Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
J Am Soc Nephrol ; 32(2): 479-494, 2021 02.
Article in English | MEDLINE | ID: covidwho-1496663

ABSTRACT

BACKGROUND: Binding of donor-specific antibodies (DSAs) to kidney allograft endothelial cells that does not activate the classic complement cascade can trigger the recruitment of innate immune effectors, including NK cells. Activated NK cells contribute to microvascular inflammation leading to chronic antibody-mediated rejection (AMR). Recipient NK cells can also trigger antibody-independent microvascular inflammation by sensing the absence of self HLA class I molecules ("missing self") on allograft endothelial cells. This translational study investigated whether the condition of missing self amplifies DSA-dependent NK cell activation to worsen chronic AMR. METHODS AND RESULTS: Among 1682 kidney transplant recipients who underwent an allograft biopsy at Lyon University Hospital between 2004 and 2017, 135 fulfilled the diagnostic criteria for AMR and were enrolled in the study. Patients with complement-fixing DSAs identified by a positive C3d binding assay (n=73, 54%) had a higher risk of transplant failure (P=0.002). Among the remaining patients with complement-independent chronic AMR (n=62, 46%), those in whom missing self was identified through donor and recipient genotyping exhibited worse allograft survival (P=0.02). In multivariable analysis, only proteinuria (HR: 7.24; P=0.01) and the presence of missing self (HR: 3.57; P=0.04) were independent predictors for transplant failure following diagnosis of chronic AMR. Cocultures of human NK cells and endothelial cells confirmed that addition of missing self to DSA-induced NK cell activation increased endothelial damage. CONCLUSIONS: The assessment of missing self at the time of diagnosis of chronic AMR identifies patients at higher risk for kidney transplant failure.


Subject(s)
Allografts/pathology , Complement Activation/physiology , Graft Rejection/etiology , Histocompatibility Antigens Class I/blood , Kidney Transplantation/adverse effects , Killer Cells, Natural/physiology , Adult , Allografts/immunology , Cell Culture Techniques , Complement C3d/metabolism , Endothelial Cells/physiology , Female , Graft Rejection/blood , Graft Rejection/pathology , Graft Survival , Humans , Killer Cells, Natural/pathology , Male , Middle Aged , Young Adult
2.
Med Sci (Paris) ; 37(4): 333-341, 2021 Apr.
Article in French | MEDLINE | ID: covidwho-1174712

ABSTRACT

The complement system is an essential component of the innate immune system. Its excessive activation during COVID-19 contributes to cytokine storm, disease-specific endothelial inflammation (endotheliitis) and thrombosis that comes with the disease. Targeted therapies of complement inhibition in COVID-19, in particular blocking the C5a-C5aR1 axis have to be taken into account in the establishment of potential biomarkers and development of therapeutic strategies in the most severe forms of the disease.


TITLE: Implication de la cascade du complément dans les formes sévères de COVID-19. ABSTRACT: Le système du complément est un composant essentiel du système immunitaire inné. Son activation excessive au cours de la COVID-19 participe à l'orage cytokinique, à l'inflammation endothéliale (endothélite) et aux thromboses qui accompagnent la maladie. Bloquer le complément, notamment l'axe C5a-C5aR1, par des thérapies spécifiques représente un espoir thérapeutique dans les formes les plus sévères de la maladie.


Subject(s)
COVID-19/immunology , COVID-19/pathology , Complement Activation/physiology , Complement System Proteins/physiology , Animals , COVID-19/metabolism , Complement C5a/immunology , Complement C5a/metabolism , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/pathology , Humans , Inflammation/complications , Inflammation/immunology , Inflammation/pathology , SARS-CoV-2/immunology , Severity of Illness Index , Signal Transduction/immunology
3.
Front Immunol ; 11: 594849, 2020.
Article in English | MEDLINE | ID: covidwho-1083385

ABSTRACT

Most patients who became critically ill following infection with COVID-19 develop severe acute respiratory syndrome (SARS) attributed to a maladaptive or inadequate immune response. The complement system is an important component of the innate immune system that is involved in the opsonization of viruses but also in triggering further immune cell responses. Complement activation was seen in plasma adsorber material that clogged during the treatment of critically ill patients with COVID-19. Apart from the lung, the kidney is the second most common organ affected by COVID-19. Using immunohistochemistry for complement factors C1q, MASP-2, C3c, C3d, C4d, and C5b-9 we investigated the involvement of the complement system in six kidney biopsies with acute kidney failure in different clinical settings and three kidneys from autopsy material of patients with COVID-19. Renal tissue was analyzed for signs of renal injury by detection of thrombus formation using CD61, endothelial cell rarefaction using the marker E-26 transformation specific-related gene (ERG-) and proliferation using proliferating cell nuclear antigen (PCNA)-staining. SARS-CoV-2 was detected by in situ hybridization and immunohistochemistry. Biopsies from patients with hemolytic uremic syndrome (HUS, n = 5), severe acute tubular injury (ATI, n = 7), zero biopsies with disseminated intravascular coagulation (DIC, n = 7) and 1 year protocol biopsies from renal transplants (Ctrl, n = 7) served as controls. In the material clogging plasma adsorbers used for extracorporeal therapy of patients with COVID-19 C3 was the dominant protein but collectin 11 and MASP-2 were also identified. SARS-CoV-2 was sporadically present in varying numbers in some biopsies from patients with COVID-19. The highest frequency of CD61-positive platelets was found in peritubular capillaries and arteries of COVID-19 infected renal specimens as compared to all controls. Apart from COVID-19 specimens, MASP-2 was detected in glomeruli with DIC and ATI. In contrast, the classical pathway (i.e. C1q) was hardly seen in COVID-19 biopsies. Both C3 cleavage products C3c and C3d were strongly detected in renal arteries but also occurs in glomerular capillaries of COVID-19 biopsies, while tubular C3d was stronger than C3c in biopsies from COVID-19 patients. The membrane attack complex C5b-9, demonstrating terminal pathway activation, was predominantly deposited in COVID-19 biopsies in peritubular capillaries, renal arterioles, and tubular basement membrane with similar or even higher frequency compared to controls. In conclusion, various complement pathways were activated in COVID-19 kidneys, the lectin pathway mainly in peritubular capillaries and in part the classical pathway in renal arteries whereas the alternative pathway seem to be crucial for tubular complement activation. Therefore, activation of the complement system might be involved in the worsening of renal injury. Complement inhibition might thus be a promising treatment option to prevent deregulated activation and subsequent collateral tissue injury.


Subject(s)
Acute Kidney Injury/immunology , Acute Kidney Injury/virology , COVID-19/complications , COVID-19/immunology , Complement Activation/physiology , Adult , Aged , Female , Humans , Male , Middle Aged , SARS-CoV-2
4.
Cytokine Growth Factor Rev ; 58: 75-81, 2021 04.
Article in English | MEDLINE | ID: covidwho-971009

ABSTRACT

Emerging evidence has documented that multisystem organ failure in coronavirus disease 2019 (COVID-19) patients is strongly associated with various coagulopathies. Treatments for COVID-19-associated coagulopathy are still a clinical challenge. An advancement in the knowledge of mechanisms of the excessive or inappropriate activation of the complement cascade involved in the genesis of COVID-19-associated coagulopathy might be a fundamental approach for developing novel classes of anticoagulant drugs. In this context, there is emerging evidence indicating that C5a, a component of the complement system, and its receptors (C5aRs) play a critical role in the genesis of the COVID-19-associated hypercoagulable state. Thus, this review describes the mechanisms by which C5a/C5aR signaling participates in the cascade of events involved in the pathophysiology of COVID-19-associated coagulopathy. Furthermore, it highlights the current possibilities for the development of a novel therapeutic approach for COVID-19 patients that targets C5a/C5aRs signaling.


Subject(s)
COVID-19/therapy , Complement C5a/physiology , Complement C5a/therapeutic use , Thrombophilia/therapy , Animals , COVID-19/blood , COVID-19/complications , COVID-19/epidemiology , Complement Activation/physiology , Humans , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Signal Transduction , Thrombophilia/epidemiology , Thrombophilia/etiology
5.
Transl Res ; 231: 55-63, 2021 05.
Article in English | MEDLINE | ID: covidwho-939331

ABSTRACT

Although some evidence showed the activation of complement systems in COVID-19 patients, proinflammatory status and lectin pathway remain unclear. Thus, the present study aimed to demonstrate the role of MBL and ficolin-3 in the complement system activation and compared to pandemic Influenza A virus H1N1 subtype infection (H1N1pdm09) and control patients. A total of 27 lungs formalin-fixed paraffin-embedded samples (10 from H1N1 group, 6 from the COVID-19 group, and 11 from the control group) were analyzed by immunohistochemistry using anti-IL-6, TNF-alfa, CD163, MBL e FCN3 antibodies. Genotyping of target polymorphisms in the MBL2 gene was performed by real-time PCR. Proinflammatory cytokines such as IL-6 and TNF-alpha presented higher tissue expression in the COVID-19 group compared to H1N1 and control groups. The same results were observed for ICAM-1 tissue expression. Increased expression of the FCN3 was observed in the COVID-19 group and H1N1 group compared to the control group. The MBL tissue expression was higher in the COVID-19 group compared to H1N1 and control groups. The genotypes AA for rs180040 (G/A), GG for rs1800451 (G/A) and CC for rs5030737 (T/C) showed a higher prevalence in the COVID-19 group. The intense activation of the lectin pathway, with particular emphasis on the MBL pathway, together with endothelial dysfunction and a massive proinflammatory cytokines production, possibly lead to a worse outcome in patients infected with SARS-Cov-2. Moreover, 3 SNPs of our study presented genotypes that might be correlated with high MBL tissue expression in the COVID-19 pulmonary samples.


Subject(s)
COVID-19/pathology , Lectins/metabolism , Lung Injury/metabolism , Lung Injury/pathology , SARS-CoV-2 , Adult , Aged , Aged, 80 and over , Autopsy , Case-Control Studies , Complement Activation/physiology , Cytokines/genetics , Cytokines/metabolism , Female , Genotype , Humans , Immunohistochemistry , Influenza A Virus, H1N1 Subtype , Influenza, Human/metabolism , Influenza, Human/pathology , Lung/pathology , Lung/virology , Lung Injury/virology , Male , Middle Aged , Polymorphism, Single Nucleotide , Young Adult
6.
J Autoimmun ; 116: 102560, 2021 01.
Article in English | MEDLINE | ID: covidwho-899086

ABSTRACT

BACKGROUND: Animal models and few clinical reports suggest the involvement of the complement system in the onset of severe manifestations of coronavirus disease-2019 (COVID-19). However, complement contribution to endotheliopathy and hypercoagulability has not been elucidated yet. OBJECTIVE: To evaluate the association among complement activation, endothelial damage and disease severity or activity in COVID-19 patients. METHODS: In this single-centre cohort study, 148 patients with COVID-19 of different severity were evaluated upon hospital admission and 30 days later. Markers of complement activation (SC5b-9 and C5a) and endothelial perturbation (von Willebrand factor [vWF], tissue-type plasminogen activator [t-PA], plasminogen activator inhibitor-1 [PAI-1], soluble thrombomodulin [sTM], and soluble endothelial selectin [sE-selectin]) were measured in plasma. RESULTS: The patients had high plasma levels of SC5b-9 and C5a (p = 0.0001 for both) and vWF, t-PA and PAI-1 (p = 0.0001 for all). Their SC5b-9 levels correlated with those of vWF (r = 0.517, p = 0.0001) and paralleled disease severity (severe vs mild p = 0.0001, severe vs moderate p = 0.026 and moderate vs mild p = 0.001). The levels of sE-selectin were significantly increased only in the patients with severe disease. After 30 days, plasma SC5b-9, C5a and vWF levels had significantly decreased (p = 0.0001 for all), and 43% of the evaluated patients had normal levels. CONCLUSIONS: Complement activation is boosted during the progression of COVID-19 and dampened during remission, thus indicating its role in the pathophysiology of the disease. The association between complement activation and the biomarkers of endothelial damage suggests that complement may contribute to tissue injury and could be the target of specific therapy.


Subject(s)
Biomarkers/blood , COVID-19/blood , Complement Activation/physiology , Endothelium, Vascular/physiopathology , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Humans , Male , Middle Aged , SARS-CoV-2
7.
J Immunol ; 205(6): 1488-1495, 2020 09 15.
Article in English | MEDLINE | ID: covidwho-662455

ABSTRACT

Coronavirus disease of 2019 (COVID-19) is a highly contagious respiratory infection that is caused by the severe acute respiratory syndrome coronavirus 2. Although most people are immunocompetent to the virus, a small group fail to mount an effective antiviral response and develop chronic infections that trigger hyperinflammation. This results in major complications, including acute respiratory distress syndrome, disseminated intravascular coagulation, and multiorgan failure, which all carry poor prognoses. Emerging evidence suggests that the complement system plays a key role in this inflammatory reaction. Indeed, patients with severe COVID-19 show prominent complement activation in their lung, skin, and sera, and those individuals who were treated with complement inhibitors all recovered with no adverse reactions. These and other studies hint at complement's therapeutic potential in these sequalae, and thus, to support drug development, in this review, we provide a summary of COVID-19 and review complement's role in COVID-19 acute respiratory distress syndrome and coagulopathy.


Subject(s)
Blood Coagulation Disorders/virology , Complement Activation/physiology , Coronavirus Infections/complications , Pneumonia, Viral/complications , Respiratory Distress Syndrome/virology , Animals , Betacoronavirus/immunology , Blood Coagulation/drug effects , Blood Coagulation Disorders/immunology , COVID-19 , Complement Activation/drug effects , Complement Inactivating Agents/therapeutic use , Complement System Proteins/drug effects , Coronavirus Infections/blood , Coronavirus Infections/immunology , Humans , Inflammation/immunology , Inflammation/virology , Pandemics , Pneumonia, Viral/blood , Pneumonia, Viral/immunology , SARS-CoV-2
8.
Transl Res ; 220: 1-13, 2020 06.
Article in English | MEDLINE | ID: covidwho-60383

ABSTRACT

Acute respiratory failure and a systemic coagulopathy are critical aspects of the morbidity and mortality characterizing infection with severe acute respiratory distress syndrome-associated coronavirus-2, the etiologic agent of Coronavirus disease 2019 (COVID-19). We examined skin and lung tissues from 5 patients with severe COVID-19 characterized by respiratory failure (n= 5) and purpuric skin rash (n = 3). COVID-19 pneumonitis was predominantly a pauci-inflammatory septal capillary injury with significant septal capillary mural and luminal fibrin deposition and permeation of the interalveolar septa by neutrophils. No viral cytopathic changes were observed and the diffuse alveolar damage (DAD) with hyaline membranes, inflammation, and type II pneumocyte hyperplasia, hallmarks of classic acute respiratory distress syndrome, were not prominent. These pulmonary findings were accompanied by significant deposits of terminal complement components C5b-9 (membrane attack complex), C4d, and mannose binding lectin (MBL)-associated serine protease (MASP)2, in the microvasculature, consistent with sustained, systemic activation of the complement pathways. The purpuric skin lesions similarly showed a pauci-inflammatory thrombogenic vasculopathy, with deposition of C5b-9 and C4d in both grossly involved and normally-appearing skin. In addition, there was co-localization of COVID-19 spike glycoproteins with C4d and C5b-9 in the interalveolar septa and the cutaneous microvasculature of 2 cases examined. In conclusion, at least a subset of sustained, severe COVID-19 may define a type of catastrophic microvascular injury syndrome mediated by activation of complement pathways and an associated procoagulant state. It provides a foundation for further exploration of the pathophysiologic importance of complement in COVID-19, and could suggest targets for specific intervention.


Subject(s)
Betacoronavirus , Complement System Proteins/metabolism , Coronavirus Infections/complications , Microvessels/pathology , Pneumonia, Viral/complications , Respiratory Insufficiency/etiology , Thrombosis/etiology , Adult , Aged , COVID-19 , Complement Activation/physiology , Coronavirus Infections/pathology , Female , Humans , Male , Microvessels/virology , Middle Aged , Pandemics , Pneumonia, Viral/pathology , Purpura/etiology , Purpura/pathology , Purpura/virology , Respiratory Insufficiency/pathology , SARS-CoV-2 , Thrombosis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL