Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Med Microbiol Immunol ; 210(2-3): 101-109, 2021 Jun.
Article in English | MEDLINE | ID: covidwho-1163038

ABSTRACT

The current outbreak of coronavirus disease 2019 (COVID-19) has affected people around the world. Typically, COVID-19 originates in the lung, but lately it can extend to other organs and lead to tissue injury and multiorgan failure in severe patients, such as acute respiratory distress syndrome (ARDS), kidney failure and sepsis or systemic inflammation. Given that COVID-19 has been detected in a range of other organs, the COVID-19-associated disease is an alert of aberrant activation of host immune response which drives un-controlled inflammation that affects multiple organs. Complement is a vital component of innate immunity where it forms the first line of defense against potentially harmful microbes, but its role in COVID-19 is still not clear. Notably, the abnormal activation and continuous deposits of complement components were identified in the pre-clinical samples from COVID-19 patients, which have been confirmed in animal models. Recent evidence has revealed that the administration of complement inhibitors leads to relieve inflammatory response in ARDS. Hence, we speculate that the targeting complement system could be a potential treatment option for organ damage in COVID-19 patients.


Subject(s)
COVID-19/immunology , Complement System Proteins/immunology , Immunity, Innate , Inflammation/immunology , Respiratory Distress Syndrome/immunology , Animals , COVID-19/pathology , Complement Inactivating Agents/pharmacology , Humans , Inflammation/virology , Lung/immunology , Lung/virology , Respiratory Distress Syndrome/virology
2.
Clin Immunol ; 226: 108716, 2021 05.
Article in English | MEDLINE | ID: covidwho-1152310

ABSTRACT

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.


Subject(s)
COVID-19/immunology , Complement Activation/immunology , Complement Pathway, Alternative/immunology , Lung Injury/immunology , Aged , Aged, 80 and over , COVID-19/complications , Complement Inactivating Agents/pharmacology , Complement Inactivating Agents/therapeutic use , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Immunohistochemistry , Lung/diagnostic imaging , Lung/immunology , Lung/pathology , Lung Injury/complications , Lung Injury/pathology , Lung Injury/virology , Male , Middle Aged
3.
Pharmacol Rev ; 73(2): 792-827, 2021 04.
Article in English | MEDLINE | ID: covidwho-1125571

ABSTRACT

The complement system was discovered at the end of the 19th century as a heat-labile plasma component that "complemented" the antibodies in killing microbes, hence the name "complement." Complement is also part of the innate immune system, protecting the host by recognition of pathogen-associated molecular patterns. However, complement is multifunctional far beyond infectious defense. It contributes to organ development, such as sculpting neuron synapses, promoting tissue regeneration and repair, and rapidly engaging and synergizing with a number of processes, including hemostasis leading to thromboinflammation. Complement is a double-edged sword. Although it usually protects the host, it may cause tissue damage when dysregulated or overactivated, such as in the systemic inflammatory reaction seen in trauma and sepsis and severe coronavirus disease 2019 (COVID-19). Damage-associated molecular patterns generated during ischemia-reperfusion injuries (myocardial infarction, stroke, and transplant dysfunction) and in chronic neurologic and rheumatic disease activate complement, thereby increasing damaging inflammation. Despite the long list of diseases with potential for ameliorating complement modulation, only a few rare diseases are approved for clinical treatment targeting complement. Those currently being efficiently treated include paroxysmal nocturnal hemoglobinuria, atypical hemolytic-uremic syndrome, myasthenia gravis, and neuromyelitis optica spectrum disorders. Rare diseases, unfortunately, preclude robust clinical trials. The increasing evidence for complement as a pathogenetic driver in many more common diseases suggests an opportunity for future complement therapy, which, however, requires robust clinical trials; one ongoing example is COVID-19 disease. The current review aims to discuss complement in disease pathogenesis and discuss future pharmacological strategies to treat these diseases with complement-targeted therapies. SIGNIFICANCE STATEMENT: The complement system is the host's defense friend by protecting it from invading pathogens, promoting tissue repair, and maintaining homeostasis. Complement is a double-edged sword, since when dysregulated or overactivated it becomes the host's enemy, leading to tissue damage, organ failure, and, in worst case, death. A number of acute and chronic diseases are candidates for pharmacological treatment to avoid complement-dependent damage, ranging from the well established treatment for rare diseases to possible future treatment of large patient groups like the pandemic coronavirus disease 2019.


