Your browser doesn't support javascript.
Montrer: 20 | 50 | 100
Résultats 1 - 6 de 6
Filtre
1.
Pathol Res Pract ; 247: 154519, 2023 Jul.
Article Dans Anglais | MEDLINE | ID: covidwho-2314785

Résumé

We explored the pathological changes and the activation of local complement system in COVID-19 pneumonia. Lung paraffin sections of COVID-19 infected patients were analyzed by HE (hematoxylin-eosin) staining. The deposition of complement C3, the deposition of C3b/iC3b/C3d and C5b-9, and the expression of complement regulatory proteins, CD59, CD46 and CD55 were detected by immunohistochemistry. In COVID-19 patients' lung tissues, fibrin exudation, mixed with erythrocyte, alveolar macrophage and shed pneumocyte are usually observed in the alveoli. The formation of an "alveolar emboli" structure may contribute to thrombosis and consolidation in lung tissue. In addition, we also found that compared to normal tissue, the lung tissues of COVID-19 patients displayed the hyper-activation of complement that is represented by extensive deposition of C3, C3b/iC3b/C3d and C5b-9, and the increased expression level of complement regulatory proteins CD55, and especially CD59 but not CD46. The thrombosis and consolidation in lung tissues may contribute to the pathogenesis of COVID-19. The increased expression of CD55 and CD59 may reflect a feedback of self-protection on the complement hyper-activation. Further, the increased C3 deposition and the strongly activated complement system in lung tissues may suggest the rationale of complement-targeted therapeutics in conquering COVID-19.


Sujets)
COVID-19 , Complexe d'attaque membranaire du complément , Humains , Antigènes CD46 , Antigènes CD55 , Poumon , Complément C3b
2.
J Biol Chem ; 299(3): 102930, 2023 03.
Article Dans Anglais | MEDLINE | ID: covidwho-2180107

Résumé

Hyperactivation of the complement system, a major component of innate immunity, has been recognized as one of the core clinical features in severe covid-19 patients. However, how the virus escapes the targeted elimination by the network of activated complement pathways still remains an enigma. Here, we identified SARS-CoV-2-encoded ORF8 protein as one of the major binding partners of human complement C3/C3b components and their metabolites. Our results demonstrated that preincubation of ORF8 with C3/C3b in the fluid phase has two immediate functional consequences in the alternative pathway; this preincubation inhibits factor I-mediated proteolysis and blocks factor B zymogen activation into active Bb. ORF8 binding results in the occlusion of both factor H and factor B from C3b, rendering the complexes resistant to factor I-mediated proteolysis and inhibition of pro-C3-convertase (C3bB) formation, respectively. We also confirmed the complement inhibitory activity of ORF8 in our hemolysis-based assay, where ORF8 prevented human serum-induced lysis of rabbit erythrocytes with an IC50 value of about 2.3 µM. This inhibitory characteristic of ORF8 was also supported by in-silico protein-protein docking analysis, as it appeared to establish primary interactions with the ß-chain of C3b, orienting itself near the C3b CUB (C1r/C1s, Uegf, Bmp1) domain like a peptidomimetic compound, sterically hindering the binding of essential cofactors required for complement amplification. Thus, ORF8 has characteristics to act as an inhibitor of critical regulatory steps in the alternative pathway, converging to hasten the decay of C3-convertase and thereby, attenuating the complement amplification loop.


Sujets)
COVID-19 , Facteur B du complément , Animaux , Humains , Lapins , Activation du complément , Complement C3-C5 Convertases/métabolisme , Complément C3b/métabolisme , Facteur B du complément/métabolisme , Facteur H du complément/métabolisme , Voie alterne d'activation du complément/physiologie , SARS-CoV-2/métabolisme , Liaison aux protéines , Simulation numérique
3.
Front Immunol ; 13: 946522, 2022.
Article Dans Anglais | MEDLINE | ID: covidwho-2022727

Résumé

Numerous publications have underlined the link between complement C5a and the clinical course of COVID-19. We previously reported that levels of C5a remain high in the group of severely ill patients up to 90 days after hospital discharge. We have now evaluated which complement pathway fuels the elevated levels of C5a during hospitalization and follow-up. The alternative pathway (AP) activation marker C3bBbP and the soluble fraction of C4d, a footprint of the classical/lectin (CP/LP) pathway, were assessed by immunoenzymatic assay in a total of 188 serial samples from 49 patients infected with SARS-CoV-2. Unlike C5a, neither C3bBbP nor C4d readouts rose proportionally to the severity of the disease. Detailed correlation analyses in hospitalization and follow-up samples collected from patients of different disease severity showed significant positive correlations of AP and CP/LP markers with C5a in certain groups, except for the follow-up samples of the patients who suffered from highly severe COVID-19 and presented the highest C5a readouts. In conclusion, there is not a clear link between persistently high levels of C5a after hospital discharge and markers of upstream complement activation, suggesting the existence of a non-canonical source of C5a in patients with a severe course of COVID-19.


Sujets)
COVID-19 , Activation du complément , Complément C3b , Complément C4b , Complément C5a , Facteur B du complément , Fragments peptidiques , Marqueurs biologiques/sang , COVID-19/sang , COVID-19/immunologie , Activation du complément/immunologie , Complément C3b/immunologie , Complément C4b/immunologie , Complément C5a/analyse , Complément C5a/immunologie , Facteur B du complément/immunologie , Protéines du système du complément/immunologie , Humains , Fragments peptidiques/immunologie , SARS-CoV-2
4.
Nat Immunol ; 23(1): 62-74, 2022 01.
Article Dans Anglais | MEDLINE | ID: covidwho-1514418

Résumé

The molecular mechanisms governing orderly shutdown and retraction of CD4+ type 1 helper T (TH1) cell responses remain poorly understood. Here we show that complement triggers contraction of TH1 responses by inducing intrinsic expression of the vitamin D (VitD) receptor and the VitD-activating enzyme CYP27B1, permitting T cells to both activate and respond to VitD. VitD then initiated the transition from pro-inflammatory interferon-γ+ TH1 cells to suppressive interleukin-10+ cells. This process was primed by dynamic changes in the epigenetic landscape of CD4+ T cells, generating super-enhancers and recruiting several transcription factors, notably c-JUN, STAT3 and BACH2, which together with VitD receptor shaped the transcriptional response to VitD. Accordingly, VitD did not induce interleukin-10 expression in cells with dysfunctional BACH2 or STAT3. Bronchoalveolar lavage fluid CD4+ T cells of patients with COVID-19 were TH1-skewed and showed de-repression of genes downregulated by VitD, from either lack of substrate (VitD deficiency) and/or abnormal regulation of this system.


Sujets)
Interféron gamma/immunologie , Interleukine-10/immunologie , SARS-CoV-2/immunologie , Lymphocytes auxiliaires Th1/immunologie , Vitamine D/métabolisme , 25-Hydroxyvitamine D3 1-alpha-hydroxylase/métabolisme , Facteurs de transcription à motif basique et à glissière à leucines/métabolisme , Liquide de lavage bronchoalvéolaire/cytologie , COVID-19/immunologie , COVID-19/anatomopathologie , Complément C3a/immunologie , Complément C3b/immunologie , Humains , JNK Mitogen-Activated Protein Kinases/métabolisme , Activation des lymphocytes/immunologie , Récepteur calcitriol/métabolisme , /immunologie , /anatomopathologie , /virologie , Facteur de transcription STAT-3/métabolisme , Transduction du signal/immunologie , Transcription génétique/génétique
5.
Am J Physiol Lung Cell Mol Physiol ; 321(2): L485-L489, 2021 08 01.
Article Dans Anglais | MEDLINE | ID: covidwho-1299247

Résumé

COVID-19, the disease caused by the SARS-CoV-2 virus, can progress to multisystem organ failure and viral sepsis characterized by respiratory failure, arrhythmias, thromboembolic complications, and shock with high mortality. Autopsy and preclinical evidence implicate aberrant complement activation in endothelial injury and organ failure. Erythrocytes express complement receptors and are capable of binding immune complexes; therefore, we investigated complement activation in patients with COVID-19 using erythrocytes as a tool to diagnose complement activation. We discovered enhanced C3b and C4d deposition on erythrocytes in COVID-19 sepsis patients and non-COVID sepsis patients compared with healthy controls, supporting the role of complement in sepsis-associated organ injury. Our data suggest that erythrocytes may contribute to a precision medicine approach to sepsis and have diagnostic value in monitoring complement dysregulation in COVID-19-sepsis and non-COVID sepsis and identifying patients who may benefit from complement targeted therapies.


Sujets)
COVID-19/complications , Activation du complément/immunologie , Complément C3b/immunologie , Complément C4b/immunologie , Érythrocytes/immunologie , Fragments peptidiques/immunologie , Insuffisance respiratoire/diagnostic , Sepsie/diagnostic , COVID-19/immunologie , COVID-19/virologie , Complément C3b/métabolisme , Complément C4b/métabolisme , Érythrocytes/métabolisme , Érythrocytes/virologie , Femelle , Humains , Mâle , Adulte d'âge moyen , Fragments peptidiques/métabolisme , Insuffisance respiratoire/immunologie , Insuffisance respiratoire/métabolisme , Insuffisance respiratoire/virologie , SARS-CoV-2/isolement et purification , Sepsie/immunologie , Sepsie/métabolisme , Sepsie/virologie
6.
EMBO Mol Med ; 12(8): e12642, 2020 08 07.
Article Dans Anglais | MEDLINE | ID: covidwho-607958

Résumé

A novel coronavirus, SARS-CoV-2, has recently emerged in China and spread internationally, posing a health emergency to the global community. COVID-19 caused by SARS-CoV-2 is associated with an acute respiratory illness that varies from mild to the life-threatening acute respiratory distress syndrome (ARDS). The complement system is part of the innate immune arsenal against pathogens, in which many viruses can evade or employ to mediate cell entry. The immunopathology and acute lung injury orchestrated through the influx of pro-inflammatory macrophages and neutrophils can be directly activated by complement components to prime an overzealous cytokine storm. The manifestations of severe COVID-19 such as the ARDS, sepsis and multiorgan failure have an established relationship with activation of the complement cascade. We have collected evidence from all the current studies we are aware of on SARS-CoV-2 immunopathogenesis and the preceding literature on SARS-CoV-1 and MERS-CoV infection linking severe COVID-19 disease directly with dysfunction of the complement pathways. This information lends support for a therapeutic anti-inflammatory strategy against complement, where a number of clinically ready potential therapeutic agents are available.


Sujets)
Betacoronavirus , Activation du complément/effets des médicaments et des substances chimiques , Inhibiteurs du complément/usage thérapeutique , Infections à coronavirus/traitement médicamenteux , Pandémies , Pneumopathie virale/traitement médicamenteux , Adulte , Pneumocytes/immunologie , Pneumocytes/métabolisme , Pneumocytes/virologie , Angiotensin-converting enzyme 2 , Animaux , Betacoronavirus/physiologie , COVID-19 , Enfant , Complément C3b/antagonistes et inhibiteurs , Complément C3b/physiologie , Inhibiteurs du complément/pharmacologie , Infections à coronavirus/immunologie , Syndrome de libération de cytokines/traitement médicamenteux , Syndrome de libération de cytokines/étiologie , Syndrome de libération de cytokines/immunologie , Glycosylation , Humains , Immunité innée , Ligands , Souris , Modèles animaux , Modèles moléculaires , Reconnaissance automatique des formes , Peptidyl-Dipeptidase A/métabolisme , Pneumopathie virale/immunologie , Conformation des protéines , Maturation post-traductionnelle des protéines , Récepteurs viraux/métabolisme , /étiologie , /immunologie , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/métabolisme ,
SÉLECTION CITATIONS
Détails de la recherche