Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 177
Filtrar
1.
Am J Respir Crit Care Med ; 205(5): 507-519, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34878969

RESUMO

Rationale: Alveolar and endothelial injury may be differentially associated with coronavirus disease (COVID-19) severity over time. Objectives: To describe alveolar and endothelial injury dynamics and associations with COVID-19 severity, cardiorenovascular injury, and outcomes. Methods: This single-center observational study enrolled patients with COVID-19 requiring respiratory support at emergency department presentation. More than 40 markers of alveolar (including receptor for advanced glycation endproducts [RAGE]), endothelial (including angiopoietin-2), and cardiorenovascular injury (including renin, kidney injury molecule-1, and troponin-I) were serially compared between invasively and spontaneously ventilated patients using mixed-effects repeated-measures models. Ventilatory ratios were calculated for intubated patients. Associations of biomarkers with modified World Health Organization scale at Day 28 were determined with multivariable proportional-odds regression. Measurements and Main Results: Of 225 patients, 74 (33%) received invasive ventilation at Day 0. RAGE was 1.80-fold higher in invasive ventilation patients at Day 0 (95% confidence interval [CI], 1.50-2.17) versus spontaneous ventilation, but decreased over time in all patients. Changes in alveolar markers did not correlate with changes in endothelial, cardiac, or renal injury markers. In contrast, endothelial markers were similar to lower at Day 0 for invasive ventilation versus spontaneous ventilation, but then increased over time only among intubated patients. In intubated patients, angiopoietin-2 was similar (fold difference, 1.02; 95% CI, 0.89-1.17) to nonintubated patients at Day 0 but 1.80-fold higher (95% CI, 1.56-2.06) at Day 3; cardiorenovascular injury markers showed similar patterns. Endothelial markers were not consistently associated with ventilatory ratios. Endothelial markers were more often significantly associated with 28-day outcomes than alveolar markers. Conclusions: Alveolar injury markers increase early. Endothelial injury markers increase later and are associated with cardiorenovascular injury and 28-day outcome. Alveolar and endothelial injury likely contribute at different times to disease progression in severe COVID-19.


Assuntos
Células Epiteliais Alveolares , COVID-19/fisiopatologia , Endotélio/lesões , Gravidade do Paciente , Alvéolos Pulmonares/lesões , Síndrome do Desconforto Respiratório/fisiopatologia , Adulto , Idoso , Biomarcadores/análise , Resultados de Cuidados Críticos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Sistema Renina-Angiotensina , Respiração Artificial , SARS-CoV-2
2.
Comput Math Methods Med ; 2021: 7710129, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34471421

RESUMO

OBJECTIVE: We aimed to explore the level of PS, cell viability, inflammatory factors, and apoptosis in neonatal respiratory distress syndrome (ARDS). Besides, we explored the potential relationship between ACE2, SIRT1/eNOS pathway, and hypoxia-induced AT II cell damage. METHODS: The hUC-MSC-derived AT II cells were verified by IF and ICC, whereas qRT-PCR was used for PS and AT II cell marker (CK-8 and KGF). The AT II cell damage model was established by hypoxia exposure. The enhanced expression of ACE2 was tested after transfection with pcDNA3.1-ACE2 by western blot. The effects of hypoxia and ACE2 on AT II cells were evaluated by MTT, western blot, ELISA, and flow cytometry. The involvement of the SIRT1/eNOS pathway in AT II cell's protective functions against NRDS was verified with the addition of SIRT1 inhibitor EX527. RESULTS: Based on the successful differentiation of AT II cells from hUC-MSCs and the buildup of AT II cell damage model, the overexpressed ACE2 impeded the hypoxia-induced cellular damage of AT II cells. It also counteracted the inhibitory effects of hypoxia on the secretion of PS. Overexpression of ACE2 rescued the cell viability and suppressed the secretion of inflammatory cytokines and the apoptosis of AT II cells triggered by hypoxia. And ACE2 activated the SIRT1/eNOS pathway to play its cell-protective and anti-inflammatory roles. CONCLUSION: Our findings provided information that ACE2 prevented AT II cells from inflammatory damage through activating the SIRT1/eNOS pathway, which suggested that ACE2 might become a novel protective agent applied in the protection and treatment of NRDS.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Alvéolos Pulmonares/lesões , Alvéolos Pulmonares/metabolismo , Surfactantes Pulmonares/metabolismo , Sirtuína 1/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Apoptose , Carbazóis/farmacologia , Diferenciação Celular , Hipóxia Celular , Sobrevivência Celular , Células Cultivadas , Biologia Computacional , Feminino , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Redes e Vias Metabólicas/efeitos dos fármacos , Alvéolos Pulmonares/efeitos dos fármacos , Síndrome do Desconforto Respiratório do Recém-Nascido/etiologia , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/prevenção & controle , Sirtuína 1/antagonistas & inibidores , Regulação para Cima
3.
Am J Physiol Lung Cell Mol Physiol ; 321(4): L641-L652, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34405719

RESUMO

The receptor for advanced glycation end-products (RAGE) has been implicated in the pathophysiology of chronic obstructive pulmonary disease (COPD). However, it is still unknown whether RAGE directly contributes to alveolar epithelial damage and abnormal repair responses. We hypothesize that RAGE activation not only induces lung tissue damage but also hampers alveolar epithelial repair responses. The effects of the RAGE ligands LL-37 and HMGB1 were examined on airway inflammation and alveolar tissue damage in wild-type and RAGE-deficient mice and on lung damage and repair responses using murine precision cut lung slices (PCLS) and organoids. In addition, their effects were studied on the repair response of human alveolar epithelial A549 cells, using siRNA knockdown of RAGE and treatment with the RAGE inhibitor FPS-ZM1. We observed that intranasal installation of LL-37 and HMGB1 induces RAGE-dependent inflammation and severe alveolar tissue damage in mice within 6 h, with stronger effects in a mouse strain susceptible for emphysema compared with a nonsusceptible strain. In PCLS, RAGE inhibition reduced the recovery from elastase-induced alveolar tissue damage. In organoids, RAGE ligands reduced the organoid-forming efficiency and epithelial differentiation into pneumocyte-organoids. Finally, in A549 cells, we confirmed the role of RAGE in impaired repair responses upon exposure to LL-37. Together, our data indicate that activation of RAGE by its ligands LL-37 and HMGB1 induces acute lung tissue damage and that this impedes alveolar epithelial repair, illustrating the therapeutic potential of RAGE inhibitors for lung tissue repair in emphysema.


Assuntos
Células Epiteliais Alveolares/patologia , Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteína HMGB1/metabolismo , Alvéolos Pulmonares/lesões , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Células A549 , Animais , Benzamidas/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Organoides/efeitos dos fármacos , Elastase Pancreática/toxicidade , Doença Pulmonar Obstrutiva Crônica/patologia , Receptor para Produtos Finais de Glicação Avançada/antagonistas & inibidores , Regeneração/fisiologia , Catelicidinas
4.
Innate Immun ; 27(4): 294-312, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34000873

RESUMO

In China, baicalin is the main active component of Scutellaria baicalensis, which has been used in the treatment of inflammation-related diseases, such as inflammation-induced acute lung injury. However, its specific mechanism remains unclear. This study examined the protective effect of baicalin on LPS-induced inflammation injury of alveolar epithelial cell line A549 and explored its protective mechanism. Compared with the LPS-induced group, the proliferation inhibition rates of alveolar type II epithelial cell line A549 intervened by different concentrations of baicalin decreased significantly, as did the levels of inflammatory factors IL-6, IL-1ß, prostaglandin 2 and TNF-α in the supernatant. The expression levels of inflammatory proteins inducible NO synthase (iNOS), NF-κB65, phosphorylated ERK (p-ERK1/2), and phosphorylated c-Jun N-terminal kinase (p-JNK1) significantly decreased, as did the protein expression of follistatin-like protein 1 (FSTL1). In contrast, expression of miR-200b-3p significantly increased in a dose-dependent manner. These results suggested that baicalin could significantly inhibit the expression of inflammation-related proteins and improve LPS-induced inflammatory injury in alveolar type II epithelial cells. The mechanism may be related to the inhibition of ERK/JNK inflammatory pathway activation by increasing the expression of miR-200b-3p. Thus, FSTL1 is the regulatory target of miR-200b-3p.


Assuntos
Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Flavonoides/uso terapêutico , Proteínas Relacionadas à Folistatina/efeitos dos fármacos , Lipopolissacarídeos , MicroRNAs/biossíntese , Alvéolos Pulmonares/lesões , Transdução de Sinais/efeitos dos fármacos , Células A549 , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Inflamação/induzido quimicamente , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , MicroRNAs/efeitos dos fármacos , MicroRNAs/genética , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia
6.
Am J Emerg Med ; 46: 625-627, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33257145

RESUMO

Envenoming syndrome induced by massive Vespa basalis stings is a critical condition. Severe systemic reaction may present with hemolytic activity and rhabdomyolysis, leading diffuse alveolar hemorrhage, adult respiratory distress syndrome, coagulopathy, and multiple organs failure. In severe envenoming syndrome population, extracorporeal membrane oxygenation (ECMO) may be considered for unstable hemodynamic status. However, few studies reported ECMO in venom-induced disseminated intravascular coagulation patients. Here, we provide a case presented with pulmonary hemorrhage due to multiple Vespa basalis stings tried to rescue by veno-arterial extracorporeal membrane oxygenation. We also highlight that early recognition of venom-induced disseminated intravascular coagulation by checking coagulation profile in high risk patients may prevent from poor outcome.


Assuntos
Hemorragia/etiologia , Rabdomiólise/etiologia , Venenos de Vespas/efeitos adversos , Idoso , Hemolíticos , Hemorragia/fisiopatologia , Humanos , Masculino , Alvéolos Pulmonares/lesões , Alvéolos Pulmonares/fisiopatologia , Rabdomiólise/fisiopatologia , Venenos de Vespas/uso terapêutico
7.
Sci Rep ; 10(1): 5499, 2020 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-32218519

RESUMO

Research on acute and chronic lung diseases would greatly benefit from reproducible availability of alveolar epithelial cells (AEC). Primary alveolar epithelial cells can be derived from human lung tissue but the quality of these cells is highly donor dependent. Here, we demonstrated that culture of EpCAM+ cells derived from human induced pluripotent stem cells (hiPSC) at the physiological air-liquid interface (ALI) resulted in type 2 AEC-like cells (iAEC2) with alveolar characteristics. iAEC2 cells expressed native AEC2 markers (surfactant proteins and LPCAT-1) and contained lamellar bodies. ALI-iAEC2 were used to study alveolar repair over a period of 2 weeks following mechanical wounding of the cultures and the responses were compared with those obtained using primary AEC2 (pAEC2) isolated from resected lung tissue. Addition of the Wnt/ß-catenin activator CHIR99021 reduced wound closure in the iAEC2 cultures but not pAEC2 cultures. This was accompanied by decreased surfactant protein expression and accumulation of podoplanin-positive cells at the wound edge. These results demonstrated the feasibility of studying alveolar repair using hiPSC-AEC2 cultured at the ALI and indicated that this model can be used in the future to study modulation of alveolar repair by (pharmaceutical) compounds.


Assuntos
Células Epiteliais Alveolares/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Modelos Biológicos , Células Epiteliais Alveolares/citologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Células Cultivadas , Humanos , Técnicas In Vitro , Células-Tronco Pluripotentes Induzidas/citologia , Alvéolos Pulmonares/lesões , Alvéolos Pulmonares/fisiologia , Alvéolos Pulmonares/fisiopatologia , Regeneração/fisiologia , Cicatrização/fisiologia
9.
Crit Care ; 23(1): 283, 2019 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-31426849

RESUMO

BACKGROUND: Altered coagulation and alveolar injury are the hallmarks of acute respiratory distress syndrome (ARDS). However, whether the biomarkers that reflect pathophysiology differ depending on the etiology of ARDS has not been examined. This study aimed to investigate the biomarker profiles of coagulopathy and alveolar epithelial injury in two subtypes of ARDS: patients with direct common risk factors (dARDS) and those with idiopathic or immune-related diseases (iARDS), which are classified as "ARDS without common risk factors" based on the Berlin definition. METHODS: This retrospective, observational study included adult patients who were admitted to the intensive care unit (ICU) at a university hospital with a diagnosis of ARDS with no indirect risk factors. Plasma biomarkers (thrombin-antithrombin complex [TAT], plasminogen activator inhibitor [PAI]-1, protein C [PC] activity, procalcitonin [PCT], surfactant protein [SP]-D, and KL-6) were routinely measured during the first 5 days of the patient's ICU stay. RESULTS: Among 138 eligible patients with ARDS, 51 were excluded based on the exclusion criteria (n = 41) or other causes of ARDS (n = 10). Of the remaining 87 patients, 56 were identified as having dARDS and 31 as having iARDS. Among the iARDS patients, TAT (marker of thrombin generation) and PAI-1 (marker of inhibited fibrinolysis) were increased, and PC activity was above normal. In contrast, PC activity was significantly decreased, and TAT or PAI-1 was present at much higher levels in dARDS compared with iARDS patients. Significant differences were also observed in PCT, SP-D, and KL-6 between patients with dARDS and iARDS. The receiver operating characteristic (ROC) analysis showed that areas under the ROC curve for PC activity, PAI-1, PCT, SP-D, and KL-6 were similarly high for distinguishing between dARDS and iARDS (PC 0.86, P = 0.33; PAI-1 0.89, P = 0.95; PCT 0.89, P = 0.66; and SP-D 0.88, P = 0.16 vs. KL-6 0.90, respectively). CONCLUSIONS: Coagulopathy and alveolar epithelial injury were observed in both patients with dARDS and with iARDS. However, their biomarker profiles were significantly different between the two groups. The different patterns of PAI-1, PC activity, SP-D, and KL-6 may help in differentiating between these ARDS subtypes.


Assuntos
Biomarcadores/análise , Alvéolos Pulmonares/lesões , Síndrome do Desconforto Respiratório/sangue , Idoso , Biomarcadores/sangue , Feminino , Humanos , Japão , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Alvéolos Pulmonares/fisiopatologia , Síndrome do Desconforto Respiratório/fisiopatologia , Estudos Retrospectivos , Fatores de Risco
10.
J Clin Invest ; 129(6): 2514-2526, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30946031

RESUMO

The lung is a specialized barrier organ that must tightly regulate interstitial fluid clearance and prevent infection in order to maintain effective gas exchange. Lymphatic vessels are important for these functions in other organs, but their roles in the lung have not been fully defined. In the present study, we addressed how the lymphatic vasculature participates in lung homeostasis. Studies using mice carrying a lymphatic reporter allele revealeded that, in contrast to other organs, lung lymphatic collecting vessels lack smooth muscle cells entirely, suggesting that forward lymph flow is highly dependent on movement and changes in pressure associated with respiration. Functional studies using CLEC2-deficient mice in which lymph flow is impaired due to loss of lympho-venous hemostasis or using inducible lung-specific ablation of lymphatic endothelial cells in a lung transplant model revealeded that loss of lymphatic function leads to an inflammatory state characterized by the formation of tertiary lymphoid organs (TLOs). In addition, impaired lymphatic flow in mice resulteds in hypoxia and features of lung injury that resemble emphysema. These findings reveal both a lung-specific mechanism of lymphatic physiology and a lung-specific consequence of lymphatic dysfunction that may contribute to chronic lung diseases that arise in association with TLO formation.


Assuntos
Células Endoteliais , Lectinas Tipo C/deficiência , Alvéolos Pulmonares , Enfisema Pulmonar , Estruturas Linfoides Terciárias , Animais , Células Endoteliais/imunologia , Células Endoteliais/patologia , Lectinas Tipo C/imunologia , Camundongos , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/lesões , Alvéolos Pulmonares/patologia , Enfisema Pulmonar/genética , Enfisema Pulmonar/imunologia , Enfisema Pulmonar/patologia , Estruturas Linfoides Terciárias/imunologia , Estruturas Linfoides Terciárias/patologia
12.
Arch. bronconeumol. (Ed. impr.) ; 55(1): 31-37, ene. 2019. ilus, tab
Artigo em Inglês | IBECS | ID: ibc-175189

RESUMO

Acute respiratory distress syndrome (ARDS) is a frequent and life-threatening entity. Recently, it has been demonstrated that diffuse alveolar damage (DAD), which is considered the histological hallmark in spite of presenting itself in only half of living patients with ARDS, exerts a relevant effect in the ARDS outcome. Despite the fact that the bronchial tree constitutes approximately 1% of the lung volume, discovering a relation between DAD and bronchial tree findings could be of paramount importance for a few reasons; (a) it could improve the description of ARDS with DAD as a clinical-pathological entity, (b) it could subrogate DAD findings with the advantage of their more accessible and safer analysis and (c) it could allow the discovery of new therapeutic targets. This narrative review is focused on pathological airway changes associated to Diffuse Alveolar Damage in the context of Acute Respiratory Distress Syndrome. It is organized into five sections: main anatomical and functional features of the human airway, why it is necessary to study airway features associated to DAD in patients with ARDS, pathological airway changes associated with DAD in animal models of ARDS, pathological airway changes associated with DAD in patients with ARDS, and the newest techniques for studying the histology of the bronchial tree and lung parenchyma


El síndrome de distrés respiratorio agudo (SDRA) es una patología frecuente con riesgo asociado para la vida de los pacientes. Recientemente, se ha demostrado que el daño alveolar difuso (DAD), considerado como la manifestación histológica característica del síndrome, además de presentarse en la mitad de los pacientes que padecen SDRA, posee un papel relevante en la evolución de este. A pesar del hecho de que el árbol bronquial constituye aproximadamente un 1% del volumen pulmonar, el descubrimiento de una asociación entre el DAD y hallazgos en el árbol bronquial podría ser muy importante por algunas razones: (a) podría mejorar el diagnóstico del SDRA con DAD como entidad clínico-patológica, (b) podría subrogar marcadores de DAD, cuyo análisis sea más seguro y accesible, (c) podría favorecer el descubrimiento de nuevas dianas terapéuticas. Esta revisión narrativa se centra en las alteraciones patológicas de las vías respiratorias asociadas al daño alveolar difuso en el contexto del síndrome de distrés respiratorio agudo. El texto se compone de cinco secciones: principales características anatómicas y funcionales de las vías respiratorias en humanos, por qué es necesario estudiar las características de las vías respiratorias asociadas al daño alveolar difuso en los pacientes con síndrome de distrés respiratorio agudo, alteraciones patológicas de las vías respiratorias asociadas con DAD en modelos animales de SDRA, alteraciones patológicas de las vías respiratorias asociadas con DAD en pacientes con SDRA y nuevas técnicas para el estudio histológico del árbol bronquial y el parénquima pulmonar


Assuntos
Humanos , Síndrome do Desconforto Respiratório/complicações , Alvéolos Pulmonares/lesões , Síndrome do Desconforto Respiratório/fisiopatologia , Alvéolos Pulmonares/fisiopatologia
14.
Immunopharmacol Immunotoxicol ; 41(3): 370-379, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30442050

RESUMO

Lung cancer continues to be the leading cause of cancer deaths and more than one million lung cancer patients will die every year worldwide. Radiotherapy (RT) plays an important role in lung cancer treatment, but the side effects of RT are pneumonitis and pulmonary fibrosis. RT-induced lung injury causes damage to alveolar-epithelial cells and vascular endothelial cells. Macrophages play an important role in the development of pulmonary fibrosis despite its role in immune response. These injury activated macrophages develop into classically activated M1 macrophage or alternative activated M2 macrophage. It secretes cytokines, interleukins, interferons, and nitric oxide. Several pro-inflammatory lipids and pro-apoptotic proteins cause lipotoxicity such as LDL, FC, DAG, and FFA. The overall findings in this review conclude the importance of macrophages in inducing toxic/inflammatory effects during RT of lung cancer, which is clinically vital to treat the radiation-induced fibrosis.


Assuntos
Metabolismo dos Lipídeos , Neoplasias Pulmonares , Macrófagos Alveolares , Alvéolos Pulmonares , Fibrose Pulmonar , Pneumonite por Radiação , Animais , Citocinas/imunologia , Humanos , Metabolismo dos Lipídeos/imunologia , Metabolismo dos Lipídeos/efeitos da radiação , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/radioterapia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/patologia , Óxido Nítrico/imunologia , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/lesões , Alvéolos Pulmonares/patologia , Fibrose Pulmonar/imunologia , Fibrose Pulmonar/patologia , Pneumonite por Radiação/imunologia , Pneumonite por Radiação/patologia , Radioterapia/efeitos adversos
16.
Am J Respir Cell Mol Biol ; 59(5): 635-647, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29958015

RESUMO

Studies showed that TRIM72 is essential for repair of alveolar cell membrane disruptions, and exogenous recombinant human TRIM72 protein (rhT72) demonstrated tissue-mending properties in animal models of tissue injury. Here we examine the mechanisms of rhT72-mediated lung cell protection in vitro and test the efficacy of inhaled rhT72 in reducing tissue pathology in a mouse model of ventilator-induced lung injury. In vitro lung cell injury was induced by glass beads and stretching. Ventilator-induced lung injury was modeled by injurious ventilation at 30 ml/kg tidal volume. Affinity-purified rhT72 or control proteins were added into culture medium or applied through nebulization. Cellular uptake and in vivo distribution of rhT72 were detected by imaging and immunostaining. Exogenous rhT72 maintains membrane integrity of alveolar epithelial cells subjected to glass bead injury in a dose-dependent manner. Inhaled rhT72 decreases the number of fatally injured alveolar cells, and ameliorates tissue-damaging indicators and cell injury markers after injurious ventilation. Using in vitro stretching assays, we reveal that rhT72 improves both cellular resilience to membrane wounding and membrane repair after injury. Image analysis detected rhT72 uptake by rat alveolar epithelial cells, which can be inhibited by a cholesterol-disrupting agent. In addition, inhaled rhT72 distributes to the distal lungs, where it colocalizes with phosphatidylserine detection on nonpermeabilized lung slices to label wounded cells. In conclusion, our study showed that inhaled rhT72 accumulates in injured lungs and protects lung tissue from ventilator injury, the mechanisms of which include improving cell resilience to membrane wounding, localizing to injured membrane, and augmenting membrane repair.


Assuntos
Proteínas de Transporte/administração & dosagem , Alvéolos Pulmonares/metabolismo , Proteínas Recombinantes/administração & dosagem , Respiração Artificial/efeitos adversos , Lesão Pulmonar Induzida por Ventilação Mecânica/prevenção & controle , Cicatrização , Administração por Inalação , Animais , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Proteínas de Membrana , Camundongos , Alvéolos Pulmonares/lesões , Alvéolos Pulmonares/patologia , Ratos , Lesão Pulmonar Induzida por Ventilação Mecânica/etiologia , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia
17.
Exp Biol Med (Maywood) ; 243(5): 395-407, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29402133

RESUMO

Plasmodium falciparum, the most virulent malaria parasite species, causes severe symptoms especially acute lung injury (ALI), of which characterized by alveolar epithelium and endothelium destruction and accelerated to blood-gas-barrier breakdown. Parasitized erythrocytes, endothelial cells, monocytes, and cytokines are all involved in this mechanism, but hemozoin (HZ), the parasitic waste from heme detoxification, also mainly contributes. In addition, it is not clear why type II pneumocyte proliferation, alveolar restorative stage, is rare in malaria-associated ALI. To address this, in vitro culture of A549 cells with Plasmodium HZ or with interleukin (IL)-1ß triggered by HZ and monocytes (HZ-IL-1ß) was conducted to determine their alveolar apoptotic effect using ethidium bromide/acridine orange staining, annexin-V-FITC/propidium iodide staining, and electron mircroscopic study. Caspase recruitment domain-containing protein 9 ( CARD9), the apoptotic regulator gene, and IL-1ß were quantified by reverse-transcriptase PCR. Junctional cellular defects were characterized by immunohistochemical staining of E-cadherin. The results revealed that cellular apoptosis and CARD9 expression levels were extremely high 24 h after induction by HZ-IL-1ß when compared to the HZ- and non-treated groups. E-cadherin was markedly down-regulated by HZ-IL-1ß and HZ treatments. CARD9 expression was positively correlated with IL-1ß expression and the number of apoptotic cells. Interestingly, the localization of HZ in the vesicular surfactant of apoptotic pneumocyte was also identified and submitted to be a cause of alveolar resolution abnormality. Thus, HZ triggers monocytes to produce IL-1ß and induces pneumocyte type II apoptosis through CARD9 pathway in association with down-regulated E-cadherin, which probably impairs alveolar resolution in malaria-associated ALI. Impact statement The present work shows the physical and immunomodulatory properties of hemozoin on the induction of pneumocyte apoptosis in relation to IL-1ß production through the CARD9 pathway. This occurrence may be a possible pathway for the retardation of lung resolution leading to blood-gas-barrier breakdown. Our findings lead to the understanding of the host-parasite relationship focusing on the dysfunction in ALI induced by HZ, a possible pathway of the recovering lung epithelial retardation in malaria-associated ARDS.


Assuntos
Lesão Pulmonar Aguda/parasitologia , Células Epiteliais Alveolares/patologia , Apoptose/fisiologia , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Hemeproteínas/metabolismo , Interleucina-1beta/metabolismo , Alvéolos Pulmonares/lesões , Células A549 , Lesão Pulmonar Aguda/patologia , Células Epiteliais Alveolares/parasitologia , Antígenos CD/biossíntese , Caderinas/biossíntese , Linhagem Celular Tumoral , Humanos , Pulmão/metabolismo , Malária Falciparum/patologia , Plasmodium falciparum/patogenicidade , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/parasitologia , Células THP-1
18.
Crit Care ; 21(1): 254, 2017 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-29052522

RESUMO

BACKGROUND: Although diffuse alveolar damage (DAD) is considered the typical histological pattern of acute respiratory distress syndrome (ARDS), only half of patients exhibit this morphological hallmark. Patients with DAD may have higher mortality than those without DAD. Therefore, we aimed to identify the factors associated with DAD in patients with ARDS. METHODS: We analyzed autopsy samples of 356 patients who had ARDS at the time of death. DAD was assessed by two pathologists, and ARDS criteria were evaluated by two intensivists. Criteria for severe ARDS included the degree of hypoxemia and the ancillary variables of the current Berlin definition assessed within 48 h before death: radiographic severity, high positive end-expiratory pressure (PEEP) level, and physiological variables (i.e., altered respiratory system compliance and large anatomic dead space). RESULTS: After multivariable analysis, high PEEP levels, physiological variables, and opacities involving only three quadrants on chest radiographs were not associated with DAD. The four markers independently associated with DAD were (1) duration of evolution (OR 3.29 [1.95-5.55] for patients with ARDS ≥ 3 days, p < 0.001), (2) degree of hypoxemia (OR 3.92 [1.48-10.3] for moderate ARDS and 6.18 [2.34-16.3] for severe ARDS, p < 0.01 for both), (3) increased dynamic driving pressure (OR 1.06 [1.04-1.09], p = 0.007), and (4) radiographic severity (OR 2.91 [1.47-5.75] for patients with diffuse opacities involving the four quadrants, p = 0.002). DAD was found in two-thirds of patients with a ratio of partial pressure of arterial oxygen to fraction of inspired oxygen ≤ 100 mmHg and opacities involving the four quadrants. CONCLUSIONS: In addition to severe hypoxemia, diffuse opacities involving the four quadrants were a strong marker of DAD.


Assuntos
Alvéolos Pulmonares/lesões , Síndrome do Desconforto Respiratório/fisiopatologia , Idoso , Idoso de 80 Anos ou mais , Autopsia/métodos , Feminino , Técnicas Histológicas/métodos , Humanos , Hipóxia/etiologia , Hipóxia/fisiopatologia , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Respiração com Pressão Positiva/métodos , Respiração com Pressão Positiva/normas , Alvéolos Pulmonares/fisiopatologia , Síndrome do Desconforto Respiratório/patologia , Estudos Retrospectivos , Espanha , Estatísticas não Paramétricas
19.
Cell Rep ; 19(2): 246-254, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28402849

RESUMO

There is evidence that certain club cells (CCs) in the murine airways associated with neuroepithelial bodies (NEBs) and terminal bronchioles are resistant to the xenobiotic naphthalene (Nap) and repopulate the airways after Nap injury. The identity and significance of these progenitors (variant CCs, v-CCs) have remained elusive. A recent screen for CC markers identified rare Uroplakin3a (Upk3a)-expressing cells (U-CCs) with a v-CC-like distribution. Here, we employ lineage analysis in the uninjured and chemically injured lungs to investigate the role of U-CCs as epithelial progenitors. U-CCs proliferate and generate CCs and ciliated cells in uninjured airways long-term and, like v-CCs, after Nap. U-CCs have a higher propensity to generate ciliated cells than non-U-CCs. Although U-CCs do not contribute to alveolar maintenance long-term, they generate alveolar type I and type II cells after Bleomycin (Bleo)-induced alveolar injury. Finally, we report that Upk3a+ cells exist in the NEB microenvironment of the human lung and are aberrantly expanded in conditions associated with neuroendocrine hyperplasias.


Assuntos
Bronquíolos/metabolismo , Microambiente Celular/genética , Células-Tronco/metabolismo , Uroplaquina III/biossíntese , Animais , Bleomicina/toxicidade , Bronquíolos/efeitos dos fármacos , Bronquíolos/lesões , Linhagem da Célula/efeitos dos fármacos , Microambiente Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Camundongos , Naftalenos/toxicidade , Corpos Neuroepiteliais/metabolismo , Corpos Neuroepiteliais/patologia , Alvéolos Pulmonares/lesões , Células-Tronco/efeitos dos fármacos , Uroplaquina III/genética , Cicatrização
20.
J Appl Physiol (1985) ; 122(4): 739-751, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-27979983

RESUMO

Edematous lungs contain regions with heterogeneous alveolar flooding. Liquid is trapped in flooded alveoli by a pressure barrier-higher liquid pressure at the border than in the center of flooded alveoli-that is proportional to surface tension, T Stress is concentrated between aerated and flooded alveoli, to a degree proportional to T Mechanical ventilation, by cyclically increasing T, injuriously exacerbates stress concentrations. Overcoming the pressure barrier to redistribute liquid more homogeneously between alveoli should reduce stress concentration prevalence and ventilation injury. In isolated rat lungs, we test whether accelerated deflation can overcome the pressure barrier and catapult liquid out of flooded alveoli. We generate a local edema model with normal T by microinfusing liquid into surface alveoli. We generate a global edema model with high T by establishing hydrostatic edema, which does not alter T, and then gently ventilating the edematous lungs, which increases T at 15 cmH2O transpulmonary pressure by 52%. Thus ventilation of globally edematous lungs increases T, which should increase stress concentrations and, with positive feedback, cause escalating ventilation injury. In the local model, when the pressure barrier is moderate, accelerated deflation causes liquid to escape from flooded alveoli and redistribute more equitably. Flooding heterogeneity tends to decrease. In the global model, accelerated deflation causes liquid escape, but-because of elevated T-the liquid jumps to nearby, aerated alveoli. Flooding heterogeneity is unaltered. In pulmonary edema with normal T, early ventilation with accelerated deflation might reduce the positive feedback mechanism through which ventilation injury increases over time.NEW & NOTEWORTHY We introduce, in the isolated rat lung, a new model of pulmonary edema with elevated surface tension. We first generate hydrostatic edema and then ventilate gently to increase surface tension. We investigate the mechanical mechanisms through which 1) ventilation injures edematous lungs and 2) ventilation with accelerated deflation might lessen ventilation injury.


Assuntos
Alvéolos Pulmonares/fisiopatologia , Edema Pulmonar/fisiopatologia , Animais , Modelos Animais de Doenças , Masculino , Pressão , Alvéolos Pulmonares/lesões , Ventilação Pulmonar/fisiologia , Ratos , Ratos Sprague-Dawley , Respiração , Mecânica Respiratória/fisiologia , Tensão Superficial
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...