Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 222
Filtrar
1.
Int J Mol Sci ; 25(10)2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38791530

RESUMEN

Neutrophil-derived proteases are critical to the pathology of many inflammatory lung diseases, both chronic and acute. These abundant enzymes play roles in key neutrophil functions, such as neutrophil extracellular trap formation and reactive oxygen species release. They may also be released, inducing tissue damage and loss of tissue function. Historically, the neutrophil serine proteases (NSPs) have been the main subject of neutrophil protease research. Despite highly promising cell-based and animal model work, clinical trials involving the inhibition of NSPs have shown mixed results in lung disease patients. As such, the cutting edge of neutrophil-derived protease research has shifted to proteases that have had little-to-no research in neutrophils to date. These include the cysteine and serine cathepsins, the metzincins and the calpains, among others. This review aims to outline the previous work carried out on NSPs, including the shortcomings of some of the inhibitor-orientated clinical trials. Our growing understanding of other proteases involved in neutrophil function and neutrophilic lung inflammation will then be discussed. Additionally, the potential of targeting these more obscure neutrophil proteases will be highlighted, as they may represent new targets for inhibitor-based treatments of neutrophil-mediated lung inflammation.


Asunto(s)
Neutrófilos , Neumonía , Humanos , Neutrófilos/metabolismo , Neutrófilos/enzimología , Neutrófilos/inmunología , Animales , Neumonía/metabolismo , Neumonía/enzimología , Neumonía/patología , Serina Proteasas/metabolismo , Péptido Hidrolasas/metabolismo
2.
Am J Physiol Regul Integr Comp Physiol ; 322(2): R126-R135, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34984926

RESUMEN

Many lung diseases are caused by an excessive inflammatory response, and inflammatory lung diseases are often modeled using lipopolysaccharide (LPS) in mice. Cyclooxygenase-2 (COX-2) encoded by the Ptgs2 gene is induced in response to inflammatory stimuli including LPS. The objective of this study was to test the hypothesis that mice deficient in COX-2 (Ptgs2-/-) will be protected from LPS-induced lung injury. Wild-type (WT; CD1 mice) and Ptgs2-/- mice (on a CD1 background) were treated with LPS or vehicle for 24 h. LPS treatment resulted in histological evidence of lung injury, which was attenuated in the Ptgs2-/- mice. LPS treatment increased the mRNA levels for tumor necrosis factor-α, interleukin-10, and monocyte chemoattractant protein-1 in the lungs of WT mice, and the LPS-induced increases in these levels were attenuated in the Ptgs2-/- mice. The protein levels of active caspase-3 and caspase-9 were lower in the LPS-treated lungs of Ptgs2-/- mice than in LPS-treated WT mice, as were the number of terminal deoxynucleotide transferase dUTP nick end labeling-positive cells in lung sections. LPS exposure resulted in a greater lung wet-to-dry weight ratio (W/D) in WT mice, suggestive of pulmonary edema, while in LPS-treated Ptgs2-/- mice, the W/D was not different from controls and less than in LPS-treated WT mice. These results demonstrate that COX-2 is involved in the inflammatory response to LPS and suggest that COX-2 not only acts as a downstream participant in the inflammatory response, but also acts as a regulator of the inflammatory response likely through a feed-forward mechanism following LPS stimulation.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Apoptosis , Ciclooxigenasa 2/deficiencia , Pulmón/enzimología , Neumonía/prevención & control , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/patología , Animales , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Ciclooxigenasa 2/genética , Modelos Animales de Enfermedad , Femenino , Interleucina-10/genética , Interleucina-10/metabolismo , Lipopolisacáridos , Pulmón/patología , Masculino , Ratones Noqueados , Neumonía/inducido químicamente , Neumonía/enzimología , Neumonía/patología , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
3.
IUBMB Life ; 74(2): 131-142, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34590785

RESUMEN

Chronic respiratory diseases (CRDs), including pulmonary fibrosis, chronic obstructive pulmonary disease (COPD), lung cancer, and asthma, are significant global health problems due to their prevalence and rising incidence. The roles of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs) in controlling tyrosine phosphorylation of targeting proteins modulate multiple physiological cellular responses and contribute to the pathogenesis of CRDs. Src homology-2 domain-containing PTP2 (SHP2) plays a pivotal role in modulating downstream growth factor receptor signaling and cytoplasmic PTKs, including MAPK/ERK, PI3K/AKT, and JAK/STAT pathways, to regulate cell survival and proliferation. In addition, SHP2 mutation and activation are commonly implicated in tumorigenesis. However, little is known about SHP2 in chronic pulmonary inflammation and fibrosis. This review discusses the potential involvement of SHP2 deregulation in chronic pulmonary inflammation and fibrosis, as well as the therapeutic effects of targeting SHP2 in CRDs.


Asunto(s)
Neumonía , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Enfermedades Respiratorias , Fibrosis , Humanos , Neumonía/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Enfermedades Respiratorias/epidemiología , Transducción de Señal
4.
Respir Res ; 22(1): 267, 2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34666758

