Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Medicinas Complementares
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Med Res ; 29(1): 234, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38622728

RESUMO

BACKGROUND: Influenza is an acute respiratory infection caused by influenza virus. Maxing Shigan Decoction (MXSGD) is a commonly used traditional Chinese medicine prescription for the prevention and treatment of influenza. However, its mechanism remains unclear. METHOD: The mice model of influenza A virus pneumonia was established by nasal inoculation. After 3 days of intervention, the lung index was calculated, and the pathological changes of lung tissue were detected by HE staining. Firstly, transcriptomics technology was used to analyze the differential genes and important pathways in mouse lung tissue regulated by MXSGD. Then, real-time fluorescent quantitative PCR (RT-PCR) was used to verify the changes in mRNA expression in lung tissues. Finally, intestinal microbiome and intestinal metabolomics were performed to explore the effect of MXSGD on gut microbiota. RESULTS: The lung inflammatory cell infiltration in the MXSGD group was significantly reduced (p < 0.05). The results of bioinformatics analysis for transcriptomics results show that these genes are mainly involved in inflammatory factors and inflammation-related signal pathways mediated inflammation biological modules, etc. Intestinal microbiome showed that the intestinal flora Actinobacteriota level and Desulfobacterota level increased in MXSGD group, while Planctomycetota in MXSGD group decreased. Metabolites were mainly involved in primary bile acid biosynthesis, thiamine metabolism, etc. This suggests that MXSGD has a microbial-gut-lung axis regulation effect on mice with influenza A virus pneumonia. CONCLUSION: MXSGD may play an anti-inflammatory and immunoregulatory role by regulating intestinal microbiome and intestinal metabolic small molecules, and ultimately play a role in the treatment of influenza A virus pneumonia.


Assuntos
Alphainfluenzavirus , Medicamentos de Ervas Chinesas , Vírus da Influenza A , Influenza Humana , Orthomyxoviridae , Pneumonia , Camundongos , Animais , Humanos , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Influenza Humana/tratamento farmacológico , Influenza Humana/genética , Pneumonia/tratamento farmacológico , Pneumonia/genética , Inflamação , Biologia de Sistemas , Perfilação da Expressão Gênica
2.
Am J Physiol Lung Cell Mol Physiol ; 324(4): L536-L549, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36852927

RESUMO

Interstitial macrophages (IMs) reside in the lung tissue surrounding key structures including airways, vessels, and alveoli. Recent work has described IM heterogeneity during homeostasis, however, there are limited data on IMs during inflammation. We sought to characterize IM origin, subsets, and transcriptomic profiles during homeostasis and lipopolysaccharide (LPS) induced acute lung inflammation. During homeostasis, we used three complementary methods, spectral flow cytometry, single-cell RNA-sequencing, and gene regulatory network enrichment, to demonstrate that IMs can be divided into two core subsets distinguished by surface and transcriptional expression of folate receptor ß (Folr2/FRß). These subsets inhabited distinct niches within the lung interstitium. Within FRß+ IMs we identified a subpopulation marked by coexpression of LYVE1. During acute LPS-induced inflammation, lung IM numbers expand. Lineage tracing revealed IM expansion was due to recruitment of monocyte-derived IMs. At the peak of inflammation, recruited IMs were comprised two unique subsets defined by expression of genes associated with interferon signaling and glycolytic pathways. As recruited IMs matured, they adopted the overall transcriptional state of FRß- resident IMs but retained expression in several origin-specific genes, such as IL-1ß. FRß+ IMs were of near-pure resident origin. Taken together our data show that during LPS-induced inflammation, there are distinct populations of IMs that likely have unique functions. FRΒ+ IMs comprise a stable, resident population, whereas FRß- ΙΜs represent a mixed population of resident and recruited IMs.


Assuntos
Receptor 2 de Folato , Pneumonia , Humanos , Monócitos/metabolismo , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/metabolismo , Macrófagos/metabolismo , Pneumonia/induzido quimicamente , Pneumonia/genética , Pneumonia/metabolismo , Inflamação/genética , Inflamação/metabolismo , Análise de Sequência de RNA/métodos , Receptor 2 de Folato/metabolismo
3.
Zhongguo Zhong Yao Za Zhi ; 46(22): 5867-5876, 2021 Nov.
Artigo em Chinês | MEDLINE | ID: mdl-34951177

RESUMO

Network pharmacology and the mouse model of viral pneumonia caused by influenza virus FM_1 were employed to explore the main active components and the mechanism of Pulsatilla chinensis against the inflammatory injury of influenza virus-induced pneumonia. The components and targets of P. chinensis were searched from TCMSP, and the targets associated with influenza virus-induced pneumonia were searched from GeneCards. The common targets between P. chinensis and influenza virus-induced pneumonia were identified with Venn diagram established in Venny 2.1. The herb-component-disease-target(H-C-D-T) network was constructed by Cytoscape 3.7.2. The above data were imported into STRING for PPI network analysis. Gene Ontology(GO) enrichment and KEGG pathway enrichment were performed with DAVID. BALB/cAnN mice were infected with the influenza virus FM_1 by nasal drip to gene-rate the mouse model of pneumonia. Immunohistochemistry was adopted to the expression profiling of inflammatory cytokines in the lung tissues of mice in the blank group, model group, and P. chinensis group 1, 3, 5, and 7 days after infection. The pathological changes of lung and trachea of mice in blank group, model group, and P. chinensis group were observed with light microscope and scanning electron microscope at all the time points. The network pharmacological analysis indicated that 9 compounds of P. chinensis were screened out, with a total of 57 targets, 22 of which were overlapped with those of influenza virus-induced pneumonia. A total of 112 GO terms(P<0.05) were enriched, including 81 terms of biological processes, 11 terms of cell components, and 20 terms of molecular functions. A total of 53 KEGG signaling pathways(P<0.05) were enriched, including TNF signaling pathway, influenza A signaling pathway, NF-κB signaling pathway, MAPK signaling pathway and other signaling pathways related to influenza/inflammation. In the P. chinensis group, the expression of TNF-α and IL-1 in the lung tissue was down-regulated on the 3 rd day after infection, and that of IL-6 in the lung tissue was down-regulated on the 5 th day after infection. Light microscopy and scanning electron microscopy showed that P. chinensis significantly alleviated the pathological damage of lung and trachea compared with the model group. This study reflects the multi-components, multi-targets, and multi-pathways of P. chinensis against influenza virus-induced pneumonia. P. chinensis may reduce the production of proinflammatory cytokines and mediators and block the pro-inflammatory signaling pathways to alleviate viral pneumonia, which provides reference for future research.