Subject(s)
COVID-19/epidemiology , COVID-19/physiopathology , Complement System Proteins/physiology , Rare Diseases/physiopathology , Collectins/metabolism , Complement Activating Enzymes/metabolism , Complement C3/metabolism , Complement Inactivating Agents/pharmacology , Genetic Therapy/methods , Humans , Inflammation Mediators/metabolism , Lectins/metabolism , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Pandemics , SARS-CoV-2 , Synapses/metabolism
4.
Life Sci ; 272: 119245, 2021 May 01.
Article in English | MEDLINE | ID: covidwho-1087130

ABSTRACT

In the past 20 years, infections caused by coronaviruses SARS-CoV, MERS-CoV and SARS-CoV-2 have posed a threat to public health since they may cause severe acute respiratory syndrome (SARS) in humans. The Complement System is activated during viral infection, being a central protagonist of innate and acquired immunity. Here, we report some interactions between these three coronaviruses and the Complement System, highlighting the central role of C3 with the severity of these infections. Although it can be protective, its role during coronavirus infections seems to be contradictory. For example, during SARS-CoV-2 infection, Complement System can control the viral infection in asymptomatic or mild cases; however, it can also intensify local and systemic damage in some of severe COVID-19 patients, due to its potent proinflammatory effect. In this last condition, the activation of the Complement System also amplifies the cytokine storm and the pathogenicity of coronavirus infection. Experimental treatment with Complement inhibitors has been an enthusiastic field of intense investigation in search of a promising additional therapy in severe COVID-19 patients.


Subject(s)
COVID-19/immunology , Complement System Proteins/immunology , SARS-CoV-2/immunology , Animals , COVID-19/complications , COVID-19/pathology , Complement Activation/drug effects , Complement C3/immunology , Complement Inactivating Agents/pharmacology , Complement Inactivating Agents/therapeutic use , Coronavirus Infections/complications , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Cytokine Release Syndrome/complications , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/pathology , Humans , Inflammation/complications , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Middle East Respiratory Syndrome Coronavirus/immunology , Severe acute respiratory syndrome-related coronavirus/immunology , Severe Acute Respiratory Syndrome/complications , Severe Acute Respiratory Syndrome/drug therapy , Severe Acute Respiratory Syndrome/immunology , Severe Acute Respiratory Syndrome/pathology , COVID-19 Drug Treatment
7.
Blood ; 136(18): 2080-2089, 2020 10 29.
Article in English | MEDLINE | ID: covidwho-740364

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a highly contagious respiratory virus that can lead to venous/arterial thrombosis, stroke, renal failure, myocardial infarction, thrombocytopenia, and other end-organ damage. Animal models demonstrating end-organ protection in C3-deficient mice and evidence of complement activation in humans have led to the hypothesis that SARS-CoV-2 triggers complement-mediated endothelial damage, but the mechanism is unclear. Here, we demonstrate that the SARS-CoV-2 spike protein (subunit 1 and 2), but not the N protein, directly activates the alternative pathway of complement (APC). Complement-dependent killing using the modified Ham test is blocked by either C5 or factor D inhibition. C3 fragments and C5b-9 are deposited on TF1PIGAnull target cells, and complement factor Bb is increased in the supernatant from spike protein-treated cells. C5 inhibition prevents the accumulation of C5b-9 on cells, but not C3c; however, factor D inhibition prevents both C3c and C5b-9 accumulation. Addition of factor H mitigates the complement attack. In conclusion, SARS-CoV-2 spike proteins convert nonactivator surfaces to activator surfaces by preventing the inactivation of the cell-surface APC convertase. APC activation may explain many of the clinical manifestations (microangiopathy, thrombocytopenia, renal injury, and thrombophilia) of COVID-19 that are also observed in other complement-driven diseases such as atypical hemolytic uremic syndrome and catastrophic antiphospholipid antibody syndrome. C5 inhibition prevents accumulation of C5b-9 in vitro but does not prevent upstream complement activation in response to SARS-CoV-2 spike proteins.


Subject(s)
Betacoronavirus , Complement Factor D/antagonists & inhibitors , Complement Inactivating Agents/pharmacology , Complement Pathway, Alternative/drug effects , Spike Glycoprotein, Coronavirus/pharmacology , Cell Line , Complement Activation/drug effects , Complement C3/metabolism , Complement C5/antagonists & inhibitors , Complement Factor H/metabolism , Complement Membrane Attack Complex/metabolism , Humans , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/physiology
8.
J Thromb Haemost ; 18(9): 2110-2117, 2020 09.
Article in English | MEDLINE | ID: covidwho-623519

ABSTRACT

COVID-19 is frequently accompanied by a hypercoagulable inflammatory state with microangiopathic pulmonary changes that can precede the diffuse alveolar damage characteristic of typical acute respiratory distress syndrome (ARDS) seen in other severe pathogenic infections. Parallels with systemic inflammatory disorders such as atypical hemolytic uremic syndrome (aHUS) have implicated the complement pathway in the pathogenesis of COVID-19, and particularly the anaphylatoxins C3a and C5a released from cleavage of C3 and C5, respectively. C5a is a potent cell signalling protein that activates a cytokine storm-a hyper-inflammatory phenomenon-within hours of infection and the innate immune response. However, excess C5a can result in a pro-inflammatory environment orchestrated through a plethora of mechanisms that propagate lung injury, lymphocyte exhaustion, and an immune paresis. Furthermore, disruption of the homeostatic interactions between complement and extrinsic and intrinsic coagulation pathways contributes to a net pro-coagulant state in the microvasculature of critical organs. Fatal COVID-19 has been associated with a systemic inflammatory response accompanied by a pro-coagulant state and organ damage, particularly microvascular thrombi in the lungs and kidneys. Pathologic studies report strong evidence of complement activation. C5 blockade reduces inflammatory cytokines and their manifestations in animal studies, and has shown benefits in patients with aHUS, prompting investigation of this approach in the treatment of COVID-19. This review describes the role of the complement pathway and particularly C5a and its aberrations in highly pathogenic virus infections, and therefore its potential as a therapeutic target in COVID-19.


Subject(s)
Blood Coagulation , COVID-19/immunology , Complement Activation , Complement C3a/metabolism , Complement C5a/metabolism , Inflammation/metabolism , Animals , Atypical Hemolytic Uremic Syndrome/complications , Atypical Hemolytic Uremic Syndrome/immunology , COVID-19/complications , COVID-19/pathology , Complement Inactivating Agents/pharmacology , Cytokines/metabolism , Hemoglobinuria, Paroxysmal/complications , Hemoglobinuria, Paroxysmal/immunology , Homeostasis , Humans , Immunity, Innate , Lung Diseases , Lung Injury , Signal Transduction
9.
EMBO Mol Med ; 12(8): e12642, 2020 08 07.
Article in English | MEDLINE | ID: covidwho-607958

ABSTRACT

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.


Subject(s)
Betacoronavirus , Complement Activation/drug effects , Complement Inactivating Agents/therapeutic use , Coronavirus Infections/drug therapy , Pandemics , Pneumonia, Viral/drug therapy , Adult , Alveolar Epithelial Cells/immunology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/virology , Angiotensin-Converting Enzyme 2 , Animals , Betacoronavirus/physiology , COVID-19 , Child , Complement C3b/antagonists & inhibitors , Complement C3b/physiology , Complement Inactivating Agents/pharmacology , Coronavirus Infections/immunology , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Glycosylation , Humans , Immunity, Innate , Ligands , Mice , Models, Animal , Models, Molecular , Pattern Recognition, Automated , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/immunology , Protein Conformation , Protein Processing, Post-Translational , Receptors, Virus/metabolism , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/immunology , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , COVID-19 Drug Treatment
SELECTION OF CITATIONS
SEARCH DETAIL