon nanodiamonds (ECNs) is able to induce oxidative/nitrosative stress, imbalance of energy metabolism, and mitochondrial dysfunction in microglial and alveolar basal epithelial cells. Therefore, the complete understanding of their "real" biosafety, along with their possible combination with other molecules mimicking the in vivo milieu, possibly allowing the modulation of their side effects becomes of utmost importance. Based on the above, the focus of the present work was to investigate whether the cellular alterations induced by an apparently non-toxic concentration of ECNs could be counteracted by their incorporation into a synthetic lung surfactant (DPPC:POPG in 7:3 molar ratio). By using two different cell lines (alveolar (A549) and microglial (BV-2)), we were able to show that the presence of lung surfactant decreased the production of ECNs-induced nitric oxide, total reactive oxygen species, and malondialdehyde, as well as counteracted reduced glutathione depletion (A549 cells only), ameliorated cell energy status (ATP and total pool of nicotinic coenzymes), and improved mitochondrial phosphorylating capacity. Overall, our results on alveolar basal epithelial and microglial cell lines clearly depict the benefits coming from the incorporation of carbon nanoparticles into a lung surfactant (mimicking its in vivo lipid composition), creating the basis for the investigation of this combination in vivo.


Assuntos
Microglia/efeitos dos fármacos , Nanopartículas/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Alvéolos Pulmonares/efeitos dos fármacos , Surfactantes Pulmonares/metabolismo , 1,2-Dipalmitoilfosfatidilcolina/química , 1,2-Dipalmitoilfosfatidilcolina/metabolismo , Células A549 , Animais , Carbono/química , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Glutationa/metabolismo , Humanos , Camundongos , Microglia/citologia , Microglia/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Nanopartículas/administração & dosagem , Nanopartículas/química , Fosfatidilgliceróis/química , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/metabolismo , Surfactantes Pulmonares/química , Espécies Reativas de Oxigênio/metabolismo , Testes de Toxicidade Subcrônica/métodos
19.
Cell Biochem Biophys ; 79(2): 311-320, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33683657

RESUMO

The proliferation of fetal alveolar type II cells (FATIICs) was impaired in bronchopulmonary dysplasia (BPD), which is modulated by hyperoxia and inflammatory response. Interleukin 24 (IL-24), a cytokine produced by certain cell types, plays an essential role in inflammation and host protection against infection. However, the ability of FATIICs to produce IL-24 remains unclear, and the role of IL-24 in BPD progression is yet to be determined. With reverse transcription quantitative polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay, the authors evaluated whether FATIICs produce IL-24 in physiological conditions. The authors quantified IL-24 expression in the lungs of newborn rat pups exposed to hyperoxia (70% oxygen) and in FATIICs isolated on embryonic day 19 that were exposed to 95% oxygen or lipopolysaccharide (LPS). The role of IL-24 in FATIICs, cell proliferation, cell apoptosis, and cell cycle were further evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and flow cytometric analysis. Also, they assessed caspase-3 and SOCS3 mRNA in IL-24 siRNA-treated cells by using RT-qPCR. During culture, IL-24 mRNA and protein levels in FATIICs gradually decreased with FATIIC differentiation. IL-24 expression increased significantly in rat lungs exposed to hyperoxia and FATIICs exposed to oxygen or LPS. Recombinant IL-24 enhanced cell proliferation by decreasing the proportion of apoptotic cells and increasing the proportion of cells in the S phase. The IL-24 siRNA-treated cells expressed more caspase-3 mRNA. Furthermore, suppressor of cytokine signaling 3 (SOCS3) mRNA was significantly decreased in rats and FATIICs exposed to oxygen, whereas it dramatically increased in FATIICs exposed to LPS. The IL-24 siRNA-treated cells expressed more SOCS3 mRNA. These studies suggest IL-24 is a pulmonary target cytokine in BPD, and may possibly regulate SOCS3 in oxidative stress and inflammation of the lung.


Assuntos
Displasia Broncopulmonar/patologia , Interleucinas/metabolismo , Animais , Displasia Broncopulmonar/metabolismo , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Feminino , Hiperóxia , Interleucinas/antagonistas & inibidores , Interleucinas/genética , Lipopolissacarídeos/farmacologia , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos , Proteína 3 Supressora da Sinalização de Citocinas/genética , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Regulação para Cima/efeitos dos fármacos
20.
J Am Heart Assoc ; 10(5): e018952, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33634705

RESUMO

Background Preclinical studies suggest that volatile anesthetics decrease infarct volume and improve the outcome of ischemic stroke. This study aims to determine their effect during noncardiac surgery on postoperative ischemic stroke incidence. Methods and Results This was a retrospective cohort study of surgical patients undergoing general anesthesia at 2 tertiary care centers in Boston, MA, between October 2005 and September 2017. Exclusion criteria comprised brain death, age <18 years, cardiac surgery, and missing covariate data. The exposure was defined as median age-adjusted minimum alveolar concentration of all intraoperative measurements of desflurane, sevoflurane, and isoflurane. The primary outcome was postoperative ischemic stroke within 30 days. Among 314 932 patients, 1957 (0.6%) experienced the primary outcome. Higher doses of volatile anesthetics had a protective effect on postoperative ischemic stroke incidence (adjusted odds ratio per 1 minimum alveolar concentration increase 0.49, 95% CI, 0.40-0.59, P<0.001). In Cox proportional hazards regression, the effect was observed for 17 postoperative days (postoperative day 1: hazard ratio (HR), 0.56; 95% CI, 0.48-0.65; versus day 17: HR, 0.85; 95% CI, 0.74-0.99). Volatile anesthetics were also associated with lower stroke severity: Every 1-unit increase in minimum alveolar concentration was associated with a 0.006-unit decrease in the National Institutes of Health Stroke Scale (95% CI, -0.01 to -0.002, P=0.002). The effects were robust throughout various sensitivity analyses including adjustment for anesthesia providers as random effect. Conclusions Among patients undergoing noncardiac surgery, volatile anesthetics showed a dose-dependent protective effect on the incidence and severity of early postoperative ischemic stroke.


Assuntos
Anestesia Geral/efeitos adversos , Desflurano/efeitos adversos , AVC Isquêmico/epidemiologia , Isoflurano/efeitos adversos , Complicações Pós-Operatórias/epidemiologia , Alvéolos Pulmonares/metabolismo , Sevoflurano/efeitos adversos , Anestésicos Inalatórios/efeitos adversos , Anestésicos Inalatórios/farmacocinética , Desflurano/farmacocinética , Relação Dose-Resposta a Droga , Feminino , Seguimentos , Humanos , Incidência , AVC Isquêmico/diagnóstico , AVC Isquêmico/etiologia , Isoflurano/farmacocinética , Masculino , Massachusetts/epidemiologia , Pessoa de Meia-Idade , Complicações Pós-Operatórias/diagnóstico , Complicações Pós-Operatórias/etiologia , Alvéolos Pulmonares/efeitos dos fármacos , Estudos Retrospectivos , Índice de Gravidade de Doença , Sevoflurano/farmacocinética , Volatilização
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...