RESUMEN

BACKGROUND: The non-cancerous functions of Akt in the airway are understudied. In some tissues, Akt phosphorylates and activates endothelial nitric oxide synthase (eNOS) to produce nitric oxide (NO) that has anti-inflammatory effects. NO production has antibacterial and antiviral effects in the airway, and increasing NO may be a useful anti-pathogen strategy. Akt also stimulates the nuclear factor erythroid 2-related factor 2 (Nrf-2) transcription factor, which transcribes antioxidant genes. Therefore, we hypothesized that activation of the Akt/eNOS pathway, which also activates Nrf-2, may have protective effects in human airway cells against injury. METHODS: To directly test the effects of Akt signaling in the airway, we treated A549 and 16HBE cells as well as primary bronchial, nasal, and type II alveolar epithelial cells with small molecule Akt activator SC79. We examined the effects of SC79 on eNOS activation, NO production, Nrf-2 target levels, and interleukin-8 (IL-8) transcription during exposure to TNF-α or Pseudomonas flagellin (TLR5 agonist). Additionally, air-liquid interface bronchial cultures were treated with cadmium, an oxidative stressor that causes airway barrier breakdown. RESULTS: SC79 induced a ~ twofold induction of p-eNOS and Nrf-2 protein levels blocked by PI3K inhibitor LY294002. Live cell imaging revealed SC79 increased acute NO production. Quantitative RT-PCR showed a ~ twofold increase in Nrf-2 target gene transcription. TNF-α or flagellin-induced IL-8 levels were also significantly reduced with SC79 treatment. Moreover, the transepithelial electrical resistance decrease observed with cadmium was ameliorated by SC79, likely by an acute increase in tight junction protein ZO-1 levels. CONCLUSIONS: Together, the data presented here demonstrate SC79 activation of Akt induces potentially anti-pathogenic NO production, antioxidant gene transcription, reduces IL-8 transcription, and may protect against oxidative barrier dysfunction in a wide range of airway epithelial cells.


Asunto(s)
Acetatos/farmacología , Antiinflamatorios/farmacología , Benzopiranos/farmacología , Activadores de Enzimas/farmacología , Células Epiteliales/efectos de los fármacos , Interleucina-8/metabolismo , Pulmón/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Óxido Nítrico/metabolismo , Neumonía/prevención & control , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células A549 , Impedancia Eléctrica , Activación Enzimática , Células Epiteliales/enzimología , Células Epiteliales/inmunología , Humanos , Interleucina-8/genética , Pulmón/enzimología , Pulmón/inmunología , Factor 2 Relacionado con NF-E2/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Neumonía/enzimología , Neumonía/genética , Neumonía/inmunología , Transducción de Señal , Transcripción Genética , Proteína de la Zonula Occludens-1/metabolismo
5.
Toxicol Appl Pharmacol ; 432: 115757, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34673086

RESUMEN

An increasing number of studies have shown that arsenic exposure increases the risk of lung cancer as well as a variety of non-malignant respiratory diseases, including bronchitis and tracheobronchitis. HMGB1 is widely expressed in a variety of tissues and cells and is involved in the pathological processes of many lung diseases through binding to the corresponding receptors and activating the downstream signaling pathways. However, the exact role of HMGB1/RAGE in arsenic-induced lung injury remains unknown. The aim of this study was to investigate whether HMGB1/RAGE and its activated downstream pathways are involved in the process of arsenic exposure-induced lung injury in rats. In this study, an animal model of oral exposure to arsenic was induced using 2.5, 5 and 10 mg/kg NaAsO2. The results showed that capillary permeability (LDH, TP, ACP, and AKP) was increased in the arsenic exposure groups, resulting in cell damage; this was accompanied by acute inflammation marked by significant neutrophil infiltration. Meanwhile, obvious histopathological damage, including thickening of the lung epithelium, increased infiltration of inflammatory cells, rupture of the alveolar wall, swelling of the mitochondria, and chromatin agglutination was observed by H&E staining and transmission electron microscopy. Furthermore, the results confirmed that the expressions of HMGB1 and RAGE in lung tissue were enhanced, and protein expression of PI3K, p-AKT, IL-1ß, IL-18, and MMP-9 was increased in lung homogenates from the arsenic-exposed groups compared to the control group. Finally, Masson's staining results revealed arsenic-induced fibrosis and collagen deposition. Moreover, a significant increase in key fibrosis factors, including TGF-ß1, p-SMAD2, p-SMAD3, and SMAD4 was observed in the lung homogenates in arsenic-exposed groups. In conclusion, the current study demonstrates that sub-chronic arsenic exposure triggers the inflammatory response and collagen fiber deposition in rat lung tissue. The potential mechanism may be closely related to activation of the pro-inflammatory-related HMGB1/RAGE pathway and initiation of the PI3K/AKT and TGF-ß1/SMAD pathways.


Asunto(s)
Proteína HMGB1/metabolismo , Pulmón/enzimología , Fosfatidilinositol 3-Quinasa/metabolismo , Neumonía/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fibrosis Pulmonar/enzimología , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Arsenitos , Colágeno/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Pulmón/ultraestructura , Masculino , Fosforilación , Neumonía/inducido químicamente , Neumonía/patología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Ratas Wistar , Transducción de Señal , Compuestos de Sodio
6.
Inflammation ; 44(6): 2170-2179, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34109517

RESUMEN

Acute ethanol intoxication increases the risk of sepsis and aggravates the symptoms of sepsis and lung injury. Therefore, this study aimed to explore whether sphingosine kinase 1 (SphK1)/sphingosine-1-phosphate (S1P)/S1P receptor 1 (S1PR1) signaling pathway functions in lung injury caused by acute ethanol intoxication-enhanced sepsis, as well as its underlying mechanism. The acute ethanol intoxication model was simulated by intraperitoneally administering mice with 32% ethanol solution, and cecal ligation and puncture (CLP) was used to construct the sepsis model. The lung tissue damage was observed by hematoxylin-eosin (H&E) staining, and the wet-to-dry (W/D) ratio was used to evaluate the degree of pulmonary edema. Inflammatory cell counting and protein concentration in bronchoalveolar lavage fluid (BALF) were, respectively, detected by hemocytometer and bicinchoninic acid (BCA) method. The levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1ß, and IL-18 in BALF were detected by their commercial enzyme-linked immunosorbent assay (ELISA) kits. The myeloperoxidase (MPO) activity and expression of apoptosis-related proteins and SphK1/S1P/S1PR1 pathway-related proteins were, respectively, analyzed by MPO ELISA kit and Western blot analysis. The cell apoptosis in lung tissues was observed by TUNEL assay. Acute ethanol intoxication (EtOH) decreased the survival rate of mice and exacerbated the lung injury caused by sepsis through increasing pulmonary vascular permeability, neutrophil infiltration, release of inflammatory factors, and cell apoptosis. In addition, EtOH could activate the SphK1/S1P/S1PR1 pathway in CLP mice. However, PF-543, as a specific inhibitor of SphK1, could partially reverse the deleterious effects on lung injury of CLP mice. PF-543 alleviated lung injury caused by sepsis in acute ethanol intoxication rats by suppressing the SphK1/S1P/S1PR1 signaling pathway.


Asunto(s)
Intoxicación Alcohólica/complicaciones , Inhibidores Enzimáticos/farmacología , Lesión Pulmonar/prevención & control , Pulmón/efectos de los fármacos , Lisofosfolípidos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Pirrolidinas/farmacología , Sepsis/tratamiento farmacológico , Receptores de Esfingosina-1-Fosfato/metabolismo , Esfingosina/análogos & derivados , Sulfonas/farmacología , Animales , Apoptosis/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Pulmón/enzimología , Pulmón/patología , Lesión Pulmonar/enzimología , Lesión Pulmonar/etiología , Lesión Pulmonar/patología , Masculino , Metanol/farmacología , Ratones Endogámicos C57BL , Infiltración Neutrófila/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Neumonía/enzimología , Neumonía/etiología , Neumonía/patología , Neumonía/prevención & control , Edema Pulmonar/enzimología , Edema Pulmonar/etiología , Edema Pulmonar/patología , Edema Pulmonar/prevención & control , Sepsis/complicaciones , Sepsis/enzimología , Transducción de Señal , Esfingosina/metabolismo
7.
FASEB J ; 35(7): e21519, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34137477

RESUMEN

Globally, COPD remains a major cause of disability and death. In the United States alone, it is estimated that approximately 14 million people suffer from the disease. Given the high disease burden and requirement for chronic, long-term medical care associated with COPD, it is essential that new disease modifying agents are developed to complement the symptomatic therapeutics currently available. In the present report, we have identified a potentially novel therapeutic agent through the use of a high throughput screen based on the knowledge that cigarette smoke induces the proteolytic enzyme MMP1 leading to destruction of the lung in COPD. A construct utilizing the cigarette responsive promoter element of MMP-1 was conjugated to a luciferase reporter and utilized in an in vitro assay to screen the NIH Molecular Libraries Small Molecule Repository to identify putative targets that suppressed luciferase expression in response to cigarette smoke extract (CSE). Selective serotonin reuptake inhibitors potently inhibited luciferase expression and were further validated. SSRI treatment suppressed MMP-1 production in small airway epithelial cells exposed to (CSE) in vitro as well as in smoke exposed rabbits. In addition, SSRI treatment inhibited inflammatory cytokine production while rescuing cigarette smoke induced downregulation in vivo of the anti-inflammatory lipid transporter ABCA1, previously shown by our laboratory to be lung protective. Importantly, SSRI treatment prevented lung destruction in smoke exposed rabbits as measured by morphometry. These studies support further investigation into SSRIs as a novel therapeutic for COPD may be warranted.


Asunto(s)
Fumar Cigarrillos/efectos adversos , Células Epiteliales/efectos de los fármacos , Pulmón/efectos de los fármacos , Metaloproteinasa 1 de la Matriz/química , Neumonía/tratamiento farmacológico , Enfisema Pulmonar/tratamiento farmacológico , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Animales , Células Cultivadas , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Pulmón/metabolismo , Pulmón/patología , Neumonía/inducido químicamente , Neumonía/enzimología , Neumonía/patología , Enfisema Pulmonar/inducido químicamente , Enfisema Pulmonar/enzimología , Enfisema Pulmonar/patología , Conejos , Serotonina/metabolismo
8.
Am J Respir Cell Mol Biol ; 64(5): 629-640, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33662226

RESUMEN

Deficiency of ASM (acid sphingomyelinase) causes the lysosomal storage Niemann-Pick disease (NPD). Patients with NPD type B may develop progressive interstitial lung disease with frequent respiratory infections. Although several investigations using the ASM-deficient (ASMKO) mouse NPD model revealed inflammation and foamy macrophages, there is little insight into the pathogenesis of NPD-associated lung disease. Using ASMKO mice, we report that ASM deficiency is associated with a complex inflammatory phenotype characterized by marked accumulation of monocyte-derived CD11b+ macrophages and expansion of airspace/alveolar CD11c+ CD11b- macrophages, both with increased size, granularity, and foaminess. Both the alternative and classical pathways were activated, with decreased in situ phagocytosis of opsonized (Fc-coated) targets, preserved clearance of apoptotic cells (efferocytosis), secretion of Th2 cytokines, increased CD11c+/CD11b+ cells, and more than a twofold increase in lung and plasma proinflammatory cytokines. Macrophages, neutrophils, eosinophils, and noninflammatory lung cells of ASMKO lungs also exhibited marked accumulation of chitinase-like protein Ym1/2, which formed large eosinophilic polygonal Charcot-Leyden-like crystals. In addition to providing insight into novel features of lung inflammation that may be associated with NPD, our report provides a novel connection between ASM and the development of crystal-associated lung inflammation with alterations in macrophage biology.


Asunto(s)
Glicoproteínas/inmunología , Lisofosfolipasa/inmunología , Macrófagos Alveolares/inmunología , Macrófagos/inmunología , Enfermedad de Niemann-Pick Tipo A/inmunología , Enfermedad de Niemann-Pick Tipo B/inmunología , Neumonía/inmunología , Esfingomielina Fosfodiesterasa/inmunología , Animales , Antígenos CD11/genética , Antígenos CD11/inmunología , Antígeno CD11b/genética , Antígeno CD11b/inmunología , Tamaño de la Célula , Quitinasas/genética , Quitinasas/inmunología , Modelos Animales de Enfermedad , Eosinófilos/inmunología , Eosinófilos/patología , Femenino , Expresión Génica , Glicoproteínas/genética , Humanos , Lectinas/genética , Lectinas/inmunología , Pulmón/inmunología , Pulmón/patología , Lisofosfolipasa/genética , Macrófagos/patología , Macrófagos Alveolares/patología , Masculino , Ratones , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/patología , Enfermedad de Niemann-Pick Tipo A/enzimología , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo A/patología , Enfermedad de Niemann-Pick Tipo B/enzimología , Enfermedad de Niemann-Pick Tipo B/genética , Enfermedad de Niemann-Pick Tipo B/patología , Fagocitosis , Neumonía/enzimología , Neumonía/genética , Neumonía/patología , Esfingomielina Fosfodiesterasa/deficiencia , Esfingomielina Fosfodiesterasa/genética , Balance Th1 - Th2/genética , beta-N-Acetilhexosaminidasas/genética , beta-N-Acetilhexosaminidasas/inmunología
9.
Nat Rev Immunol ; 21(7): 411-425, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33514947

RESUMEN

Haem oxygenase 1 (HO-1), an inducible enzyme responsible for the breakdown of haem, is primarily considered an antioxidant, and has long been overlooked by immunologists. However, research over the past two decades in particular has demonstrated that HO-1 also exhibits numerous anti-inflammatory properties. These emerging immunomodulatory functions have made HO-1 an appealing target for treatment of diseases characterized by high levels of chronic inflammation. In this Review, we present an introduction to HO-1 for immunologists, including an overview of its roles in iron metabolism and antioxidant defence, and the factors which regulate its expression. We discuss the impact of HO-1 induction in specific immune cell populations and provide new insights into the immunomodulation that accompanies haem catabolism, including its relationship to immunometabolism. Furthermore, we highlight the therapeutic potential of HO-1 induction to treat chronic inflammatory and autoimmune diseases, and the issues faced when trying to translate such therapies to the clinic. Finally, we examine a number of alternative, safer strategies that are under investigation to harness the therapeutic potential of HO-1, including the use of phytochemicals, novel HO-1 inducers and carbon monoxide-based therapies.


Asunto(s)
Antioxidantes/metabolismo , Hemo-Oxigenasa 1/metabolismo , Inflamación/enzimología , Animales , Antiinflamatorios/inmunología , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Monóxido de Carbono/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Inducción Enzimática/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/inmunología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/enzimología , Enfermedades Inflamatorias del Intestino/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Modelos Biológicos , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/enzimología , Esclerosis Múltiple/inmunología , Fitoquímicos/uso terapéutico , Neumonía/tratamiento farmacológico , Neumonía/enzimología , Neumonía/inmunología , Psoriasis/tratamiento farmacológico , Psoriasis/enzimología , Psoriasis/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Inmunología del Trasplante
10.
Int Immunopharmacol ; 90: 107187, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33249045

RESUMEN

Sophoricoside (SOP), an isoflavone glycoside isolated from seed of Sophora japonica L., has been reported to have various pharmacological activities, including anti-cancer, anti-allergy and anti-inflammation. However, the effect of SOP on lipopolysaccharides (LPS)-acute lung injury (ALI) is completely unclear. Here, we found that SOP pretreatment significantly ameliorated LPS-induced pathological damage, tissue permeability, neutrophil infiltration and the production of pro-inflammatory cytokines (TNF-α, IL-1ß and IL-6) in a murine model of ALI. Besides, SOP reduced the production of pro-inflammatory mediators such as iNOS, NO and inflammatory cytokines including TNF-α, IL-1ß and IL-6 in LPS-stimulated RAW264.7 cells and bone marrow derived macrophages. Interestingly, treatment with SOP exhibited no effect on the activation of NF-κB and MAPKs in macrophages but prominently accelerated the expression and nuclear translocation of Nrf2. By using ML385, a specific Nrf2 inhibitor, we found that inhibition of Nrf2 abolished the inhibitory effect of SOP on LPS-induced iNOS expression, NO production as well as pro-inflammatory cytokine generation. SOP also activated AMPK, an upstream protein of Nrf2, under LPS stimuli. Furthermore, we demonstrated that the accelerated expression of Nrf2 induced by SOP was reversed by interference with the AMPK inhibitor Compound C. Taken together, our results suggested that SOP attenuated LPS-induced ALI in AMPK/Nrf2 dependent manner and indicated that SOP might be a potential therapeutic candidate for treating ALI/ARDS.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Antiinflamatorios/farmacología , Benzopiranos/farmacología , Pulmón/efectos de los fármacos , Macrófagos/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Neumonía/prevención & control , Proteínas Quinasas Activadas por AMP/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/patología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Lipopolisacáridos , Pulmón/enzimología , Pulmón/patología , Macrófagos/enzimología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Neumonía/inducido químicamente , Neumonía/enzimología , Neumonía/patología , Células RAW 264.7 , Transducción de Señal
11.
Med Sci Monit ; 26: e924482, 2020 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-32844782

RESUMEN

BACKGROUND Acute pneumonia is a severe inflammatory disease of the respiratory system. Drugs used to treat acute pneumonia often have strong side effects. Recent studies have shown that tetrahydropyrimidines, ZL-5015 has anti-inflammatory and antitumor effects. However, whether ZL-5015 can relieve symptoms of acute pneumonia is unclear. MATERIAL AND METHODS In this study, we used lipo-polysaccharide (LPS) to stimulate SD rats to simulate conditions of acute pneumonia. Diverse doses of ZL-5015 were used for treatment of these rats. After the rates were sacrificed, serum, lung tissue, and bronchoalveolar lavage fluid were collected for the next study. Hematoxylin-eosin (H&E) staining then was used to detect pathologic changes in lung tissues. Enzyme-linked immunosorbent assay was performed to assess levels of inflammatory factors in serum. Commercial kits were used to assess levels of reactive oxygen species (ROS) in bronchoalveolar lavage fluid. RESULTS Treatment of ZL-5015 relieved stenosis of the alveolar space and pulmonary edema. Furthermore, levels of inflammatory factors (TNF-alpha, IL-1ß and IL-18) in the lung tissues and serum were downregulated after treatment with ZL-5015. Production of ROS also was suppressed after application of ZL-5015. Moreover, inhibition of expression of NRF-2 and HO-1 was relieved after treatment with ZL-5015. The therapeutic effect of ZL-5015 showed a dose-response relationship. CONCLUSIONS ZL-5015 alleviated LPS-induced inflammatory injury and oxidative damage by activating the NRF-2/HO-1 pathway.


Asunto(s)
Ésteres/farmacología , Hemo-Oxigenasa 1/metabolismo , Lipopolisacáridos/toxicidad , Factor 2 Relacionado con NF-E2/metabolismo , Neumonía/prevención & control , Pirimidinas/farmacología , Enfermedad Aguda , Animales , Modelos Animales de Enfermedad , Neumonía/enzimología , Neumonía/metabolismo , Ratas
12.
J Ethnopharmacol ; 261: 113095, 2020 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-32531410

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Chronic obstructive pulmonary disease (COPD) is a respiratory inflammatory disease. Unlike asthma, COPD is insensitive to glucocorticoid treatment; thus, it is of great importance to find alternative medications, including Chinese medicine, to suppress inflammation. Bu-Shen-Fang-Chuan formula (BSFCF) is commonly used for the treatment of COPD in China. However, the mechanisms of BSFCF in COPD treatment are still unclear. AIM OF THE STUDY: To verify the anti-inflammatory efficacy of BSFCF in COPD and to explore the possible mechanisms underlying its anti-inflammatory efficacy based on the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt)-Nuclear factor erythroid 2-related factor 2 (Nrf2) and Nuclear factor (NF)-κB signalling pathways. MATERIALS AND METHODS: A rat model of COPD was established by chronic exposure to cigarette smoke (CS) for 6 months. Bronchoalveolar lavage fluid (BALF) and blood were obtained to detect inflammatory cytokines. Lung samples were harvested, and part of each sample was fixed for subsequent H&E staining and immunohistochemical (IHC) analysis. The remaining lung tissues were used for RNA sequencing analysis and western blotting. RESULTS: BSFCF significantly reduced inflammatory infiltration in the lungs of CS-exposed rats and decreased the concentrations of tumor necrosis factor (TNF)-α and interleukin (IL)-6 in both the BALF and serum. Additionally, BSFCF evidently attenuated NF-κB activation and downregulation of glucocorticoid receptor (GR) caused by CS. Furthermore, BSFCF increased the activation of PI3K/Akt-Nrf2 signalling in response to CS. CONCLUSIONS: BSFCF attenuated CS-induced inflammation in COPD, which was partially achieved through the PI3K/Akt-Nrf2 and NF-κB signalling pathways.


Asunto(s)
Antiinflamatorios/farmacología , Fumar Cigarrillos , Medicamentos Herbarios Chinos/farmacología , Pulmón/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Neumonía/prevención & control , Proteínas Proto-Oncogénicas c-akt/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/prevención & control , Animales , Líquido del Lavado Bronquioalveolar/química , Citocinas/metabolismo , Modelos Animales de Enfermedad , Pulmón/enzimología , Pulmón/patología , Masculino , Fosfatidilinositol 3-Quinasa , Fosforilación , Neumonía/enzimología , Neumonía/etiología , Neumonía/patología , Enfermedad Pulmonar Obstructiva Crónica/enzimología , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/patología , Ratas Sprague-Dawley , Receptores de Glucocorticoides/metabolismo , Transducción de Señal
13.
Life Sci ; 256: 117896, 2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32504758

RESUMEN

AIMS: Numerous studies indicate that toll-like receptor 2 (TLR2) led to divergent effects in asthma. The occurrence of autophagy in asthma pathogenesis is still incompletely understood. Here, we aimed to investigate the role of TLR2 and the underlying mechanisms in allergic airway inflammation and autophagy activation. MAIN METHODS: C57BL/6 and TLR2 knockout (TLR2-/-) mice were subjected to an ovalbumin (OVA)-immunized allergic airway model, and were treated with SP600125. Differential cell counts in bronchoalveolar lavage fluid were determined by Wright's staining. Histological analysis of airway inflammation was determined by haematoxylin and eosin (H&E) and periodic acid-Schiff (PAS) staining. The levels of OVA-specific immunoglobulin E (IgE), tumor necrosis factor α (TNF-α) and interleukin 10 (IL-10) were detected by enzyme-linked immunosorbent assay (ELISA). Proteins expression in lung tissues was detected by western blot, expression of TLR2 was further observed by immunofluorescence. Autophagy activation was determined by western blot and transmission electron microscopy (TEM). KEY FINDINGS: TLR2 expression was increased upon OVA challenge, and TLR2 deficiency was associated with decreased allergic airway inflammation. Meanwhile, TLR2 deficiency weakened autophagy activation. Moreover, inhibition of c-Jun N-terminal kinase (JNK) by SP600125 also suppressed OVA-induced allergic airway inflammation and autophagy activation. Interestingly, treating TLR2-/- mice with SP600125 showed similar OVA-induced allergic airway inflammation and autophagy activation compared to that in vehicle-treated TLR2-/- mice. SIGNIFICANCE: TLR2 might contribute to the maintenance of allergic airway inflammation through JNK signaling pathway accompanying with autophagy activation. These findings may provide a novel signal target for prevention of allergic airway inflammation.


Asunto(s)
Autofagia , Hipersensibilidad/enzimología , Hipersensibilidad/patología , Sistema de Señalización de MAP Quinasas , Ovalbúmina/efectos adversos , Neumonía/enzimología , Neumonía/patología , Receptor Toll-Like 2/metabolismo , Animales , Modelos Animales de Enfermedad , Células Caliciformes/patología , Inmunoglobulina E/sangre , Pulmón/patología , Pulmón/ultraestructura , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Neumonía/sangre , Proteínas Proto-Oncogénicas c-akt/metabolismo
14.
Naunyn Schmiedebergs Arch Pharmacol ; 393(9): 1659-1670, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32377772

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease with limited treatment options. Zingerone found in ginger (Zingiber officinale L.) has many pharmacological effects, especially antiinflammatory and antioxidant activity. However, the effect of zingerone on pulmonary fibrosis (PF) is not fully known. The aim of this study was to investigate the effect of zingerone on bleomycin (BLM)-induced PF and its underlying mechanisms. Wistar-albino rats were given single dose of BLM (5 mg/kg, intratracheal) or vehicle (saline). In treatment groups, zingerone (50 and 100 mg/kg, p.o.) was administered orally for 14 days after BLM administration. Rats and lung tissue were weighed to determine lung index. Antioxidant, antiinflammatory effects, and hydroxyproline content of zingerone were determined by ELISA method. Pulmonary inflammation, collagen deposition, and fibrosis score were determined with Hematoxylin-Eosin (HxE) and Masson's trichrome staining. Transforming growth factor-beta 1 (TGF-ß1) and inducible nitric oxide synthase (iNOS) expressions were detected immunohistochemically. BLM administration increased lipid peroxidation (MDA) and decreased superoxide dismutase (SOD) and glutathione peroxidase (GPx) activity. In addition, BLM caused increased levels of tumor necrosis factor alpha (TNF-α) and interleukin-1ß (IL-1ß) in bronchoalveolar lavage fluid (BALF) and accumulation of collagen bundles. Zingerone administration decreased collagen accumulation, TNF-α and IL-1ß levels, MDA level, TGF-ß1, and iNOS expression and increased SOD and GPx activity. Histopathological findings supported the results. These results show that zingerone (50 and 100 mg/kg) at both doses significantly contributes to healing of PF by improving inflammation, oxidative stress, and histopathological alterations and by affecting TGF-ß1 and iNOS signaling pathways.


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Guayacol/análogos & derivados , Mediadores de Inflamación/metabolismo , Pulmón/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estrés Oxidativo/efectos de los fármacos , Neumonía/prevención & control , Fibrosis Pulmonar/prevención & control , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Bleomicina , Modelos Animales de Enfermedad , Guayacol/farmacología , Peroxidación de Lípido/efectos de los fármacos , Pulmón/enzimología , Pulmón/patología , Neumonía/inducido químicamente , Neumonía/enzimología , Neumonía/patología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/enzimología , Fibrosis Pulmonar/patología , Ratas Wistar , Transducción de Señal
15.
Biosci Rep ; 40(5)2020 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-32420586

RESUMEN

The present study aims to reveal the molecular mechanism of peroxisome proliferator-activated receptor γ (PPARγ) on sepsis-induced acute lung injury (ALI). To do that, the rat injury model was established using cecal ligation and perforation (CLP) method, followed by different treatments, and the rats were divided into Sham group, CLP group, CLP + rosiglitazone (PPARγ agonist) group, CLP + GW9662 (PPARγ inhibitor) group, CLP + bpV (phosphatase and tensin homolog (PTEN) inhibitor) group, CLP + GW9662 + bpV group. Compared with Sham group, the mRNA and protein expression levels of PPARγ were down-regulated, the inflammation levels were elevated, and the apoptosis was increased in CLP group. After treatment with rosiglitazone, the protein expression level of PPARγ was significantly up-regulated, the phosphorylation level of PTEN/ß-catenin pathway was decreased, the PTEN/ß-catenin pathway was inhibited, the lung injury, inflammation and apoptosis were reduced. The opposite effect was observed after treatment with GW9662. Besides, bpV inhibited PTEN/ß-catenin pathway, and relieved the lung tissue injury. The overexpression of PPARγ reduced inflammatory response and inhibited apoptosis in sepsis-induced ALI. Furthermore, PPARγ relieved the sepsis-induced ALI by inhibiting the PTEN/ß-catenin pathway.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Pulmón/efectos de los fármacos , PPAR gamma/agonistas , Fosfohidrolasa PTEN/metabolismo , Rosiglitazona/farmacología , Sepsis/tratamiento farmacológico , beta Catenina/metabolismo , Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/microbiología , Lesión Pulmonar Aguda/patología , Anilidas/farmacología , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Pulmón/enzimología , Pulmón/microbiología , Pulmón/patología , Masculino , PPAR gamma/antagonistas & inhibidores , PPAR gamma/genética , PPAR gamma/metabolismo , Fosforilación , Neumonía/enzimología , Neumonía/microbiología , Neumonía/prevención & control , Edema Pulmonar/enzimología , Edema Pulmonar/microbiología , Edema Pulmonar/prevención & control , Ratas Sprague-Dawley , Sepsis/enzimología , Sepsis/microbiología , Transducción de Señal
16.
Pediatr Res ; 87(3): 494-500, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31578032

RESUMEN

BACKGROUND: Intrauterine inflammation affects fetal lung development. BTB and CNC homology 1 (Bach1) is a transcriptional repressor of heme oxygenase-1 (HO-1) and interleukin-6 (IL-6) genes. We investigated the role of Bach1 in the development of fetal mouse lungs exposed to lipopolysaccharide (LPS) using a whole fetal lung tissue culture system. METHODS: We isolated and cultured embryonic day 12.5 fetal mouse lungs from pregnant Bach1 knockout (-/-) and wild-type (WT) mice. Airway branching morphogenesis was assessed by microscopically counting peripheral lung buds after incubation with/without LPS. Expression levels of genes related to inflammation and oxidative stress were evaluated using quantitative PCR. Zinc protoporphyrin, HO-1-specific inhibitor, was used. RESULTS: Branching morphogenesis was observed in Bach1-/- and WT fetal mice lungs without LPS exposure; after exposure to LPS, the number of peripheral lung buds was suppressed in Bach1-/- group only. Basal messenger RNA (mRNA) and protein expression of HO-1 was significantly higher in Bach1-/- group than in WT group; IL-6 and monocyte chemoattractant protein-1 mRNA expression was significantly increased after LPS exposure in both groups. Zinc protoporphyrin mitigated the LPS-induced suppression of branching morphogenesis in Bach1-/- mice. CONCLUSION: The ablation of Bach1 suppresses airway branching morphogenesis after LPS exposure by increased basal expression levels of HO-1.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Hemo-Oxigenasa 1/metabolismo , Pulmón/enzimología , Proteínas de la Membrana/metabolismo , Neumonía/enzimología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/deficiencia , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Hemo-Oxigenasa 1/genética , Interleucina-6/genética , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Pulmón/embriología , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis , Técnicas de Cultivo de Órganos , Neumonía/embriología , Embarazo , Transducción de Señal , Regulación hacia Arriba
17.
Toxicol Lett ; 313: 178-187, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31284023

RESUMEN

Long-term inhalation of crystalline silica particles leads to silicosis characterized by pulmonary inflammation and interstitial fibrosis. The growth arrest-specific protein 6 (Gas6) and its tyrosine receptor Mer have been implicated to involve in the regulation of inflammation, innate immunity and tissue repair. However, the role of Gas6 or Mer in silica-induced lung inflammation and fibrosis has not been investigated previously. In this study, we observed a remarkable increase of Gas6 in bronchoalveolar lavage fluid (BALF) from wild-type C57BL/6 mice after silica intratracheal administration. Then, we investigated whether genetic loss of Gas6 or Mer could attenuate silica-induced lung inflammation and fibrosis. Our results showed that Gas6-/- and Mer-/- mice exhibited reduced lung inflammation response from days 7 to 84 after silica exposure. We also uncovered an overexpression of the suppressor of cytokine signaling protein 1 in silica-treated deficient mice. Moreover, Gas6 or Mer deficiency attenuated silica-induced collagen deposition by inhibiting the expression of transforming growth factor-ß. We conclude that gene absence of Gas6 or Mer is protective against silica-induced lung inflammation and fibrosis in mice. Targeting Gas6/Mer pathway may be a potential therapeutic approach to treat pulmonary fibrosis in patients with silicosis.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/deficiencia , Pulmón/enzimología , Neumonía/prevención & control , Fibrosis Pulmonar/prevención & control , Silicosis/prevención & control , Tirosina Quinasa c-Mer/deficiencia , Animales , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Péptidos y Proteínas de Señalización Intercelular/genética , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/enzimología , Neumonía/genética , Neumonía/patología , Fibrosis Pulmonar/enzimología , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Transducción de Señal , Silicosis/enzimología , Silicosis/genética , Silicosis/patología , Proteína 1 Supresora de la Señalización de Citocinas/genética , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Tirosina Quinasa c-Mer/genética
18.
Artículo en Inglés | MEDLINE | ID: mdl-31190784

RESUMEN

Background: Exposure to environmental particulate matter (PM) ≤2.5 µm in diameter (PM2.5) and smoking are common contributors to COPD, and pertinent research implicates both factors in pulmonary inflammation. Using in vivo mouse and in vitro human cellular models, we investigated the joint impact of PM2.5 pollution, and cigarette smoke (CS) in mice or cigarette-smoke extract (CSE) in cells on COPD inflammation, and explored potential mechanisms. Methods: Tissue changes in lungs of C57BL/6 mice exposed to PM2.5 and CS were studied by light microscopy, H&E, immunochemistry, and immunofluorescence-stained sections. Levels of inflammatory factors induced by PM2.5/CS in mice and PM2.5/CSE in 16HBE cells were also monitored by quantitative reverse-transcription (qRT)-PCR and ELISA. Expression of genes related to the Wnt5a-signaling pathway was assessed at transcriptional and protein levels using immunofluorescence, qRT-PCR, and Western blotting. Results: Inflammatory response to combined exposure of PM2.5 and CS or CSE in mouse and 16HBE cells surpassed responses incited separately. Although separate PM2.5 and CS/CSE exposure upregulated the expression of Wnt5a (a member of the Wnt-secreted glycoprotein family), combined PM2.5 and CS/CSE exposure produced a steeper rise in Wnt5a levels. Use of a Wnt5a antagonist (BOX5) successfully blocked related inflammatory effects. ERK phosphorylation appeared to mediate the effects of Wnt5a in the COPD model, promoting PM2.5 aggravation of CS/CSE-induced airway inflammation. Conclusion: Our findings suggest that combined PM2.5 and CS/CSE exposure induce airway inflammation and Wnt5a expression in vivo in mice and in vitro in 16HBE cells. Furthermore, PM2.5 seems to aggravate CS/CSE-induced inflammation via the Wnt5a-ERK pathway in the context of COPD.


Asunto(s)
Fumar Cigarrillos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Pulmón/efectos de los fármacos , Material Particulado/toxicidad , Neumonía/etiología , Enfermedad Pulmonar Obstructiva Crónica/etiología , Vía de Señalización Wnt/efectos de los fármacos , Proteína Wnt-5a/metabolismo , Animales , Línea Celular , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Humanos , Pulmón/enzimología , Masculino , Ratones Endogámicos C57BL , Tamaño de la Partícula , Neumonía/enzimología , Neumonía/genética , Enfermedad Pulmonar Obstructiva Crónica/enzimología , Enfermedad Pulmonar Obstructiva Crónica/genética , Proteína Wnt-5a/genética
19.
PLoS One ; 14(5): e0216569, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31067276

RESUMEN

Although accepted agents in chorioamnionitis and preterm birth, the role of Ureaplasma species (spp.) in inflammation-driven morbidities of prematurity, including the development of bronchopulmonary dysplasia, remains controversial. To add to scarce in vitro data addressing the pro-inflammatory capacity of Ureaplasma spp., pulmonary epithelial-like A549 cells and human pulmonary microvascular endothelial cells (HPMEC) were incubated with Ureaplasma (U.) urealyticum, U. parvum, and Escherichia coli lipopolysaccharide (LPS). Ureaplasma isolates down-regulated caspase mRNA levels in A549 cells (caspase 8: p<0.001, 9: p<0.001, vs. broth), while increasing caspase protein expression, enzyme activity, and cell death in HPMEC (active caspase 3: p<0.05, caspase 8: p<0.05, active caspase 9: p<0.05, viability: p<0.05). LPS, contrarily, induced caspase mRNA expression in HPMEC (caspase 3: p<0.01, 4: p<0.001, 5: p<0.001, 8: p<0.001, vs. control), but not in A549 cells, and did not affect enzyme activity or protein levels in either cell line. LPS, but neither Ureaplasma isolate, enhanced mRNA expression of pro-inflammatory interleukin (IL)-6 in both A549 (p<0.05, vs. control) and HPMEC (p<0.001) as well as tumor necrosis factor-α (p<0.01), IL-1ß (p<0.001), and IL-8 (p<0.05) in HPMEC. We are therefore the first to demonstrate a differential modulation of pulmonary caspases by Ureaplasma spp. in vitro. Ureaplasma-driven enhanced protein expression and activity of caspases in pulmonary endothelial cells result in cell death and may cause structural damage. Down-regulated caspase mRNA in pulmonary epithelial cells, contrarily, may indicate Ureaplasma-induced inhibition of apoptosis and prevent effective immune responses. Both may ultimately contribute to chronic Ureaplasma colonization and long-term pulmonary inflammation.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Citocinas/metabolismo , Endotelio Vascular/enzimología , Células Epiteliales/enzimología , Neumonía/etiología , Infecciones por Ureaplasma/complicaciones , Células A549 , Células Cultivadas , Humanos , Neumonía/enzimología , Neumonía/patología , Ureaplasma/aislamiento & purificación
20.
Toxicol Lett ; 310: 61-69, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31018152

RESUMEN

PM2.5 is the main particulate air pollutant that is capable of inducing airway injury. Previous studies have indicated that Rac1 is involved in cigarette smoke-induced lung inflammation and lipopolysaccharide-mediated pulmonary injury. However, the contribution of Rac1 activity to PM2.5-induced lung inflammation remains largely unclear. Here, we investigated the regulation of Rac1 in PM2.5-induced inflammation in mouse airways and human bronchial epithelial cells (16HBE). The lungs of mice exposed to PM2.5 showed increased IL-1ß expression and an accumulation of inflammatory cells, thereby indicating high Rac1 activity. The exposure of 16HBE cells to PM2.5 resulted in elevated Rac1 levels, as well as an increased release of IL-1ß. Particularly, the selective inhibition of Rac1 ameliorated the IL-1ß release and inflammation in model lungs. Histological assessment showed that treatment with a Rac1 inhibitor, NSC23766, reduced the infiltration of neutrophils and macrophages into the airway lumen. Moreover, the selective inhibition or knockdown of Rac1 decreased IL-1ß release in 16HBE cells induced by PM2.5, which correlated with PM2.5-induced Rac1-regulated AKT signaling. Our data suggest an important role for Rac1 in the pathological alterations associated with PM2.5-mediated lung inflammation. Rac1 may be a promising therapeutic target for the treatment of the inflammatory diseases induced by PM2.5 inhalation.


Asunto(s)
Aminoquinolinas/farmacología , Antiinflamatorios/farmacología , Pulmón/efectos de los fármacos , Neuropéptidos/antagonistas & inhibidores , Material Particulado/toxicidad , Neumonía/prevención & control , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Animales , Línea Celular , Humanos , Exposición por Inhalación/efectos adversos , Interleucina-1beta/metabolismo , Pulmón/enzimología , Pulmón/patología , Masculino , Ratones Endogámicos ICR , Neuropéptidos/metabolismo , Tamaño de la Partícula , Neumonía/inducido químicamente , Neumonía/enzimología , Neumonía/patología , Interferencia de ARN , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA