Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Medicinas Complementárias
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
mBio ; 12(4): e0097021, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34340553

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic has caused significant morbidity and mortality on a global scale. The etiologic agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), initiates host cell entry when its spike protein (S) binds to its receptor, angiotensin-converting enzyme 2 (ACE2). In airway epithelia, the spike protein is cleaved by the cell surface protease TMPRSS2, facilitating membrane fusion and entry at the cell surface. This dependence on TMPRSS2 and related proteases suggests that protease inhibitors might limit SARS-CoV-2 infection in the respiratory tract. Here, we tested two serine protease inhibitors, camostat mesylate and nafamostat mesylate, for their ability to inhibit entry of SARS-CoV-2 and that of a second pathogenic coronavirus, Middle East respiratory syndrome coronavirus (MERS-CoV). Both camostat and nafamostat reduced infection in primary human airway epithelia and in the Calu-3 2B4 cell line, with nafamostat exhibiting greater potency. We then assessed whether nafamostat was protective against SARS-CoV-2 in vivo using two mouse models. In mice sensitized to SARS-CoV-2 infection by transduction with human ACE2, intranasal nafamostat treatment prior to or shortly after SARS-CoV-2 infection significantly reduced weight loss and lung tissue titers. Similarly, prophylactic intranasal treatment with nafamostat reduced weight loss, viral burden, and mortality in K18-hACE2 transgenic mice. These findings establish nafamostat as a candidate for the prevention or treatment of SARS-CoV-2 infection and disease pathogenesis. IMPORTANCE The causative agent of COVID-19, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), requires host cell surface proteases for membrane fusion and entry into airway epithelia. We tested the hypothesis that inhibitors of these proteases, the serine protease inhibitors camostat and nafamostat, block infection by SARS-CoV-2. We found that both camostat and nafamostat reduce infection in human airway epithelia, with nafamostat showing greater potency. We then asked whether nafamostat protects mice against SARS-CoV-2 infection and subsequent COVID-19 lung disease. We performed infections in mice made susceptible to SARS-CoV-2 infection by introducing the human version of ACE2, the SARS-CoV-2 receptor, into their airway epithelia. We observed that pretreating these mice with nafamostat prior to SARS-CoV-2 infection resulted in better outcomes, in the form of less virus-induced weight loss, viral replication, and mortality than that observed in the untreated control mice. These results provide preclinical evidence for the efficacy of nafamostat in treating and/or preventing COVID-19.


Asunto(s)
Benzamidinas/farmacología , Ésteres/farmacología , Guanidinas/farmacología , SARS-CoV-2/efectos de los fármacos , Serina Endopeptidasas/metabolismo , Inhibidores de Serina Proteinasa/farmacología , Internalización del Virus/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Tratamiento Farmacológico de COVID-19
2.
J Mol Med (Berl) ; 98(10): 1493-1503, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32948884

RESUMEN

The phytotherapeutic compound EPs® 7630, an extract manufactured from Pelargonium sidoides roots, is frequently used for the treatment of airway infections. Nevertheless, the knowledge of the mode of action of EPs® 7630 is still sparse. Our study aimed at further elucidating the underlying pharmacological mechanisms by focusing on antimicrobial defense mechanisms of EPs® 7630. While investigating the influence of EPs® 7630 on lymphokine production by PBMCs, we found that EPs® 7630 is a novel inducer of IL-22 and IL-17. This cytokine-inducing effect was most pronounced for IL-22 and clearly dose-dependent starting from 1 µg/ml of the extract. Furthermore, EPs® 7630 pretreatment selectively enhanced the IL-22 and IL-17 production capacity of CD3/28-activated PBMCs while strongly limiting the IFN-γ production capacity of innate lymphoid cells. The relevance of EPs® 7630-induced IL-22 production was proven in vitro and in vivo, where IL-22 provoked a strong increase of the antimicrobial protein S100A9 in lung epithelial cells and pulmonary tissue, respectively. A detailed analysis of IL-22 induction modi revealed no direct influence of EPs® 7630 on the basal or anti-CD3/CD28 antibody-induced IL-22 production by CD4+ memory T cells. In fact, EPs® 7630-induced IL-22 production by CD4+ memory T cells was found to be essentially dependent on soluble mediators (IL-1/IL-23) as well as on direct cellular contact with monocytes. In summary, our study reveals a new immune-modulating function of EPs® 7630 that might confer IL-22 and IL-17-induced protection from bacterial airway infection. KEY MESSAGES: EPs® 7630 selectively strengthens IL-22 and IL-17 production of memory T cells. EPs® 7630 limits the IFN-y production capacity of innate lymphoid cells. EPs® 7630-caused IL-22 production by T cells is essentially dependent on monocytes. IL-22 increase antimicrobial proteins (AMPs) in airway epithelium. EPs® 7630 might protect against airway infection by induction of AMP-inducers.


