Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 265
Filtrar
1.
Respir Res ; 25(1): 330, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39227918

RESUMEN

BACKGROUND: Vascular endothelial damage is involved in the development and exacerbation of ventilator-induced lung injury (VILI). Pulmonary endothelial glycocalyx and neutrophil extracellular traps (NETs) are endothelial protective and damaging factors, respectively; however, their dynamics in VILI and the effects of recombinant thrombomodulin and antithrombin on these dynamics remain unclear. We hypothesized that glycocalyx degradation and NETs are induced by VILI and suppressed by recombinant thrombomodulin, recombinant antithrombin, or their combination. METHODS: VILI was induced in male C57BL/6J mice by intraperitoneal lipopolysaccharide injection (20 mg/kg) and high tidal volume ventilation (20 mL/kg). In the intervention groups, recombinant thrombomodulin, recombinant antithrombin, or their combination was administered at the start of mechanical ventilation. Glycocalyx degradation was quantified by measuring serum syndecan-1, fluorescence-labeled lectin intensity, and glycocalyx-occupied area in the pulmonary vascular lumen. Double-stranded DNA in the bronchoalveolar fluid and fluorescent areas of citrullinated histone H3 and myeloperoxidase were quantified as NET formation. RESULTS: Serum syndecan-1 increased, and lectin fluorescence intensity decreased in VILI. Electron microscopy revealed decreases in glycocalyx-occupied areas within pulmonary microvessels in VILI. Double-stranded DNA levels in the bronchoalveolar lavage fluid and the fluorescent area of citrullinated histone H3 and myeloperoxidase in lung tissues increased in VILI. Recombinant thrombomodulin, recombinant antithrombin, and their combination reduced glycocalyx injury and NET marker levels. There was little difference in glycocalyx injury and NET makers between the intervention groups. CONCLUSION: VILI induced glycocalyx degradation and NET formation. Recombinant thrombomodulin and recombinant antithrombin attenuated glycocalyx degradation and NETs in our VILI model. The effect of their combination did not differ from that of either drug alone. Recombinant thrombomodulin and antithrombin have the potential to be therapeutic agents for biotrauma in VILI.


Asunto(s)
Antitrombinas , Endotoxemia , Trampas Extracelulares , Glicocálix , Ratones Endogámicos C57BL , Proteínas Recombinantes , Trombomodulina , Lesión Pulmonar Inducida por Ventilación Mecánica , Animales , Glicocálix/metabolismo , Glicocálix/efectos de los fármacos , Glicocálix/patología , Trombomodulina/metabolismo , Trombomodulina/administración & dosificación , Trampas Extracelulares/metabolismo , Trampas Extracelulares/efectos de los fármacos , Masculino , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Ratones , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/patología , Lesión Pulmonar Inducida por Ventilación Mecánica/tratamiento farmacológico , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control , Endotoxemia/metabolismo , Endotoxemia/patología , Endotoxemia/tratamiento farmacológico , Endotoxemia/inducido químicamente , Antitrombinas/farmacología , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Modelos Animales de Enfermedad , Sindecano-1/metabolismo
2.
Discov Med ; 36(187): 1600-1609, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39190375

RESUMEN

BACKGROUND: Macrophages are activated in ventilator-induced lung injury (VILI), accompanied by macrophage pyroptosis. Remimazolam (Re) plays a role in inhibiting macrophage activation. In this study, we aimed to investigate the mechanism of Re in VILI. METHODS: A VILI model (20 mL/kg mechanical ventilation) was created using C57BL/6 mice. Alveolar macrophages were isolated from bronchoalveolar lavage fluid (BALF) and received mechanical stretching to simulate the mechanical ventilation in vitro. VILI model mice were treated with Re (16 mg/kg) to assess the alveolar structure, wet/dry (W/D) weight ratio, endothelial barrier antigen (EBA) permeability index, BALF protein content, inflammatory factors, macrophage pyroptosis, pyroptosis-related factors, and translocator protein (TSPO) level using a series of biological experiments. Whether Re alleviated macrophage pyroptosis by regulating TSPO was determined by rescue experiments. RESULTS: Re alleviated VILI, as evidenced by improvement of abnormal morphology of lung tissues during VILI and decreases in the lung W/D weight ratio, lung EBA permeability index, and BALF protein content. Re attenuated pulmonary inflammation and macrophage pyroptosis during VILI via down-regulation of inflammatory factors (myeloperoxidase, malondialchehyche, 8-hydroxy-2 deoxyguanosine, interleukin-6, tumor necrosis factor-α, macrophage inflammatory protein-2, interleukin-1ß, and interleukin-18), and pyroptosis factors (cleaved gasdermin D (GSDMD)/GSDMD value, NOD-like receptor thermal protein domain associated protein 3 (NLRP3), and caspase-1). Re activated TSPO in macrophages. TSPO overexpression rescued the cell stretch-inhibited macrophage viability and cell stretch-induced macrophage pyroptosis. CONCLUSION: Re alleviates VILI by activating TSPO to inhibit macrophage pyroptosis.


Asunto(s)
Ratones Endogámicos C57BL , Piroptosis , Lesión Pulmonar Inducida por Ventilación Mecánica , Animales , Lesión Pulmonar Inducida por Ventilación Mecánica/patología , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/tratamiento farmacológico , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control , Piroptosis/efectos de los fármacos , Ratones , Masculino , Receptores de GABA/metabolismo , Modelos Animales de Enfermedad , Líquido del Lavado Bronquioalveolar/química , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/patología
3.
Int Immunopharmacol ; 139: 112725, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39059100

RESUMEN

PURPOSE: To investigate esketamine's impact on inflammation and oxidative stress in ventilated chronic obstructive pulmonary disease (COPD) rats, examining its regulatory mechanisms. METHODS: Rats were divided into four groups: control group (Con), COPD model group (M), COPD model with saline treatment group (M+S), and COPD model with esketamine treatment group (M+K), with 12 rats in each group. After two months, all rats underwent anesthesia and mechanical ventilation. Group M+K received 5 mg/kg esketamine intravenously, while Group M+S received the same volume of saline. Lung tissues were collected for analysis two hours later, including airway peak pressure, wet-to-dry(W/D) ratio, lung permeability index(LPI), hematoxylin and eosin(H&E) staining, and transmission electron microscopy(TEM). Tumor necrosis factor-alpha(TNF-α), interleukin-6(IL-6), interleukin-8(IL-8), and interleukin-10(IL-10) levels were determined by enzyme-linked immunosorbent assay(ELISA); phosphorylated Nuclear Factor Kappa B(p-NF-κB), mitogen-activated protein kinase 14(p38), phosphorylated p38 (p-p38), c-Jun N-terminal kinase(JNK), and phosphorylated JNK (p-JNK) expressions by Western blotting and immunohistochemistry; and malondialdehyde(MDA), myeloperoxidase(MPO), and superoxide dismutase(SOD) levels were also measured by corresponding biochemical assays. RESULTS: Lung specimens from groups M, M+S, and M+K manifested hallmark histopathological features of COPD. Compared with group Con, group M displayed increased peak airway pressure, W/D ratio, and LPI. In group M+K, compared with group M, esketamine significantly reduced the W/D ratio, LPI, and concentrations of pro-inflammatory cytokines TNF-α, IL-6, and IL-8 while concurrently elevating IL-10 levels. Furthermore, the treatment attenuated the activation of the NF-κB and MAPK pathways, indicated by decreased levels of p-NF-κB, p-p38, and p-JNK.Additionally, compared to group M, group M+K showed decreased MDA and MPO levels and increased SOD levels in lung tissue. CONCLUSION: Esketamine attenuates mechanical ventilation-induced lung injury in COPD rat models by inhibiting the MAPK/NF-κB signaling pathway and reducing oxidative stress.


Asunto(s)
Citocinas , Ketamina , Pulmón , FN-kappa B , Estrés Oxidativo , Enfermedad Pulmonar Obstructiva Crónica , Ratas Sprague-Dawley , Transducción de Señal , Animales , Ketamina/uso terapéutico , Ketamina/farmacología , Estrés Oxidativo/efectos de los fármacos , FN-kappa B/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Masculino , Citocinas/metabolismo , Ratas , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/inmunología , Transducción de Señal/efectos de los fármacos , Lesión Pulmonar Inducida por Ventilación Mecánica/tratamiento farmacológico , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/patología , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Modelos Animales de Enfermedad , Respiración Artificial/efectos adversos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo
4.
Mol Med ; 30(1): 68, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38778274

RESUMEN

BACKGROUND: Acute respiratory distress syndrome (ARDS) is characterized by alveolar edema that can progress to septal fibrosis. Mechanical ventilation can augment lung injury, termed ventilator-induced lung injury (VILI). Connective tissue growth factor (CTGF), a mediator of fibrosis, is increased in ARDS patients. Blocking CTGF inhibits fibrosis and possibly vascular leakage. This study investigated whether neutralizing CTGF reduces pulmonary edema in VILI. METHODS: Following LPS administration, rats were mechanically ventilated for 6 h with low (6 mL/kg; low VT) or moderate (10 mL/kg; mod VT) tidal volume and treated with a neutralizing CTGF antibody (FG-3154) or placebo lgG (vehicle). Control rats without LPS were ventilated for 6 h with low VT. Lung wet-to-dry weight ratio, FITC-labeled dextran permeability, histopathology, and soluble RAGE were determined. RESULTS: VILI was characterized by reduced PaO2/FiO2 ratio (low VT: 540 [381-661] vs. control: 693 [620-754], p < 0.05), increased wet-to-dry weight ratio (low VT: 4.8 [4.6-4.9] vs. control: 4.5 [4.4-4.6], p < 0.05), pneumonia (low VT: 30 [0-58] vs. control: 0 [0-0]%, p < 0.05) and interstitial inflammation (low VT: 2 [1-3] vs. control: 1 [0-1], p < 0.05). FG-3154 did not affect wet-to-dry weight ratio (mod VT + FG-3154: 4.8 [4.7-5.0] vs. mod VT + vehicle: 4.8 [4.8-5.0], p > 0.99), extravasated dextrans (mod VT + FG-3154: 0.06 [0.04-0.09] vs. mod VT + vehicle: 0.04 [0.03-0.09] µg/mg tissue, p > 0.99), sRAGE (mod VT + FG-3154: 1865 [1628-2252] vs. mod VT + vehicle: 1885 [1695-2159] pg/mL, p > 0.99) or histopathology. CONCLUSIONS: 'Double hit' VILI was characterized by inflammation, impaired oxygenation, pulmonary edema and histopathological lung injury. Blocking CTGF does not improve oxygenation nor reduce pulmonary edema in rats with VILI.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo , Edema Pulmonar , Lesión Pulmonar Inducida por Ventilación Mecánica , Animales , Lesión Pulmonar Inducida por Ventilación Mecánica/tratamiento farmacológico , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/patología , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/antagonistas & inhibidores , Ratas , Masculino , Edema Pulmonar/etiología , Edema Pulmonar/metabolismo , Anticuerpos Neutralizantes/farmacología , Ratas Sprague-Dawley , Pulmón/patología , Pulmón/metabolismo , Modelos Animales de Enfermedad , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores
5.
Am J Physiol Lung Cell Mol Physiol ; 327(1): L19-L39, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38712429

RESUMEN

Mechanical ventilation can cause ventilation-induced lung injury (VILI). The concept of stress concentrations suggests that surfactant dysfunction-induced microatelectases might impose injurious stresses on adjacent, open alveoli and function as germinal centers for injury propagation. The aim of the present study was to quantify the histopathological pattern of VILI progression and to test the hypothesis that injury progresses at the interface between microatelectases and ventilated lung parenchyma during low-positive end-expiratory pressure (PEEP) ventilation. Bleomycin was used to induce lung injury with microatelectases in rats. Lungs were then mechanically ventilated for up to 6 h at PEEP = 1 cmH2O and compared with bleomycin-treated group ventilated protectively with PEEP = 5 cmH2O to minimize microatelectases. Lung mechanics were measured during ventilation. Afterward, lungs were fixed at end-inspiration or end-expiration for design-based stereology. Before VILI, bleomycin challenge reduced the number of open alveoli [N(alvair,par)] by 29%. No differences between end-inspiration and end-expiration were observed. Collapsed alveoli clustered in areas with a radius of up to 56 µm. After PEEP = 5 cmH2O ventilation for 6 h, N(alvair,par) remained stable while PEEP = 1 cmH2O ventilation led to an additional loss of aerated alveoli by 26%, mainly due to collapse, with a small fraction partly edema filled. Alveolar loss strongly correlated to worsening of tissue elastance, quasistatic compliance, and inspiratory capacity. The radius of areas of collapsed alveoli increased to 94 µm, suggesting growth of the microatelectases. These data provide evidence that alveoli become unstable in neighborhood of microatelectases, which most likely occurs due to stress concentration-induced local vascular leak and surfactant dysfunction.NEW & NOTEWORTHY Low-volume mechanical ventilation in the presence of high surface tension-induced microatelectases leads to the degradation of lung mechanical function via the progressive loss of alveoli. Microatelectases grow at the interfaces of collapsed and open alveoli. Here, stress concentrations might cause injury and alveolar instability. Accumulation of small amounts of alveolar edema can be found in a fraction of partly collapsed alveoli but, in this model, alveolar flooding is not a major driver for degradation of lung mechanics.


Asunto(s)
Respiración con Presión Positiva , Alveolos Pulmonares , Lesión Pulmonar Inducida por Ventilación Mecánica , Animales , Alveolos Pulmonares/patología , Alveolos Pulmonares/fisiopatología , Ratas , Masculino , Respiración con Presión Positiva/métodos , Respiración con Presión Positiva/efectos adversos , Lesión Pulmonar Inducida por Ventilación Mecánica/patología , Lesión Pulmonar Inducida por Ventilación Mecánica/fisiopatología , Bleomicina/toxicidad , Bleomicina/efectos adversos , Ratas Sprague-Dawley , Pulmón/patología , Pulmón/fisiopatología , Respiración Artificial/efectos adversos , Respiración Artificial/métodos , Mecánica Respiratoria , Atelectasia Pulmonar/patología , Atelectasia Pulmonar/fisiopatología
6.
Am J Respir Cell Mol Biol ; 71(1): 43-52, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38767348

RESUMEN

Mechanical ventilation contributes to the morbidity and mortality of patients in intensive care, likely through the exacerbation and dissemination of inflammation. Despite the proximity of the pleural cavity to the lungs and exposure to physical forces, little attention has been paid to its potential as an inflammatory source during ventilation. Here, we investigate the pleural cavity as a novel site of inflammation during ventilator-induced lung injury. Mice were subjected to low or high tidal volume ventilation strategies for up to 3 hours. Ventilation with a high tidal volume significantly increased cytokine and total protein levels in BAL and pleural lavage fluid. In contrast, acid aspiration, explored as an alternative model of injury, only promoted intraalveolar inflammation, with no effect on the pleural space. Resident pleural macrophages demonstrated enhanced activation after injurious ventilation, including upregulated ICAM-1 and IL-1ß expression, and the release of extracellular vesicles. In vivo ventilation and in vitro stretch of pleural mesothelial cells promoted ATP secretion, whereas purinergic receptor inhibition substantially attenuated extracellular vesicles and cytokine levels in the pleural space. Finally, labeled protein rapidly translocated from the pleural cavity into the circulation during high tidal volume ventilation, to a significantly greater extent than that of protein translocation from the alveolar space. Overall, we conclude that injurious ventilation induces pleural cavity inflammation mediated through purinergic pathway signaling and likely enhances the dissemination of mediators into the vasculature. This previously unidentified consequence of mechanical ventilation potentially implicates the pleural space as a focus of research and novel avenue for intervention in critical care.


Asunto(s)
Ratones Endogámicos C57BL , Cavidad Pleural , Lesión Pulmonar Inducida por Ventilación Mecánica , Animales , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/patología , Cavidad Pleural/metabolismo , Cavidad Pleural/patología , Inflamación/patología , Inflamación/metabolismo , Ratones , Respiración Artificial/efectos adversos , Volumen de Ventilación Pulmonar , Macrófagos/metabolismo , Macrófagos/patología , Adenosina Trifosfato/metabolismo , Vesículas Extracelulares/metabolismo , Masculino , Citocinas/metabolismo , Líquido del Lavado Bronquioalveolar , Modelos Animales de Enfermedad , Interleucina-1beta/metabolismo
8.
Am J Physiol Lung Cell Mol Physiol ; 326(6): L687-L697, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38563965

RESUMEN

Chronic cigarette smoke exposure decreases lung expression of WWOX which is known to protect the endothelial barrier during infectious models of acute respiratory distress syndrome (ARDS). Proteomic analysis of WWOX-silenced endothelial cells (ECs) was done using tandem mass tag mass spectrometry (TMT-MS). WWOX-silenced ECs as well as those isolated from endothelial cell Wwox knockout (EC Wwox KO) mice were subjected to cyclic stretch (18% elongation, 0.5 Hz, 4 h). Cellular lysates and media supernatant were harvested for assays of cellular signaling, protein expression, and cytokine release. These were repeated with dual silencing of WWOX and zyxin. Control and EC Wwox KO mice were subjected to high tidal volume ventilation. Bronchoalveolar lavage fluid and mouse lung tissue were harvested for cellular signaling, cytokine secretion, and histological assays. TMT-MS revealed upregulation of zyxin expression during WWOX knockdown which predicted a heightened inflammatory response to mechanical stretch. WWOX-silenced ECs and ECs isolated from EC Wwox mice displayed significantly increased cyclic stretch-mediated secretion of various cytokines (IL-6, KC/IL-8, IL-1ß, and MCP-1) relative to controls. This was associated with increased ERK and JNK phosphorylation but decreased p38 mitogen-activated kinases (MAPK) phosphorylation. EC Wwox KO mice subjected to VILI sustained a greater degree of injury than corresponding controls. Silencing of zyxin during WWOX knockdown abrogated stretch-induced increases in IL-8 secretion but not in IL-6. Loss of WWOX function in ECs is associated with a heightened inflammatory response during mechanical stretch that is associated with increased MAPK phosphorylation and appears, in part, to be dependent on the upregulation of zyxin.NEW & NOTEWORTHY Prior tobacco smoke exposure is associated with an increased risk of acute respiratory distress syndrome (ARDS) during critical illness. Our laboratory is investigating one of the gene expression changes that occurs in the lung following smoke exposure: WWOX downregulation. Here we describe changes in protein expression associated with WWOX knockdown and its influence on ventilator-induced ARDS in a mouse model.


Asunto(s)
Células Endoteliales , Inflamación , Ratones Noqueados , Lesión Pulmonar Inducida por Ventilación Mecánica , Oxidorreductasa que Contiene Dominios WW , Animales , Oxidorreductasa que Contiene Dominios WW/metabolismo , Oxidorreductasa que Contiene Dominios WW/genética , Ratones , Células Endoteliales/metabolismo , Células Endoteliales/patología , Inflamación/metabolismo , Inflamación/patología , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/patología , Lesión Pulmonar Inducida por Ventilación Mecánica/genética , Citocinas/metabolismo , Ratones Endogámicos C57BL , Técnicas de Silenciamiento del Gen , Masculino , Pulmón/metabolismo , Pulmón/patología , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética
9.
Respir Res ; 25(1): 37, 2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-38238778

RESUMEN

Acute respiratory distress syndrome (ARDS) alters the dynamics of lung inflation during mechanical ventilation. Repetitive alveolar collapse and expansion (RACE) predisposes the lung to ventilator-induced lung injury (VILI). Two broad approaches are currently used to minimize VILI: (1) low tidal volume (LVT) with low-moderate positive end-expiratory pressure (PEEP); and (2) open lung approach (OLA). The LVT approach attempts to protect already open lung tissue from overdistension, while simultaneously resting collapsed tissue by excluding it from the cycle of mechanical ventilation. By contrast, the OLA attempts to reinflate potentially recruitable lung, usually over a period of seconds to minutes using higher PEEP used to prevent progressive loss of end-expiratory lung volume (EELV) and RACE. However, even with these protective strategies, clinical studies have shown that ARDS-related mortality remains unacceptably high with a scarcity of effective interventions over the last two decades. One of the main limitations these varied interventions demonstrate to benefit is the observed clinical and pathologic heterogeneity in ARDS. We have developed an alternative ventilation strategy known as the Time Controlled Adaptive Ventilation (TCAV) method of applying the Airway Pressure Release Ventilation (APRV) mode, which takes advantage of the heterogeneous time- and pressure-dependent collapse and reopening of lung units. The TCAV method is a closed-loop system where the expiratory duration personalizes VT and EELV. Personalization of TCAV is informed and tuned with changes in respiratory system compliance (CRS) measured by the slope of the expiratory flow curve during passive exhalation. Two potentially beneficial features of TCAV are: (i) the expiratory duration is personalized to a given patient's lung physiology, which promotes alveolar stabilization by halting the progressive collapse of alveoli, thereby minimizing the time for the reopened lung to collapse again in the next expiration, and (ii) an extended inspiratory phase at a fixed inflation pressure after alveolar stabilization gradually reopens a small amount of tissue with each breath. Subsequently, densely collapsed regions are slowly ratcheted open over a period of hours, or even days. Thus, TCAV has the potential to minimize VILI, reducing ARDS-related morbidity and mortality.


Asunto(s)
Síndrome de Dificultad Respiratoria , Lesión Pulmonar Inducida por Ventilación Mecánica , Humanos , Respiración Artificial/métodos , Pulmón/patología , Alveolos Pulmonares/patología , Síndrome de Dificultad Respiratoria/diagnóstico , Síndrome de Dificultad Respiratoria/terapia , Síndrome de Dificultad Respiratoria/patología , Presión de las Vías Aéreas Positiva Contínua/métodos , Volumen de Ventilación Pulmonar , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control , Lesión Pulmonar Inducida por Ventilación Mecánica/patología
10.
Nan Fang Yi Ke Da Xue Xue Bao ; 43(11): 1886-1891, 2023 Nov 20.
Artículo en Chino | MEDLINE | ID: mdl-38081606

RESUMEN

OBJECTIVE: To explore the mechanism underlying the protective effect of sevoflurane against ventilator-induced lung injury (VILI). METHODS: Thirty-two SD rats were randomized into mechanical ventilation (MV) group, MV+sevoflurane group (MS group), MV+sevoflurane+transient receptor potential vanillate subtype 4 (TRPV4) agonist group (MST group) and MV+ sevoflurane + vehicle group (MSV group). Arachidonic acid (AA) in the lung tissues was quantified with ELISA. TRPV4, cytoplasmic phospholipase A2 (C-PLA2) and myosin light chain kinase (MLCK) protein expressions were detected by Western blotting. Lung injury in the rats was evaluated by assessing MLCK protein expression level, pulmonary permeability index, lung wet/dry ratio, leukocyte count in the bronchoalveolar lavage fluid (BALF), myeloperoxidase content in lung tissue, and histological score of the lungs. RESULTS: The rats in MV group showed significantly increased TRPV4 and C-PLA2 expression levels in the lung tissues with increased lung permeability and obvious lung inflammation compared with those in the other 3 groups (P < 0.05). No significant differences were found in the parameters associated with lung injuries between MS group and MSV group. Compared with those in MST group, the rats in MS group and MSV group showed significantly reduced AA production and TRPV4 and C-PLA2 expressions in the lungs (P < 0.05) with alleviated lung hyper-permeability and inflammation (P < 0.05). CONCLUSION: Sevoflurane protects against VILI in rats by down-regulating the TRPV4/C-PLA2 signaling pathway.


Asunto(s)
Canales Catiónicos TRPV , Lesión Pulmonar Inducida por Ventilación Mecánica , Ratas , Animales , Sevoflurano , Canales Catiónicos TRPV/metabolismo , Ratas Sprague-Dawley , Pulmón/patología , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/patología , Líquido del Lavado Bronquioalveolar , Fosfolipasas A2/metabolismo , Transducción de Señal
11.
Immun Inflamm Dis ; 11(10): e1062, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37904713

RESUMEN

BACKGROUND: Mechanical ventilation is an important means of respiratory support and treatment for various diseases. However, its use can lead to serious complications, especially ventilator-induced lung injury (VILI). The mechanisms underlying this disease are complex, but activation of inflammatory signalling pathways results in activation of cytokines and inflammatory mediators, which play key roles in VILI. Recent studies have demonstrated that nod-like receptor protein 3 (NLRP3) inflammasome activation mediates VILI and also accompanied by cell proliferation and transdifferentiation to compensate for alveolar membrane damage. Type I alveolar epithelial cells (AECs I), which are involved in the formation of the blood-air barrier, are vulnerable to damage but cannot proliferate by themselves; thus, replacing AECs I relies on type II alveolar epithelial cells (AECs II). OBJECTIVE: The review aims to introduce the mechanisms of NLRP3 inflammasome activation and its inhibitors, as well as the mechanisms that regulate cell proliferation and transdifferentiation. METHODS: A large number of relevant literature was searched, then the key content was summarized and figures were also made. RESULTS: The mechanism of NLRP3 inflammasome activation has been further explored, including but not limited to pathogenic and aseptic inflammatory signals, such as, pathogenic molecular patterns and host-derived danger-associated molecular patterns activate toll-like receptor 4/nuclear factor-kappaB pathway or reactive oxygen species, cyclic stretch, adenosine triphosphate induce K+ efflux through P2X7, Ca2+ inflow, mitochondrial damage, etc, eventually induce NIMA-related kinase 7/NLRP3 binding and NLRP3 inflammasome activation. Not only that, the review also described in detail the inhibitors of NLRP3 inflammasome. And the mechanisms regulating cell proliferation and transdifferentiation are complex and unclear, including the Wnt/ß-catenin, Yap/Taz, BMP/Smad and Notch signalling pathways. CONCLUSIONS: NLRP3 inflammasome activation mediated VILI, and VILI is alleviated after interfering with its activation, and inflammation and repair exist simultaneously in VILI. Clarifying these mechanisms is expected to provide theoretical guidance for alleviating VILI by inhibiting the inflammatory response and accelerating alveolar epithelial cell regeneration in the early stage.


Asunto(s)
Inflamasomas , Lesión Pulmonar Inducida por Ventilación Mecánica , Humanos , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas NLR , Transdiferenciación Celular , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/patología , Proliferación Celular
12.
Biomed Pharmacother ; 165: 115237, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37516020

RESUMEN

Mechanical ventilation (MV) may negatively affect the lungs and cause the release of inflammatory mediators, resulting in extra-pulmonary organ dysfunction. Studies have revealed systemically elevated levels of proinflammatory cytokines in animal models of ventilator-induced lung injury (VILI); however, whether these cytokines have an effect on gut injury and the mechanisms involved remain unknown. In this study, VILI was generated in mice with high tidal volume mechanical ventilation (20 ml/kg). Tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and IL-6 concentrations in serum and gut measured by ELISA showed significant elevation in the VILI mice. Significant increases in gut injury and PANoptosis were observed in the VILI mice, which were positively correlated with the serum levels of TNF-α, IL-1ß, and IL-6. The VILI mice displayed intestinal barrier defects, decreased expressions of occludin and zonula occludin-1 (ZO-1), and increased expression of claudin-2 and the activation of myosin light chain (MLC). Importantly, intratracheal administration of Imp7 siRNA nanoparticle effectively inhibited cytokines production and protected mice from VILI-induced gut injury. These data provide evidence of systemic cytokines contributing to gut injury following VILI and highlight the possibility of targeting cytokines inhibition via Imp7 siRNA nanoparticle as a potential therapeutic intervention for alleviating gut injury following VILI.


Asunto(s)
Citocinas , Lesión Pulmonar Inducida por Ventilación Mecánica , Ratones , Animales , Citocinas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-6/metabolismo , ARN Interferente Pequeño/metabolismo , Ocludina/metabolismo , Pulmón/patología , Lesión Pulmonar Inducida por Ventilación Mecánica/tratamiento farmacológico , Lesión Pulmonar Inducida por Ventilación Mecánica/patología , Ratones Endogámicos C57BL
13.
Crit Care ; 27(1): 239, 2023 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-37328874

RESUMEN

BACKGROUND: Animal models of acute respiratory distress syndrome (ARDS) do not completely resemble human ARDS, struggling translational research. We aimed to characterize a porcine model of ARDS induced by pneumonia-the most common risk factor in humans-and analyze the additional effect of ventilator-induced lung injury (VILI). METHODS: Bronchoscopy-guided instillation of a multidrug-resistant Pseudomonas aeruginosa strain was performed in ten healthy pigs. In six animals (pneumonia-with-VILI group), pulmonary damage was further increased by VILI applied 3 h before instillation and until ARDS was diagnosed by PaO2/FiO2 < 150 mmHg. Four animals (pneumonia-without-VILI group) were protectively ventilated 3 h before inoculum and thereafter. Gas exchange, respiratory mechanics, hemodynamics, microbiological studies and inflammatory markers were analyzed during the 96-h experiment. During necropsy, lobar samples were also analyzed. RESULTS: All animals from pneumonia-with-VILI group reached Berlin criteria for ARDS diagnosis until the end of experiment. The mean duration under ARDS diagnosis was 46.8 ± 7.7 h; the lowest PaO2/FiO2 was 83 ± 5.45 mmHg. The group of pigs that were not subjected to VILI did not meet ARDS criteria, even when presenting with bilateral pneumonia. Animals developing ARDS presented hemodynamic instability as well as severe hypercapnia despite high-minute ventilation. Unlike the pneumonia-without-VILI group, the ARDS animals presented lower static compliance (p = 0.011) and increased pulmonary permeability (p = 0.013). The highest burden of P. aeruginosa was found at pneumonia diagnosis in all animals, as well as a high inflammatory response shown by a release of interleukin (IL)-6 and IL-8. At histological examination, only animals comprising the pneumonia-with-VILI group presented signs consistent with diffuse alveolar damage. CONCLUSIONS: In conclusion, we established an accurate pulmonary sepsis-induced ARDS model.


Asunto(s)
Neumonía , Síndrome de Dificultad Respiratoria , Lesión Pulmonar Inducida por Ventilación Mecánica , Humanos , Porcinos , Animales , Síndrome de Dificultad Respiratoria/diagnóstico , Pulmón/patología , Neumonía/complicaciones , Lesión Pulmonar Inducida por Ventilación Mecánica/complicaciones , Lesión Pulmonar Inducida por Ventilación Mecánica/patología , Mecánica Respiratoria , Respiración Artificial/efectos adversos
14.
Int Immunopharmacol ; 120: 110356, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37244115

RESUMEN

Ventilator-induced lung injury (VILI) has become an increasingly common complication in the clinic concerning mechanical ventilation. Previous research showed that VILI is the result of a response to cascade inflammation; however, the inflammatory mechanism involved remains unclear. As a newly recognized form of cell death, ferroptosis can release damage-related molecules (DAMPs) to trigger and amplify the inflammatory response and is involved in several inflammatory diseases. The present study aimed to investigate a previously unrecognized role of ferroptosis in VILI. A mouse model of VILI and a model of cyclic stretching (CS)-induced lung epithelial cell injury were established. Mice and cells were pretreated with ferrostain-1, an inhibitor of ferroptosis. Lung tissue and cells were then harvested to determine lung injury, inflammatory responses, indicators and protein expression associated with ferroptosis. Compared to the control group, mice subjected to high tidal volumes (HTV) for 4 h showed more severe pulmonary edema and inflammation and the activation of ferroptosis. Ferrostain-1 significantly ameliorated histological injury and inflammation in the VILI mouse and alleviated CS-induced lung epithelial cell injury. Mechanistically, ferrostain-1 markedly limited the activation of ferroptosis and recovered functionality of the SLC7A11/GPX4 axis both in vitro and in vivo, thus demonstrating its potential as a novel therapeutic target for VILI.


Asunto(s)
Ferroptosis , Lesión Pulmonar Inducida por Ventilación Mecánica , Ratones , Animales , Pulmón/patología , Inflamación/tratamiento farmacológico , Lesión Pulmonar Inducida por Ventilación Mecánica/patología
15.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 35(2): 135-139, 2023 Feb.
Artículo en Chino | MEDLINE | ID: mdl-36916372

RESUMEN

OBJECTIVE: To observe the effect of ventilator-induced lung injury (VILI) on blood-brain barrier permeability in rats. METHODS: Forty-eight healthy clean male Sprague-Dawley (SD) rats were randomly divided into sham operation (Sham) group, low tidal volume (LVT) mechanical ventilation group (LVT group), normal tidal volume (NVT) mechanical ventilation group (NVT group) and high tidal volume (HVT) mechanical ventilation group (HVT group) with 12 rats in each group. After anesthesia, rats in the Sham group were intubated and kept spontaneous breathing. The rats in different tidal volume (VT) groups were mechanically ventilated by endotracheal intubation with VT of 6 mL/kg (LVT group), 10 mL/kg (NVT group), and 20 mL/kg (HVT group), respectively. The inspiration-expiration ratio of the three groups was 1:1, the ventilation frequency was 40 times/min, and the ventilation time was 3 hours. At the end of the experiment, the bronchoalveolar lavage fluid (BALF) of rats was collected, and the levels of pro-inflammatory factors [tumor necrosis factor-α (TNF-α), interleukins (IL-1ß and IL-6)] in BALF were detected by enzyme-linked immunosorbent assay (ELISA). The lung tissues of rats were collected, and the lung wet/dry weight (W/D) ratio was calculated. The pathological changes of lung tissues were observed under light microscopy after hematoxylin-eosin (HE) staining, and lung injury scores were performed. The brain tissue of rats was taken to measure the brain water content, and the Evans blue (EB) content of brain tissue was measured to reflect the permeability of the blood-brain barrier. The tight junction proteins in the brain tissues were detected by Western blotting. RESULTS: After 3 hours of mechanical ventilation, with the increase of VT, the degree of lung injury in VILI rats gradually increased. When VT reached 20 mL/kg, lung tissue structure was significantly injured, alveolar wall edema, alveolar congestion, lung interstitial thickening, a large number of inflammatory cells infiltrated, and the lung injury score, lung W/D ratio, and the levels of TNF-α, IL-1ß and IL-6 in BALF were significantly higher than those in the Sham group [lung injury score: 10.6±1.1 vs. 1.4±1.0, lung W/D ratio: 6.6±0.8 vs. 3.7±0.6, TNF-α (ng/L): 832.9±97.9 vs. 103.8±23.3, IL-1ß (ng/L): 68.9±14.1 vs. 15.7±2.6, IL-6 (ng/L): 70.8±16.4 vs. 20.3±5.4, all P < 0.05]. Lung injury in rats was accompanied by aggravating brain injury. When VT reached 20 mL/kg, brain water content and EB content in brain tissue were significantly higher than those in the Sham group [brain water content: (85.4±3.6)% vs. (68.7±2.7)%, EB content in brain tissue (µg/g): 887±78 vs. 97±14, both P < 0.05], and the protein expressions of claudin-5, occluding and zonula occluden-1 (ZO-1) in the brain tissue were significantly lower than those in the Sham group [claudin-5 protein (claudin-5/ß-actin): 0.67±0.12 vs. 1.45±0.19, occludin protein (occludin/ß-actin): 0.48±0.11 vs. 0.99±0.21, ZO-1 protein (ZO-1/ß-actin): 0.13±0.03 vs. 0.63±0.12, all P < 0.05]. CONCLUSIONS: VILI can induce brain edema and increase blood-brain barrier permeability in rats, which may be related to the down-regulation of tight junction protein expression in the brain tissue.


Asunto(s)
Factor de Necrosis Tumoral alfa , Lesión Pulmonar Inducida por Ventilación Mecánica , Ratas , Masculino , Animales , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Actinas/metabolismo , Claudina-5/metabolismo , Ocludina/metabolismo , Pulmón/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/patología
16.
Int Immunopharmacol ; 114: 109462, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36476487

RESUMEN

Ventilator-induced lung injury (VILI) is a lung injury induced or aggravated by mechanical ventilation. Transforming growth factor (TGF)-ß1 is a cytokine that mediates immune function, enabling inflammatory attenuation and tissue repair. Here, we hypothesized that it plays an important role in the attenuation of VILI and inflammation. Ventilation with high tidal volume was performed on C57BL/6 mice to establish a VILI model. After 4 h of ventilation, mice were sacrificed (end of ventilation [EOV]) or extubated for resuscitation at 4 h (post-ventilation 4 h [PV4h]), 8 h (PV8h) and 24 h post-ventilation (PV1d). Recombinant mouse TGF-ß1 (rTGF-ß1) and the neutralization antibody of TGF-ß1 (nTAb) were used in vivo to examine the effect of TGF-ß1 on immune function and inflammatory attenuation in VILI mice. Lung injury was exacerbated at the same trend as the interleukin (IL)-1ß level, peaking at PV1d, whereas IL-6 and tumor necrosis factor (TNF)-α levels gradually reduced. Most active phagosomes, swollen round mitochondria, and cavitating lamellar bodies were observed at PV4h. The CD4+ T cells were significantly increased from PV4h to PV1d, and the CD8a + T cells were higher in the PV4h and PV1d groups; furthermore, the mice in the PV8h group showed highest proportion of CD4+CD8a+ T cells and CD4+/CD8a+ ratio. CD19 + and CD5 + CD19 + B cells in VILI mice began to increase at PV1d. The pulmonary expression of latent and monomer TGF-ß1 increased at PV4h and PV8h. Treatment of rTGF-ß1 only induced high expression of latent and monomer TGF-ß1 at EOV to decrease pulmonary levels of IL-1ß, IL-6, and TNF-α; however, lung injury attenuated from EOV to PV1d. TGF-ß1 induced the delayed elevation of CD4+/CD8a+ T cells ratio and activation of pulmonary CD4+CD8a+ double-positive T cells under certain conditions. Elastic fibers and celluloses, although relatively less proteoglycan, were observed with the overexpression of TGF-ß1 at PV4h and PV8h. In conclusion, TGF-ß1 attenuates the inflammatory response and lung injury of VILI via immune function regulation.


Asunto(s)
Factor de Crecimiento Transformador beta1 , Lesión Pulmonar Inducida por Ventilación Mecánica , Ratones , Animales , Factor de Crecimiento Transformador beta1/metabolismo , Interleucina-6/metabolismo , Ratones Endogámicos C57BL , Pulmón/patología , Lesión Pulmonar Inducida por Ventilación Mecánica/patología , Inflamación/metabolismo , Inmunidad
17.
Crit Care ; 26(1): 274, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36100846

RESUMEN

Acute neuropsychiatric impairments occur in over 70% of patients with acute lung injury. Mechanical ventilation is a well-known precipitant of acute lung injury and is strongly associated with the development of acute delirium and anxiety phenotypes. In prior studies, we demonstrated that IL-6 mediates neuropathological changes in the frontal cortex and hippocampus of animals with mechanical ventilation-induced brain injury; however, the effect of systemic IL-6 inhibition on structural and functional acute neuropsychiatric phenotypes is not known. We hypothesized that a murine model of mechanical ventilation-induced acute lung injury (VILI) would induce neural injury to the amygdala and hippocampus, brain regions that are implicated in diverse neuropsychiatric conditions, and corresponding delirium- and anxiety-like functional impairments. Furthermore, we hypothesized that these structural and functional changes would reverse with systemic IL-6 inhibition. VILI was induced using high tidal volume (35 cc/kg) mechanical ventilation. Cleaved caspase-3 (CC3) expression was quantified as a neural injury marker and found to be significantly increased in the VILI group compared to spontaneously breathing or anesthetized and mechanically ventilated mice with 10 cc/kg tidal volume. VILI mice treated with systemic IL-6 inhibition had significantly reduced amygdalar and hippocampal CC3 expression compared to saline-treated animals and demonstrated amelioration in acute neuropsychiatric behaviors in open field, elevated plus maze, and Y-maze tests. Overall, these data provide evidence of a pathogenic role of systemic IL-6 in mediating structural and functional acute neuropsychiatric symptoms in VILI and provide preclinical justification to assess IL-6 inhibition as a potential intervention to ameliorate acute neuropsychiatric phenotypes following VILI.


Asunto(s)
Lesión Pulmonar Aguda , Delirio , Lesión Pulmonar Inducida por Ventilación Mecánica , Lesión Pulmonar Aguda/complicaciones , Lesión Pulmonar Aguda/tratamiento farmacológico , Animales , Delirio/complicaciones , Modelos Animales de Enfermedad , Interleucina-6 , Ratones , Fenotipo , Lesión Pulmonar Inducida por Ventilación Mecánica/patología
18.
Metabolomics ; 18(8): 66, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35925420

RESUMEN

INTRODUCTION: Nowadays,the mechanical ventilation (MV) aims to rest the respiratory muscles while providing adequate gas exchange, and it has been a part of basic life support during general anesthesia as well as in critically ill patients with and without respiratory failure. However, MV itself has the potential to cause or worsen lung injury, which is also known as ventilator-induced lung injury (VILI). Thus, the early diagnosis of VILI is of great importance for the prevention and treatment of VILI. OBJECTIVE: This study aimed to investigate the metabolomes in the lung and plasma of mice receiving mechanical ventilation (MV). METHODS: Healthy mice were randomly assigned into control group; (2) high volume tidal (HV) group (30 ml/kg); (3) low volume tidal (LV) group (6 ml/kg). After ventilation for 4 h, mice were sacrificed and the lung tissue and plasma were collected. The lung and plasma were processed for the metabolomics analysis. We also performed histopathological examination on the lung tissue. RESULTS: We detected moderate inflammatory damage with alveolar septal thickening in the HV group compared with the normal and LV groups.The metabolomics analysis results showed MV altered the metabolism which was characterized by the dysregulation of γ-amino butyric acid (GABA) system and urea cycle (desregulations in plasma and lung guanidinosuccinic acid, argininosuccinic acid, succinic acid semialdehyde and lung GABA ), Disturbance of citric acid cycle (CAC) (increased plasma glutamine and lung phosphoenol pyruvate) and redox imbalance (desregulations in plasma and/or lung ascorbic acid, chenodeoxycholic acid, uric acid, oleic acid, stearidonic acid, palmitoleic acid and docosahexaenoic acid). Moreover, the lung and plasma metabolomes were also significantly different between LV and HV groups. CONCLUSIONS: Some lung and plasma metabolites related to the GABA system and urea cycle, citric acid cycle and redox balance were significantly altered, and they may be employed for the evaluation of VILI and serve as targets in the treatment of VILI.


Asunto(s)
Metabolómica , Lesión Pulmonar Inducida por Ventilación Mecánica , Animales , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Urea/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/patología , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control , Ácido gamma-Aminobutírico/metabolismo
19.
Am J Physiol Lung Cell Mol Physiol ; 323(3): L281-L296, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35700201

RESUMEN

Supportive mechanical ventilation is a necessary lifesaving treatment for acute respiratory distress syndrome (ARDS). This intervention often leads to injury exacerbation by ventilator-induced lung injury (VILI). Patterns of injury in ARDS and VILI are recognized to be heterogeneous; however, quantification of these injury distributions remains incomplete. Developing a more detailed understanding of injury heterogeneity, particularly how it varies in space and time, can help elucidate the mechanisms of VILI pathogenesis. Ultimately, this knowledge can be used to develop protective ventilation strategies that slow disease progression. To expand existing knowledge of VILI heterogeneity, we document the spatial evolution of cellular injury distribution and leukocyte infiltration, on the micro- and macroscales, during protective and injurious mechanical ventilation. We ventilated naïve mice using either high inspiratory pressure and zero positive end-expiratory pressure ventilation or low tidal volume with positive end-expiratory pressure. Distributions of cellular injury, identified with propidium iodide staining, were microscopically analyzed at three levels of injury severity. Cellular injury initiated in diffuse, quasi-random patterns, and progressed through expansion of high-density regions of injured cells termed "injury clusters." The density profile of the expanding injury regions suggests that stress shielding occurs, protecting the already injured regions from further damage. Spatial distribution of leukocytes did not correlate with that of cellular injury or ventilation-induced changes in lung function. These results suggest that protective ventilation protocols should protect the interface between healthy and injured regions to stymie injury propagation.


Asunto(s)
Síndrome de Dificultad Respiratoria , Lesión Pulmonar Inducida por Ventilación Mecánica , Animales , Leucocitos , Ratones , Respiración con Presión Positiva/métodos , Respiración Artificial/efectos adversos , Respiración Artificial/métodos , Volumen de Ventilación Pulmonar , Lesión Pulmonar Inducida por Ventilación Mecánica/patología
20.
Chem Biol Drug Des ; 100(2): 218-229, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35434894

RESUMEN

Punicalagin is recorded to be a potent anti-inflammatory drug, while its effect on inflammation existing in ventilator-induced lung injury (VILI) requires further verification. Rats were pretreated with punicalagin, followed by VILI modeling. Lung histopathological examination was performed with hematoxylin-eosin staining accompanied by the lung injury score. The lung wet/dry (W/D) weight ratio and total bronchoalveolar lavage fluid (BALF) protein level were measured. After transfection with protease-activated receptor-2 (PAR2) overexpression plasmids, mouse alveolar epithelial MLE-12 cells were treated with punicalagin and then subjected to cyclic stretching. Punicalagin's cytotoxicity to MLE-12 cells were measured by MTT assay. The levels of inflammatory cytokines (tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-6), PAR2, NLR family pyrin domain containing-3 (NLRP3), and apoptosis-associated speck-like protein containing a CARD (ASC) in the BALF, lung tissues or cells were analyzed by enzyme-linked immune-sorbent assay (ELISA), qRT-PCR or/and western blot. Punicalagin treatment attenuated VILI-induced lung histopathological changes and counteracted VILI-induced increases in the lung injury score, W/D weight ratio and total protein level in BALF. Also, punicalagin treatment counteracted in vivo VILI/cyclic stretching-induced increases in the levels of PAR2, inflammatory cytokines, NLRP3, and ASC. PAR2 overexpression potentiated the cyclic stretching-induced effects, while punicalagin treatment revoked this PAR2 overexpression-induced potentiation effect. In turn, PAR2 overexpression partly resisted the punicalagin treatment-induced counteractive effects on the cyclic stretching-induced effects. Punicalagin suppresses inflammation in VILI through PAR2 inhibition-induced inhibition of NLRP3 inflammasome activation.


Asunto(s)
Taninos Hidrolizables , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Receptor PAR-2 , Lesión Pulmonar Inducida por Ventilación Mecánica , Animales , Citocinas/metabolismo , Taninos Hidrolizables/farmacología , Inflamasomas/metabolismo , Inflamación/tratamiento farmacológico , Pulmón/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Dominio Pirina , Ratas , Receptor PAR-2/antagonistas & inhibidores , Receptor PAR-2/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/tratamiento farmacológico , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...