Assuntos
Medicamentos de Ervas Chinesas , Orthomyxoviridae , Pneumonia , Pulsatilla , Animais , Camundongos , Farmacologia em Rede , Pneumonia/tratamento farmacológico , Pneumonia/genética
4.
Cell Rep ; 37(6): 109921, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34758300

RESUMO

Regulatory T (Treg) cells are critical for immunological tolerance and immune homeostasis. Treg cells strongly rely on mitochondrial metabolism and show a lower level of glycolysis. However, little is known about the role of lipid metabolism in the regulation of Treg cell homeostasis. Some members of the ACSL family of acyl-coenzyme A (CoA) synthases are expressed in T cells, but their function remains unclear. A combination of RNA-sequencing and proteome analyses shows that Acsbg1, a member of ACSL, is selectively expressed in Treg cells. We show that the genetic deletion of Acsbg1 not only causes mitochondrial dysfunction, but it also dampens other metabolic pathways. The extrinsic supplementation of Acsbg1-deficient Treg cells with oleoyl-CoA restores the phenotype of the Treg metabolic signature. Furthermore, this pathway in ST2+ effector Treg cells enhances immunosuppressive capacity in airway inflammation. Thus, Acsbg1 serves as a metabolic checkpoint governing Treg cell homeostasis and the resolution of lung inflammation.


Assuntos
Coenzima A Ligases/metabolismo , Metabolismo Energético , Pulmão/enzimologia , Mitocôndrias/enzimologia , Pneumonia/enzimologia , Linfócitos T Reguladores/enzimologia , Animais , Coenzima A Ligases/genética , Modelos Animais de Doenças , Ácidos Graxos/metabolismo , Regulação Enzimológica da Expressão Gênica , Homeostase , Interleucina-33 , Pulmão/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/imunologia , Biogênese de Organelas , Pneumonia/genética , Pneumonia/imunologia , Transdução de Sinais , Linfócitos T Reguladores/imunologia
5.
Toxicol Appl Pharmacol ; 431: 115730, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34601004

RESUMO

Pre-existing conditions modulate sensitivity to numerous xenobiotic exposures such as air pollution. Specifically, individuals suffering from metabolic syndrome (MetS) demonstrate enhanced acute inflammatory responses following particulate matter inhalation. The mechanisms associated with these exacerbated inflammatory responses are unknown, impairing interventional strategies and our understanding of susceptible populations. We hypothesize MetS-associated lipid dysregulation influences mediators of inflammatory resolution signaling contributing to increased acute pulmonary toxicity. To evaluate this hypothesis, healthy and MetS mouse models were treated with either 18-hydroxy eicosapentaenoic acid (18-HEPE), 14-hydroxy docosahexaenoic acid (14-HDHA), 17-hydroxy docosahexaenoic acid (17-HDHA), or saline (control) via intraperitoneal injection prior to oropharyngeal aspiration of silver nanoparticles (AgNP). In mice receiving saline treatment, AgNP exposure resulted in an acute pulmonary inflammatory response that was exacerbated in MetS mice. A targeted lipid assessment demonstrated 18-HEPE, 14-HDHA, and 17-HDHA treatments altered lung levels of specialized pro-resolving lipid mediators (SPMs). 14-HDHA and 17-HDHA treatments more efficiently reduced the exacerbated acute inflammatory response in AgNP exposed MetS mice as compared to 18-HEPE. This included decreased neutrophilic influx, diminished induction of inflammatory cytokines/chemokines, and reduced alterations in SPMs. Examination of SPM receptors determined baseline reductions in MetS mice compared to healthy as well as decreases due to AgNP exposure. Overall, these results demonstrate AgNP exposure disrupts inflammatory resolution, specifically 14-HDHA and 17-HDHA derived SPMs, in MetS contributing to exacerbated acute inflammatory responses. Our findings identify a potential mechanism responsible for enhanced susceptibility in MetS that can be targeted for interventional therapeutic approaches.


Assuntos
Mediadores da Inflamação/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Síndrome Metabólica/complicações , Nanopartículas Metálicas/toxicidade , Pneumonia/induzido quimicamente , Compostos de Prata/toxicidade , Animais , Anti-Inflamatórios/farmacologia , Citocinas/genética , Citocinas/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/farmacologia , Regulação da Expressão Gênica , Ácidos Hidroxieicosatetraenoicos/farmacologia , Metabolismo dos Lipídeos/genética , Pulmão/metabolismo , Masculino , Síndrome Metabólica/tratamento farmacológico , Síndrome Metabólica/genética , Síndrome Metabólica/metabolismo , Camundongos Endogâmicos C57BL , Pneumonia/genética , Pneumonia/metabolismo , Pneumonia/prevenção & controle , Transdução de Sinais
6.
Respir Res ; 22(1): 99, 2021 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-33823870

RESUMO

BACKGROUND: COVID-19 pneumonia has been associated with severe acute hypoxia, sepsis-like states, thrombosis and chronic sequelae including persisting hypoxia and fibrosis. The molecular hypoxia response pathway has been associated with such pathologies and our recent observations on anti-hypoxic and anti-inflammatory effects of whole aqueous extract of Adhatoda Vasica (AV) prompted us to explore its effects on relevant preclinical mouse models. METHODS: In this study, we tested the effect of whole aqueous extract of AV, in murine models of bleomycin induced pulmonary fibrosis, Cecum Ligation and Puncture (CLP) induced sepsis, and siRNA induced hypoxia-thrombosis phenotype. The effect on lung of AV treated naïve mice was also studied at transcriptome level. We also determined if the extract may have any effect on SARS-CoV2 replication. RESULTS: Oral administration AV extract attenuates increased airway inflammation, levels of transforming growth factor-ß1 (TGF-ß1), IL-6, HIF-1α and improves the overall survival rates of mice in the models of pulmonary fibrosis and sepsis and rescues the siRNA induced inflammation and associated blood coagulation phenotypes in mice. We observed downregulation of hypoxia, inflammation, TGF-ß1, and angiogenesis genes and upregulation of adaptive immunity-related genes in the lung transcriptome. AV treatment also reduced the viral load in Vero cells infected with SARS-CoV2. CONCLUSION: Our results provide a scientific rationale for this ayurvedic herbal medicine in ameliorating the hypoxia-hyperinflammation features and highlights the repurposing potential of AV in COVID-19-like conditions.


Assuntos
Anti-Inflamatórios/farmacologia , Tratamento Farmacológico da COVID-19 , Reposicionamento de Medicamentos , Hipóxia/tratamento farmacológico , Justicia , Pulmão/efeitos dos fármacos , Extratos Vegetais/farmacologia , Pneumonia/prevenção & controle , Fibrose Pulmonar/tratamento farmacológico , Sepse/tratamento farmacológico , Animais , Anti-Inflamatórios/isolamento & purificação , Bleomicina , COVID-19/metabolismo , COVID-19/virologia , Ceco/microbiologia , Ceco/cirurgia , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Hipóxia/genética , Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Mediadores da Inflamação/metabolismo , Justicia/química , Ligadura , Pulmão/metabolismo , Pulmão/microbiologia , Pulmão/patologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Extratos Vegetais/isolamento & purificação , Pneumonia/genética , Pneumonia/metabolismo , Pneumonia/microbiologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Fibrose Pulmonar/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Sepse/genética , Sepse/metabolismo , Sepse/microbiologia , Transcriptoma
7.
Artigo em Chinês | WPRIM | ID: wpr-921708

RESUMO

Network pharmacology and the mouse model of viral pneumonia caused by influenza virus FM_1 were employed to explore the main active components and the mechanism of Pulsatilla chinensis against the inflammatory injury of influenza virus-induced pneumonia. The components and targets of P. chinensis were searched from TCMSP, and the targets associated with influenza virus-induced pneumonia were searched from GeneCards. The common targets between P. chinensis and influenza virus-induced pneumonia were identified with Venn diagram established in Venny 2.1. The herb-component-disease-target(H-C-D-T) network was constructed by Cytoscape 3.7.2. The above data were imported into STRING for PPI network analysis. Gene Ontology(GO) enrichment and KEGG pathway enrichment were performed with DAVID. BALB/cAnN mice were infected with the influenza virus FM_1 by nasal drip to gene-rate the mouse model of pneumonia. Immunohistochemistry was adopted to the expression profiling of inflammatory cytokines in the lung tissues of mice in the blank group, model group, and P. chinensis group 1, 3, 5, and 7 days after infection. The pathological changes of lung and trachea of mice in blank group, model group, and P. chinensis group were observed with light microscope and scanning electron microscope at all the time points. The network pharmacological analysis indicated that 9 compounds of P. chinensis were screened out, with a total of 57 targets, 22 of which were overlapped with those of influenza virus-induced pneumonia. A total of 112 GO terms(P<0.05) were enriched, including 81 terms of biological processes, 11 terms of cell components, and 20 terms of molecular functions. A total of 53 KEGG signaling pathways(P<0.05) were enriched, including TNF signaling pathway, influenza A signaling pathway, NF-κB signaling pathway, MAPK signaling pathway and other signaling pathways related to influenza/inflammation. In the P. chinensis group, the expression of TNF-α and IL-1 in the lung tissue was down-regulated on the 3 rd day after infection, and that of IL-6 in the lung tissue was down-regulated on the 5 th day after infection. Light microscopy and scanning electron microscopy showed that P. chinensis significantly alleviated the pathological damage of lung and trachea compared with the model group. This study reflects the multi-components, multi-targets, and multi-pathways of P. chinensis against influenza virus-induced pneumonia. P. chinensis may reduce the production of proinflammatory cytokines and mediators and block the pro-inflammatory signaling pathways to alleviate viral pneumonia, which provides reference for future research.


Assuntos
Animais , Camundongos , Medicamentos de Ervas Chinesas , Farmacologia em Rede , Orthomyxoviridae , Pneumonia/genética , Pulsatilla
8.
J Tradit Chin Med ; 40(5): 792-802, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33000580

RESUMO

OBJECTIVE: To investigate the efficacy of active compounds of Chanqin (CQ) granules on PM2.5-induced airway neurogenic inflammation in vivo, and to elucidate the underlying mechanisms of action. METHODS: The Traditional Chinese Medicine systems pharmacology (TCMSP) database was searched, and the results were combined with oral bioavailability and drug analysis to identify the compounds in CQ granules. The pharmacophore modeling approach was used to predict the compound targets, and the diseases corresponding to the targets were obtained by searching the therapeutic target database (TTD), pharmacogenomics knowledgebase (PharmGKB) and DrugBank databases. Cytoscape software was used to construct the network pharmacological charts for Component-Target and Target-Disease interactions of the CQ granules. Then, the mechanisms of action and effectiveness of CQ granules for the treatment of PM2.5-induced airway neurogenic inflammation were analyzed. RESULTS: A total of 195 compounds and 171 targets were obtained from the analyses. A total of 569 corresponding diseases were identified for these targets. Component-target and target-disease networks were constructed. The possible mechanisms and effective components in CQ granules for treating airway neurogenic inflammation were analyzed. Quercetin, kaempferol and isorhamnetin, beta-sitosterol and sitosterol, which are typically found in the formulation, have extensive pharmacological activities, including anti-inflammatory, antioxidant and antiviral actions and neuroprotective properties. Among these targets, androgen receptor, estrogen receptor, prostaglandin G/H synthase 2, and inducible nitric oxide synthase play important pathological roles, including the induction of neurogenic inflammation. CQ granules may have therapeutic effectiveness for numerous diseases in addition to respiratory diseases, including neoplasms, digestive system diseases, cardiovascular diseases, respiratory tract diseases and nervous system diseases. In vivo, CQ granules are effective in treating pulmonary inflammation and downregulate neuropeptides in the bronchoalveolar lavage fluid after PM2.5 exposure. CQ granules significantly decreased the levels of neurokinin A, neurokinin B and calcitonin gene-related peptide in the lung and dorsal root ganglia. CQ also significantly suppressed the upregulation of p-extracellular regulated protein kinase 1/2 and p-methyl ethyl ketone 1/2 induced by PM2.5 exposure. CONCLUSION: CQ granules have potential for the treatment of neurogenic inflammation induced by PM2.5 in vivo, and the mechanism might involve downregulation of neuropeptides in the BALF, lung and dorsal root ganglia.


Assuntos
Medicamentos de Ervas Chinesas/administração & dosagem , Medicamentos de Ervas Chinesas/química , Inflamação Neurogênica/tratamento farmacológico , Material Particulado/toxicidade , Animais , Humanos , Masculino , Inflamação Neurogênica/etiologia , Inflamação Neurogênica/genética , Inflamação Neurogênica/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Pneumonia/tratamento farmacológico , Pneumonia/genética , Pneumonia/metabolismo , Ratos , Ratos Sprague-Dawley
9.
J Tradit Chin Med ; 40(5): 803-811, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33000581

RESUMO

OBJECTIVE: To investigate synergistic effect of Reduning (RDN) injection plus ribavirin against severe pneumonia induced by H1N1 influenza A virus in mice. METHODS: We established a mouse model of severe pneumonia induced by influenza A virus by infecting Balb/c mice with CA07 virus. We randomly assigned the infected mice into four groups, and treated them with normal saline (NS group), RDN (injection, 86.6 mg/kg), ribavirin (injection, 66.6 mg/kg) or double Ribavirin plus RDN group, the same dosage as used in the single treatments) for 5 d. Lung index and lung pathology were recorded or calculated in terms of the curative effective. Cytokines, NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome related protein including caspase-associated recruitment domain (CARD) domain Apoptosis-associated speck-like protein containing a caspase recruitment domain(ASC), caspase-1 and NOD-like receptor family, pyrin domain containing 3 (NLRP3), and reactive oxygen species were simultaneously investigated. RESULTS: RDN plus ribavirin treatment, not RDN or ribavirin alone, provided a significant survival benefit to the influenza A virus-infected mice. The combination treatment protected the mice against severe influenza infection by attenuating the severe lung injury. The combined treatment also reduced the viral titers in mouse lungs and lung index, downregulated their immunocytokine levels, including IL-1ß and IL-18, and down regulated the NLRP3, especially the transcription and translation of caspase-1. Meanwhile NS group had significantly higher reactive oxygen species (ROS) expression which could was dramatically reduced by the treatment of RDN plus ribavirin. CONCLUSION: Our study showed that RDN combined with ribavirin could protect the mice, and reduce the lung immunopathologic damage caused by severe influenza pneumonia. The mechanism could be that it reduced ROS produce and inhibited NLRP3 inflammasome activation so that mainly lower the downstream inflammatory cytokines IL-1ß and IL-18.


Assuntos
Medicamentos de Ervas Chinesas/administração & dosagem , Vírus da Influenza A Subtipo H1N1/fisiologia , Pneumonia/tratamento farmacológico , Ribavirina/administração & dosagem , Animais , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Influenza Humana/complicações , Influenza Humana/virologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucina-8/genética , Interleucina-8/imunologia , Pulmão/imunologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Pneumonia/etiologia , Pneumonia/genética , Pneumonia/imunologia
10.
J Food Biochem ; 44(12): e13510, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33025599

RESUMO

Pneumonia refers to a death-causing infection. Astragali Radix (AR) and Atractylodis Macrocephalae Rhizoma (AMR) are widely used as traditional tonic and promising edible immunomodulatory herbal medicine, but the systemic mechanism is not well understood. Therefore, a strategy based on network pharmacology and molecular docking was designed to explore the systemic mechanism of AR-AMR acting on pneumonia. After a series of bioinformatics assays, seven kernel targets were obtained, including TNF, IL6, IFNG, IL1B, IL10, IL4, and TLR9. And seven key compounds were identified as the synergy components of AR-AMR acting on pneumonia, the four key compounds belonging to AR were (3R)-3-(2-hydroxy-3,4-dimethoxyphenyl)-7-chromanol, formononetin, quercetin, and kaempferol, the three key compounds belonging to AMR were atractylone, 14-acetyl-12-senecioyl-2E, 8E, 10E-atractylentriol, and α-Amyrin. The crucial pathways were mainly related to three modules, including immune diseases, infectious disease, and organismal systems. Collectively, these observations strongly suggest that the molecular mechanisms of AR-AMR regulating pneumonia were closely related to the correlation between inflammation and immune response. PRACTICAL APPLICATIONS: Astragali radix and Atractylodis macrocephalae rhizoma can be used as "medicine-food homology" for dietary supplement. AR and AMR are widely used as a traditional tonic and promising edible immunomodulatory herbal medicine. The AR-AMR herb pairs are used for compatibility many times in the recommended prescriptions in COVID-19 develop pneumonia in China. However, the ingredients and mechanisms of AR-AMR acting on Pneumonia via immunomodulation are unclear. In this paper, bioinformatics and network biology were used to systematically explore the mechanisms of the AR-AMR herb pairs in treatment of pneumonia, and further analyze the correlation mechanism between it and COVID-19 develop pneumonia. To sum up, our study reveals the interrelationships between components, targets, and corresponding biological processes of AR-AMR acting on pneumonia. Understanding these relationships may provide guidance and theoretical basis for the further application of AR-AMR herb pairs.


Assuntos
Medicamentos de Ervas Chinesas/química , Pneumonia/imunologia , Astragalus propinquus , COVID-19/imunologia , Citocinas/genética , Citocinas/imunologia , Medicamentos de Ervas Chinesas/farmacologia , Humanos , Imunomodulação/efeitos dos fármacos , Simulação de Acoplamento Molecular , Pneumonia/tratamento farmacológico , Pneumonia/genética , Rizoma/química , Tratamento Farmacológico da COVID-19
11.
Am J Physiol Lung Cell Mol Physiol ; 318(5): L921-L930, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32159972

RESUMO

The incidence of asthma has increased from 5.5% to near 8% of the population, which is a major health concern. The hallmarks of asthma include eosinophilic airway inflammation that is associated with chronic airway remodeling. Allergic airway inflammation is characterized by a complex interplay of resident and inflammatory cells. MicroRNAs (miRNAs) are small noncoding RNAs that function as posttranscriptional modulators of gene expression. However, the role of miRNAs, specifically miR-451, in the regulation of allergic airway inflammation is unexplored. Our previous findings showed that oxidant stress regulates miR-451 gene expression in macrophages during an inflammatory process. In this paper, we examined the role of miR-451 in regulating macrophage phenotype using an experimental poly-allergenic murine model of allergic airway inflammation. We found that miR-451 contributes to the allergic induction of CCL17 in the lung and plays a key role in proasthmatic macrophage activation. Remarkably, administration of a Sirtuin 2 (Sirt2) inhibitor diminished alternate macrophage activation and markedly abrogated triple-allergen [dust mite, ragweed, Aspergillus fumigatus (DRA)]-induced lung inflammation. These data demonstrate a role for miR-451 in modulating allergic inflammation by influencing allergen-mediated macrophages phenotype.


Assuntos
Asma/genética , Macrófagos Alveolares/imunologia , MicroRNAs/genética , Pneumonia/genética , Sirtuína 2/genética , Alérgenos/administração & dosagem , Animais , Anti-Inflamatórios/farmacologia , Antígenos de Plantas/administração & dosagem , Aspergillus/química , Aspergillus/imunologia , Asma/induzido quimicamente , Asma/patologia , Asma/terapia , Quimiocina CCL17/genética , Quimiocina CCL17/imunologia , Modelos Animais de Doenças , Fungos/química , Fungos/imunologia , Furanos/farmacologia , Regulação da Expressão Gênica , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/imunologia , Extratos Vegetais/administração & dosagem , Pneumonia/induzido quimicamente , Pneumonia/patologia , Pneumonia/terapia , Pyroglyphidae/química , Pyroglyphidae/imunologia , Quinolinas/farmacologia , Transdução de Sinais , Sirtuína 2/antagonistas & inibidores , Sirtuína 2/imunologia
12.
FASEB J ; 33(5): 6442-6455, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30776316

RESUMO

Cellular stress or injury induces release of endogenous danger signals such as ATP, which plays a central role in activating immune cells. ATP is essential for the release of nonclassically secreted cytokines such as IL-1ß but, paradoxically, has been reported to inhibit the release of classically secreted cytokines such as TNF. Here, we reveal that ATP does switch off soluble TNF (17 kDa) release from LPS-treated macrophages, but rather than inhibiting the entire TNF secretion, ATP packages membrane TNF (26 kDa) within microvesicles (MVs). Secretion of membrane TNF within MVs bypasses the conventional endoplasmic reticulum- and Golgi transport-dependent pathway and is mediated by acid sphingomyelinase. These membrane TNF-carrying MVs are biologically more potent than soluble TNF in vivo, producing significant lung inflammation in mice. Thus, ATP critically alters TNF trafficking and secretion from macrophages, inducing novel unconventional membrane TNF signaling via MVs without direct cell-to-cell contact. These data have crucial implications for this key cytokine, particularly when therapeutically targeting TNF in acute inflammatory diseases.-Soni, S., O'Dea, K. P., Tan, Y. Y., Cho, K., Abe, E., Romano, R., Cui, J., Ma, D., Sarathchandra, P., Wilson, M. R., Takata, M. ATP redirects cytokine trafficking and promotes novel membrane TNF signaling via microvesicles.


Assuntos
Trifosfato de Adenosina/imunologia , Membrana Celular/imunologia , Vesículas Extracelulares/imunologia , Macrófagos/imunologia , Pneumonia/imunologia , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/imunologia , Doença Aguda , Trifosfato de Adenosina/genética , Animais , Comunicação Celular/genética , Comunicação Celular/imunologia , Membrana Celular/genética , Retículo Endoplasmático/genética , Retículo Endoplasmático/imunologia , Vesículas Extracelulares/genética , Complexo de Golgi/genética , Complexo de Golgi/imunologia , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/imunologia , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Knockout , Pneumonia/induzido quimicamente , Pneumonia/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fator de Necrose Tumoral alfa/genética
13.
BMC Complement Altern Med ; 19(1): 15, 2019 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-30630473

RESUMO

BACKGROUND: Asian traditional herbal remedies are typically a concoction of a major and several complementary herbs. While balancing out any adverse effect of the major herb, the complementary herbs could dilute the efficacy of the major herb, resulting in a suboptimal therapeutic effect of an herbal remedy. Here, we formulated Chung-Sang (CS) by collating five major herbs, which are used against inflammatory diseases, and tested whether an experimental formula composed of only major herbs is effective in suppressing inflammation without significant side effects. METHODS: The 50% ethanol extract of CS (eCS) was fingerprinted by HPLC. Cytotoxicity to RAW 264.7 cells was determined by an MTT assay and a flow cytometer. Nuclear NF-κB and Nrf2 were analyzed by western blot. Ubiquitinated Nrf2 was similarly analyzed following immunoprecipitation of Nrf2. Acute lung inflammation and sepsis were induced in C57BL/6 mice. The effects of eCS on lung disease were measured by HE staining of lung sections, a differential cell counting of bronchoalveolar lavage fluid, a myeloperoxidase (MPO) assay, a real-time qPCR, and Kaplan-Meier survival of mice. RESULTS: eCS neither elicited cytotoxicity nor reactive oxygen species. While not suppressing NF-κB, eCS activated Nrf2, reduced the ubiquitination of Nrf2, and consequently induced the expression of Nrf2-dependent genes. In an acute lung inflammation mouse model, an intratracheal (i.t.) eCS suppressed neutrophil infiltration, the expression of inflammatory cytokine genes, and MPO activity. In a sepsis mouse model, a single i.t. eCS was sufficient to significantly decrease mouse mortality. CONCLUSIONS: eCS could suppress severe lung inflammation in mice. This effect seemed to associate with eCS activating Nrf2. Our findings suggest that herbal remedies consisting of only major herbs are worth considering.


Assuntos
Anti-Inflamatórios/administração & dosagem , Fator 2 Relacionado a NF-E2/imunologia , Extratos Vegetais/administração & dosagem , Pneumonia/tratamento farmacológico , Animais , Anti-Inflamatórios/isolamento & purificação , Composição de Medicamentos , Humanos , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/genética , NF-kappa B/genética , NF-kappa B/imunologia , Infiltração de Neutrófilos/efeitos dos fármacos , Extratos Vegetais/química , Extratos Vegetais/isolamento & purificação , Pneumonia/genética , Pneumonia/imunologia , Células RAW 264.7
14.
Biomed Pharmacother ; 102: 1092-1098, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29710526

RESUMO

BACKGROUND: Xianyu decoction (XD), a Chinese experience recipe, shows inhibitory effects on lung cancer. However, the potential functions of XD on pneumonia were unknown. This study aimed to investigate the effect of XD on inflammatory response of childhood pneumonia. METHODS: Human lung bronchial epithelial cell line BEAS-2B was cultured in different doses of LPS with or without XD treatment. The expression of miR-15a and IKBKB were altered by transfection assay. RT-PCR and western blot were used to evaluate the effects of XD and miR-15a mimic/inhibitor on the expression levels of miR-15a, IKBKB, p65 and IκBα. ELISA was used to determine the levels of CRP, IL-6 and IL-8. RESULTS: High expression of miR-15a was observed in serum and cell model of pneumonia. miR-15a promoted the expression of inflammatory cytokines IL-6, IL-8, CRP and IKBKB in vitro. XD treatment downregulated the level of miR-15a in pneumonia children. In addition, XD reduced the expression of inflammatory cytokines and the phosphorylation levels of p65 and IκBα by inhibition of miR-15a and IKBKB expression in LPS-stimulated BEAS-2B cells. CONCLUSION: XD downregulated the level of miR-15a in serum of pneumonia children. Additionally, XD inhibited inflammatory response in LPS-stimulated BEAS-2B cells possibly by blocking IKBKB/NF-κB signal pathway which was regulated by miR-15a.


Assuntos
Brônquios/patologia , Medicamentos de Ervas Chinesas/uso terapêutico , Células Epiteliais/patologia , Inflamação/patologia , Pulmão/patologia , Linhagem Celular , Citocinas/metabolismo , Regulação para Baixo/genética , Medicamentos de Ervas Chinesas/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Proteínas I-kappa B/metabolismo , Inflamação/complicações , Inflamação/tratamento farmacológico , Inflamação/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Modelos Biológicos , Pneumonia/complicações , Pneumonia/tratamento farmacológico , Pneumonia/genética , Pneumonia/patologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
15.
J Appl Toxicol ; 38(3): 385-397, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29094763

RESUMO

Our recent studies revealed a dose-dependent proinflammatory response to copper oxide nanoparticles (CuO NPs) in rats following short-term inhalation exposure for five consecutive days. Here transcriptomics approaches were applied using the same model to assess global gene expression in lung tissues obtained 1 day post-exposure and after a recovery period of 22 days from rats exposed to clean air or 6 hour equivalent doses of 3.3 mg m-3 (low dose) and 13.2 mg m-3 (high dose). Microarray analyses yielded about 1000 differentially expressed genes in the high-dose group and 200 in low-dose compared to the clean air control group, and less than 20 after the recovery period. Pathway analysis indicated cell proliferation/survival and inflammation as the main processes triggered by exposure to CuO NPs. We did not find significant perturbations of pathways related to oxidative stress. Upregulation of epithelial cell transforming protein 2 (Ect2), a known oncogene, was noted and ECT2 protein was upregulated in the lungs of exposed animals. Proliferation of alveolar epithelial cells was demonstrated based on Ki67 expression. The gene encoding monocyte chemoattractant protein 1 (or CCL2) was also upregulated and this was confirmed by immunohistochemistry. However, no aberrant DNA methylation of inflammation-associated genes was observed. In conclusion, we have found that inhalation of CuO NPs in rats causes upregulation of the oncoprotein ECT2 and the chemokine CCL2 and other proinflammatory markers as well as proliferation in bronchoalveolar epithelium after a short-term inhalation exposure. Thus, pathways known to be associated with neoplastic processes and inflammation were affected in this model.