Asunto(s)
Antiinfecciosos/farmacología , Interleucinas/biosíntesis , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Extractos Vegetales/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Citocinas/biosíntesis , Humanos , Inmunidad Innata/efectos de los fármacos , Memoria Inmunológica , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Masculino , Ratones , Monocitos/inmunología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Linfocitos T/inmunología , Interleucina-22
3.
Am J Rhinol Allergy ; 34(4): 471-481, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32046501

RESUMEN

BACKGROUND: Respiratory epithelium is a key defense against inhaled pathogens. Vitamin D3 (VD) has been suggested to modulate airway inflammation; however, its effect on innate airway defenses, the physical barrier, mucociliary apparatus, and cytokine release remains unclear. OBJECTIVE: To investigate the outcomes of VD application prior to challenge in an in vitro model of human sinonasal epithelium, through assessment of epithelial transepithelial resistance (TER), cilia beat frequency (CBF), and interleukin (IL)-6 release, and secondarily to determine whether topical VD is beneficial to patients with inflammatory sinonasal pathology. METHODS: Primary human sinonasal epithelial cells from patients with eosinophilic chronic rhinosinusitis (eCRS) and healthy controls were cultured in air-liquid interface (ALI). Well-differentiated cultures from each patient were pretreated for 24 hours with 4 different VD doses. Toxicity was quantified at 24 hours in unchallenged ALI by lactate dehydrogenase (LDH) assay. Innate responses were assessed by measuring TER and CBF before and up to 24 hours after house dust mite Dermatophagoides pteronyssinus challenge. IL-6 release was evaluated 24-hour postchallenge. RESULTS: Fifteen patients (53 ± 13.5 years, 60% females, 53% eCRS) representing 120 ALI wells were assessed. VD (0, 25, 50, 150 IU/mL) released less LDH than vehicle, indicating noncytotoxicity (0.15 ± 0.02; 0.15 ± 0.00; 0.14 ± 0.02; 0.11 ± 0.01 vs 0.17 ± 0.03, P = .004). VD increased TER for eCRS wells at 5 minutes (50 IU/mL: Δ6.76 ± 3.93 vs Δ3.87 ± 2.46, P = .04) and 24 hours (50 IU/mL: Δ0.88 ± 0.49 vs Δ0.40 ± 0.42, P = .02; 150 IU/mL: Δ1.06 ± 0.58 vs Δ0.47 ± 0.46, P = .01). CBF increased at 1 hour for eCRS wells (50 IU/mL: Δ0.62 ± 0.14 vs Δ0.41 ± 0.13, P = .001; 150 IU/ml: Δ0.60 ± 0.13 vs Δ0.38 ± 0.11, P < .001). IL-6 release was similar between normal and eCRS wells. CONCLUSION: Topical VD supplementation in eCRS patients may be beneficial for innate epithelial defenses. VD is noncytotoxic and does not adversely affect the physical barrier, mucociliary apparatus, or IL-6 release. Further studies should clarify its potential as a therapeutic agent.


Asunto(s)
Cilios/patología , Eosinófilos/inmunología , Hipersensibilidad/terapia , Inflamación/terapia , Mucosa Nasal/patología , Senos Paranasales/patología , Mucosa Respiratoria/patología , Rinitis/terapia , Sinusitis/terapia , Vitamina D/farmacología , Administración Tópica , Adulto , Anciano , Animales , Antígenos Dermatofagoides/inmunología , Células Cultivadas , Enfermedad Crónica , Femenino , Humanos , Inmunomodulación , Interleucina-6/metabolismo , Masculino , Persona de Mediana Edad , Pyroglyphidae , Rinitis/patología , Sinusitis/patología
4.
J Ethnopharmacol ; 249: 112425, 2020 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-31765763

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Mucus hypersecretion (MH) is recognized as a key pathophysiological and clinical feature of many airway inflammatory diseases. MUC5AC is a major component of airway mucus. Tanreqing injection (TRQ) is a widely used herbal formula for the treatment of respiratory inflammations for years in China. However, a holistic network pharmacology approach to understanding its therapeutic mechanisms against MH has not been pursued. AIM OF THE STUDY: This study aimed to explore the systems-level potential active compounds and therapeutic mechanisms of TRQ in the treatment of MH. MATERIALS AND METHODS: We established systems pharmacology-based strategies comprising compound screenings, target predictions, and pathway identifications to speculate the potential active compounds and therapeutic targets of TRQ. We also applied compound-target and target-disease network analyses to evaluate the possible action mechanisms of TRQ. Then, lipopolysaccharide (LPS)-induced Sprague-Dawley (SD) rat model was constructed to assess the effect of TRQ in the treatment of MH and to validate the possible molecular mechanisms as predicted in systems pharmacology approach. RESULTS: The comprehensive compound collection successfully generated 55 compound candidates from TRQ. Among them, 11 compounds with high relevance to the potential targets were defined as representative and potential active ingredients in TRQ formula. Target identification revealed 172 potential targets, including pro-inflammatory cytokines of tumor necrosis factor α (TNF-α), interleukin (IL)-6, and IL-8. Pathway analyses uncovered the possible action of TRQ in the regulation of IL-17 signaling pathway and its downstream protein MUC5AC. Then in vivo experiment indicated that TRQ could significantly inhibit LPS stimulated MUC5AC over-production as well as the expression of TNF-α, IL-6, IL-8, and IL-17A, in both protein and mRNA levels. CONCLUSIONS: Based on the systems pharmacology method and in vivo experiment, our work provided a general knowledge on the potential active compounds and possible therapeutic targets of TRQ formula in its anti-MH process. This work might suggest directions for further research on TRQ and provide more insight into better understanding the chemical and pharmacological mechanisms of complex herbal prescriptions in a network perspective.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Etnofarmacología/métodos , Moco/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Mucosa Respiratoria/efectos de los fármacos , Animales , Análisis de Datos , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/uso terapéutico , Humanos , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/inmunología , Pulmón/efectos de los fármacos , Pulmón/patología , Masculino , Mucina 5AC/metabolismo , Mapeo de Interacción de Proteínas/métodos , Mapas de Interacción de Proteínas/efectos de los fármacos , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/patología , Ratas , Ratas Sprague-Dawley , Mucosa Respiratoria/patología , Programas Informáticos , Máquina de Vectores de Soporte
5.
Clin Sci (Lond) ; 133(13): 1523-1536, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31270147

RESUMEN

Cigarette smoke (CS) is the major cause of chronic obstructive pulmonary disease (COPD). CS heightens inflammation, oxidative stress and apoptosis. Ergosterol is the main bioactive ingredient in Cordyceps sinensis (C. sinensis), a traditional medicinal herb for various diseases. The objective of this work was to investigate the effects of ergosterol on anti-inflammatory and antioxidative stress as well as anti-apoptosis in a cigarette smoke extract (CSE)-induced COPD model both in vitro and in vivo Our results demonstrate that CSE induced inflammatory and oxidative stress and apoptosis with the involvement of the Bcl-2 family proteins via the nuclear factor kappa B (NF-κB)/p65 pathway in both 16HBE cells and Balb/c mice. CSE induced epithelial cell death and increased the expression of nitric oxide (NO), interleukin-6 (IL-6), tumor necrosis factor α (TNF-α), malondialdehyde (MAD) and the apoptosis-related proteins cleaved caspase 3/7/9 and cleaved-poly-(ADP)-ribose polymerase (PARP) both in vitro and in vivo, whereas decreased the levels of superoxide dismutase (SOD) and catalase (CAT). Treatment of 16HBE cells and Balb/c mice with ergosterol inhibited CSE-induced inflammatory and oxidative stress and apoptosis by inhibiting the activation of NF-κB/p65. Ergosterol suppressed apoptosis by inhibiting the expression of the apoptosis-related proteins both in vitro and in vivo Moreover, the usage of QNZ (an inhibitor of NF-κB) also partly demonstrated that NF-κB/p65 pathway was involved in the ergosterol protective progress. These results show that ergosterol suppressed COPD inflammatory and oxidative stress and apoptosis through the NF-κB/p65 pathway, suggesting that ergosterol may be partially responsible for the therapeutic effects of cultured C. sinensis on COPD patients.


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Ergosterol/farmacología , Mediadores de Inflamación/metabolismo , Pulmón/efectos de los fármacos , Estrés Oxidativo , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Mucosa Respiratoria/efectos de los fármacos , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Oxidación-Reducción , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Transducción de Señal , Humo , Productos de Tabaco
6.
Sci Rep ; 9(1): 4787, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30886217

RESUMEN

Pollens are well-known triggers of respiratory allergies and asthma. The pollen burden in today's ambient air is constantly increasing due to rising climate change and air pollution. How pollens interact with the respiratory mucosa remains largely unknown due to a lack of representative model systems. We here demonstrate how pollen proteases of Kentucky bluegrass, white birch and hazel selectively destroy integrity and anchorage of columnar respiratory epithelial cells, but not of basal cells, in both ex vivo respiratory mucosal explants and in vitro primary equine respiratory epithelial cells (EREC). In turn, this pollen protease-induced damage to respiratory epithelial cell anchorage resulted in increased infection by the host-specific and ancestral alphaherpesvirus equine herpesvirus type 1 (EHV1). Pollen proteases of all three plant species were characterized by zymography and those of white birch were fully identified for the first time as serine proteases of the subtilase family and meiotic prophase aminopeptidase 1 using mass spectrometry-based proteomics. Together, our findings demonstrate that pollen proteases selectively and irreversibly damage integrity and anchorage of columnar respiratory epithelial cells. In turn, alphaherpesviruses benefit from this partial loss-of-barrier function, resulting in increased infection of the respiratory epithelium.


Asunto(s)
Infecciones por Herpesviridae/etiología , Proteínas de Plantas/metabolismo , Polen/toxicidad , Mucosa Respiratoria/virología , Serina Proteasas/metabolismo , Animales , Betula , Células Cultivadas , Corylus , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Herpesvirus Équido 1/patogenicidad , Caballos , Poaceae , Polen/enzimología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología
7.
Allergol Immunopathol (Madr) ; 47(2): 185-193, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30528469

RESUMEN

INTRODUCTION: Asthma is a chronic inflammatory disease of the airways. In this study, we evaluated the anti-inflammatory effects of myrtenol on the inflammatory indices in the pulmonary parenchyma and airways and on the inflammatory and oxidative indices of the bronchoalveolar lavage fluid (BALF) of asthmatic rats. METHODS: The allergic asthma was induced by sensitization (two weeks) followed by the inhalation of ovalbumin (four weeks). Animals were divided into two main groups: (1) Histopathology, and (2) measurement of inflammatory and oxidative biomarkers in the BALF. Each main group was subdivided into four subgroups: Control, Asthma, Asthma+Dexamethasone and Asthma+Myrtenol. (-)-Myrtenol (50mg/kg) or Dexamethasone (2.5mg/kg) was administered intraperitoneally once a day for one week, at the end of the inhalation period. On day 50, lung histopathologic parameters and inflammatory indices in BALF including INF-γ, IL-10, IL-1ß, and TNF-α and oxidative stress biomarkers (MDA, SOD, and GPX) were measured. RESULT: In the Asthma group, leukocyte infiltration, the thickness of smooth muscle and epithelium of airways wall and the number of goblet cells increased. Myrtenol reduced all of the above-mentioned indices except the epithelium thickness. It also inhibited the increase in BALF IL-1ß, TNF-α and MDA and increased the levels of INF-γ, IL-10 and SOD. CONCLUSION: Our results suggest that myrtenol reduced damage caused by experimental asthma by reducing the inflammatory indices, normalizing the level of interleukins and balancing oxidative stress in the lungs. It also prevented airway remodeling. Myrtenol may be suggested as a potent herbal medicine for the treatment of allergic asthma.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Antiinflamatorios/uso terapéutico , Asma/tratamiento farmacológico , Células Caliciformes/patología , Leucocitos/inmunología , Pulmón/inmunología , Monoterpenos/uso terapéutico , Mucosa Respiratoria/patología , Animales , Monoterpenos Bicíclicos , Movimiento Celular , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Ratas , Ratas Wistar , Mucosa Respiratoria/efectos de los fármacos
8.
Int Immunopharmacol ; 67: 1-12, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30530164

RESUMEN

Fraxin, the effective component of the Chinese traditional medicine Cortex Fraxini, is reported to have anti-inflammatory effects. This study assessed the anti-inflammatory effect of fraxin on the lipopolysaccharide (LPS)-induced inflammatory response in A549 cells and the protective efficacy on LPS-induced acute lung injury (ALI) in mice. Fraxin reduced LPS-induced TNF-α, IL-6 and IL-1ß production in A549 cells and alleviated the LPS-induced wet/dry (W/D) weight ratio and the effects observed via histopathological examination of the lung in vivo. Furthermore, fraxin reduced the protein concentrations in the broncho-alveolar lavage (BAL) fluid and cytokine production in the sera. Fraxin also clearly attenuated the oxidation index, including the activity of myeloperoxidase (MPO), malondialdehyde (MDA), superoxide dismutase (SOD) and glutathione (GSH). Immunohistochemistry analysis showed that fraxin suppressed LPS-induced inflammatory damage. The expression of proteins involved in the NF-κB and NLRP3 inflammatory corpuscle signalling pathways was consistent between the lung tissues and cell samples. Overall, fraxin played a protective role in LPS-induced lung injury by inhibiting the NF-κB and NLRP3 signalling pathways.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Antiinflamatorios/uso terapéutico , Cumarinas/uso terapéutico , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Mucosa Respiratoria/efectos de los fármacos , Células A549 , Lesión Pulmonar Aguda/inducido químicamente , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/inmunología , Masculino , Medicina Tradicional China , Ratones , Ratones Endogámicos , Oxidación-Reducción/efectos de los fármacos , Peroxidasa/metabolismo , Mucosa Respiratoria/patología , Transducción de Señal
9.
J Cell Biochem ; 120(3): 3833-3844, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30260006

RESUMEN

BACKGROUND: Pingchuanning decoction is a well-known traditional Chinese medicine for the treatment of airway inflammatory diseases, including asthma. However, the potential mechanism by which Pingchuanning decoction contributes to the amelioration of airway inflammation remains unknown. METHODS: A rat model of asthma was well established by inducing ovalbumin. Lipopolysaccharide-stimulated rat tracheal epithelial (RTE) cells were used as cellular model. Lung histopathology and goblet cell hyperplasia were assessed by hematoxylin-eosin (HE) and periodic acid Schiff staining, respectively. Total inflammatory cells count and RTE cell apoptosis were analyzed by flow cytometry. The autophagic activities were evaluated by immunohistochemical and immunofluorescence analysis and Western blot analysis of autophagy-related proteins. We also detected the effects of Pingchuanning decoction on phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) and high-mobility group box 1 (HMGB1)-mediated toll-like receptor 4 (TLR4)/NF-κB pathways-related proteins and inflammatory cytokines using the Western blot analysis and enzyme-linked immunosorbent assay. RESULTS: Pingchuanning decoction effectively attenuated pulmonary pathology and autophagy. Treatment with Pingchuanning decoction activated PI3K/Akt/mTOR pathway and inhibited HMGB1/TLR4/NF-κB pathway, which could be overturned by LY294002, a PI3K antagonist, or rapamycin (Rapa), an autophagy inducer. CONCLUSION: Pingchuanning decoction exerted a therapeutic effect on asthma by inhibiting autophagy via PI3K/Akt /mTOR signaling pathway.


Asunto(s)
Antiasmáticos/farmacología , Asma/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Fosfatidilinositol 3-Quinasa/genética , Proteínas Proto-Oncogénicas c-akt/genética , Serina-Treonina Quinasas TOR/genética , Animales , Asma/inducido químicamente , Asma/genética , Asma/metabolismo , Autofagia/efectos de los fármacos , Autofagia/genética , Dexametasona/farmacología , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación de la Expresión Génica , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ovalbúmina/administración & dosificación , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Resultado del Tratamiento
10.
J Cyst Fibros ; 17(6): 705-714, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29661510

RESUMEN

BACKGROUND: Progressive airway damage due to bacterial infections, especially with Pseudomonas aeruginosa remains the first cause of morbidity and mortality in CF patients. Our previous work revealed a repair delay in CF airway epithelia compared to non-CF. This delay was partially prevented after CFTR correction (with VRT-325) in the absence of infection. Our goals were now to evaluate the effect of the Orkambi combination (CFTR VX-809 corrector + VX-770 potentiator) on the repair of CF primary airway epithelia, in infectious conditions. METHODS: Primary airway epithelial cell cultures from patients with class II mutations were mechanically injured and wound healing rates and transepithelial resistances were monitored after CFTR rescue, in the absence and presence of P. aeruginosa exoproducts. RESULTS: Our data revealed that combined treatment with VX-809 and VX-770 elicited a greater beneficial impact on airway epithelial repair than VX-809 alone, in the absence of infection. The treatment with Orkambi was effective not only in airway epithelial cell cultures from patients homozygous for the F508del mutation but also from heterozygous patients carrying F508del and another class II mutation (N1303 K, I507del). The stimulatory effect of the Orkambi treatment was prevented by CFTR inhibition with GlyH101. Finally, Orkambi combination elicited a slight but significant improvement in airway epithelial repair and transepithelial resistance, despite the presence of P. aeruginosa exoproducts. CONCLUSIONS: Our findings indicate that Orkambi may favor airway epithelial integrity in CF patients with class II mutations. Complementary approaches would however be needed to further improve CFTR rescue and airway epithelial repair.


Asunto(s)
Aminofenoles/farmacología , Aminopiridinas/farmacología , Benzodioxoles/farmacología , Fibrosis Quística , Exotoxinas , Glicina/análogos & derivados , Hidrazinas/farmacología , Pseudomonas aeruginosa/fisiología , Quinolonas/farmacología , Mucosa Respiratoria , Células Cultivadas , Agonistas de los Canales de Cloruro/farmacología , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/genética , Fibrosis Quística/microbiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Combinación de Medicamentos , Glicina/farmacología , Humanos , Mutación , Regeneración/efectos de los fármacos , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/microbiología , Mucosa Respiratoria/patología
11.
Integr Cancer Ther ; 17(3): 832-843, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29484963

RESUMEN

BACKGROUND: Inonotus obliquus, also known as Chaga, is a parasitic fungus growing on birches and used in traditional medicine (especially by Khanty people) to treat various health problems. In this study, we aimed to quantify the 3 metabolites frequently cited in literature, that is, betulin, betulinic acid, and inotodiol in the Chaga recently discovered in forests located in Normandy (France), and to compare their concentrations with Ukrainian and Canadian Chaga. This study also explores the cytotoxicity of the French Chaga against cancer-derived cells and transformed cells. METHODS: A quantification method by HPLC-MS-MS (high-performance liquid chromatography-tandem mass spectrometry) of betulin, betulinic acid, and inotodiol was developed to study the French Chaga and compare the concentration of these metabolites with extracts provided from Chaga growing in Canada and Ukraine. This method was also used to identify and quantify those 3 compounds in other traditional preparations of Chaga (aqueous extract, infusion, and decoction). Among these preparations, the aqueous extract that contains betulin, betulinic acid, and inotodiol was chosen to evaluate and compare its cytotoxic activity toward human lung adenocarcinoma cells (A549 line) and human bronchial epithelial cells (BEAS-2B line). RESULTS: French Chaga contains betulin and betulinic acid at higher levels than in other Chaga, whereas the concentration of inotodiol is greater in the Canadian Chaga. Moreover, the results highlighted a cytotoxic activity of the Chaga's aqueous extract after 48 and 72 hours of exposure with a higher effect on cancer-derived cells A549 than on normal transformed cells BEAS-2B ( P = 0.025 after 48 hours of exposure and P = 0.004 after 72 hours of exposure).


Asunto(s)
Adenocarcinoma del Pulmón/patología , Bronquios/efectos de los fármacos , Citotoxinas/aislamiento & purificación , Citotoxinas/farmacología , Células Epiteliales/efectos de los fármacos , Neoplasias Pulmonares/patología , Poria/química , Células A549 , Antineoplásicos/aislamiento & purificación , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Bronquios/patología , Extractos Celulares/química , Extractos Celulares/aislamiento & purificación , Extractos Celulares/farmacología , Extractos Celulares/uso terapéutico , Células Cultivadas , Citotoxinas/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales , Células Epiteliales/fisiología , Humanos , Oncología Médica/métodos , Oncología Médica/tendencias , Medicina Tradicional/métodos , Polyporus/química , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/patología
12.
Biol Pharm Bull ; 40(9): 1416-1422, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28867724

RESUMEN

DW2008 is an anhydrous ethanol extract of Justicia procumbens produced by Dong-Wha Pharmaceutical, Inc., Co. as a candidate anti-asthmatic drug. In this study, DW2008 selectively reduced T helper 2 (Th2) cytokines in mouse splenocytes and ameliorated ovalbumin-induced airway inflammation by downregulating pulmonary infiltration of differential inflammatory cells and Th2 cytokines more than a decoction or ethanol extract of J. procumbens did in a mouse asthma model. DW2008 also significantly inhibited airway hyperresponsiveness and reduced the thickness of the airway epithelium. HPLC analysis showed that the major peaks (justicidin A and B) of DW2008 were higher than those of the other extracts. Justicidin A and B significantly suppressed Th2 cytokine levels in mouse spleen cells and exhibited a protective effect in ovalbumin-induced airway inflammation. Our findings indicate that DW2008 effectively inhibits allergic airway inflammatory reactions and airway hyperresponsiveness in a mouse model of asthma, suggesting its potential as an anti-asthmatic agent.


Asunto(s)
Antiasmáticos/farmacología , Antiinflamatorios/farmacología , Asma/inducido químicamente , Asma/patología , Citocinas/antagonistas & inhibidores , Ovalbúmina , Extractos Vegetales/farmacología , Plantas Medicinales/química , Bazo/metabolismo , Células Th2/metabolismo , Animales , Citocinas/biosíntesis , Citocinas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Femenino , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Extractos Vegetales/química , Hipersensibilidad Respiratoria/inducido químicamente , Hipersensibilidad Respiratoria/prevención & control , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/patología , Bazo/citología , Células Th2/efectos de los fármacos
13.
Molecules ; 22(9)2017 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-28892004

RESUMEN

Arenobufagin, an active component isolated from the traditional Chinese medicine Chan Su, exhibits anticancer influences in several human malignancies. However, the effects and action mechanisms of arenobufagin on non-small-cell lung cancer (NSCLC) are still unknown. In this study, we reported that arenobufagin acted through activation of Noxa-related pathways and promoted apoptotic cell death in human NSCLC cells. Our results revealed that arenobufagin-induced apoptosis was caspase-dependent, as evidenced by the fact that caspase-9, caspase-3 and poly (ADP-ribose) polymerase (PARP) were cleaved, and pretreatment with a pan-caspase inhibitor Z-VAD-FMK inhibited the pro-apoptosis effect of arenobufagin. Mechanistically, we further found that arenobufagin rapidly upregulated the expression of the pro-apoptosis protein Noxa, and abrogated the anti-apoptosis protein Mcl-1, a major binding partner of Noxa in the cell. More importantly, the knockdown of Noxa greatly blocked arenobufagin-induced cell death, highlighting the contribution of this protein in the anti-NSCLC effects of arenobufagin. Interestingly, arenobufagin also increased the expression of p53, a direct transcriptional activator for the upregulation of the Noxa protein. Taken together, our results suggest that arenobufagin is a potential anti-NSCLC agent that triggers apoptotic cell death in NSCLC cells through interfering with the Noxa-related pathway.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Bufanólidos/química , Bufanólidos/farmacología , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-bcl-2/genética , Células A549 , Clorometilcetonas de Aminoácidos/farmacología , Venenos de Anfibios/química , Antineoplásicos/aislamiento & purificación , Apoptosis/genética , Bufanólidos/aislamiento & purificación , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Medicina Tradicional China , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Transducción de Señal , Proteína p53 Supresora de Tumor/agonistas , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
14.
Physiol Rep ; 5(15)2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28774952

RESUMEN

In disease settings, vitamin D may be important for maintaining optimal lung epithelial integrity and suppressing inflammation, but less is known of its effects prior to disease onset. Female BALB/c dams were fed a vitamin D3-supplemented (2280 IU/kg, VitD+) or nonsupplemented (0 IU/kg, VitD-) diet from 3 weeks of age, and mated at 8 weeks of age. Male offspring were fed the same diet as their mother. Some offspring initially fed the VitD- diet were switched to a VitD+ diet from 8 weeks of age (VitD-/+). At 12 weeks of age, signs of low-level inflammation were observed in the bronchoalveolar lavage fluid (BALF) of VitD- mice (more macrophages and neutrophils), which were suppressed by subsequent supplementation with vitamin D3 There was no difference in the level of expression of the tight junction proteins occludin or claudin-1 in lung epithelial cells of VitD+ mice compared to VitD- mice; however, claudin-1 levels were reduced when initially vitamin D-deficient mice were fed the vitamin D3-containing diet (VitD-/+). Reduced total IgM levels were detected in BALF and serum of VitD-/+ mice compared to VitD+ mice. Lung mRNA levels of the vitamin D receptor (VDR) were greatest in VitD-/+ mice. Total IgG levels in BALF were greater in mice fed the vitamin D3-containing diet, which may be explained by increased activation of B cells in airway-draining lymph nodes. These findings suggest that supplementation of initially vitamin D-deficient mice with vitamin D3 suppresses signs of lung inflammation but has limited effects on the epithelial integrity of the lungs.


Asunto(s)
Neumonía/tratamiento farmacológico , Receptores de Calcitriol/genética , Mucosa Respiratoria/metabolismo , Deficiencia de Vitamina D/tratamiento farmacológico , Vitamina D/uso terapéutico , Vitaminas/uso terapéutico , Animales , Líquido del Lavado Bronquioalveolar , Claudina-1/genética , Claudina-1/metabolismo , Suplementos Dietéticos , Femenino , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ocludina/genética , Ocludina/metabolismo , Neumonía/complicaciones , Receptores de Calcitriol/metabolismo , Mucosa Respiratoria/patología , Vitamina D/administración & dosificación , Deficiencia de Vitamina D/complicaciones , Vitaminas/administración & dosificación
15.
J Int Med Res ; 45(1): 101-113, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27913746

RESUMEN

Asthma is a heterogeneous disease characterized by symptoms of chronic inflammation and airway structural and functional changes. It affects about 300 million people worldwide and causes 250 000 deaths annually, but its symptoms can be greatly relieved by regular use of inhaled glucocorticoids (GCs). GCs exert their function through interacting with glucocorticoid receptors (GRs). Diosgenin is a naturally occurring steroidal saponin abundantly present in many medicinal plants, including Dioscorea nipponica, which shares a similar steroidal structure with GC. In this study, ovalbumin (OVA)-induced asthmatic mice and primary tracheal epithelial cells (TECs) were used as research models. ELISAs were applied to measure the secretion of TNF-α, IL-1ß, and IL-6, while quantitative PCR and western blotting were applied to evaluate expression of GRs SLPI, TTP, GILZ, MKP-1, and NF-κB. Our data demonstrated that diosgenin suppressed the secretion of TNF-α, IL-1ß, and IL-6 by enhancing the expression of GRs, SLPI, GILZ, and MKP-1, and inhibiting the expression of HSP70. These data provide some evidence on the molecular mechanism of diosgenin, which might facilitate its clinical applications.


Asunto(s)
Antiasmáticos/farmacología , Asma/tratamiento farmacológico , Dioscorea/química , Diosgenina/farmacología , Receptores de Glucocorticoides/agonistas , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Antiasmáticos/aislamiento & purificación , Asma/inducido químicamente , Asma/inmunología , Asma/patología , Dexametasona/farmacología , Diosgenina/aislamiento & purificación , Modelos Animales de Enfermedad , Fosfatasa 1 de Especificidad Dual/genética , Fosfatasa 1 de Especificidad Dual/inmunología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/patología , Femenino , Regulación de la Expresión Génica , Glucocorticoides/farmacología , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/inmunología , Humanos , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/antagonistas & inhibidores , Interleucina-6/genética , Interleucina-6/inmunología , Ratones , Ratones Endogámicos BALB C , FN-kappa B/genética , FN-kappa B/inmunología , Ovalbúmina , Extractos Vegetales/química , Cultivo Primario de Células , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/inmunología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Inhibidor Secretorio de Peptidasas Leucocitarias/genética , Inhibidor Secretorio de Peptidasas Leucocitarias/inmunología , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
16.
Lab Invest ; 96(12): 1279-1300, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27775689

RESUMEN

Silicosis is an occupational pulmonary fibrosis caused by inhalation of silica (SiO2) and there are no ideal drugs to treat this disease. Earthworm extract (EE), a natural nutrient, has been reported to have anti-inflammatory, antioxidant, and anti-apoptosis effects. The purpose of the current study was to test the protective effects of EE against SiO2-induced pulmonary fibrosis and to explore the underlying mechanisms using both in vivo and in vitro models. We found that treatment with EE significantly reduced lung inflammation and fibrosis and improved lung structure and function in SiO2-instilled mice. Further mechanistic investigations revealed that EE administration markedly inhibited SiO2-induced oxidative stress, mitochondrial apoptotic pathway, and epithelial-mesenchymal transition in HBE and A549 cells. Furthermore, we demonstrate that Nrf2 activation partly mediates the interventional effects of EE against SiO2-induced pulmonary fibrosis. Our study has identified EE to be a potential anti-oxidative, anti-inflammatory, and anti-fibrotic drug for silicosis.


Asunto(s)
Antioxidantes/uso terapéutico , Modelos Animales de Enfermedad , Pulmón/efectos de los fármacos , Materia Medica/uso terapéutico , Oligoquetos/química , Fibrosis Pulmonar/prevención & control , Silicosis/tratamiento farmacológico , Extractos de Tejidos/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Células Cultivadas , Transición Epitelial-Mesenquimal/efectos de los fármacos , Inyecciones Intraperitoneales , Pulmón/metabolismo , Pulmón/patología , Pulmón/fisiopatología , Masculino , Materia Medica/administración & dosificación , Materia Medica/farmacología , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/agonistas , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/inmunología , Interferencia de ARN , Distribución Aleatoria , Mucosa Respiratoria/citología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Silicosis/metabolismo , Silicosis/patología , Silicosis/fisiopatología , Organismos Libres de Patógenos Específicos , Extractos de Tejidos/administración & dosificación , Extractos de Tejidos/farmacología
17.
J Nutr Biochem ; 36: 81-88, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27596528

RESUMEN

Hypoxia-induced oxidative stress and excessive proliferation of pulmonary artery smooth muscle cells (PASMCs) play important roles in the pathological process of hypoxic pulmonary hypertension (HPH). Grape seed procyanidin extract (GSPE) possesses antioxidant properties and has beneficial effects on the cardiovascular system. However, the effect of GSPE on HPH remains unclear. In this study, adult Sprague-Dawley rats were exposed to intermittent chronic hypoxia for 4 weeks to mimic a severe HPH condition. Hemodynamic and pulmonary pathomorphology data showed that chronic hypoxia significantly increased right ventricular systolic pressures (RVSP), weight of the right ventricle/left ventricle plus septum (RV/LV+S) ratio and median width of pulmonary arteries. GSPE attenuated the elevation of RVSP, RV/LV+S, and reduced the pulmonary vascular structure remodeling. GSPE also increased the levels of SOD and reduced the levels of MDA in hypoxia-induced HPH model. In addition, GSPE suppressed Nox4 mRNA levels, ROS production and PASMCs proliferation. Meanwhile, increased expression of phospho-STAT3, cyclin D1, cyclin D3 and Ki67 in PASMCs caused by hypoxia was down-regulated by GSPE. These results suggested that GSPE might potentially prevent HPH via antioxidant and antiproliferative mechanisms.


Asunto(s)
Antioxidantes/uso terapéutico , Suplementos Dietéticos , Extracto de Semillas de Uva/uso terapéutico , Hipertensión Pulmonar/prevención & control , Músculo Liso Vascular/metabolismo , Estrés Oxidativo , Proantocianidinas/uso terapéutico , Daño por Reperfusión/prevención & control , Animales , Antioxidantes/efectos adversos , Antioxidantes/metabolismo , Biomarcadores/metabolismo , Proliferación Celular , Células Cultivadas , Suplementos Dietéticos/efectos adversos , Regulación Enzimológica de la Expresión Génica , Extracto de Semillas de Uva/efectos adversos , Extracto de Semillas de Uva/metabolismo , Hipertensión Pulmonar/dietoterapia , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Peroxidación de Lípido , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Pulmón/patología , Masculino , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , NADPH Oxidasa 4 , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Proantocianidinas/efectos adversos , Proantocianidinas/metabolismo , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Distribución Aleatoria , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/dietoterapia , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Mucosa Respiratoria/irrigación sanguínea , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Remodelación Vascular
18.
Am J Physiol Lung Cell Mol Physiol ; 311(1): L101-10, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27190062

RESUMEN

Agricultural dust exposure results in significant lung inflammation, and individuals working in concentrated animal feeding operations (CAFOs) are at risk for chronic airway inflammatory diseases. Exposure of bronchial epithelial cells to aqueous extracts of hog CAFO dusts (HDE) leads to inflammatory cytokine production that is driven by protein kinase C (PKC) activation. cAMP-dependent protein kinase (PKA)-activating agents can inhibit PKC activation in epithelial cells, leading to reduced inflammatory cytokine production following HDE exposure. ß2-Adrenergic receptor agonists (ß2-agonists) activate PKA, and we hypothesized that ß2-agonists would beneficially impact HDE-induced adverse airway inflammatory consequences. Bronchial epithelial cells were cultured with the short-acting ß2-agonist salbutamol or the long-acting ß2-agonist salmeterol prior to stimulation with HDE. ß2-Agonist treatment significantly increased PKA activation and significantly decreased HDE-stimulated IL-6 and IL-8 production in a concentration- and time-dependent manner. Salbutamol treatment significantly reduced HDE-induced intracellular adhesion molecule-1 expression and neutrophil adhesion to epithelial cells. Using an established intranasal inhalation exposure model, we found that salbutamol pretreatment reduced airway neutrophil influx and IL-6, TNF-α, CXCL1, and CXCL2 release in bronchoalveolar lavage fluid following a one-time exposure to HDE. Likewise, when mice were pretreated daily with salbutamol prior to HDE exposure for 3 wk, HDE-induced neutrophil influx and inflammatory mediator production were also reduced. The severity of HDE-induced lung pathology in mice repetitively exposed to HDE for 3 wk was also decreased with daily salbutamol pretreatment. Together, these results support the need for future clinical investigations to evaluate the utility of ß2-agonist therapies in the treatment of airway inflammation associated with CAFO dust exposure.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/farmacología , Contaminantes Atmosféricos/toxicidad , Albuterol/farmacología , Neumonía/tratamiento farmacológico , Xinafoato de Salmeterol/farmacología , Animales , Línea Celular , Citocinas/metabolismo , Evaluación Preclínica de Medicamentos , Polvo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neumonía/etiología , Neumonía/inmunología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/patología
19.
Drug Metab Pers Ther ; 31(1): 47-54, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26943616

RESUMEN

BACKGROUND: Garlic capsule (GAR) and/or selenium- vitamin A, C, E (S-VACE) might be useful in the treatment of lung diseases. The present study evaluated the toxicity of lisinopril (LIS) in the lungs of male rats and the reversal effect of GAR and/or selenium-vitamins A, C, and E (S-VACE). METHODS: Group I served as the control, whereas animals in groups II, III, IV, and V received 28 mg of LIS/kg body weight by gavage. Group III was co-treated with GAR at a therapeutic dosage of 250 mg/kg body weight per day. Group IV was co-treated with S-VACE at dosage of 500 mg/kg body weight per day. Lastly, group V was co-treated with GAR and S-VACE at dosages of 250 and 500 mg/kg body weight per day, respectively. The experiment lasted for 8 days (sub-acute exposure). RESULTS: Administration of therapeutic dose of LIS to male rats depleted enzymatic antioxidants (superoxide dismutase and catalase) and cellular adenosine triphosphate content with concomitant increase in lipid peroxidation. Histopathology examination showed damage to the epithelial cells of the airways. These effects were prevented by both single and combination treatment of GAR and S-VACE in male rats with LIS-induced lung toxicity. CONCLUSIONS: We therefore concluded that the combination of GAR and S-VACE can be a novel therapy for the management of lung diseases in humans.


Asunto(s)
Ajo , Lisinopril/toxicidad , Enfermedades Pulmonares/prevención & control , Selenio/farmacología , Vitaminas/farmacología , Adenosina Trifosfato/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/toxicidad , Animales , Antioxidantes/metabolismo , Ácido Ascórbico/administración & dosificación , Ácido Ascórbico/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Peroxidación de Lípido/efectos de los fármacos , Enfermedades Pulmonares/inducido químicamente , Enfermedades Pulmonares/patología , Masculino , Ratas , Ratas Wistar , Mucosa Respiratoria/citología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/patología , Selenio/administración & dosificación , Vitamina A/administración & dosificación , Vitamina A/farmacología , Vitamina E/administración & dosificación , Vitamina E/farmacología , Vitaminas/administración & dosificación
20.
Histol Histopathol ; 31(6): 657-62, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26648111

RESUMEN

OBJECTIVE: This paper is aimed at identifying putative morphological changes induced in the rat's tracheal mucosa by chronic hyperbaric oxygen (HBO) treatment or chronic intermittent hypoxia (CIH). STUDY DESIGN: Tracheal samples were obtained from three groups of 11, 12 and 13 adult Wistar rats. The first group was submitted to 20 sessions of 100 min-long HBO treatment; the second group was submitted to eucapnic CIH for 35 days; and the third group was not submitted to any CIH or HBO therapy. METHODS: Four proximal tracheal rings were collected after sacrifice and neck dissection of the animals. The samples were processed for both light microscopy and morphometric analysis. Inflammatory leukocyte infiltration was evaluated by a semiquantitative method. Unpaired t test and Bernoulli distribution were applied to evaluate statistical differences in the data collected from the three groups. RESULTS: Both CIH and HBO promote an increase in the thickness of the epithelium and of the basement membrane of the rat tracheal mucosa, as well as an increment in the number of infiltrating leukocytes, when compared with results seen in the untreated group. In the HBO group there was a significant lack of seromucous glands, as opposed to the results obtained in the CIH group. CONCLUSIONS: Chronic HBO and CIH exposure causes only minor changes in the architecture of the tracheal mucosa of the rat. The respiratory tract of the rat showed a mild inflammatory response when subject to variations of pressure and oxygen content. Apparently these effects do not constitute a critical issue on prescribing HBO treatments and in the management of sleep apnea patients.


Asunto(s)
Oxigenoterapia Hiperbárica/efectos adversos , Hipoxia/patología , Mucosa Respiratoria/patología , Tráquea/patología , Animales , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA