Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cells ; 10(11)2021 11 07.
Artículo en Inglés | MEDLINE | ID: covidwho-1512135

RESUMEN

The bronchial vascular endothelial network plays important roles in pulmonary pathology during respiratory viral infections, including respiratory syncytial virus (RSV), influenza A(H1N1) and importantly SARS-Cov-2. All of these infections can be severe and even lethal in patients with underlying risk factors.A major obstacle in disease prevention is the lack of appropriate efficacious vaccine(s) due to continuous changes in the encoding capacity of the viral genome, exuberant responsiveness of the host immune system and lack of effective antiviral drugs. Current management of these severe respiratory viral infections is limited to supportive clinical care. The primary cause of morbidity and mortality is respiratory failure, partially due to endothelial pulmonary complications, including edema. The latter is induced by the loss of alveolar epithelium integrity and by pathological changes in the endothelial vascular network that regulates blood flow, blood fluidity, exchange of fluids, electrolytes, various macromolecules and responses to signals triggered by oxygenation, and controls trafficking of leukocyte immune cells. This overview outlines the latest understanding of the implications of pulmonary vascular endothelium involvement in respiratory distress syndrome secondary to viral infections. In addition, the roles of infection-induced cytokines, growth factors, and epigenetic reprogramming in endothelial permeability, as well as emerging treatment options to decrease disease burden, are discussed.


Asunto(s)
Células Endoteliales/patología , Estrés Oxidativo , Síndrome de Dificultad Respiratoria/patología , Virosis/patología , Epigénesis Genética , Humanos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Edema Pulmonar/genética , Edema Pulmonar/patología , Edema Pulmonar/virología , Síndrome de Dificultad Respiratoria/genética , Síndrome de Dificultad Respiratoria/virología , Virus Sincitiales Respiratorios/patogenicidad , SARS-CoV-2/patogenicidad , Virosis/genética , Virosis/virología
2.
Nat Struct Mol Biol ; 28(9): 755-761, 2021 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1406396

RESUMEN

Bradykinin and kallidin are endogenous kinin peptide hormones that belong to the kallikrein-kinin system and are essential to the regulation of blood pressure, inflammation, coagulation and pain control. Des-Arg10-kallidin, the carboxy-terminal des-Arg metabolite of kallidin, and bradykinin selectively activate two G protein-coupled receptors, type 1 and type 2 bradykinin receptors (B1R and B2R), respectively. The hyperactivation of bradykinin receptors, termed 'bradykinin storm', is associated with pulmonary edema in COVID-19 patients, suggesting that bradykinin receptors are important targets for COVID-19 intervention. Here we report two G protein-coupled complex structures of human B1R and B2R bound to des-Arg10-kallidin and bradykinin, respectively. Combined with functional analysis, our structures reveal the mechanism of ligand selectivity and specific activation of the bradykinin receptor. These findings also provide a framework for guiding drug design targeting bradykinin receptors for the treatment of inflammation, cardiovascular disorders and COVID-19.


Asunto(s)
Bradiquinina/metabolismo , COVID-19/patología , Calidina/metabolismo , Receptores de Bradiquinina/metabolismo , Microscopía por Crioelectrón , Activación Enzimática/fisiología , Humanos , Estructura Terciaria de Proteína , Edema Pulmonar/patología , Edema Pulmonar/virología , SARS-CoV-2
3.
SLAS Discov ; 26(9): 1079-1090, 2021 10.
Artículo en Inglés | MEDLINE | ID: covidwho-1314244

RESUMEN

The recent renascence of phenotypic drug discovery (PDD) is catalyzed by its ability to identify first-in-class drugs and deliver results when the exact molecular mechanism is partially obscure. Acute respiratory distress syndrome (ARDS) is a severe, life-threatening condition with a high mortality rate that has increased in frequency due to the COVID-19 pandemic. Despite decades of laboratory and clinical study, no efficient pharmacological therapy for ARDS has been found. An increase in endothelial permeability is the primary event in ARDS onset, causing the development of pulmonary edema that leads to respiratory failure. Currently, the detailed molecular mechanisms regulating endothelial permeability are poorly understood. Therefore, the use of the PDD approach in the search for efficient ARDS treatment can be more productive than classic target-based drug discovery (TDD), but its use requires a new cell-based assay compatible with high-throughput (HTS) and high-content (HCS) screening. Here we report the development of a new plate-based image cytometry method to measure endothelial barrier function. The incorporation of image cytometry in combination with digital image analysis substantially decreases assay variability and increases the signal window. This new method simultaneously allows for rapid measurement of cell monolayer permeability and cytological analysis. The time-course of permeability increase in human pulmonary artery endothelial cells (HPAECs) in response to the thrombin and tumor necrosis factor α treatment correlates with previously published data obtained by transendothelial resistance (TER) measurements. Furthermore, the proposed image cytometry method can be easily adapted for HTS/HCS applications.


Asunto(s)
COVID-19/diagnóstico por imagen , Ensayos Analíticos de Alto Rendimiento/métodos , Citometría de Imagen/métodos , Síndrome de Dificultad Respiratoria/diagnóstico por imagen , COVID-19/diagnóstico , COVID-19/virología , Permeabilidad de la Membrana Celular/genética , Descubrimiento de Drogas , Células Endoteliales/ultraestructura , Células Endoteliales/virología , Humanos , Procesamiento de Imagen Asistido por Computador , Pandemias/prevención & control , Fenotipo , Arteria Pulmonar/diagnóstico por imagen , Arteria Pulmonar/patología , Arteria Pulmonar/virología , Edema Pulmonar/diagnóstico , Edema Pulmonar/diagnóstico por imagen , Edema Pulmonar/virología , Síndrome de Dificultad Respiratoria/diagnóstico , Síndrome de Dificultad Respiratoria/virología , Insuficiencia Respiratoria/diagnóstico , Insuficiencia Respiratoria/diagnóstico por imagen , Insuficiencia Respiratoria/virología , SARS-CoV-2/patogenicidad , Trombina/farmacología , Factor de Necrosis Tumoral alfa/farmacología
4.
Sci Rep ; 11(1): 11524, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1253988

RESUMEN

Nearly 5% of patients suffering from COVID-19 develop acute respiratory distress syndrome (ARDS). Extravascular lung water index (EVLWI) is a marker of pulmonary oedema which is associated with mortality in ARDS. In this study, we evaluate whether EVLWI is higher in patients with COVID-19 associated ARDS as compared to COVID-19 negative, ventilated patients with ARDS and whether EVLWI has the potential to monitor disease progression. EVLWI and cardiac function were monitored by transpulmonary thermodilution in 25 patients with COVID-19 ARDS subsequent to intubation and compared to a control group of 49 non-COVID-19 ARDS patients. At intubation, EVLWI was noticeably elevated and significantly higher in COVID-19 patients than in the control group (17 (11-38) vs. 11 (6-26) mL/kg; p < 0.001). High pulmonary vascular permeability index values (2.9 (1.0-5.2) versus 1.9 (1.0-5.2); p = 0.003) suggested a non-cardiogenic pulmonary oedema. By contrast, the cardiac parameters SVI, GEF and GEDVI were comparable in both cohorts. High EVLWI values were associated with viral persistence, prolonged intensive care treatment and in-hospital mortality (23.2 ± 6.7% vs. 30.3 ± 6.0%, p = 0.025). Also, EVLWI showed a significant between-subjects (r = - 0.60; p = 0.001) and within-subjects correlation (r = - 0.27; p = 0.028) to Horowitz index. Compared to non COVID-19 ARDS, COVID-19 results in markedly elevated EVLWI-values in patients with ARDS. High EVLWI reflects a non-cardiogenic pulmonary oedema in COVID-19 ARDS and could serve as parameter to monitor ARDS progression on ICU.


Asunto(s)
COVID-19/complicaciones , Agua Pulmonar Extravascular/inmunología , Edema Pulmonar/mortalidad , Síndrome de Dificultad Respiratoria/mortalidad , Adulto , Anciano , Anciano de 80 o más Años , COVID-19/diagnóstico , COVID-19/inmunología , COVID-19/mortalidad , Permeabilidad Capilar , Progresión de la Enfermedad , Agua Pulmonar Extravascular/virología , Femenino , Mortalidad Hospitalaria , Humanos , Unidades de Cuidados Intensivos/estadística & datos numéricos , Tiempo de Internación/estadística & datos numéricos , Pulmón/irrigación sanguínea , Pulmón/fisiopatología , Masculino , Persona de Mediana Edad , Monitoreo Fisiológico/métodos , Monitoreo Fisiológico/estadística & datos numéricos , Pronóstico , Edema Pulmonar/diagnóstico , Edema Pulmonar/inmunología , Edema Pulmonar/virología , Respiración Artificial , Síndrome de Dificultad Respiratoria/diagnóstico , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/terapia , Medición de Riesgo/métodos , SARS-CoV-2/aislamiento & purificación , Índice de Severidad de la Enfermedad , Termodilución/métodos , Termodilución/estadística & datos numéricos , Adulto Joven
5.
Nephrol Ther ; 17(4): 226-232, 2021 Aug.
Artículo en Francés | MEDLINE | ID: covidwho-1074879

RESUMEN

BACKGROUND: The effect of COVID-19 pandemic on end stage renal disease patient who should initiated dialysis are limited in Sub-Saharan Africa is unknown. We sought to describe the epidemiologic and clinical profile of newly admitted patient in chronic haemodialysis during the COVID-19 pandemic in Cameroon and evaluate their survival between 90days of dialysis initiation. MATERIAL AND METHOD: We conducted a cohort study of 6months from April to October 2020. End stage renal disease patients newly admitted in the haemodialysis facility of the General Hospital of Douala were included. Patients with confirmed or suspected COVID-19 were identified. Socio-demographic, clinical and biological data at dialysis initiation as well as mortality between the 90days of dialysis initiation were registered. RESULTS: A total of 57 incident patients were recorded from April to October 2020 with a monthly mean of 9.5 patients. The mean age was 46.95±13.12years. Twenty-four COVID-19 were identified with a frequency of 49% among emergency admission. Pulmonary œdema (79.2% vs. 42.4%; P=0.006) and uremic encephalopathy (83.4% vs. 53.6%; P=0.022) were more common in COVID-19. The overall survival at 90days was 48% with a tendency to poor survival among COVID-19 and patients with low socioeconomic level. In Cox regression, low socioeconomic level increase the risk of instant death by 3.08. CONCLUSION: SARS-CoV2 seem to increase nephrology emergency and poor survival in haemodialysis at 90days.


Asunto(s)
COVID-19/mortalidad , Hospitalización , Fallo Renal Crónico/mortalidad , Diálisis Renal , Encefalopatías/epidemiología , Encefalopatías/etiología , Camerún/epidemiología , Femenino , Hospitales Generales , Humanos , Incidencia , Fallo Renal Crónico/terapia , Masculino , Persona de Mediana Edad , Pandemias , Estudios Prospectivos , Edema Pulmonar/epidemiología , Edema Pulmonar/virología , Clase Social , Uremia/epidemiología , Uremia/virología
7.
Vascul Pharmacol ; 137: 106829, 2021 04.
Artículo en Inglés | MEDLINE | ID: covidwho-1014876

RESUMEN

Angiotensin-converting enzyme 2 (ACE2) is an important player of the renin-angiotensin-aldosterone system (RAAS) in regulating the conversion of angiotensin II into angiotensin (1-7). While expressed on the surface of human cells, such as lung, heart, kidney, neurons, and endothelial cells (EC), ACE2 is the entry receptor for SARS-CoV-2. Here, we would like to highlight that ACE2 is predominant on the EC membrane. Many of coronavirus disease 2019 (COVID-19) symptoms have been associated with the large recruitment of immune cells, directly affecting EC. Additionally, cytokines, hypoxia, and complement activation can trigger the activation of EC leading to the coagulation cascade. The EC dysfunction plus the inflammation due to SARS-CoV-2 infection may lead to abnormal coagulation, actively participating in thrombo-inflammatory processes resulting in vasculopathy and indicating poor prognosis in patients with COVID-19. Considering the intrinsic relationship between EC and the pathophysiology of SARS-CoV-2, EC-associated therapies such as anticoagulants, fibrinolytic drugs, immunomodulators, and molecular therapies have been proposed. In this review, we will discuss the role of EC in the lung inflammation and edema, in the disseminate coagulation process, ACE2 positive cancer patients, and current and future EC-associated therapies to treat COVID-19.


Asunto(s)
COVID-19/virología , Enfermedades Cardiovasculares/virología , Endotelio Vascular/virología , Inflamación/virología , SARS-CoV-2/patogenicidad , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Coagulación Sanguínea , COVID-19/complicaciones , COVID-19/terapia , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/terapia , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Interacciones Huésped-Patógeno , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/terapia , Mediadores de Inflamación/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Edema Pulmonar/metabolismo , Edema Pulmonar/patología , Edema Pulmonar/virología , Transducción de Señal
8.
Front Immunol ; 11: 574862, 2020.
Artículo en Inglés | MEDLINE | ID: covidwho-845301

RESUMEN

It is currently believed that innate immunity is unable to prevent the spread of SARS-CoV-2 from the upper airways to the alveoli of high-risk groups of patients. SARS-CoV-2 replication in ACE-2-expressing pneumocytes can drive the diffuse alveolar injury through the cytokine storm and immunothrombosis by upregulating the transcription of chemokine/cytokines, unlike several other respiratory viruses. Here we report histopathology data obtained in post-mortem lung biopsies of COVID-19, showing the increased density of perivascular and septal mast cells (MCs) and IL-4-expressing cells (n = 6), in contrast to the numbers found in pandemic H1N1-induced pneumonia (n = 10) or Control specimens (n = 10). Noteworthy, COVID-19 lung biopsies showed a higher density of CD117+ cells, suggesting that c-kit positive MCs progenitors were recruited earlier to the alveolar septa. These findings suggest that MC proliferation/differentiation in the alveolar septa might be harnessed by the shift toward IL-4 expression in the inflamed alveolar septa. Future studies may clarify whether the fibrin-dependent generation of the hyaline membrane, processes that require the diffusion of procoagulative plasma factors into the alveolar lumen and the endothelial dysfunction, are preceded by MC-driven formation of interstitial edema in the alveolar septa.


Asunto(s)
Betacoronavirus/inmunología , Infecciones por Coronavirus/inmunología , Mastocitos/inmunología , Neumonía Viral/inmunología , Alveolos Pulmonares/inmunología , Edema Pulmonar/inmunología , Trombosis/inmunología , Adulto , Anciano , Anciano de 80 o más Años , COVID-19 , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Femenino , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Gripe Humana/patología , Gripe Humana/virología , Interleucina-4/inmunología , Masculino , Mastocitos/patología , Persona de Mediana Edad , Pandemias , Neumonía Viral/patología , Neumonía Viral/virología , Proteínas Proto-Oncogénicas c-kit/inmunología , Alveolos Pulmonares/patología , Alveolos Pulmonares/virología , Edema Pulmonar/patología , Edema Pulmonar/virología , SARS-CoV-2 , Trombosis/patología , Trombosis/virología
9.
Trends Immunol ; 41(10): 856-859, 2020 10.
Artículo en Inglés | MEDLINE | ID: covidwho-703987

RESUMEN

Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and mainly affects the lungs. Sarcoidosis is an autoinflammatory disease characterized by the diffusion of granulomas in the lungs and other organs. Here, we discuss how the two diseases might involve some common mechanistic cellular pathways around the regulation of autophagy.


Asunto(s)
Autofagia/efectos de los fármacos , Betacoronavirus/patogenicidad , Infecciones por Coronavirus/tratamiento farmacológico , Neumonía Viral/tratamiento farmacológico , Edema Pulmonar/tratamiento farmacológico , Sarcoidosis/tratamiento farmacológico , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Autofagia/genética , Azitromicina/uso terapéutico , Betacoronavirus/crecimiento & desarrollo , COVID-19 , Cloroquina/uso terapéutico , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/virología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Isoniazida/uso terapéutico , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/virología , Pandemias , Neumonía Viral/epidemiología , Neumonía Viral/genética , Neumonía Viral/virología , Edema Pulmonar/epidemiología , Edema Pulmonar/genética , Edema Pulmonar/virología , Rifampin/uso terapéutico , SARS-CoV-2 , Sarcoidosis/epidemiología , Sarcoidosis/genética , Sarcoidosis/virología , Síndrome Respiratorio Agudo Grave/epidemiología , Síndrome Respiratorio Agudo Grave/genética , Síndrome Respiratorio Agudo Grave/virología , Índice de Severidad de la Enfermedad
10.
Am J Physiol Lung Cell Mol Physiol ; 318(6): L1239-L1243, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: covidwho-246452

RESUMEN

Lethality of coronavirus disease (COVID-19) during the 2020 pandemic, currently still in the exponentially accelerating phase in most countries, is critically driven by disruption of the alveolo-capillary barrier of the lung, leading to lung edema as a direct consequence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We argue for inhibition of the transient receptor potential vanilloid 4 (TRPV4) calcium-permeable ion channel as a strategy to address this issue, based on the rationale that TRPV4 inhibition is protective in various preclinical models of lung edema and that TRPV4 hyperactivation potently damages the alveolo-capillary barrier, with lethal outcome. We believe that TRPV4 inhibition has a powerful prospect at protecting this vital barrier in COVID-19 patients, even to rescue a damaged barrier. A clinical trial using a selective TRPV4 inhibitor demonstrated a benign safety profile in healthy volunteers and in patients suffering from cardiogenic lung edema. We argue for expeditious clinical testing of this inhibitor in COVID-19 patients with respiratory malfunction and at risk for lung edema. Perplexingly, among the currently pursued therapeutic strategies against COVID-19, none is designed to directly protect the alveolo-capillary barrier. Successful protection of the alveolo-capillary barrier will not only reduce COVID-19 lethality but will also preempt a distressing healthcare scenario with insufficient capacity to provide ventilator-assisted respiration.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus , Pulmón/virología , Pandemias , Neumonía Viral , Edema Pulmonar/prevención & control , Canales Catiónicos TRPV/antagonistas & inhibidores , COVID-19 , Calcio/metabolismo , Infecciones por Coronavirus/virología , Humanos , Pulmón/metabolismo , Neumonía Viral/virología , Edema Pulmonar/virología , Respiración Artificial , SARS-CoV-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA