Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
EBioMedicine ; 85: 104296, 2022 Nov.
Article in English | MEDLINE | ID: covidwho-2322217

ABSTRACT

BACKGROUND: COVID-19 is characterized by a heterogeneous clinical presentation, ranging from mild symptoms to severe courses of disease. 9-20% of hospitalized patients with severe lung disease die from COVID-19 and a substantial number of survivors develop long-COVID. Our objective was to provide comprehensive insights into the pathophysiology of severe COVID-19 and to identify liquid biomarkers for disease severity and therapy response. METHODS: We studied a total of 85 lungs (n = 31 COVID autopsy samples; n = 7 influenza A autopsy samples; n = 18 interstitial lung disease explants; n = 24 healthy controls) using the highest resolution Synchrotron radiation-based hierarchical phase-contrast tomography, scanning electron microscopy of microvascular corrosion casts, immunohistochemistry, matrix-assisted laser desorption ionization mass spectrometry imaging, and analysis of mRNA expression and biological pathways. Plasma samples from all disease groups were used for liquid biomarker determination using ELISA. The anatomic/molecular data were analyzed as a function of patients' hospitalization time. FINDINGS: The observed patchy/mosaic appearance of COVID-19 in conventional lung imaging resulted from microvascular occlusion and secondary lobular ischemia. The length of hospitalization was associated with increased intussusceptive angiogenesis. This was associated with enhanced angiogenic, and fibrotic gene expression demonstrated by molecular profiling and metabolomic analysis. Increased plasma fibrosis markers correlated with their pulmonary tissue transcript levels and predicted disease severity. Plasma analysis confirmed distinct fibrosis biomarkers (TSP2, GDF15, IGFBP7, Pro-C3) that predicted the fatal trajectory in COVID-19. INTERPRETATION: Pulmonary severe COVID-19 is a consequence of secondary lobular microischemia and fibrotic remodelling, resulting in a distinctive form of fibrotic interstitial lung disease that contributes to long-COVID. FUNDING: This project was made possible by a number of funders. The full list can be found within the Declaration of interests / Acknowledgements section at the end of the manuscript.


Subject(s)
COVID-19 , Lung Diseases, Interstitial , Humans , Lung/diagnostic imaging , Lung/pathology , Lung Diseases, Interstitial/pathology , Fibrosis , Biomarkers/analysis , Ischemia/pathology , Post-Acute COVID-19 Syndrome
3.
Am J Respir Crit Care Med ; 206(2): 229-230, 2022 07 15.
Article in English | MEDLINE | ID: covidwho-2227053
5.
Morphologie ; 106(354, Supplement):S38, 2022.
Article in French | ScienceDirect | ID: covidwho-1983708

ABSTRACT

Objet L’architecture capillaire et la circulation bronchique habituelle semble sensiblement modifiée dans le cadre des pneumopathies à SARS-CoV-2, associés à des thromboses multiples [1], [2]. L’imagerie en contraste de phase par source synchrotron (sPCI) permet d’étudier précisément l’ensemble des tissus organiques à une résolution microscopique et de façon non destructive. Le but de cette étude était de comparer l’anatomie vasculaire bronchique entre un poumon sain et un poumon de patients infectés par la COVID-19. Méthodes Trois poumons témoins ont été prélevés au Laboratoire d’Anatomie Des Alpes Françaises puis comparés à trois poumons de patients infectés par le SARS-CoV-2, provenant de la banque d’organe de l’Université Witten/Herdecke (Allemagne). Après préparation, les poumons ont été imagés au Synchrotron Européen de Grenoble à 26μm, 6μm et 2μm sans injection de produit de contraste [3]. La vascularisation a été étudiée sur les coupes tomodensitométriques 2D et sur les reconstructions tridimensionnelles, puis sur coupes histologiques et via des injections-corrosions. Le projet a été financé par la Chan Zuckerberg Initiative. Résultats La circulation bronchique, qui provient de l’aorte thoracique et des artères intercostales, est modifiée par le processus inflammatoire et hypoxique. L’étude de l’anatomie microscopique bronchique en sPCI a permis d’établir la présence de nombreuses d’anastomoses de moins de 50μm entre la circulation bronchique et l’artère lobulaire dans les poumons de patients infectés par la COVID-19, entraînant un shunt doit-gauche intra-pulmonaire. Par ailleurs, une angiogenèse anarchique majeure a été détectée au niveau des plexus alvéolaires des zones atteintes par l’infection, au dépend des artères intra-lobulaires, par rapport aux poumons témoins. Conclusion L’imagerie sPCI réalisée a permis la première visualisation tridimensionnelle d’un shunt bronchio-pulmonaire dans la COVID-19 ainsi que les phénomènes de néovascularisations excessives associés.

6.
Front Immunol ; 13: 879157, 2022.
Article in English | MEDLINE | ID: covidwho-1933664

ABSTRACT

During the COVID-19 pandemic, vaccination is the most important countermeasure. Pharmacovigilance concerns however emerged with very rare, but potentially disastrous thrombotic complications following vaccination with ChAdOx1. Platelet factor-4 antibody mediated vaccine-induced immune thrombotic thrombocytopenia (VITT) was described as an underlying mechanism of these thrombotic events. Recent work moreover suggests that mechanisms of immunothrombosis including neutrophil extracellular trap (NET) formation might be critical for thrombogenesis during VITT. In this study, we investigated blood and thrombus specimens of a female patient who suffered severe stroke due to VITT after vaccination with ChAdOx1 in comparison to 13 control stroke patients with similar clinical characteristics. We analyzed cerebral thrombi using histological examination, staining of complement factors, NET-markers, DNase and LL-37. In blood samples at the hyper-acute phase of stroke and 7 days later, we determined cell-free DNA, myeloperoxidase-histone complexes, DNase activity, myeloperoxidase activity, LL-37 and inflammatory cytokines. NET markers were identified in thrombi of all patients. Interestingly, the thrombus of the VITT-patient exclusively revealed complement factors and high amounts of DNase and LL-37. High DNase activity was also measured in blood, implying a disturbed NET-regulation. Furthermore, serum of the VITT-patient inhibited reactive oxygen species-dependent NET-release by phorbol-myristate-acetate to a lesser degree compared to controls, indicating either less efficient NET-inhibition or enhanced NET-induction in the blood of the VITT-patient. Additionally, the changes in specific cytokines over time were emphasized in the VITT-patient as well. In conclusion, insufficient resolution of NETs, e.g. by endogenous DNases or protection of NETs against degradation by embedded factors like the antimicrobial peptide LL-37 might thus be an important factor in the pathology of VITT besides increased NET-formation. On the basis of these findings, we discuss the potential implications of the mechanisms of disturbed NETs-degradation for diagnostic and therapeutic approaches in VITT-related thrombogenesis, other auto-immune disorders and beyond.


Subject(s)
COVID-19 , Extracellular Traps , Purpura, Thrombocytopenic, Idiopathic , Stroke , Thrombocytopenia , Thrombosis , Vaccines , Deoxyribonuclease I/metabolism , Deoxyribonucleases , Female , Humans , Neutrophils , Pandemics , Peroxidase/metabolism , Platelet Factor 4/metabolism , Purpura, Thrombocytopenic, Idiopathic/metabolism , Stroke/etiology , Stroke/metabolism , Thrombocytopenia/chemically induced , Thrombocytopenia/metabolism , Thrombosis/etiology , Thrombosis/metabolism , Vaccines/metabolism
7.
PLoS Pathog ; 18(5): e1010471, 2022 05.
Article in English | MEDLINE | ID: covidwho-1833668

ABSTRACT

The ability to treat severe viral infections is limited by our understanding of the mechanisms behind virus-induced immunopathology. While the role of type I interferons (IFNs) in early control of viral replication is clear, less is known about how IFNs can regulate the development of immunopathology and affect disease outcomes. Here, we report that absence of type I IFN receptor (IFNAR) is associated with extensive immunopathology following mucosal viral infection. This pathology occurred independent of viral load or type II immunity but required the presence of macrophages and IL-6. The depletion of macrophages and inhibition of IL-6 signaling significantly abrogated immunopathology. Tissue destruction was mediated by macrophage-derived matrix metalloproteinases (MMPs), as MMP inhibition by doxycycline and Ro 28-2653 reduced the severity of tissue pathology. Analysis of post-mortem COVID-19 patient lungs also displayed significant upregulation of the expression of MMPs and accumulation of macrophages. Overall, we demonstrate that IFNs inhibit macrophage-mediated MMP production to prevent virus-induced immunopathology and uncover MMPs as a therapeutic target towards viral infections.


Subject(s)
COVID-19 , Interferon Type I , Orthomyxoviridae Infections , Humans , Interleukin-6/metabolism , Macrophages/metabolism , Proteolysis
8.
Int J Mol Sci ; 23(3)2022 Jan 29.
Article in English | MEDLINE | ID: covidwho-1667195

ABSTRACT

(1) Background: In COVID-19 survivors there is an increased prevalence of pulmonary fibrosis of which the underlying molecular mechanisms are poorly understood; (2) Methods: In this multicentric study, n = 12 patients who succumbed to COVID-19 due to progressive respiratory failure were assigned to an early and late group (death within ≤7 and >7 days of hospitalization, respectively) and compared to n = 11 healthy controls; mRNA and protein expression as well as biological pathway analysis were performed to gain insights into the evolution of pulmonary fibrogenesis in COVID-19; (3) Results: Median duration of hospitalization until death was 3 (IQR25-75, 3-3.75) and 14 (12.5-14) days in the early and late group, respectively. Fifty-eight out of 770 analyzed genes showed a significantly altered expression signature in COVID-19 compared to controls in a time-dependent manner. The entire study group showed an increased expression of BST2 and IL1R1, independent of hospitalization time. In the early group there was increased activity of inflammation-related genes and pathways, while fibrosis-related genes (particularly PDGFRB) and pathways dominated in the late group; (4) Conclusions: After the first week of hospitalization, there is a shift from pro-inflammatory to fibrogenic activity in severe COVID-19. IL1R1 and PDGFRB may serve as potential therapeutic targets in future studies.


Subject(s)
COVID-19/genetics , COVID-19/metabolism , Pulmonary Fibrosis/pathology , Aged , COVID-19/mortality , Female , Hospital Mortality/trends , Hospitalization , Humans , Lung/pathology , Male , Middle Aged , Pulmonary Fibrosis/metabolism , Respiratory Insufficiency/pathology , SARS-CoV-2/pathogenicity
11.
Pathologe ; 42(2): 164-171, 2021 Mar.
Article in German | MEDLINE | ID: covidwho-1235729

ABSTRACT

Viral respiratory diseases constitute the most common reasons for hospitalization with more than half of all acute illnesses worldwide. Progressive respiratory failure with pronounced diffuse alveolar damage has been identified as the primary cause of death in COVID-19. COVID-19 pneumonia shares common histopathological hallmarks with influenza (H1N1)-related ARDS, like diffuse alveolar damage (DAD) with edema, hemorrhage, and intra-alveolar fibrin deposition. The lungs with COVID-19 pneumonia revealed perivascular inflammation, an endothelial injury, microangiopathy, and an aberrant blood vessel neoformation by intussusceptive angiogenesis. While this pronounced angiocentric inflammation is likely be found - to varying degrees - in numerous other organs, e.g., the heart, COVID-19 is hypothesized to be not just a pulmonary, but rather a systemic "vascular disease."


Subject(s)
COVID-19 , Influenza A Virus, H1N1 Subtype , Heart , Humans , Lung , SARS-CoV-2
SELECTION OF CITATIONS
SEARCH DETAIL