Assuntos
Proliferação de Células/efeitos dos fármacos , Cobre/toxicidade , Células Epiteliais/efeitos dos fármacos , Perfilação da Expressão Gênica/métodos , Nanopartículas Metálicas , Pneumonia/induzido quimicamente , Alvéolos Pulmonares/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Administração por Inalação , Animais , Proliferação de Células/genética , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Cobre/administração & dosagem , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Regulação da Expressão Gênica , Hiperplasia , Mediadores da Inflamação/metabolismo , Masculino , Pneumonia/genética , Pneumonia/metabolismo , Pneumonia/patologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Ratos Wistar , Fatores de Tempo
16.
Int J Mol Med ; 40(6): 1932-1940, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29039495

RESUMO

Neem (Azadirachta indica A. Juss.) leaf has been reported to exert anti-inflammatory, antibacterial and antioxidant effects. The purpose of this study was to investigate the protective effects of neem leaf extract (NLE) against cigarette smoke (CS)- and lipopolysaccharide (LPS)-induced pulmonary inflammation. Treatment with NLE significantly attenuated the infiltration of inflammatory cells, such as neutrophils and macrophages in bronchoalveolar lavage fluid (BALF). NLE also reduced the production of reactive oxygen species and the activity of neutrophil elastase in BALF. Moreover, NLE attenuated the release of pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 in BALF. NLE inhibited the recruitment of inflammatory cells and the expression of monocyte chemoattractant protein-1 (MCP-1) in the lungs of mice with CS- and LPS-induced pulmonary inflammation. NLE also decreased the expression of inducible nitric oxide synthase (iNOS) in the lungs of the mice CS- and LPS-induced pulmonary inflammation. Furthermore, treatment with NLE significantly attenuated the activation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) in the lungs mice exposed to CS and LPS. NLE also inhibited the phosphorylation of nuclear factor (NF)-κB and inhibitor of NF-κB (IκB) in the lungs of mice expose to CS and LPS. These findings thus suggest that NLE has potential for use in the treatment of chronic obstructive pulmonary disease.


Assuntos
Azadirachta/química , Extratos Vegetais/administração & dosagem , Pneumonia/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Animais , Líquido da Lavagem Broncoalveolar/química , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-6/genética , Lipopolissacarídeos/toxicidade , Pulmão/efeitos dos fármacos , Pulmão/patologia , Camundongos , Extratos Vegetais/química , Folhas de Planta/química , Pneumonia/induzido quimicamente , Pneumonia/genética , Pneumonia/patologia , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/patologia , Espécies Reativas de Oxigênio/metabolismo , Fumar/efeitos adversos , Fator de Necrose Tumoral alfa/genética
17.
Toxicol Sci ; 160(1): 4-14, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29036520

RESUMO

The lung is constantly exposed to ambient pollutants such as ambient fine particulate matter (PM2.5), making it one of the most frequent locations of inflammation in the body. Given the establishment of crucial role of inflammation in the pathogenesis of cardiometabolic diseases, pulmonary inflammation is thus widely believed to be an important risk factor for cardiometabolic diseases. However, the causality between them has not yet been well established. To determine if pulmonary inflammation is sufficient to cause adverse cardiometabolic effects, SFTPC-rtTA+/-tetO-cre+/-pROSA-inhibitor κB kinase 2(IKK2)ca+/- (LungIKK2ca) and littermate SFTPC-rtTA+/-tetO-cre-/-pROSA-IKK2ca+/- wildtype (WT) mice were fed with doxycycline diet to induce constitutively active Ikk2 (Ikk2ca) overexpression in the lung and their pulmonary, systemic, adipose, and hypothalamic inflammations, vascular function, and glucose homeostasis were assessed. Feeding with doxycycline diet resulted in IKK2ca overexpression in the lungs of LungIKK2ca but not WT mice. This induction of IKK2ca was accompanied by marked pulmonary inflammation as evidenced by significant increases in bronchoalveolar lavage fluid leukocytes, pulmonary macrophage infiltration, and pulmonary mRNA expression of tumor necrosis factor α (Tnfα) and interleukin-6 (Il-6). This pulmonary inflammation due to lung-specific overexpression of IKK2ca was sufficient to increase circulating TNFα and IL-6 levels, adipose expression of Tnfα and Il-6 mRNA, aortic endothelial dysfunction, and systemic insulin resistance. Unexpectedly, no significant alteration in hypothalamic expression of Tnfα and Il-6 mRNA and glucose intolerance were observed in these mice. Pulmonary inflammation is sufficient to induce systemic inflammation, endothelial dysfunction, and insulin resistance, but not hypothalamic inflammation and glucose intolerance.


Assuntos
Tecido Adiposo/enzimologia , Encefalite/enzimologia , Intolerância à Glucose/enzimologia , Hipotálamo/enzimologia , Quinase I-kappa B/metabolismo , Pulmão/enzimologia , Paniculite/enzimologia , Pneumonia/enzimologia , Animais , Aorta/enzimologia , Aorta/fisiopatologia , Glicemia/metabolismo , Encefalite/genética , Endotélio Vascular/enzimologia , Endotélio Vascular/fisiopatologia , Ativação Enzimática , Predisposição Genética para Doença , Intolerância à Glucose/sangue , Intolerância à Glucose/genética , Quinase I-kappa B/genética , Resistência à Insulina , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmão/patologia , Camundongos Transgênicos , Paniculite/genética , Fenótipo , Pneumonia/genética , Pneumonia/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
18.
J Tradit Chin Med ; 36(2): 181-6, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27400472

RESUMO

OBJECTIVE: To investigate the effects of ribavirin administration combined with Reduning in a mouse model of influenza A (H1N1)-induced severe pneumonia. METHODS: Influenza A/Beijing/501/2009 (H1N1)-infected C57BL/6 mice were randomly divided into four experimental groups treated with either a mock injection of phosphate-buffered saline (PBS), ribavirin (66.6 mg/kg daily) or Reduning (86.6 mg/ kg daily), or a combination of both, for 7 days. Mice were monitored for clinical signs and survival, and body weight was measured daily for 14 days. Virus titer, lung wet-to-dry ratios, pathology and cytokines including interleukin (IL)-6, IL-10, and interferon (IFN)-γ were assayed on different days. RESULTS: In the untreated group injected with phosphate buffer saline, all the mice died of the infection. The survival rate of mice treated with Reduning was only 10%, whereas 100% of the ribavirin- and the combination-treated mice survived. Low lung viral loads indicated that ribavirin significantly inhibited virus replication, whereas Reduning did not. Lung wet-to-dry ratios demonstrated that both ribavirin and Reduning, administered together or separately, reduced acute lung edema compared with results in the untreated group. Pathology analyses also showed that treatment with a combination of both drugs relieved pathological lesions, whereas the single drug treatment did not. Levels of IL-6, IL-10 and IFN-γ in mice treated with ribavirin or the combination of both ribavirin and Reduning were all significantly lower than in the untreated group, especially in the combination-treated group. In addition, Reduning administration significantly decreased both IL-6 and IL-10 production but had no effect on IFN-γ. CONCLUSION: Due to the synergistic effect of antiviral and antiinflammation, the combination of ribavirin and Reduning could be an effective treatment for severe H1N1 which was considered to be significant to delayed antiviral and drug resistant.


Assuntos
Antivirais/administração & dosagem , Medicamentos de Ervas Chinesas/administração & dosagem , Vírus da Influenza A Subtipo H1N1/fisiologia , Influenza Humana/tratamento farmacológico , Pneumonia/tratamento farmacológico , Ribavirina/administração & dosagem , Animais , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Humanos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Influenza Humana/genética , Influenza Humana/imunologia , Influenza Humana/virologia , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia/genética , Pneumonia/imunologia , Pneumonia/virologia
19.
PLoS One ; 11(1): e0146102, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26752192

RESUMO

INTRODUCTION: Macrophage migration inhibitory factor (MIF) is an inflammatory cytokine associated with acute and chronic inflammatory disorders and corticosteroid insensitivity. Its expression in the airways of patients with chronic obstructive pulmonary disease (COPD), a relatively steroid insensitive inflammatory disease is unclear, however. METHODS: Sputum, bronchoalveolar lavage (BAL) macrophages and serum were obtained from non-smokers, smokers and COPD patients. To mimic oxidative stress-induced COPD, mice were exposed to ozone for six-weeks and treated with ISO-1, a MIF inhibitor, and/or dexamethasone before each exposure. BAL fluid and lung tissue were collected after the final exposure. Airway hyperresponsiveness (AHR) and lung function were measured using whole body plethysmography. HIF-1α binding to the Mif promoter was determined by Chromatin Immunoprecipitation assays. RESULTS: MIF levels in sputum and BAL macrophages from COPD patients were higher than those from non-smokers, with healthy smokers having intermediate levels. MIF expression correlated with that of HIF-1α in all patients groups and in ozone-exposed mice. BAL cell counts, cytokine mRNA and protein expression in lungs and BAL, including MIF, were elevated in ozone-exposed mice and had increased AHR. Dexamethasone had no effect on these parameters in the mouse but ISO-1 attenuated cell recruitment, cytokine release and AHR. CONCLUSION: MIF and HIF-1α levels are elevated in COPD BAL macrophages and inhibition of MIF function blocks corticosteroid-insensitive lung inflammation and AHR. Inhibition of MIF may provide a novel anti-inflammatory approach in COPD.


Assuntos
Corticosteroides/uso terapêutico , Isoxazóis/uso terapêutico , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Pneumonia/complicações , Doença Pulmonar Obstrutiva Crônica/complicações , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Hipersensibilidade Respiratória/complicações , Adulto , Idoso , Animais , Líquido da Lavagem Broncoalveolar , Contagem de Células , Citocinas/metabolismo , Dexametasona/farmacologia , Modelos Animais de Doenças , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Pulmão/patologia , Fatores Inibidores da Migração de Macrófagos/genética , Fatores Inibidores da Migração de Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Pessoa de Meia-Idade , Ozônio , Pneumonia/genética , Pneumonia/patologia , Pneumonia/fisiopatologia , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Testes de Função Respiratória , Hipersensibilidade Respiratória/tratamento farmacológico , Hipersensibilidade Respiratória/genética , Hipersensibilidade Respiratória/fisiopatologia , Fumar/efeitos adversos , Escarro/efeitos dos fármacos , Escarro/metabolismo
20.
PLoS One ; 10(11): e0142816, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26569396

RESUMO

Lung diseases constitute an important public health problem and its growing level of concern has led to efforts for the development of new therapies, particularly for the control of lung inflammation. Low Level Laser Therapy (LLLT) has been highlighted as a non-invasive therapy with few side effects, but its mechanisms need to be better understood and explored. Considering that pollution causes several harmful effects on human health, including lung inflammation, in this study, we have used formaldehyde (FA), an environmental and occupational pollutant, for the induction of neutrophilic lung inflammation. Our objective was to investigate the local and systemic effects of LLLT after FA exposure. Male Wistar rats were exposed to FA (1%) or vehicle (distillated water) during 3 consecutive days and treated or not with LLLT (1 and 5 hours after each FA exposure). Non-manipulated rats were used as control. 24 h after the last FA exposure, we analyzed the local and systemic effects of LLLT. The treatment with LLLT reduced the development of neutrophilic lung inflammation induced by FA, as observed by the reduced number of leukocytes, mast cells degranulated, and a decreased myeloperoxidase activity in the lung. Moreover, LLLT also reduced the microvascular lung permeability in the parenchyma and the intrapulmonary bronchi. Alterations on the profile of inflammatory cytokines were evidenced by the reduced levels of IL-6 and TNF-α and the elevated levels of IL-10 in the lung. Together, our results showed that LLLT abolishes FA-induced neutrophilic lung inflammation by a reduction of the inflammatory cytokines and mast cell degranulation. This study may provide important information about the mechanisms of LLLT in lung inflammation induced by a pollutant.


Assuntos
Formaldeído/efeitos adversos , Terapia com Luz de Baixa Intensidade , Pneumonia/etiologia , Pneumonia/radioterapia , Hipersensibilidade Respiratória/complicações , Animais , Células da Medula Óssea/metabolismo , Líquido da Lavagem Broncoalveolar , Degranulação Celular , Regulação da Expressão Gênica , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmão/irrigação sanguínea , Pulmão/patologia , Masculino , Mastócitos/metabolismo , Microvasos/patologia , Neutrófilos/metabolismo , Permeabilidade , Pneumonia/genética , Ratos Wistar , Hipersensibilidade Respiratória/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA