Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nature ; 628(8009): 835-843, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38600381

RESUMEN

Severe influenza A virus (IAV) infections can result in hyper-inflammation, lung injury and acute respiratory distress syndrome1-5 (ARDS), for which there are no effective pharmacological therapies. Necroptosis is an attractive entry point for therapeutic intervention in ARDS and related inflammatory conditions because it drives pathogenic lung inflammation and lethality during severe IAV infection6-8 and can potentially be targeted by receptor interacting protein kinase 3 (RIPK3) inhibitors. Here we show that a newly developed RIPK3 inhibitor, UH15-38, potently and selectively blocked IAV-triggered necroptosis in alveolar epithelial cells in vivo. UH15-38 ameliorated lung inflammation and prevented mortality following infection with laboratory-adapted and pandemic strains of IAV, without compromising antiviral adaptive immune responses or impeding viral clearance. UH15-38 displayed robust therapeutic efficacy even when administered late in the course of infection, suggesting that RIPK3 blockade may provide clinical benefit in patients with IAV-driven ARDS and other hyper-inflammatory pathologies.


Asunto(s)
Lesión Pulmonar , Necroptosis , Infecciones por Orthomyxoviridae , Inhibidores de Proteínas Quinasas , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Animales , Femenino , Humanos , Masculino , Ratones , Células Epiteliales Alveolares/patología , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/virología , Células Epiteliales Alveolares/metabolismo , Virus de la Influenza A/clasificación , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/inmunología , Virus de la Influenza A/patogenicidad , Lesión Pulmonar/complicaciones , Lesión Pulmonar/patología , Lesión Pulmonar/prevención & control , Lesión Pulmonar/virología , Ratones Endogámicos C57BL , Necroptosis/efectos de los fármacos , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/mortalidad , Infecciones por Orthomyxoviridae/virología , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Síndrome de Dificultad Respiratoria/complicaciones , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/prevención & control , Síndrome de Dificultad Respiratoria/virología
2.
Int J Mol Sci ; 25(2)2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-38256231

RESUMEN

The pandemic of coronavirus disease 2019 (COVID-19) has been the foremost modern global public health challenge. The airway is the primary target in severe acute respiratory distress syndrome coronavirus 2 (SARS-CoV-2) infection, with substantial cell death and lung injury being signature hallmarks of exposure. The viral factors that contribute to cell death and lung injury remain incompletely understood. Thus, this study investigated the role of open reading frame 7b (Orf7b), an accessory protein of the virus, in causing lung injury. In screening viral proteins, we identified Orf7b as one of the major viral factors that mediates lung epithelial cell death. Overexpression of Orf7b leads to apoptosis and ferroptosis in lung epithelial cells, and inhibitors of apoptosis and ferroptosis ablate Orf7b-induced cell death. Orf7b upregulates the transcription regulator, c-Myc, which is integral in the activation of lung cell death pathways. Depletion of c-Myc alleviates both apoptotic and ferroptotic cell deaths and lung injury in mouse models. Our study suggests a major role of Orf7b in the cell death and lung injury attributable to COVID-19 exposure, supporting it as a potential therapeutic target.


Asunto(s)
COVID-19 , Ferroptosis , Lesión Pulmonar , Proteínas Virales , Animales , Ratones , Apoptosis , Lesión Pulmonar/virología , Sistemas de Lectura Abierta , SARS-CoV-2 , Proteínas Virales/genética
3.
Ter Arkh ; 96(3): 218-227, 2024 Apr 16.
Artículo en Ruso | MEDLINE | ID: mdl-38713035

RESUMEN

AIM: To study the clinical and histological profile of lung tissue in patients with persistent pulmonary disease, respiratory symptoms and CT findings after SARS-CoV-2 infection. MATERIALS AND METHODS: The study included 15 patients (7 females and 8 males) with a mean age of 57.7 years. All patients underwent laboratory tests, chest computed tomography, echocardiography, and pulmonary function tests. Pulmonary tissue and bronchoalveolar lavage samples were obtained by fibrobronchoscopy, transbronchial forceps (2 patients), and lung cryobiopsy (11 patients); open biopsy was performed in 2 patients. Cellular composition, herpesvirus DNA, SARS-CoV-2, Mycobacterium tuberculosis complex, galactomannan optical density index, and bacterial and fungal microflora growth were determined in bronchoalveolar lavage. SARS-CoV-2 was also identified in samples from the nasal mucosa, throat and feces using a polymerase chain reaction. RESULTS: The results showed no true pulmonary fibrosis in patients recovered from SARS-CoV-2 infection with persistent respiratory symptoms, functional impairment, and CT findings after SARS-CoV-2 infection. The observed changes comply with the current and/or resolving infection and inflammatory process. CONCLUSION: Thus, no true pulmonary fibrosis was found in patients after SARS-CoV-2 infection with persistent respiratory symptoms, functional impairment, and CT findings. The observed changes comply with the current and/or resolving infection and inflammatory process.


Asunto(s)
COVID-19 , SARS-CoV-2 , Tomografía Computarizada por Rayos X , Humanos , COVID-19/diagnóstico , COVID-19/complicaciones , Masculino , Femenino , Persona de Mediana Edad , Tomografía Computarizada por Rayos X/métodos , Pulmón/diagnóstico por imagen , Pulmón/patología , Lesión Pulmonar/virología , Lesión Pulmonar/etiología , Lesión Pulmonar/diagnóstico , Pruebas de Función Respiratoria/métodos
4.
Physiology (Bethesda) ; 37(2): 88-100, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34698589

RESUMEN

The lung is the major target organ of SARS-CoV-2 infection, which causes COVID-19. Here, we outline the multistep mechanisms of lung epithelial and endothelial injury induced by SARS-CoV-2: direct viral infection, chemokine/cytokine-mediated damage, and immune cell-mediated lung injury. Finally, we discuss the recent progress in terms of antiviral therapeutics as well as the development of anti-inflammatory or immunomodulatory therapeutic approaches. This review also provides a systematic overview of the models for studying SARS-CoV-2 infection and discusses how an understanding of mechanisms of lung injury will help identify potential targets for future drug development to mitigate lung injury.


Asunto(s)
COVID-19 , Lesión Pulmonar , Antivirales/uso terapéutico , COVID-19/complicaciones , Humanos , Pulmón , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/virología , SARS-CoV-2
5.
Clin Microbiol Rev ; 34(3)2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-33789928

RESUMEN

Several viruses target the human respiratory tract, causing different clinical manifestations spanning from mild upper airway involvement to life-threatening acute respiratory distress syndrome (ARDS). As dramatically evident in the ongoing SARS-CoV-2 pandemic, the clinical picture is not always easily predictable due to the combined effect of direct viral and indirect patient-specific immune-mediated damage. In this review, we discuss the main RNA (orthomyxoviruses, paramyxoviruses, and coronaviruses) and DNA (adenoviruses, herpesviruses, and bocaviruses) viruses with respiratory tropism and their mechanisms of direct and indirect cell damage. We analyze the thin line existing between a protective immune response, capable of limiting viral replication, and an unbalanced, dysregulated immune activation often leading to the most severe complication. Our comprehension of the molecular mechanisms involved is increasing and this should pave the way for the development and clinical use of new tailored immune-based antiviral strategies.


Asunto(s)
Virus ADN , Lesión Pulmonar , Virus ARN , Infecciones del Sistema Respiratorio , Virosis , Adulto , Anciano , Antivirales/uso terapéutico , COVID-19 , Niño , Preescolar , Femenino , Humanos , Factores Inmunológicos/uso terapéutico , Lactante , Recién Nacido , Interferones/uso terapéutico , Pulmón/inmunología , Pulmón/virología , Lesión Pulmonar/diagnóstico , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/inmunología , Lesión Pulmonar/virología , Masculino , Persona de Mediana Edad , Pandemias , SARS-CoV-2
6.
Eur J Clin Invest ; 52(9): e13827, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35753029

RESUMEN

BACKGROUND: COVID-19 global pandemic started in late 2019 with the first wave. In this cross-sectional and observational study, we evaluated the associations between the biomarkers, COVID-19 pneumonia severity and 1-year mortality. METHODS: A sample of 276 polymerase chain reaction (PCR)-positive patients for SARS-CoV-2 was included. Computerized tomography severity score (CT-SS) was used to assess the severity of COVID-19 pneumonia in 222 cases. Multivariate analyses were performed to find the predictors of CT-SS, severe CT-SS (≥20) and 1-year mortality. Biomarkers of ferritin, high-sensitive C-reactive protein (CRP), lactate dehydrogenase (LDH), cardiac troponin (cTn), neutrophil-to-lymphocyte ratio (NLR), uric acid (UA) and d-dimer were routinely measured. RESULTS: Severe CT-SS (>20) was observed in 86 (31.2%) cases. Mortality was observed in 75 (27.2%) patients at 1 year. LDH displayed the highest predictive accuracy for severe CT-SS (AUC 0.741, sensitivity = 81% and specificity = 68%, cut-off value: 360 mg/dl). Linear regression analysis displayed that LDH predicted CT-SS [B = 11 (95% CI for B = 5-17, p < .001)]. Age was the most significant parameter that was associated with severe CT-SS (OR 0.96, 95% CI 0.92-0.99, p = .015). d-dimer was the only biomarker that predicted with 1-year mortality (OR 1.62, 95% CI 1.08-2.42, p = .020). CONCLUSION: LDH is a sensitive and specific biomarker to determine patients with severe lung injury in COVID-19. d-dimer is the only biomarker that predicts 1-year mortality. Neither LDH nor CT-SS is associated with 1-year mortality.


Asunto(s)
COVID-19 , Lesión Pulmonar , Biomarcadores/sangre , COVID-19/diagnóstico , COVID-19/mortalidad , Estudios Transversales , Productos de Degradación de Fibrina-Fibrinógeno/análisis , Humanos , L-Lactato Deshidrogenasa/sangre , Lesión Pulmonar/virología , Estudios Retrospectivos , SARS-CoV-2 , Índice de Severidad de la Enfermedad
7.
Exp Physiol ; 107(7): 683-693, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34541721

RESUMEN

NEW FINDINGS: What is the topic of this review? This review presents the fundamental concepts of respiratory physiology and pathophysiology, with particular reference to lung mechanics and the pulmonary phenotype associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and subsequent coronavirus disease 2019 (COVID-19) pneumonia. What advances does it highlight? The review provides a critical summary of the main physiological aspects to be considered for safe and effective mechanical ventilation in patients with severe COVID-19 in the intensive care unit. ABSTRACT: Severe respiratory failure from coronavirus disease 2019 (COVID-19) pneumonia not responding to non-invasive respiratory support requires mechanical ventilation. Although ventilation can be a life-saving therapy, it can cause further lung injury if airway pressure and flow and their timing are not tailored to the respiratory system mechanics of the individual patient. The pathophysiology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can lead to a pattern of lung injury in patients with severe COVID-19 pneumonia typically associated with two distinct phenotypes, along a temporal and pathophysiological continuum, characterized by different levels of elastance, ventilation-to-perfusion ratio, right-to-left shunt, lung weight and recruitability. Understanding the underlying pathophysiology, duration of symptoms, radiological characteristics and lung mechanics at the individual patient level is crucial for the appropriate choice of mechanical ventilation settings to optimize gas exchange and prevent further lung injury. By critical analysis of the literature, we propose fundamental physiological and mechanical criteria for the selection of ventilation settings for COVID-19 patients in intensive care units. In particular, the choice of tidal volume should be based on obtaining a driving pressure < 14 cmH2 O, ensuring the avoidance of hypoventilation in patients with preserved compliance and of excessive strain in patients with smaller lung volumes and lower lung compliance. The level of positive end-expiratory pressure (PEEP) should be informed by the measurement of the potential for lung recruitability, where patients with greater recruitability potential may benefit from higher PEEP levels. Prone positioning is often beneficial and should be considered early. The rationale for the proposed mechanical ventilation settings criteria is presented and discussed.


Asunto(s)
COVID-19/terapia , Lesión Pulmonar/virología , Respiración Artificial , Síndrome de Dificultad Respiratoria/virología , SARS-CoV-2 , COVID-19/fisiopatología , Humanos , Unidades de Cuidados Intensivos/normas , Lesión Pulmonar/terapia , Respiración Artificial/efectos adversos , Respiración Artificial/normas , Síndrome de Dificultad Respiratoria/terapia , Mecánica Respiratoria/fisiología , Volumen de Ventilación Pulmonar/fisiología
8.
Clin Immunol ; 226: 108716, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33774179

RESUMEN

Lung inflammation and damage is prominent in people infected with SARS-Cov-2 and a major determinant of morbidity and mortality. We report the deposition of complement components in the lungs of people who succumbed to COVID-19 consistent with the activation of the classical and the alternative pathways. Our study provides strong rationale for the expansion of trials involving the use of complement inhibitors to treat patients with COVID-19.


Asunto(s)
COVID-19/inmunología , Activación de Complemento/inmunología , Vía Alternativa del Complemento/inmunología , Lesión Pulmonar/inmunología , Anciano , Anciano de 80 o más Años , COVID-19/complicaciones , Inactivadores del Complemento/farmacología , Inactivadores del Complemento/uso terapéutico , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Humanos , Inmunohistoquímica , Pulmón/diagnóstico por imagen , Pulmón/inmunología , Pulmón/patología , Lesión Pulmonar/complicaciones , Lesión Pulmonar/patología , Lesión Pulmonar/virología , Masculino , Persona de Mediana Edad
9.
Am J Pathol ; 190(10): 2013-2017, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32735889

RESUMEN

Coronavirus disease 2019 has markedly varied clinical presentations, with most patients being asymptomatic or having mild symptoms. However, severe acute respiratory disease, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is common and associated with mortality in patients who require hospitalization. The etiology of susceptibility to severe lung injury remains unclear. Angiotensin II, converted by angiotensin-converting enzyme (ACE) from angiotensin I and metabolized by ACE 2 (ACE2), plays a pivotal role in the pathogenesis of lung injury. ACE2 is identified as an essential receptor for SARS-CoV-2 to enter the cell. The binding of ACE2 and SARS-CoV-2 leads to the exhaustion and down-regulation of ACE2. The interaction and imbalance between ACE and ACE2 result in an unopposed angiotensin II. Considering that the ACE insertion (I)/deletion (D) gene polymorphism contributes to the ACE level variability in general population, in which mean ACE level in DD carriers is approximately twice that in II carriers, we propose a hypothesis of genetic predisposition to severe lung injury in patients with coronavirus disease 2019. It is plausible that the ACE inhibitors and ACE receptor blockers may have the potential to prevent and to treat the acute lung injury after SARS-CoV-2 infection, especially for those with the ACE genotype associated with high ACE level.


Asunto(s)
Infecciones por Coronavirus , Predisposición Genética a la Enfermedad , Lesión Pulmonar/etiología , Pandemias , Peptidil-Dipeptidasa A/genética , Neumonía Viral , Angiotensina I/metabolismo , Angiotensina II/metabolismo , Antagonistas de Receptores de Angiotensina/farmacología , Enzima Convertidora de Angiotensina 2 , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Betacoronavirus/patogenicidad , COVID-19 , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/fisiopatología , Frecuencia de los Genes , Genotipo , Humanos , Lesión Pulmonar/virología , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/fisiopatología , Polimorfismo Genético , Receptores Virales/metabolismo , Sistema Renina-Angiotensina/genética , Sistema Renina-Angiotensina/fisiología , SARS-CoV-2
10.
J Med Virol ; 93(10): 6008-6015, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34232533

RESUMEN

INTRODUCTION: Coronavirus disease-2019 (COVID-19) is a respiratory disease whose clinical manifestation ranges from asymptomatic to severe respiratory failure. The purpose of this study was to investigate the place of serum surfactant-D (SP-D) and angiopoetin-2 (Ang-2) levels in predicting severity of disease in patients diagnosed with COVID-19. METHODS: Sixty-four patients diagnosed with COVID-19 between September 2020 and February 2021, 50 patients diagnosed with community-acquired pneumonia and a 50-member healthy control group were included in the study. Plasma samples and clinical data were collected within 72 h after admission, during hospital stay. Serum SP-D and Ang-2 concentrations were measured using the enzyme-linked immunosorbent assay. RESULTS: SP-D and Ang-2 levels were significantly higher in the mild-moderate pneumonia and severe/critical patient groups compared to the asymptomatic and noncomplicated COVID-19 patients (p < 0.001 for all groups). Serum SP-D and Ang-2 levels of severe-critical COVID-19 patients were significantly higher than CAP patients (p < 0.001). Powerful correlation was present between clinical severity of COVID-19 and SP-D and Ang-2 levels (r = 0.885 p < 0.001 and r = 0.913 p < 0.001, respectively). Cut-off values of 37.7 ng/ml (AUC = 0.763, p < 0.001, 95% confidence interval [CI] = 0.667-0.860) for SP-D and 4208.3 pg/ml (AUC = 0.659, p = 0.004, 95% CI = 0.554-0.763) for Ang-2 were identified as predictors of COVID-19 disease at receiver operating characteristic curve analysis. CONCLUSION: SP-D and Ang-2 are predictive factors in differentiating COVID-19 patients and determining severity of disease. These data may be important for the initiation of treatment in the early stage of the disease in patients with COVID-19.


Asunto(s)
Angiopoyetina 2/metabolismo , COVID-19/diagnóstico , COVID-19/metabolismo , Lesión Pulmonar/metabolismo , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Adulto , Anciano , Biomarcadores/sangre , Infecciones Comunitarias Adquiridas/diagnóstico , Infecciones Comunitarias Adquiridas/virología , Pruebas Diagnósticas de Rutina , Femenino , Humanos , Lesión Pulmonar/virología , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Curva ROC , Índice de Severidad de la Enfermedad
11.
Crit Care ; 25(1): 276, 2021 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-34348797

RESUMEN

BACKGROUND: Typical features differentiate COVID-19-associated lung injury from acute respiratory distress syndrome. The clinical role of chest computed tomography (CT) in describing the progression of COVID-19-associated lung injury remains to be clarified. We investigated in COVID-19 patients the regional distribution of lung injury and the influence of clinical and laboratory features on its progression. METHODS: This was a prospective study. For each CT, twenty images, evenly spaced along the cranio-caudal axis, were selected. For regional analysis, each CT image was divided into three concentric subpleural regions of interest and four quadrants. Hyper-, normally, hypo- and non-inflated lung compartments were defined. Nonparametric tests were used for hypothesis testing (α = 0.05). Spearman correlation test was used to detect correlations between lung compartments and clinical features. RESULTS: Twenty-three out of 111 recruited patients were eligible for further analysis. Five hundred-sixty CT images were analyzed. Lung injury, composed by hypo- and non-inflated areas, was significantly more represented in subpleural than in core lung regions. A secondary, centripetal spread of lung injury was associated with exposure to mechanical ventilation (p < 0.04), longer spontaneous breathing (more than 14 days, p < 0.05) and non-protective tidal volume (p < 0.04). Positive fluid balance (p < 0.01), high plasma D-dimers (p < 0.01) and ferritin (p < 0.04) were associated with increased lung injury. CONCLUSIONS: In a cohort of COVID-19 patients with severe respiratory failure, a predominant subpleural distribution of lung injury is observed. Prolonged spontaneous breathing and high tidal volumes, both causes of patient self-induced lung injury, are associated to an extensive involvement of more central regions. Positive fluid balance, inflammation and thrombosis are associated with lung injury. Trial registration Study registered a priori the 20th of March, 2020. Clinical Trials ID NCT04316884.


Asunto(s)
COVID-19/diagnóstico por imagen , Lesión Pulmonar/diagnóstico por imagen , Anciano , COVID-19/complicaciones , Femenino , Humanos , Lesión Pulmonar/virología , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Respiración Artificial , Suecia , Volumen de Ventilación Pulmonar , Tomografía Computarizada por Rayos X
12.
Nature ; 517(7536): 621-5, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25533958

RESUMEN

Broadly, tissue regeneration is achieved in two ways: by proliferation of common differentiated cells and/or by deployment of specialized stem/progenitor cells. Which of these pathways applies is both organ- and injury-specific. Current models in the lung posit that epithelial repair can be attributed to cells expressing mature lineage markers. By contrast, here we define the regenerative role of previously uncharacterized, rare lineage-negative epithelial stem/progenitor (LNEP) cells present within normal distal lung. Quiescent LNEPs activate a ΔNp63 (a p63 splice variant) and cytokeratin 5 remodelling program after influenza or bleomycin injury in mice. Activated cells proliferate and migrate widely to occupy heavily injured areas depleted of mature lineages, at which point they differentiate towards mature epithelium. Lineage tracing revealed scant contribution of pre-existing mature epithelial cells in such repair, whereas orthotopic transplantation of LNEPs, isolated by a definitive surface profile identified through single-cell sequencing, directly demonstrated the proliferative capacity and multipotency of this population. LNEPs require Notch signalling to activate the ΔNp63 and cytokeratin 5 program, and subsequent Notch blockade promotes an alveolar cell fate. Persistent Notch signalling after injury led to parenchymal 'micro-honeycombing' (alveolar cysts), indicative of failed regeneration. Lungs from patients with fibrosis show analogous honeycomb cysts with evidence of hyperactive Notch signalling. Our findings indicate that distinct stem/progenitor cell pools repopulate injured tissue depending on the extent of the injury, and the outcomes of regeneration or fibrosis may depend in part on the dynamics of LNEP Notch signalling.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/patología , Lesión Pulmonar/patología , Pulmón/citología , Pulmón/patología , Repitelización , Células Madre/citología , Animales , Bleomicina , Linaje de la Célula , Proliferación Celular , Separación Celular , Quistes/metabolismo , Quistes/patología , Células Epiteliales/metabolismo , Femenino , Humanos , Queratina-5/metabolismo , Pulmón/fisiología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/virología , Masculino , Ratones , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Trasplante de Células Madre , Células Madre/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
13.
Int J Med Sci ; 18(1): 29-41, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33390771

RESUMEN

Rationale: Previous studies of coronavirus disease 2019 (COVID-19) were mainly focused on cross-sectional analysis. In this study, we sought to evaluate the dynamic changes of immunological and radiographic features, and the association with the outcome of pulmonary lesions in COVID-19 patients. Methods: Peripheral blood samples and radiographic data were collected longitudinally for up to 8 weeks from 158 laboratory-confirmed COVID-19 patients. The chest computed tomography (CT) scans were scored based on a semi-quantification assessment according to the extent of pulmonary abnormalities; the temporal change of the immunological and radiographic features was analyzed. Results: Compared with mild and moderate patients, severe patients had significantly decreased counts of lymphocytes, CD4+ T cells, CD8+ T cells, and CD19+ B cells but dramatically elevated counts of neutrophils and levels of interleukin (IL)-6. Sequential monitoring showed a sustained increase in lymphocytes counts and significantly decreased levels of IL-6 in severe patients during the disease course. Notably, patients with persistent pulmonary lesions (CT score ≥ 5 in week 8) showed high levels of IL-6 during the follow-up period, compared with those with recovery lesions (CT score < 5 in week 8). More importantly, the peak expression of IL-6 prior to the aggravated lung injury was mainly found in patients with persistent lesions, and multivariate analysis showed that IL-6 level upon admission was an independent factor associated with the persistent pulmonary injury. Conclusion: Prolonged elevation of IL-6 is associated with persistent pulmonary lesions in COVID-19 patients. Sequential monitoring and timely intervention of IL-6 may favor the clinical management of COVID-19.


Asunto(s)
COVID-19/inmunología , Interleucina-6/sangre , Lesión Pulmonar/sangre , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , COVID-19/sangre , COVID-19/complicaciones , COVID-19/diagnóstico por imagen , Femenino , Humanos , Estudios Longitudinales , Lesión Pulmonar/diagnóstico por imagen , Lesión Pulmonar/virología , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Radiografía Torácica , Estudios Retrospectivos , SARS-CoV-2 , Tomografía Computarizada por Rayos X , Adulto Joven
14.
Phytother Res ; 35(9): 4988-5006, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33928690

RESUMEN

The SARS-CoV-2 virus, responsible for COVID-19, spread rapidly worldwide and became a pandemic in 2020. In some patients, the virus remains in the respiratory tract, causing pneumonia, respiratory failure, acute respiratory distress syndrome (ARDS), and sepsis, leading to death. Natural flavonoids (aglycone and glycosides) possess broad biological activities encompassing antiinflammatory, antiviral, antitumoral, antiallergic, antiplatelet, and antioxidant effects. While many studies have focused on the effects of natural flavonoids in experimental models, reports based on clinical trials are still insufficient. In this review, we highlight the effects of flavonoids in controlling pulmonary diseases, particularly the acute respiratory distress syndrome, a consequence of COVID-19, and their potential use in coronavirus-related diseases. Furthermore, we also focus on establishing a relationship between biological potential and chemical aspects of related flavonoids and discuss several possible mechanisms of action, pointing out some possible effects on COVID-19.


Asunto(s)
COVID-19 , Flavonoides , Lesión Pulmonar , COVID-19/complicaciones , Flavonoides/farmacología , Humanos , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/virología , Pandemias
15.
Int J Mol Sci ; 22(2)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33478107

RESUMEN

A complete understanding of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) physiopathology and related histopathologic lesions is necessary to improve treatment and outcome of coronavirus disease 2019 (COVID-19) patients. Many studies have focused on autopsy findings in COVID-19-related deaths to try and define any possible specific pattern. Histopathologic alterations are principally found within lungs and blood vessels, and these abnormalities also seem to have the highest clinical impact. Nevertheless, many of the morphological data collected so far are non-specific, fickle, and possibly associated with other co-existing factors. The aim of this minireview is to describe the main histopathological features related to COVID-19 and the mechanism known as "cytokine storm".


Asunto(s)
COVID-19/inmunología , COVID-19/patología , Lesión Pulmonar/inmunología , Lesión Pulmonar/virología , Enzima Convertidora de Angiotensina 2/metabolismo , Autopsia , COVID-19/diagnóstico por imagen , Síndrome de Liberación de Citoquinas , Citocinas/sangre , Humanos , Lesión Pulmonar/diagnóstico por imagen , Lesión Pulmonar/patología , SARS-CoV-2/aislamiento & purificación
16.
Am J Physiol Lung Cell Mol Physiol ; 318(2): L215-L225, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31774305

RESUMEN

Cell death is increasingly recognized as a driving factor in the development of acute lung injury. Necroptosis, an immunogenic regulated cell death program important in innate immunity, has been implicated in the development of lung injury in a diverse range of conditions. Characterized by lytic cell death and consequent extracellular release of endogenous inflammatory mediators, necroptosis can be both beneficial and deleterious to the host, depending on the context. Here, we review recent investigations linking necroptosis and the development of experimental lung injury. We assess the consequences of necroptosis during bacterial pneumonia, viral infection, sepsis, and sterile injury, highlighting increasing evidence from in vitro studies, animal models, and clinical studies that implicates necroptosis in the pathogenesis of ARDS. Lastly, we highlight current challenges in translating laboratory findings to the bedside.


Asunto(s)
Lesión Pulmonar/patología , Necroptosis , Animales , Modelos Animales de Enfermedad , Humanos , Lesión Pulmonar/microbiología , Lesión Pulmonar/virología , Modelos Biológicos , Infecciones del Sistema Respiratorio/complicaciones , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/virología
17.
Am J Physiol Lung Cell Mol Physiol ; 319(2): L325-L336, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32639866

RESUMEN

A dysregulation of the renin-angiotensin system (RAS) has been involved in the genesis of lung injury and acute respiratory distress syndrome from different causes, including several viral infections. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection of pneumocytes, the hallmark of the pandemic coronavirus disease 2019 (COVID-19) involving both alveolar interstitium and capillaries, is linked to angiotensin-converting enzyme 2 (ACE2) binding and its functional downregulation. ACE2 is a key enzyme for the balance between the two main arms of the RAS: the ACE/angiotensin (Ang) II/Ang II type 1 receptor axis ("classic RAS") and the ACE2/Ang(1-7)/Mas receptor (MasR) axis ("anti-RAS"). The ACE2 downregulation, as a result of SARS-coronaviruses binding, enhances the classic RAS, leading to lung damage and inflammation with leaky pulmonary blood vessels and fibrosis, when the attenuation mediated by the anti-RAS arm is reduced. ACE inhibitors (ACE-I) and Ang II type 1 receptor blockers (ARB), effective in cardiovascular diseases, were found to prevent and counteract acute lung injury in several experimental models by restoring the balance between these two opposing arms. The evidence of RAS arm disequilibrium in COVID-19 and the hypothesis of a beneficial role of RAS modulation supported by preclinical and clinical studies are the focus of the present review. Preclinical and clinical studies on drugs balancing RAS arms might be the right way to counter COVID-19.


Asunto(s)
Antagonistas de Receptores de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Infecciones por Coronavirus/patología , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/patología , Alveolos Pulmonares/patología , Síndrome de Dificultad Respiratoria/patología , Células Epiteliales Alveolares/virología , Enzima Convertidora de Angiotensina 2 , Betacoronavirus/metabolismo , COVID-19 , Infecciones por Coronavirus/tratamiento farmacológico , Regulación hacia Abajo , Humanos , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/prevención & control , Lesión Pulmonar/virología , Pandemias , Neumonía Viral/tratamiento farmacológico , Proto-Oncogenes Mas , Alveolos Pulmonares/irrigación sanguínea , Alveolos Pulmonares/virología , Receptores de Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/efectos de los fármacos , Síndrome de Dificultad Respiratoria/virología , SARS-CoV-2
18.
Eur Respir J ; 55(6)2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32265310

RESUMEN

The severe respiratory consequences of the coronavirus disease 2019 (COVID-19) pandemic have prompted urgent need for novel therapies. Cell-based approaches, primarily using mesenchymal stem (stromal) cells (MSCs), have demonstrated safety and possible efficacy in patients with acute respiratory distress syndrome (ARDS), although they are not yet well studied in respiratory virus-induced ARDS. Limited pre-clinical data suggest that systemic MSC administration can significantly reduce respiratory virus (influenza strains H5N1 and H9N2)-induced lung injury; however, there are no available data in models of coronavirus respiratory infection.There is a rapidly increasing number of clinical investigations of cell-based therapy approaches for COVID-19. These utilise a range of different cell sources, doses, dosing strategies and targeted patient populations. To provide a rational strategy to maximise potential therapeutic use, it is critically important to understand the relevant pre-clinical studies and postulated mechanisms of MSC actions in respiratory virus-induced lung injuries. This review presents these, along with consideration of current clinical investigations.


Asunto(s)
Infecciones por Coronavirus/terapia , Medios de Cultivo Condicionados , Gripe Humana/terapia , Lesión Pulmonar/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Neumonía Viral/terapia , Síndrome de Dificultad Respiratoria/terapia , Enzima Convertidora de Angiotensina 2 , Animales , Betacoronavirus , COVID-19 , Tratamiento Basado en Trasplante de Células y Tejidos , Vesículas Extracelulares/trasplante , Humanos , Subtipo H5N1 del Virus de la Influenza A , Subtipo H9N2 del Virus de la Influenza A , Lesión Pulmonar/virología , Células Madre Mesenquimatosas/metabolismo , Infecciones por Orthomyxoviridae/terapia , Pandemias , Peptidil-Dipeptidasa A/metabolismo , SARS-CoV-2 , Serina Endopeptidasas/metabolismo
19.
PLoS Pathog ; 14(11): e1007428, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30422993

RESUMEN

Severe influenza A virus infection causes high mortality and morbidity worldwide due to delayed antiviral treatment and inducing overwhelming immune responses, which contribute to immunopathological lung injury. Sirolimus, an inhibitor of mammalian target of rapamycin (mTOR), was effective in improving clinical outcomes in patients with severe H1N1 infection; however, the mechanisms by which it attenuates acute lung injury have not been elucidated. Here, delayed oseltamivir treatment was used to mimic clinical settings on lethal influenza A (H1N1) pdm09 virus (pH1N1) infection mice model. We revealed that delayed oseltamivir plus sirolimus treatment protects mice against lethal pH1N1 infection by attenuating severe lung damage. Mechanistically, the combined treatment reduced viral titer and pH1N1-induced mTOR activation. Subsequently, it suppressed the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome-mediated secretion of interleukin (IL)-1ß and IL-18. It was noted that decreased NLRP3 inflammasome activation was associated with inhibited nuclear factor (NF)-κB activation, reduced reactive oxygen species production and increased autophagy. Additionally, the combined treatment reduced the expression of other proinflammatory cytokines and chemokines, and decreased inflammatory cell infiltration in lung tissue and bronchioalveolar lavage fluid. Consistently, it inhibited the mTOR-NF-κB-NLRP3 inflammasome-IL-1ß axis in a lung epithelial cell line. These results demonstrated that combined treatment with sirolimus and oseltamivir attenuates pH1N1-induced severe lung injury, which is correlated with suppressed mTOR-NLRP3-IL-1ß axis and reduced viral titer. Therefore, treatment with sirolimus as an adjuvant along with oseltamivir may be a promising immunomodulatory strategy for managing severe influenza.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/virología , Oseltamivir/farmacología , Sirolimus/farmacología , Animales , Líquido del Lavado Bronquioalveolar/citología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Perros , Quimioterapia Combinada/métodos , Células Epiteliales , Femenino , Inflamasomas/efectos de los fármacos , Inflamasomas/inmunología , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Interleucina-18/inmunología , Interleucina-1beta/inmunología , Pulmón/patología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Proteína con Dominio Pirina 3 de la Familia NLR/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
20.
Am J Pathol ; 189(8): 1513-1525, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31108100

RESUMEN

Cadmium (Cd) is a naturally occurring environmental toxicant that disrupts mitochondrial function at occupational exposure levels. The impacts of Cd exposure at low levels through dietary intake remain largely uncharacterized. Human respiratory syncytial virus (RSV) causes severe morbidity, which can require hospitalization and result in death in young children and elderly populations. The impacts of environmental Cd exposure on the severity of RSV disease are unknown. Herein, we used a mouse model to examine whether Cd pre-exposure at a level of dietary intake potentiates pulmonary inflammation on subsequent infection with RSV. Mice were given Cd or saline in drinking water for 28 days. Subsets of these mice were infected with RSV at 5 days before the end of the study. Cd pre-exposure caused relatively subtle changes in lung; however, it elevated the IL-4 level and altered metabolites associated with fatty acid metabolism. After RSV infection, mice pre-exposed to Cd had elevated lung RSV titer and increased inflammation, as measured by histopathology, immune cell infiltration, cytokines, and chemokines. RSV infection after Cd pre-exposure also caused widespread perturbation in metabolism of glycerophospholipids and amino acids (Trp, Met, and Cys, branched-chain amino acids), as well as carnitine shuttle associated with mitochondrial energy metabolism. The results show that Cd burden by dietary intake potentiates RSV infection and severe disease with associated mitochondrial metabolic disruption.


Asunto(s)
Cadmio/toxicidad , Exposición a Riesgos Ambientales/efectos adversos , Lesión Pulmonar , Infecciones por Virus Sincitial Respiratorio , Virus Sincitiales Respiratorios/metabolismo , Animales , Humanos , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Lesión Pulmonar/virología , Ratones , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA