Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 155
Filtrar
Más filtros

Medicinas Complementárias
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Nanobiotechnology ; 22(1): 190, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38637808

RESUMEN

Acute lung injury (ALI) is generally caused by severe respiratory infection and characterized by overexuberant inflammatory responses and inefficient pathogens-containing, the two major processes wherein alveolar macrophages (AMs) play a central role. Dysfunctional mitochondria have been linked with distorted macrophages and hence lung disorders, but few treatments are currently available to correct these defects. Plant-derive nanovesicles have gained significant attention because of their therapeutic potential, but the targeting cells and the underlying mechanism remain elusive. We herein prepared the nanovesicles from Artemisia annua, a well-known medicinal plant with multiple attributes involving anti-inflammatory, anti-infection, and metabolism-regulating properties. By applying three mice models of acute lung injury caused by bacterial endotoxin, influenza A virus (IAV) and SARS-CoV-2 pseudovirus respectively, we showed that Artemisia-derived nanovesicles (ADNVs) substantially alleviated lung immunopathology and raised the survival rate of challenged mice. Macrophage depletion and adoptive transfer studies confirmed the requirement of AMs for ADNVs effects. We identified that gamma-aminobutyric acid (GABA) enclosed in the vesicles is a major molecular effector mediating the regulatory roles of ADNVs. Specifically, GABA acts on macrophages through GABA receptors, promoting mitochondrial gene programming and bioenergy generation, reducing oxidative stress and inflammatory signals, thereby enhancing the adaptability of AMs to inflammation resolution. Collectively, this study identifies a promising nanotherapeutics for alleviating lung pathology, and elucidates a mechanism whereby the canonical neurotransmitter modifies AMs and mitochondria to resume tissue homeostasis, which may have broader implications for treating critical pulmonary diseases such as COVID-19.


Asunto(s)
Lesión Pulmonar Aguda , Plantas Medicinales , Neumonía Viral , Neumonía , Ratones , Animales , Macrófagos Alveolares/metabolismo , Pulmón/metabolismo , Neumonía Viral/tratamiento farmacológico , Lesión Pulmonar Aguda/patología , Mitocondrias/patología , Ácido gamma-Aminobutírico/metabolismo , Neumonía/metabolismo
2.
J Ethnopharmacol ; 323: 117706, 2024 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-38176670

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Platycodonis Radix (PR) is a traditional herbal remedy used to prevent and treat lung inflammation, and platycodins are speculated to be the major active constituents. However, concrete experimental verification for this assertion remains absent thus far. AIM OF THE STUDY: This study aims to compare the pulmonary distribution dynamics of five platycodins and analyze their effects on cytokines. Through the grey relational analysis (GRA) between pulmonary active components and cytokines, the study ascertains platycodins as the potential effective component against lung inflammation. MATERIALS AND METHODS: A rat lung inflammation model was created using lipopolysaccharides (LPS). Pulmonary distribution dynamics were analyzed via LC-MS/MS. Cytokine changes and distribution patterns in lung tissues were studied by multi-factor reagent kit. GRA was applied to determine correlations between pulmonary components and cytokines. Finally, the anti-inflammatory properties of platycodins were further studied using LPS-induced BEAS-2B cells in vitro. RESULTS: The results showed that five platycodins (Platycodin D, Platycodin D3, Deapio Platycodin D, 3-O-ß-D-Glucopyranosyl Platycodigenin, and Platycodigenin) featured fast absorption rate, short time to peak, and slow metabolism rate. The pulmonary distribution dynamics were significantly affected within 2 h after LPS modeling. At the same time, PR altered the relationships among different cytokines induced by LPS stimulation, particularly inflammatory cytokines IL-6 and IFN-γ. The GRA results indicated good correlation between the pulmonary distribution dynamics of the five platycodins components and the changing patterns of cytokine levels, with Platycodin D3 contributing the most. Additionally, Platycodin D3 exhibited a protective role against LPS-induced inflammation by reducing the production of pro-inflammatory mediators such as IL-1ß, IL-8, and ROS, as well as increasing the expression of the anti-inflammatory mediator IL-10. CONCLUSIONS: Platycodins are the main anti-inflammatory agents in PR and there is a good correlation with cytokines. This contributes to the anti-pneumonia effect of PR.


Asunto(s)
Citocinas , Neumonía , Saponinas , Triterpenos , Ratas , Animales , Citocinas/metabolismo , Cromatografía Liquida , Lipopolisacáridos/farmacología , Espectrometría de Masas en Tándem , Pulmón , Neumonía/inducido químicamente , Neumonía/tratamiento farmacológico , Neumonía/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Antiinflamatorios/metabolismo , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/metabolismo
3.
J Ethnopharmacol ; 321: 117497, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38048893

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Chronic obstructive pulmonary disease (COPD) is a major global health concern characterized by pulmonary inflammation and airway remodeling. Traditional Chinese medicine, such as Modified Jiawei Bushen Yiqi Formula (MBYF), has been used as a complementary therapy for COPD in China. AIM OF THE STUDY: To investigate the therapeutic potential of MBYF in a rat model of COPD induced by cigarette smoke (CS) exposure and explore the underlying mechanism. MATERIALS AND METHODS: The COPD rat model was established through 24 weeks of CS exposure, with MBYF administration starting in the 9th week. Pulmonary function, histological analysis, inflammatory cell count and molecular assays were employed to assess the effects of MBYF on airway remodeling, pulmonary inflammation, neutrophils chemotaxis and the IL17 signaling pathway. RESULTS: MBYF treatment effectively delayed airway remodeling, as evidenced by improved pulmonary function parameters. Histological examination and bronchoalveolar lavage fluid analysis revealed that MBYF mitigated CS-induced pulmonary inflammation by reducing inflammatory cell infiltration. Pharmacological network analysis suggested that MBYF may act through the IL17 signaling pathway to regulate inflammatory responses. RNA-sequencing and molecular assays indicated that MBYF inhibited neutrophils chemotaxis through downregulating the CXCL1/CXCL5/CXCL8-CXCR2 axis, and suppressed IL17A, IL17F and its downstream cytokines, including IL6, TNFα, IL1ß, and COX2. Furthermore, MBYF inhibited the activation of NF-κB and MAPKs in the IL17 signaling pathway. CONCLUSION: MBYF exhibits potential as an adjunct or alternative treatment for COPD, effectively mitigating CS-induced pulmonary inflammation and airway remodeling through the inhibition of neutrophil chemotaxis and IL17 signaling pathway.


Asunto(s)
Neumonía , Enfermedad Pulmonar Obstructiva Crónica , Ratas , Animales , Neutrófilos , Quimiotaxis , Remodelación de las Vías Aéreas (Respiratorias) , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Pulmón , Neumonía/metabolismo , Transducción de Señal , Líquido del Lavado Bronquioalveolar
4.
J Tradit Chin Med ; 43(6): 1126-1139, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37946475

RESUMEN

OBJECTIVE: To explore the underlying mechanisms of the effects of Yangqing Chenfei formula (, YCF) on inflammation and fibrosis in silicosis via inhibition of macrophage polarization. METHODS: A silicotic rat model was established via a single intratracheal instillation of silica particles on the first day of week 0. Subsequently, YCF was administered intragastrically to silicotic rats during weeks 0-2 and 5-8 twice daily. The mouse-derived alveolar macrophage cell line was used to investigate the mechanisms of YCF in M1/M2 polarization. RESULTS: YCF treatment effectively inhibited lung pathological changes, including inflammatory cell infiltration and tissue damage, and increased the forced expiratory volume in the first 0.3 s, functional residual capacity, and maximal mid-expiratory flow in weeks 2 and 8. Furthermore, the treatment improved lung functions by upregulating tidal volume, pause increase, and expiratory flow at 50% tidal volume from weeks 5 to 8. Moreover, YCF could significantly suppressed the progression of inflammation and fibrosis, by reducing the levels of inflammatory cytokines, as well as collagen- I and III. YCF treatment also decreased the numbers of macrophages and M1/M2 macrophages and the level of transforming growth factor-ß (TGF-ß). Additionally, YCF5, the effective substance in YCF, decreased lipopolysaccharide and interferon-γ-induced M1 macrophage polarization in a concentration-dependent manner. The mechanism of anti-M1 polarization might be related to a decrease in extracellular signal-regulated kinase, c-JUN N-terminal kinase, P38, and P65 phosphorylation. Furthermore, YCF5 inhibited interleukin-4-induced M2 macrophages by decreasing the protein and mRNA expressions of arginase-1 and CD206 as well as the levels of profibrotic factors, such as TGF-ß and connective tissue growth factor. The mechanisms underlying the anti-M2 polarization of YCF5 were primarily associated with the inhibition of the nuclear translocation of phosphorylated signal transducer and activator of transcription 6 (p-STAT6). CONCLUSION: YCF significantly inhibits inflammation and fibrosis in silicotic rats probably via the suppression of M1/M2 macrophage polarization mediated by the inhibition of mitogen-activated protein kinase and nuclear factor kappa B signaling pathways and Janus kinase/STAT6 pathways.


Asunto(s)
Neumonía , Dióxido de Silicio , Ratas , Ratones , Animales , Dióxido de Silicio/metabolismo , Dióxido de Silicio/farmacología , Fibrosis , Inflamación/tratamiento farmacológico , Macrófagos , Neumonía/inducido químicamente , Neumonía/tratamiento farmacológico , Neumonía/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología
5.
Am J Physiol Lung Cell Mol Physiol ; 325(2): L95-L103, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37256661

RESUMEN

The development of chronic lung disease in the neonate, also known as bronchopulmonary dysplasia (BPD), is the most common long-term complication in prematurely born infants. In BPD, the disease-characteristic inflammatory response culminates in nonreversible remodeling of the developing gas exchange area, provoked by the impact of postnatal treatments such as mechanical ventilation (MV) and oxygen treatment. To evaluate the potential of prenatal treatment regimens to modulate this inflammatory response and thereby impact the vulnerability of the lung toward postnatal injury, we designed a multilayered preclinical mouse model. After administration of either prenatal vitamin D-enriched (VitD+; 1,500 IU/g food) or -deprived (VitD-; <10 IU/kg) food during gestation in C57B6 mice (the onset of mating until birth), neonatal mice were exposed to hyperoxia (FiO2 = 0.4) with or without MV for 8 h at days 5-7 of life, whereas controls spontaneously breathed room air. Prenatal vitamin D supplementation resulted in a decreased number of monocytes/macrophages in the neonatal lung undergoing postnatal injury together with reduced TGF-ß pathway activation. In consequence, neonatal mice that received a VitD+ diet during gestation demonstrated less extracellular matrix (ECM) remodeling upon lung injury, reflected by the reduction of pulmonary α-smooth muscle actin-positive fibroblasts, decreased collagen and elastin deposition, and lower amounts of interstitial tissue in the lung periphery. In conclusion, our findings support strategies that attempt to prevent vitamin D insufficiency during pregnancy as they could impact lung health in the offspring by mitigating inflammatory changes in neonatal lung injury and ameliorating subsequent remodeling of the developing gas exchange area.NEW & NOTEWORTHY Vitamin D-enriched diet during gestation resulted in reduced lung inflammation and matrix remodeling in neonatal mice exposed to clinically relevant, postnatal injury. The results underscore the need to monitor the subclinical effects of vitamin D insufficiency that impact health in the offspring when other risk factors come into play.


Asunto(s)
Displasia Broncopulmonar , Hiperoxia , Lesión Pulmonar , Neumonía , Deficiencia de Vitamina D , Humanos , Embarazo , Femenino , Recién Nacido , Animales , Ratones , Animales Recién Nacidos , Lesión Pulmonar/metabolismo , Vitamina D/farmacología , Vitamina D/metabolismo , Pulmón/metabolismo , Displasia Broncopulmonar/tratamiento farmacológico , Displasia Broncopulmonar/prevención & control , Displasia Broncopulmonar/metabolismo , Neumonía/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Hiperoxia/metabolismo , Deficiencia de Vitamina D/tratamiento farmacológico , Deficiencia de Vitamina D/metabolismo , Suplementos Dietéticos
6.
Eur Respir J ; 61(6)2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37105573

RESUMEN

BACKGROUND: Electronic cigarette (e-cigarette) vapour is gaining popularity as an alternative to tobacco smoking and can induce acute lung injury. However, the specific role of nicotine in e-cigarette vapour and its long-term effects on the airways, lung parenchyma and vasculature remain unclear. RESULTS: In vitro exposure to nicotine-containing e-cigarette vapour extract (ECVE) or to nicotine-free e-cigarette vapour extract (NF ECVE) induced changes in gene expression of epithelial cells and pulmonary arterial smooth muscle cells (PASMCs), but ECVE in particular caused functional alterations (e.g. a decrease in human and mouse PASMC proliferation by 29.3±5.3% and 44.3±8.4%, respectively). Additionally, acute inhalation of nicotine-containing e-cigarette vapour (ECV) but not nicotine-free e-cigarette vapour (NF ECV) increased pulmonary endothelial permeability in isolated lungs. Long-term in vivo exposure of mice to ECV for 8 months significantly increased the number of inflammatory cells, in particular lymphocytes, compared to control and NF ECV in the bronchoalveolar fluid (BALF) (ECV: 853.4±150.8 cells·mL-1; control: 37.0±21.1 cells·mL-1; NF ECV: 198.6±94.9 cells·mL-1) and in lung tissue (ECV: 25.7±3.3 cells·mm-3; control: 4.8±1.1 cells·mm-3; NF ECV: 14.1±2.2 cells·mm-3). BALF cytokines were predominantly increased by ECV. Moreover, ECV caused significant changes in lung structure and function (e.g. increase in airspace by 17.5±1.4% compared to control), similar to mild tobacco smoke-induced alterations, which also could be detected in the NF ECV group, albeit to a lesser degree. In contrast, the pulmonary vasculature was not significantly affected by ECV or NF ECV. CONCLUSIONS: NF ECV components induce cell type-specific effects and mild pulmonary alterations, while inclusion of nicotine induces significant endothelial damage, inflammation and parenchymal alterations.


Asunto(s)
Cigarrillo Electrónico a Vapor , Sistemas Electrónicos de Liberación de Nicotina , Neumonía , Humanos , Animales , Ratones , Nicotina/efectos adversos , Cigarrillo Electrónico a Vapor/efectos adversos , Cigarrillo Electrónico a Vapor/metabolismo , Neumonía/etiología , Neumonía/metabolismo , Pulmón/metabolismo , Extractos Vegetales/metabolismo , Extractos Vegetales/farmacología
7.
Biomed Pharmacother ; 163: 114793, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37121151

RESUMEN

Shiwei Longdanhua Granule (SWLDH) is a classic Tibetan medicine (TM) ranking in the top 20 Chinese patent medicines in prescription rate to treat respiratory diseases like pneumonia, acute and chronic tracheobronchitis, acute exacerbation of COPD and bronchial asthma in solution of inflammation, cough and phlegm obstruction in clinical practice. However, its systematic pharmacological mechanisms have not been elucidated yet. Here, we studied the therapeutic efficacy of SWLDH in treatment of acute respiratory diseases in BALB/c mice by comprehensive analysis of airway inflammation, oxidative stress, mucus hypersecretion, cough hypersensitivities and indicators associated with the development of chronic diseases. Our results show that SWLDH might exhibit its inhibitory effects on pulmonary inflammation by interference with arachidonic acid (AA) metabolism pathways. Oxidative stress that highly related to the degree of tissue injury could be alleviated by enhancing the reductive activities of glutathione redox system, thioredoxin system and the catalytic activities of catalase and superoxide dismutase (SOD) after SWLDH treatment. In addition, SWLDH could significantly abrogate the mucus hypersecretion induced bronchiole obstruction by inactivate the globlet cells and decrease the secretion of gel-forming mucins (MUC5AC and MUC5B) under pathological condition, demonstrating its mucoactive potency. SWLDH also showed reversed effects on the release of neuropeptides that are responsible for airway sensory hypersensitivity. Simultaneously observed inhibition of calcium influx, reduction in in vivo biosynthesis of acetylcholine and the recovery of the content of cyclic adenosine monophosphate (cAMP) might collaboratively contribute to cause airway smooth muscle cells (ASMCs) relexation. These findings indicated that SWLDH might exhibited antitussive potency via suppression of the urge to cough and ASMCs contraction. Moreover, SWLDH might affect airway remodeling. We found SWLDH could retard the elevation of TGF-ß1 and α-SMA, which are important indicators for hyperplasia and contraction during the progression of the chronic airway inflammatory diseases like COPD and asthma.


Asunto(s)
Asma , Hipersensibilidad , Neumonía , Enfermedad Pulmonar Obstructiva Crónica , Ratones , Animales , Tos/inducido químicamente , Tos/tratamiento farmacológico , Lipopolisacáridos/metabolismo , Neumonía/inducido químicamente , Neumonía/tratamiento farmacológico , Neumonía/metabolismo , Moco/metabolismo , Asma/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Enfermedad Crónica , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Estrés Oxidativo , Mucina 5AC/metabolismo
8.
Am J Physiol Lung Cell Mol Physiol ; 324(4): L536-L549, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36852927

RESUMEN

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.


Asunto(s)
Receptor 2 de Folato , Neumonía , Humanos , Monocitos/metabolismo , Lipopolisacáridos/farmacología , Lipopolisacáridos/metabolismo , Macrófagos/metabolismo , Neumonía/inducido químicamente , Neumonía/genética , Neumonía/metabolismo , Inflamación/genética , Inflamación/metabolismo , Análisis de Secuencia de ARN/métodos , Receptor 2 de Folato/metabolismo
9.
Nutrients ; 14(23)2022 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-36501052

RESUMEN

In the wake of the COVID-19 pandemic, lung disorders have become a major health concern for humans. Allergic asthma is the most prevalent form of asthma, and its treatments target the inflammation process. Despite significant developments in the diagnosis and management of allergic asthma, side effects are a major concern. Additionally, its extreme heterogeneity impedes the efficacy of the majority of treatments. Thus, newer, safer therapeutic substances, such as natural products, are desired. Citrus junos Tanaka has traditionally been utilized as an anti-inflammatory, sedative, antipyretic, and antitoxic substance. In this study, the protective effects of Citrus junos Tanaka peel extract (B215) against lung inflammation were examined, and efforts were made to understand the underlying protective mechanism using an HDM-induced lung inflammation murine model. The administration of B215 reduced immune cell infiltration in the lungs, plasma IgE levels, airway resistance, mucus hypersecretions, and cytokine production. These favorable effects alleviated HDM-induced lung inflammation by modulating the NF-κB signaling pathway. Hence, B215 might be a promising functional food to treat lung inflammation without adverse effects.


Asunto(s)
Asma , COVID-19 , Citrus , Neumonía , Ratones , Humanos , Animales , Pandemias , Modelos Animales de Enfermedad , COVID-19/metabolismo , Pulmón , Neumonía/tratamiento farmacológico , Neumonía/metabolismo , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Extractos Vegetales/metabolismo , Inmunidad
10.
J Ethnopharmacol ; 298: 115637, 2022 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-35970312

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Respiratory syncytial virus (RSV) is a common pathogen that causes lower respiratory tract disease in infants and the elderly, and no vaccination is presently available. Qingfei oral liquid (QF), a traditional Chinese medicine formula, has been shown in clinic to have anti-inflammatory properties. AIM OF THE STUDY: The present study investigated whether QF can suppress RSV-induced lung inflammation in mice models via fatty acid-dependent macrophage polarization. MATERIAL AND METHODS: BALB/c mice were given a low, medium, or high dose of QF intragastrically for four consecutive days following RSV infection. The lung inflammatory status was assessed using H&E staining and cytokine assays. The active components of QF and fatty acid metabolism were analyzed using ultra-high-performance liquid chromatography/tandem mass spectrometry (UPLC-MS/MS). A lipid metabolism-related pathway was found through network pharmacology and molecular docking investigations. Western blotting assays were used to determine the levels of ATP-citrate lyase (ACLY), peroxisome proliferation-activated receptor alpha (PPAR), Akt protein kinase B and its phosphorylated form in Akt signaling. Flow cytometry was used to quantify the number of macrophage subtypes (M1/M2), and immunohistochemistry was used to examine the expression of inducible nitric oxide synthase (iNOS) and arginase-1 (Arg-1). RESULTS: In the lung tissues of RSV-infected mice, QF suppressed the transcription of pro-inflammatory proteins such as interleukin-1 beta (IL-1ß), tumor necrosis factor alpha (TNF-α), and interleukin-6 (IL-6), while increasing the level of anti-inflammatory factors such as interleukin-10 (IL-10). The alterations in metabolic enzyme activity mediated by Akt signaling were linked to QF's significant reduction in lung fatty acid accumulation. Lower ACLY expression and higher PPAR expression were found after QF treatment, showing that these two enzymes were downstream targets of Akt signaling, controlling fatty acid synthesis (FAS) and fatty acid oxidation (FAO), respectively. The reprogramming of fatty acid metabolism resulted in the polarization of macrophages from M1 to M2, with lower expression of iNOS and higher expression of Arg-1. Additionally, application of an Akt agonist (SC-79) reduced QF's anti-inflammatory effects by increasing FAS and decreasing macrophage polarization. CONCLUSIONS: QF inhibited Akt-mediated FAS and polarized M1 to M2 macrophages, resulting in an anti-inflammatory impact.


Asunto(s)
Neumonía , Infecciones por Virus Sincitial Respiratorio , Animales , Antiinflamatorios/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Cromatografía Liquida , Medicamentos Herbarios Chinos , Ácidos Grasos/metabolismo , Inflamación/tratamiento farmacológico , Interleucina-6/metabolismo , Macrófagos , Ratones , Ratones Endogámicos BALB C , Simulación del Acoplamiento Molecular , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Neumonía/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Infecciones por Virus Sincitial Respiratorio/metabolismo , Transducción de Señal , Espectrometría de Masas en Tándem
11.
Respir Res ; 23(1): 40, 2022 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-35236342

RESUMEN

BACKGROUND: In chronic obstructive pulmonary disease (COPD), exacerbations cause acute inflammatory flare-ups and increase the risk for hospitalization and mortality. Exacerbations are common in all disease stages and are often caused by bacterial infections e.g., non-typeable Heamophilus influenzae (NTHi). Accumulating evidence also associates vitamin D deficiency with the severity of COPD and exacerbation frequency. However, it is still unclear whether vitamin D deficiency when combined with cigarette smoking would worsen and prolong exacerbations caused by repeated infections with the same bacterial strain. METHODS: Vitamin D sufficient (VDS) and deficient (VDD) mice were exposed to nose-only cigarette smoke (CS) for 14 weeks and oropharyngeally instilled with NTHi at week 6, 10 and 14. Three days after the last instillation, mice were assessed for lung function, tissue remodeling, inflammation and immunity. The impact of VDD and CS on inflammatory cells and immunoglobulin (Ig) production was also assessed in non-infected animals while serum Ig production against NTHi and dsDNA was measured in COPD patients before and 1 year after supplementation with Vitamin D3. RESULTS: VDD enhanced NTHi eradication, independently of CS and complete eradication was reflected by decreased anti-NTHi Ig's within the lung. In addition, VDD led to an increase in total lung capacity (TLC), lung compliance (Cchord), MMP12/TIMP1 ratio with a rise in serum Ig titers and anti-dsDNA Ig's. Interestingly, in non-infected animals, VDD exacerbated the CS-induced anti-NTHi Ig's, anti-dsDNA Ig's and inflammatory cells within the lung. In COPD patients, serum Ig production was not affected by vitamin D status but anti-NTHi IgG increased after vitamin D3 supplementation in patients who were Vitamin D insufficient before treatment. CONCLUSION: During repeated infections, VDD facilitated NTHi eradication and resolution of local lung inflammation through production of anti-NTHi Ig, independently of CS whilst it also promoted autoantibodies. In COPD patients, vitamin D supplementation could be protective against NTHi infections in vitamin D insufficient patients. Future research is needed to decipher the determinants of dual effects of VDD on adaptive immunity. TRAIL REGISTRATION: ClinicalTrials, NCT00666367. Registered 23 April 2008, https://www.clinicaltrials.gov/ct2/show/study/NCT00666367 .


Asunto(s)
Fumar Cigarrillos/efectos adversos , Infecciones por Haemophilus/complicaciones , Haemophilus influenzae/inmunología , Pulmón/microbiología , Neumonía/complicaciones , Deficiencia de Vitamina D/metabolismo , Animales , Modelos Animales de Enfermedad , Infecciones por Haemophilus/metabolismo , Infecciones por Haemophilus/microbiología , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Neumonía/metabolismo
12.
J Ethnopharmacol ; 292: 115120, 2022 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-35202713

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Asthma is the leading inflammatory disease of the airways with inadequate therapeutic options. 'Malla Sindoor' (MS) is a metal-based ethnomedicinal formulation that has been prescribed in the ancient traditional medicinal system for treating chronic inflammations. AIM OF THE STUDY: Here, we validated the anti-inflammatory and anti-asthmatic properties of traditional metallic medicine MS in asthmatic mice model and in LPS stimulated human monocytic THP-1 cells, by examining the relevant cellular, biochemical and molecular intermediates. MATERIALS AND METHODS: Scanning Electron Microscope (SEM), Electron Dispersive X-ray (EDX), and X-Ray Diffraction (XRD) were performed to characterize MS particles. Allergic asthma was induced in Balb/c mice through intraperitoneal ovalbumin (OVA) injection. Experimental groups include, normal control, disease control, Dexamethasone (2 mg/kg) and three MS treated groups: 4.3 mg/kg, 13 mg/kg, and 39 mg/kg. Quantitative PCR, inflammatory cytokines and anti-oxidant enzymes, and histological analysis were performed, in the treated mice and LPS stimulated human monocytic THP-1 cells for determining the MS efficacy. RESULTS: SEM image analysis showed the MS to be heterogenous in shape with a particle size distribution between 100 nm-1 µm. Elemental composition showed the presence of mercury (Hg), arsenic (As), and sulphur (S) along with other elements in the forms of mercury sulfide, arsenic trioxide, and their alloy crystals. OVA-challenge of the Balb/c mice resulted in the development of overt pathological features for allergic asthma including smooth muscle thickening and collagen deposition. Mice receiving MS-exhibited alleviation of allergic asthma features. BAL fluid analysis showed a decrease in the total cell count and decreases in neutrophils, monocytes, lymphocytes, and eosinophils. Further, the stimulated levels of interleukin (IL)-1ß, -6, and TNF-α cytokines and antioxidant levels were also reduced upon MS-treatment. At the molecular level, MS-treatment reduced stimulated mRNA expression levels for IL-4, -5, -10, -13, -33, and IFN-γ cytokines. Histological analysis following MS-treatment of OVA-stimulated mice lungs showed a reduction in mucus accumulation in airways, decreases in peribronchial collagen deposition, bronchial smooth muscle thickening, and attenuation of inflammatory cell infiltration. In addition, under in-vitro conditions, MS-treatment attenuated the LPS induced secretion of IL-1ß, -6, and TNF-α from THP-1 cells. CONCLUSION: Collectively, the results suggest that MS acts as an effective anti-asthmatic and anti-inflammatory agent, by regulating various cellular, biochemical and molecular intermediates.


Asunto(s)
Antiasmáticos , Antiinflamatorios , Asma , Neumonía , Animales , Antiasmáticos/farmacología , Antiinflamatorios/uso terapéutico , Antioxidantes/farmacología , Asma/tratamiento farmacológico , Asma/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Lipopolisacáridos/farmacología , Medicina Tradicional , Mercurio/toxicidad , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/farmacología , Estrés Oxidativo , Neumonía/tratamiento farmacológico , Neumonía/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
13.
Int J Chron Obstruct Pulmon Dis ; 16: 3317-3335, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34916790

RESUMEN

PURPOSE: Chronic obstructive pulmonary disease (COPD), a prevalent obstructive airway disease, has become the third most common cause of death globally. Xuanbai Chengqi decoction (XBCQ) is a traditional Chinese medicine prescription for the acute exacerbation of COPD. Here, we aimed to reveal the therapeutic effects of XBCQ administration and its molecular mechanisms mediated by Th17/Treg balance and gut microbiota. METHODS: We determined the counts of Th17 and Treg cells in the serum of 15 COPD and 10 healthy subjects. Then, cigarette smoke extract-induced COPD mice were gavaged with low, middle, and high doses of XBCQ, respectively. Weight loss, pulmonary function and inflammation, Th17/Treg ratio, and gut microbiota were measured to evaluate the efficacy of XBCQ on COPD. RESULTS: COPD patients had a higher Th17/Treg ratio in the serum than healthy controls, which was consistent with the results in the lung and colon of COPD mice. The middle dose of XBCQ (M-XBCQ) significantly decreased the weight loss and improved the pulmonary function (FEV0.2/FVC) in COPD mice. Moreover, M-XBCQ alleviated lung inflammation by rectifying the Th17/Treg imbalance, reducing the expressions of TNF-α, IL-1ß, and MMP-9, and suppressing inflammatory cells infiltration. Meanwhile, M-XBCQ greatly improved the microbial homeostasis in COPD mice by accumulating probiotic Gordonibacter and Akkermansia but inhibiting the growth of pathogenic Streptococcus, which showed significant correlations with pulmonary injury. CONCLUSION: Oral M-XBCQ could alleviate COPD exacerbations by reshaping the gut microbiota and improving the Th17/Treg balance, which aids in elucidating the mechanism through which XBCQ as a therapy for COPD.


Asunto(s)
Microbioma Gastrointestinal , Neumonía , Enfermedad Pulmonar Obstructiva Crónica , Animales , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos , Humanos , Ratones , Neumonía/tratamiento farmacológico , Neumonía/metabolismo , Neumonía/prevención & control , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Linfocitos T Reguladores , Células Th17/metabolismo
14.
Front Immunol ; 12: 701341, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34777335

RESUMEN

The essential micronutrient Selenium (Se) is co-translationally incorporated as selenocysteine into proteins. Selenoproteins contain one or more selenocysteines and are vital for optimum immunity. Interestingly, many pathogenic bacteria utilize Se for various biological processes suggesting that Se may play a role in bacterial pathogenesis. A previous study had speculated that Francisella tularensis, a facultative intracellular bacterium and the causative agent of tularemia, sequesters Se by upregulating Se-metabolism genes in type II alveolar epithelial cells. Therefore, we investigated the contribution of host vs. pathogen-associated selenoproteins in bacterial disease using F. tularensis as a model organism. We found that F. tularensis was devoid of any Se utilization traits, neither incorporated elemental Se, nor exhibited Se-dependent growth. However, 100% of Se-deficient mice (0.01 ppm Se), which express low levels of selenoproteins, succumbed to F. tularensis-live vaccine strain pulmonary challenge, whereas 50% of mice on Se-supplemented (0.4 ppm Se) and 25% of mice on Se-adequate (0.1 ppm Se) diet succumbed to infection. Median survival time for Se-deficient mice was 8 days post-infection while Se-supplemented and -adequate mice was 11.5 and >14 days post-infection, respectively. Se-deficient macrophages permitted significantly higher intracellular bacterial replication than Se-supplemented macrophages ex vivo, corroborating in vivo observations. Since Francisella replicates in alveolar macrophages during the acute phase of pneumonic infection, we hypothesized that macrophage-specific host selenoproteins may restrict replication and systemic spread of bacteria. F. tularensis infection led to an increased expression of several macrophage selenoproteins, suggesting their key role in limiting bacterial replication. Upon challenge with F. tularensis, mice lacking selenoproteins in macrophages (TrspM) displayed lower survival and increased bacterial burden in the lung and systemic tissues in comparison to WT littermate controls. Furthermore, macrophages from TrspM mice were unable to restrict bacterial replication ex vivo in comparison to macrophages from littermate controls. We herein describe a novel function of host macrophage-specific selenoproteins in restriction of intracellular bacterial replication. These data suggest that host selenoproteins may be considered as novel targets for modulating immune response to control a bacterial infection.


Asunto(s)
Francisella tularensis/inmunología , Interacciones Huésped-Patógeno/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Selenoproteínas/metabolismo , Tularemia/etiología , Tularemia/metabolismo , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Francisella tularensis/genética , Francisella tularensis/patogenicidad , Ratones , Neumonía/inmunología , Neumonía/metabolismo , Neumonía/microbiología , Neumonía/patología , Tularemia/mortalidad , Virulencia/genética , Factores de Virulencia/genética
15.
Life Sci ; 287: 120059, 2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34728227

RESUMEN

AIMS: Bleomycin, an important toxic anti-cancer agent, induces pulmonary fibrosis. The significance of oxidative stress and inflammation in promoting of bleomycin-induced idiopathic pulmonary fibrosis (IPF) has been reported. Thus, we evaluated the protective effects of carnosol as a robust natural antioxidant and anti-inflammatory agent for bleomycin-related IPF in rats. MAIN METHODS: Male Wistar rats (n = 40) were randomly assigned to five groups. Group 1 was administrated with saline (intratracheally) on day 7 and oral gavage of dimethyl sulfoxide (DMSO, 0.05%) from day 1 to day 28. Group 2 received a single dose of bleomycin (intratracheally, 7.5 UI/kg) on day 7 and oral gavage of saline for 28 days. Groups 3, 4 and 5 were administrated with bleomycin (single dose) on day 7, along with oral administration of carnosol (at doses 10, 20 and 40 mg/kg, respectively) from day 1 to day 28. The lungs were isolated to measure the histopathological and biochemical and inflammatory markers. KEY FINDINGS: Carnosol treatment significantly reduced malondialdehyde, nitric oxide, protein carbonyl, tumor necrosis factor- α, interleukin-6 levels and myeloperoxidase activity in the lungs of rats exposed to bleomycin. Also, lung glutathione content, catalase, glutathione peroxidase and superoxide dismutase activities significantly increased in the carnosol/bleomycin-treated group than the bleomycin group. Lung index, hydroxyproline content, fibrosis and histopathological changes, also significantly decreased by carnosol therapy. SIGNIFICANCE: Treatment with carnosol can modulate biochemical and histological alterations caused by bleomycin. Thus, it can be regarded as an appropriate therapeutic approach for IPF.


Asunto(s)
Abietanos/uso terapéutico , Bleomicina/toxicidad , Estrés Oxidativo/efectos de los fármacos , Neumonía/tratamiento farmacológico , Fibrosis Pulmonar/tratamiento farmacológico , Abietanos/farmacología , Animales , Antibióticos Antineoplásicos/toxicidad , Relación Dosis-Respuesta a Droga , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Estrés Oxidativo/fisiología , Neumonía/inducido químicamente , Neumonía/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Ratas , Ratas Wistar , Rosmarinus
16.
Nutrients ; 13(10)2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-34684415

RESUMEN

YG-1 extract used in this study is a mixture of Lonicera japonica, Arctic Fructus, and Scutellariae Radix. The present study was designed to investigate the effect of YG-1 extract on bronchodilatation (ex vivo) and acute bronchial and pulmonary inflammation relief (in vivo). Ex vivo: The bronchodilation reaction was confirmed by treatment with YG-1 concentration-accumulation (0.01, 0.03, 0.1, 0.3, and 1 mg/mL) in the bronchial tissue ring pre-contracted by acetylcholine (10 µM). As a result, YG-1 extract is considered to affect bronchodilation by increased cyclic adenosine monophosphate, cAMP) levels through the ß2-adrenergic receptor. In vivo: experiments were performed in C57BL/6 mice were divided into the following groups: control group; PM2.5 (fine particulate matter)-exposed group (PM2.5, 200 µg/kg/mL saline); and PM2.5-exposed + YG-1 extract (200 mg/kg/day) group. The PM2.5 (200 µg/kg/mL saline) was exposed for 1 h for 5 days using an ultrasonic nebulizer aerosol chamber to instill fine dust in the bronchi and lungs, thereby inducing acute lung and bronchial inflammation. From two days before PM2.5 exposure, YG-1 extract (200 mg/kg/day) was administered orally for 7 days. The PM2.5 exposure was involved in airway remodeling and inflammation, suggesting that YG-1 treatment improves acute bronchial and pulmonary inflammation by inhibiting the inflammatory cytokines (NLRP3/caspase-1 pathway). The application of YG-1 extract with broncho-dilating effect to acute bronchial and pulmonary inflammation animal models has great significance in developing therapeutic agents for respiratory diseases. Therefore, these results can provide essential data for the development of novel respiratory symptom relievers. Our study provides strong evidence that YG-1 extracts reduce the prevalence of respiratory symptoms and the incidence of non-specific lung diseases and improve bronchial and lung function.


Asunto(s)
Broncodilatadores/farmacología , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Extractos Vegetales/farmacología , Neumonía/metabolismo , Neumonía/patología , Animales , Biomarcadores , Broncodilatadores/administración & dosificación , Broncodilatadores/química , Cromatografía Líquida de Alta Presión , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Ratones , Estructura Molecular , Material Particulado/efectos adversos , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Neumonía/tratamiento farmacológico , Neumonía/etiología , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal/efectos de los fármacos
17.
Toxicol Appl Pharmacol ; 431: 115730, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34601004

RESUMEN

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.


Asunto(s)
Mediadores de Inflamación/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Pulmón/efectos de los fármacos , Síndrome Metabólico/complicaciones , Nanopartículas del Metal/toxicidad , Neumonía/inducido químicamente , Compuestos de Plata/toxicidad , Animales , Antiinflamatorios/farmacología , Citocinas/genética , Citocinas/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/farmacología , Regulación de la Expresión Génica , Ácidos Hidroxieicosatetraenoicos/farmacología , Metabolismo de los Lípidos/genética , Pulmón/metabolismo , Masculino , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/genética , Síndrome Metabólico/metabolismo , Ratones Endogámicos C57BL , Neumonía/genética , Neumonía/metabolismo , Neumonía/prevención & control , Transducción de Señal
18.
Biomed Pharmacother ; 143: 112101, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34474340

RESUMEN

Chronic obstructive pulmonary disease (COPD) is among the leading causes of death worldwide, and is characterized by persistent respiratory symptoms and airflow limitation due to chronic airway inflammation. Cigarette smoking is a major risk factor for COPD. This study aims to determine the therapeutic effects of polysaccharides extracted from Dendrobium officinale (DOPs), a valuable traditional Chinese Medicinal herb, on cigarette smoke (CS)-induced airway inflammation in a rat passive smoking model. Male Sprague-Dawley rats were exposed to CS or sham air (SA) as control for a 56-day period. On Day 29, rats were subdivided and given water, DOPs or N-acetylcysteine (NAC) via oral gavage on a daily basis for the remaining duration. DOPs reduced CS-induced oxidative stress as evidenced by reducing malondialdehyde (MDA) levels in the lung. DOPs also exerted potent anti-inflammatory properties as evidenced by a reduction in the number of lymphocytes and monocytes in serum, significantly attenuating infiltration of inflammatory cells in lung tissue, as well as pro-inflammatory mediators in serum, bronchoalveolar lavage (BAL) and lung. Additionally, DOPs inhibited the CS-induced activation of ERK, p38 MAPK and NF-κB signaling pathways. These findings suggest that DOPs may have potentially beneficial effects in limiting smoking-related lung oxidative stress, and inflammation mediated via the inhibition of MAPK and NF-κB signaling pathways in smokers, without or with COPD.


Asunto(s)
Antioxidantes/farmacología , Dendrobium , Pulmón/efectos de los fármacos , Extractos Vegetales/farmacología , Neumonía/prevención & control , Polisacáridos/farmacología , Humo/efectos adversos , Productos de Tabaco/efectos adversos , Animales , Antioxidantes/aislamiento & purificación , Dendrobium/química , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Extractos Vegetales/aislamiento & purificación , Neumonía/etiología , Neumonía/inmunología , Neumonía/metabolismo , Polisacáridos/aislamiento & purificación , Ratas Sprague-Dawley , Transducción de Señal
19.
Respir Res ; 22(1): 99, 2021 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-33823870

RESUMEN

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.


Asunto(s)
Antiinflamatorios/farmacología , Tratamiento Farmacológico de COVID-19 , Reposicionamiento de Medicamentos , Hipoxia/tratamiento farmacológico , Género Justicia , Pulmón/efectos de los fármacos , Extractos Vegetales/farmacología , Neumonía/prevención & control , Fibrosis Pulmonar/tratamiento farmacológico , Sepsis/tratamiento farmacológico , Animales , Antiinflamatorios/aislamiento & purificación , Bleomicina , COVID-19/metabolismo , COVID-19/virología , Ciego/microbiología , Ciego/cirugía , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hipoxia/genética , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Mediadores de Inflamación/metabolismo , Género Justicia/química , Ligadura , Pulmón/metabolismo , Pulmón/microbiología , Pulmón/patología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Extractos Vegetales/aislamiento & purificación , Neumonía/genética , Neumonía/metabolismo , Neumonía/microbiología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Sepsis/genética , Sepsis/metabolismo , Sepsis/microbiología , Transcriptoma
20.
Am J Physiol Lung Cell Mol Physiol ; 320(5): L757-L769, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33565386

RESUMEN

Severe asthma is a chronic airway disease that exhibits poor response to conventional asthma therapies. Growing evidence suggests that elevated hypoxia increases the severity of asthmatic inflammation among patients and in model systems. In this study, we elucidate the therapeutic effects and mechanistic basis of Adhatoda vasica (AV) aqueous extract on mouse models of acute allergic as well as severe asthma subtypes at physiological, histopathological, and molecular levels. Oral administration of AV extract attenuates the increased airway resistance and inflammation in acute allergic asthmatic mice and alleviates the molecular signatures of steroid (dexamethasone) resistance like IL-17A, KC (murine IL-8 homologue), and HIF-1α (hypoxia-inducible factor-1α) in severe asthmatic mice. AV inhibits HIF-1α levels through restoration of expression of its negative regulator-PHD2 (prolyl hydroxylase domain-2). Alleviation of hypoxic response mediated by AV is further confirmed in the acute and severe asthma model. AV reverses cellular hypoxia-induced mitochondrial dysfunction in human bronchial epithelial cells-evident from bioenergetic profiles and morphological analysis of mitochondria. In silico docking of AV constituents reveal higher negative binding affinity for C and O-glycosides for HIF-1α, IL-6, Janus kinase 1/3, TNF-α, and TGF-ß-key players of hypoxia inflammation. This study for the first time provides a molecular basis of action and effect of AV whole extract that is widely used in Ayurveda practice for diverse respiratory ailments. Further, through its effect on hypoxia-induced mitochondrial dysfunction, the study highlights its potential to treat severe steroid-resistant asthma.


Asunto(s)
Asma/tratamiento farmacológico , Hipoxia/complicaciones , Género Justicia/química , Mitocondrias/efectos de los fármacos , Fitoquímicos/farmacología , Extractos Vegetales/farmacología , Neumonía/prevención & control , Animales , Asma/etiología , Asma/metabolismo , Asma/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos BALB C , Mitocondrias/metabolismo , Mitocondrias/patología , Neumonía/etiología , Neumonía/metabolismo , Neumonía/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA