Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 358
Filter
1.
Mol Immunol ; 170: 19-25, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38598870

ABSTRACT

The assembly of tissue-damaging membrane attack complexes (MACs; C5b-9) is a major mechanism by which excessive complement activation causes diseases. We previously developed a mouse anti-human C6 monoclonal antibody (mAb) 1C9 that selectively inhibits the assembly of MACs in human and non-human primates. In this project, we found that 1C9 also cross-reacted with rat and guinea pig C6, and determined its binding domains on C6 using different truncated C6 proteins. We then humanized the anti-C6 mAb by molecular modeling and complementarity-determining region grafting. After screening a library of 276 humanized variants with different combinations of humanized light and heavy chains in biophysical assays, we identified clone 3713 with the best developability profile, and an increased affinity against C6 when compared with the parental 1C9 mAb. This humanized 3713 mAb inhibited human, monkey, and rat complement-mediated hemolysis in vitro, and more importantly, it significantly reduced complement-mediated hemolysis in vivo in rats. These results demonstrated the successful humanization of the anti-C6 mAb and suggested that the humanized 3713 mAb could be further developed as a new therapeutic that selectively targets MAC for certain complement-mediated pathological conditions.


Subject(s)
Antibodies, Monoclonal , Complement C6 , Hemolysis , Animals , Humans , Rats , Guinea Pigs , Mice , Hemolysis/drug effects , Hemolysis/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Complement C6/immunology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Complement Activation/immunology , Complement Activation/drug effects , Complement Membrane Attack Complex/immunology , Cross Reactions/immunology
2.
Proc Natl Acad Sci U S A ; 119(33): e2122716119, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35960843

ABSTRACT

The microenvironment of malignant melanomas defines the properties of tumor blood vessels and regulates infiltration and vascular dissemination of immune and cancer cells, respectively. Previous research in other cancer entities suggested the complement system as an essential part of the tumor microenvironment. Here, we confirm activation of the complement system in samples of melanoma patients and murine melanomas. We identified the tumor endothelium as the starting point of the complement cascade. Generation of complement-derived C5a promoted the recruitment of neutrophils. Upon contact with the vascular endothelium, neutrophils were further activated by complement membrane attack complexes (MACs). MAC-activated neutrophils release neutrophil extracellular traps (NETs). Close to the blood vessel wall, NETs opened the endothelial barrier as indicated by an enhanced vascular leakage. This facilitated the entrance of melanoma cells into the circulation and their systemic spread. Depletion of neutrophils or lack of MAC formation in complement component 6 (C6)-deficient animals protected the vascular endothelium and prevented vascular intravasation of melanoma cells. Our data suggest that inhibition of MAC-mediated neutrophil activation is a potent strategy to abolish hematogenous dissemination in melanoma.


Subject(s)
Complement Membrane Attack Complex , Endothelium, Vascular , Extracellular Traps , Melanoma , Neutrophils , Tumor Microenvironment , Animals , Complement Membrane Attack Complex/immunology , Complement System Proteins , Endothelium, Vascular/physiopathology , Humans , Melanoma/blood supply , Melanoma/immunology , Melanoma/pathology , Mice , Neutrophils/immunology , Permeability
3.
Mol Immunol ; 141: 273-279, 2022 01.
Article in English | MEDLINE | ID: mdl-34906905

ABSTRACT

BACKGROUND: Complement activation plays an important pathogenic role in numerous diseases. The ratio between an activation product and its parent protein is suggested to be more sensitive to detect complement activation than the activation product itself. In the present study we explored whether the ratio between the activation product and the parent protein for C3 (C3bc/C3) and for C5 (sC5b-9/C5) increased the sensitivity to detect complement activation in acute clinical settings compared to the activation product alone. MATERIALS AND METHODS: Samples from patients with acute heart failure following ST-elevated myocardial infarction (STEMI) and from patients with out-of-hospital cardiac arrest (OHCA) were used. C3, C3bc and C5, sC5b-9 were analysed in 629 and 672 patient samples, respectively. Healthy controls (n = 20) served to determine reference cut-off values for activation products and ratios, defined as two SD above the mean. RESULTS: Increased C3bc/C3- and sC5b-9/C5 ratios were vastly dependent on C3bc and sC5b-9. Thus, 99.5 % and 98.1 % of the increased C3bc/C3- and sC5b-9/C5 ratios were solely dependent on increased C3bc and sC5b-9, respectively. Significantly decreased C3 and C5 caused increased ratios in only 3/600 (0.5 %) and 4/319 (1.3 %) samples, respectively. Strong correlations between C3bc and C3bc/C3-ratio and between sC5b-9 and sC5b-9/C5-ratio were found in the STEMI- (r = 0.926 and r = 0.786, respectively) and the OHCA-population (r = 0.908 and r = 0.843, respectively; p < 0.0001 for all). Importantly, sC5b-9 identified worse outcome groups better than sC5b-9/C5-ratio. CONCLUSION: C3bc and sC5b-9 were sensitive markers of complement activation. The ratios of C3bc/C3 and sC5b-9/C5 did not improve detection of complement activation systemically.


Subject(s)
Complement Activation/immunology , Complement C3/immunology , Complement C3b/immunology , Complement C5/immunology , Complement Membrane Attack Complex/immunology , Peptide Fragments/immunology , Adult , Case-Control Studies , Female , Humans , Male , Middle Aged
4.
Nat Commun ; 12(1): 6086, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34667172

ABSTRACT

Unregulated complement activation causes inflammatory and immunological pathologies with consequences for human disease. To prevent bystander damage during an immune response, extracellular chaperones (clusterin and vitronectin) capture and clear soluble precursors to the membrane attack complex (sMAC). However, how these chaperones block further polymerization of MAC and prevent the complex from binding target membranes remains unclear. Here, we address that question by combining cryo electron microscopy (cryoEM) and cross-linking mass spectrometry (XL-MS) to solve the structure of sMAC. Together our data reveal how clusterin recognizes and inhibits polymerizing complement proteins by binding a negatively charged surface of sMAC. Furthermore, we show that the pore-forming C9 protein is trapped in an intermediate conformation whereby only one of its two transmembrane ß-hairpins has unfurled. This structure provides molecular details for immune pore formation and helps explain a complement control mechanism that has potential implications for how cell clearance pathways mediate immune homeostasis.


Subject(s)
Complement Membrane Attack Complex/chemistry , Complement Membrane Attack Complex/immunology , Complement C8/chemistry , Complement C8/metabolism , Complement C9/chemistry , Complement C9/immunology , Cryoelectron Microscopy , Humans , Protein Binding , Protein Conformation, alpha-Helical , Protein Domains
5.
Front Immunol ; 12: 720655, 2021.
Article in English | MEDLINE | ID: mdl-34650553

ABSTRACT

Interleukin 1ß (IL-1ß) plays a major role in inflammation and is secreted by immune cells, such as macrophages, upon recognition of danger signals. Its secretion is regulated by the inflammasome, the assembly of which results in caspase 1 activation leading to gasdermin D (GSDMD) pore formation and IL-1ß release. During inflammation, danger signals also activate the complement cascade, resulting in the formation of the membrane attack complex (MAC). Here, we report that stimulation of LPS-primed human macrophages with sub-lytic levels of MAC results in activation of the NOD-like receptor 3 (NLRP3) inflammasome and GSDMD-mediated IL-1ß release. The MAC is first internalized into endosomes and then colocalizes with inflammasome components; adapter protein apoptosis associated speck-like protein containing a CARD (ASC) and NLRP3. Pharmacological inhibitors established that MAC-triggered activation of the NLRP3 inflammasome was dependent on MAC endocytosis. Internalization of the MAC also caused dispersion of the trans-Golgi network. Thus, these data uncover a role for the MAC in activating the inflammasome and triggering IL-1ß release in human macrophages.


Subject(s)
Complement Membrane Attack Complex/immunology , Complement Membrane Attack Complex/metabolism , Inflammasomes/metabolism , Interleukin-1beta/biosynthesis , Macrophages/immunology , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Biomarkers , Cell Line , Cells, Cultured , Complement System Proteins/immunology , Endocytosis , Endosomes/metabolism , Humans , Macrophage Activation/immunology , Models, Biological , Protein Transport
6.
Mol Med ; 27(1): 35, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33832430

ABSTRACT

BACKGROUND: Complement activation is a central mechanism in systemic inflammation and remote organ dysfunction following major trauma. Data on temporal changes of complement activation early after injury is largely missing. We aimed to describe in detail the kinetics of complement activation in individual trauma patients from admission to 10 days after injury, and the association with trauma characteristics and outcome. METHODS: In a prospective cohort of 136 trauma patients, plasma samples obtained with high time resolution (admission, 2, 4, 6, 8 h, and thereafter daily) were assessed for terminal complement complex (TCC). We studied individual TCC concentration curves and calculated a summary measure to obtain the accumulated TCC response 3 to 6 h after injury (TCC-AUC3-6). Correlation analyses and multivariable linear regression analyses were used to explore associations between individual patients' admission TCC, TCC-AUC3-6, daily TCC during the intensive care unit stay, trauma characteristics, and predefined outcome measures. RESULTS: TCC concentration curves showed great variability in temporal shapes between individuals. However, the highest values were generally seen within the first 6 h after injury, before they subsided and remained elevated throughout the intensive care unit stay. Both admission TCC and TCC-AUC3-6 correlated positively with New Injury Severity Score (Spearman's rho, p-value 0.31, 0.0003 and 0.21, 0.02) and negatively with admission Base Excess (- 0.21, 0.02 and - 0.30, 0.001). Multivariable analyses confirmed that deranged physiology was an important predictor of complement activation. For patients without major head injury, admission TCC and TCC-AUC3-6 were negatively associated with ventilator-free days. TCC-AUC3-6 outperformed admission TCC as a predictor of Sequential Organ Failure Assessment score at day 0 and 4. CONCLUSIONS: Complement activation 3 to 6 h after injury was a better predictor of prolonged mechanical ventilation and multiple organ dysfunction syndrome than admission TCC. Our data suggest that the greatest surge of complement activation is found within the first 6 h after injury, and we argue that this time period should be in focus in the design of future experimental studies and clinical trials using complement inhibitors.


Subject(s)
Complement Activation , Craniocerebral Trauma/immunology , Multiple Organ Failure/immunology , Respiration, Artificial , Wounds and Injuries/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Complement Membrane Attack Complex/immunology , Craniocerebral Trauma/mortality , Female , Hospitalization , Humans , Male , Middle Aged , Prospective Studies , Severity of Illness Index , Syndrome , Time Factors , Wounds and Injuries/mortality , Young Adult
7.
Exp Mol Med ; 53(4): 572-590, 2021 04.
Article in English | MEDLINE | ID: mdl-33811247

ABSTRACT

Glomerular mesangial cell (GMC) proliferation is a histopathological alteration in human mesangioproliferative glomerulonephritis (MsPGN) or in animal models of MsPGN, e.g., the rat Thy-1 nephritis (Thy-1N) model. Although sublytic C5b-9 assembly on the GMC membrane can trigger cell proliferation, the mechanisms are still undefined. We found that sublytic C5b-9-induced rat GMC proliferation was driven by extracellular signal-regulated kinase 1/2 (ERK1/2), sry-related HMG-box 9 (SOX9), and Cyclin D1. Here, ERK1/2 phosphorylation was a result of the calcium influx-PKC-α-Raf-MEK1/2 axis activated by sublytic C5b-9, and Cyclin D1 gene transcription was enhanced by ERK1/2-dependent SOX9 binding to the Cyclin D1 promoter (-582 to -238 nt). In addition, ERK1/2 not only interacted with SOX9 in the cell nucleus to mediate its phosphorylation at serine residues 64 (a new site identified by mass spectrometry) and 181 (a known site), but also indirectly induced SOX9 acetylation by elevating the expression of general control non-repressed protein 5 (GCN5), which together resulted in Cyclin D1 synthesis and GMC proliferation. Moreover, our in vivo experiments confirmed that silencing these genes ameliorated the lesions of Thy-1N rats and reduced SOX9 phosphorylation, acetylation and Cyclin D1 expression. Furthermore, the renal tissue sections of MsPGN patients also showed higher phosphorylation or expression of ERK1/2, SOX9, and Cyclin D1. In summary, these findings suggest that sublytic C5b-9-induced GMC proliferation in rat Thy-1N requires SOX9 phosphorylation and acetylation via enhanced Cyclin D1 gene transcription, which may provide a new insight into human MsPGN pathogenesis.


Subject(s)
Complement Membrane Attack Complex/immunology , Cyclin D1/genetics , Glomerulonephritis/etiology , Glomerulonephritis/metabolism , MAP Kinase Signaling System , Mesangial Cells/immunology , Mesangial Cells/metabolism , SOX9 Transcription Factor/metabolism , Acetylation , Animals , Biomarkers , Calcium/metabolism , Calcium Signaling , Cell Proliferation , Cyclin D1/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression Regulation , Gene Knockdown Techniques , Glomerulonephritis/pathology , Male , Mesangial Cells/pathology , Models, Biological , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Rats , SOX9 Transcription Factor/genetics
8.
J Clin Invest ; 131(5)2021 03 01.
Article in English | MEDLINE | ID: mdl-33351779

ABSTRACT

Primary membranous nephropathy (pMN) is a leading cause of nephrotic syndrome in adults. In most cases, this autoimmune kidney disease is associated with autoantibodies against the M-type phospholipase A2 receptor (PLA2R1) expressed on kidney podocytes, but the mechanisms leading to glomerular damage remain elusive. Here, we developed a cell culture model using human podocytes and found that anti-PLA2R1-positive pMN patient sera or isolated IgG4, but not IgG4-depleted sera, induced proteolysis of the 2 essential podocyte proteins synaptopodin and NEPH1 in the presence of complement, resulting in perturbations of the podocyte cytoskeleton. Specific blockade of the lectin pathway prevented degradation of synaptopodin and NEPH1. Anti-PLA2R1 IgG4 directly bound mannose-binding lectin in a glycosylation-dependent manner. In a cohort of pMN patients, we identified increased levels of galactose-deficient IgG4, which correlated with anti-PLA2R1 titers and podocyte damage induced by patient sera. Assembly of the terminal C5b-9 complement complex and activation of the complement receptors C3aR1 or C5aR1 were required to induce proteolysis of synaptopodin and NEPH1 by 2 distinct proteolytic pathways mediated by cysteine and aspartic proteinases, respectively. Together, these results demonstrated a mechanism by which aberrantly glycosylated IgG4 activated the lectin pathway and induced podocyte injury in primary membranous nephropathy.


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases/immunology , Complement Pathway, Mannose-Binding Lectin/immunology , Glomerulonephritis, Membranous/immunology , Immunoglobulin G/immunology , Nephrotic Syndrome/immunology , Podocytes/immunology , Receptors, Phospholipase A2/immunology , Adult , Autoimmune Diseases/pathology , Carrier Proteins/immunology , Cell Line, Transformed , Complement Membrane Attack Complex/immunology , Glomerulonephritis, Membranous/pathology , Humans , Membrane Proteins/immunology , Microfilament Proteins/immunology , Nephrotic Syndrome/pathology , Podocytes/pathology , Receptor, Anaphylatoxin C5a/immunology , Receptors, Complement/immunology
9.
Front Immunol ; 11: 569549, 2020.
Article in English | MEDLINE | ID: mdl-33193347

ABSTRACT

The complement system has developed different strategies to clear infections by several effector mechanisms, such as opsonization, which supports phagocytosis, attracting immune cells by C3 and C5 cleavage products, or direct killing of pathogens by the formation of the membrane attack complex (MAC). As the Zika virus (ZIKV) activates the classical complement pathway and thus has to avoid clearance by the complement system, we analyzed putative viral escape mechanisms, which limit virolysis. We identified binding of the recombinant viral envelope E protein to components of the terminal pathway complement (C5b6, C7, C8, and C9) by ELISA. Western blot analyses revealed that ZIKV E protein interfered with the polymerization of C9, induced on cellular surfaces, either by purified terminal complement proteins or by normal human serum (NHS) as a source of the complement. Further, the hemolytic activity of NHS was significantly reduced in the presence of the recombinant E protein or entire viral particles. This data indicates that ZIKV reduces MAC formation and complement-mediated lysis by binding terminal complement proteins to the viral E protein.


Subject(s)
Complement Membrane Attack Complex/immunology , Complement Membrane Attack Complex/metabolism , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Zika Virus Infection/immunology , Zika Virus Infection/virology , Zika Virus/immunology , Cell Line , Cell Membrane/immunology , Cell Membrane/metabolism , Complement Activation/immunology , Complement C9/immunology , Complement C9/metabolism , Complement Pathway, Classical , Complement System Proteins/immunology , Complement System Proteins/metabolism , Host-Pathogen Interactions/immunology , Humans , Protein Binding , Protein Multimerization
10.
Front Immunol ; 11: 585108, 2020.
Article in English | MEDLINE | ID: mdl-33240274

ABSTRACT

The soluble membrane attack complex (sMAC, a.k.a., sC5b-9 or TCC) is generated on activation of complement and contains the complement proteins C5b, C6, C7, C8, C9 together with the regulatory proteins clusterin and/or vitronectin. sMAC is a member of the MACPF/cholesterol-dependent-cytolysin superfamily of pore-forming molecules that insert into lipid bilayers and disrupt cellular integrity and function. sMAC is a unique complement activation macromolecule as it is comprised of several different subunits. To date no complement-mediated function has been identified for sMAC. sMAC is present in blood and other body fluids under homeostatic conditions and there is abundant evidence documenting changes in sMAC levels during infection, autoimmune disease and trauma. Despite decades of scientific interest in sMAC, the mechanisms regulating its formation in healthy individuals and its biological functions in both health and disease remain poorly understood. Here, we review the structural differences between sMAC and its membrane counterpart, MAC, and examine sMAC immunobiology with respect to its presence in body fluids in health and disease. Finally, we discuss the diagnostic potential of sMAC for diagnostic and prognostic applications and potential utility as a companion diagnostic.


Subject(s)
Complement Activation/immunology , Complement Membrane Attack Complex/immunology , Animals , Humans
11.
Mol Immunol ; 128: 195-204, 2020 12.
Article in English | MEDLINE | ID: mdl-33142137

ABSTRACT

Membranous nephropathy (MN) is an immune complex mediated disease. Although limited to the kidney, in up to 20% of patients MN is associated with other autoimmune, infectious or malignant diseases. The initial pathogenetic event in what is still considered "primary" MN is the binding of circulating autoantibodies to proteins (autoantigens) expressed in glomerular podocytes. This antibody binding leads to the formation of immune complexes in the glomerular basement membrane. There is clinical and experimental evidence that these immune deposits lead to the activation of the complement system. Experimental studies in the MN model of Heymann's nephritis show that the terminal membrane attack complex (MAC) of the complement system induces a disturbance of the glomerular filtration barrier and leads to proteinuria, the clinical hallmark of MN. After the discovery of the phospholipase A2 receptor 1 and thrombospondin type 1 domain containing protein 7A as endogenous antigens, it is assumed that IgG4 antibodies directed against these proteins induce MN in over 85% of patients with primary MN. As a result, the role of complement in the pathogenesis of MN needs to be defined in light of these developments. In this review we describe the current knowledge on the function of the complement system in primary MN and discuss the open questions, which have to be solved for a better understanding of the potential role of complement in the pathophysiology of primary MN.


Subject(s)
Complement System Proteins/immunology , Glomerulonephritis, Membranous/immunology , Animals , Antigen-Antibody Complex/immunology , Autoantibodies/immunology , Autoantigens/immunology , Basement Membrane/immunology , Complement Membrane Attack Complex/immunology , Humans , Immunoglobulin G/immunology , Kidney/immunology , Kidney Glomerulus/immunology , Podocytes/immunology , Receptors, Phospholipase A2/immunology , Thrombospondins/immunology
12.
Clin Immunol ; 221: 108616, 2020 12.
Article in English | MEDLINE | ID: mdl-33148511

ABSTRACT

In complement-driven thrombotic microangiopathies, failure to regulate complement activation leads to end-organ damage. The modified Ham (mHam) test measures complement-mediated killing of a nucleated cell in vitro but lacks a confirmatory assay and reliable positive controls. We demonstrate that C5b-9 accumulation on the surface of TF1 PIGAnull cells correlates with cell killing in the mHam. We also show that Sialidase treatment of cells or addition of Shiga toxin 1 to human serum serve as a more reliable positive control for the mHam than cobra venom factor or lipopolysaccharide. Simultaneously performing the mHam and measuring C5b-9 accumulation either in GVB++ or GVB0 MgEGTA buffer with the addition of complement pathway specific inhibitors (anti-C5 antibody or a factor D inhibitor, ACH-145951) can be used to localize defects in complement regulation. As more targeted complement inhibitors become available, these assays may aid in the selection of personalized treatments for patients with complement-mediated diseases.


Subject(s)
Antiphospholipid Syndrome/immunology , Atypical Hemolytic Uremic Syndrome/immunology , Complement Activation/drug effects , Complement Inactivating Agents/pharmacology , Adult , Biological Assay , Cell Line, Tumor , Complement C3c/immunology , Complement C4b/immunology , Complement Membrane Attack Complex/immunology , Elapid Venoms/pharmacology , Female , Humans , Lipopolysaccharides/pharmacology , Male , Middle Aged , Neuraminidase/pharmacology , Peptide Fragments/immunology , Shiga Toxin 1/pharmacology
13.
J Clin Invest ; 130(11): 6151-6157, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32759504

ABSTRACT

Emerging data indicate that complement and neutrophils contribute to the maladaptive immune response that fuels hyperinflammation and thrombotic microangiopathy, thereby increasing coronavirus 2019 (COVID-19) mortality. Here, we investigated how complement interacts with the platelet/neutrophil extracellular traps (NETs)/thrombin axis, using COVID-19 specimens, cell-based inhibition studies, and NET/human aortic endothelial cell (HAEC) cocultures. Increased plasma levels of NETs, tissue factor (TF) activity, and sC5b-9 were detected in patients. Neutrophils of patients yielded high TF expression and released NETs carrying active TF. Treatment of control neutrophils with COVID-19 platelet-rich plasma generated TF-bearing NETs that induced thrombotic activity of HAECs. Thrombin or NETosis inhibition or C5aR1 blockade attenuated platelet-mediated NET-driven thrombogenicity. COVID-19 serum induced complement activation in vitro, consistent with high complement activity in clinical samples. Complement C3 inhibition with compstatin Cp40 disrupted TF expression in neutrophils. In conclusion, we provide a mechanistic basis for a pivotal role of complement and NETs in COVID-19 immunothrombosis. This study supports strategies against severe acute respiratory syndrome coronavirus 2 that exploit complement or NETosis inhibition.


Subject(s)
Betacoronavirus , Complement Membrane Attack Complex , Coronavirus Infections , Extracellular Traps , Neutrophils , Pandemics , Pneumonia, Viral , Thromboplastin , Thrombosis , Aged , Betacoronavirus/immunology , Betacoronavirus/metabolism , COVID-19 , Complement Activation/drug effects , Complement Membrane Attack Complex/immunology , Complement Membrane Attack Complex/metabolism , Coronavirus Infections/blood , Coronavirus Infections/immunology , Extracellular Traps/immunology , Extracellular Traps/metabolism , Female , Humans , Male , Middle Aged , Neutrophils/immunology , Neutrophils/metabolism , Peptides, Cyclic/pharmacology , Pneumonia, Viral/blood , Pneumonia, Viral/immunology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/blood , Receptor, Anaphylatoxin C5a/immunology , Respiratory Distress Syndrome/blood , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/virology , SARS-CoV-2 , Thrombin/immunology , Thrombin/metabolism , Thromboplastin/immunology , Thromboplastin/metabolism , Thrombosis/blood , Thrombosis/immunology , Thrombosis/virology
14.
Front Immunol ; 11: 1450, 2020.
Article in English | MEDLINE | ID: mdl-32733480

ABSTRACT

The complement system is a key component of innate immunity which readily responds to invading microorganisms. Activation of the complement system typically occurs via three main pathways and can induce various antimicrobial effects, including: neutralization of pathogens, regulation of inflammatory responses, promotion of chemotaxis, and enhancement of the adaptive immune response. These can be vital host responses to protect against acute, chronic, and recurrent viral infections. Consequently, many viruses (including dengue virus, West Nile virus and Nipah virus) have evolved mechanisms for evasion or dysregulation of the complement system to enhance viral infectivity and even exacerbate disease symptoms. The complement system has multifaceted roles in both innate and adaptive immunity, with both intracellular and extracellular functions, that can be relevant to all stages of viral infection. A better understanding of this virus-host interplay and its contribution to pathogenesis has previously led to: the identification of genetic factors which influence viral infection and disease outcome, the development of novel antivirals, and the production of safer, more effective vaccines. This review will discuss the antiviral effects of the complement system against numerous viruses, the mechanisms employed by these viruses to then evade or manipulate this system, and how these interactions have informed vaccine/therapeutic development. Where relevant, conflicting findings and current research gaps are highlighted to aid future developments in virology and immunology, with potential applications to the current COVID-19 pandemic.


Subject(s)
Betacoronavirus/immunology , Complement Membrane Attack Complex/immunology , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Immune Evasion , Pandemics/prevention & control , Pneumonia, Viral/immunology , Pneumonia, Viral/prevention & control , Viral Vaccines/immunology , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , COVID-19 , Complement Activation/drug effects , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Flavivirus/immunology , Flavivirus Infections/immunology , Flavivirus Infections/virology , Humans , Pneumonia, Viral/drug therapy , Pneumonia, Viral/virology , SARS-CoV-2 , Virus Internalization
15.
Cells ; 9(7)2020 07 04.
Article in English | MEDLINE | ID: mdl-32635578

ABSTRACT

We have previously shown that blood astrocytic-origin extracellular vesicles (AEVs) from Alzheimer's disease (AD) patients contain high complement levels. To test the hypothesis that circulating EVs from AD patients can induce complement-mediated neurotoxicity involving Membrane Attack Complex (MAC) formation, we assessed the effects of immunocaptured AEVs (using anti-GLAST antibody), in comparison with neuronal-origin (N)EVs (using anti-L1CAM antibody), and nonspecific CD81+ EVs (using anti-CD81 antibody), from the plasma of AD, frontotemporal lobar degeneration (FTLD), and control participants. AEVs (and, less effectively, NEVs) of AD participants induced Membrane Attack Complex (MAC) expression on recipient neurons (by immunohistochemistry), membrane disruption (by EthD-1 assay), reduced neurite density (by Tuj-1 immunohistochemistry), and decreased cell viability (by MTT assay) in rat cortical neurons and human iPSC-derived neurons. Demonstration of decreased cell viability was replicated in a separate cohort of autopsy-confirmed AD patients. These effects were not produced by CD81+ EVs from AD participants or AEVs/NEVs from FTLD or control participants, and were suppressed by the MAC inhibitor CD59 and other complement inhibitors. Our results support the stated hypothesis and should motivate future studies on the roles of neuronal MAC deposition and AEV/NEV uptake, as effectors of neurodegeneration in AD.


Subject(s)
Astrocytes/metabolism , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Neurons/metabolism , Animals , CD59 Antigens/metabolism , Case-Control Studies , Cells, Cultured , Complement Activation/immunology , Complement Membrane Attack Complex/immunology , Complement Membrane Attack Complex/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Rats
16.
Infect Immun ; 88(8)2020 07 21.
Article in English | MEDLINE | ID: mdl-32513855

ABSTRACT

The serum complement system is a first line of defense against bacterial invaders. Resistance to killing by serum enhances the capacity of Klebsiella pneumoniae to cause infection, but it is an incompletely understood virulence trait. Identifying and characterizing the factors responsible for preventing activation of, and killing by, serum complement could inform new approaches to treatment of K. pneumoniae infections. Here, we used functional genomic profiling to define the genetic basis of complement resistance in four diverse serum-resistant K. pneumoniae strains (NTUH-K2044, B5055, ATCC 43816, and RH201207), and explored their recognition by key complement components. More than 90 genes contributed to resistance in one or more strains, but only three, rfaH, lpp, and arnD, were common to all four strains. Deletion of the antiterminator rfaH, which controls the expression of capsule and O side chains, resulted in dramatic complement resistance reductions in all strains. The murein lipoprotein gene lpp promoted capsule retention through a mechanism dependent on its C-terminal lysine residue; its deletion led to modest reductions in complement resistance. Binding experiments with the complement components C3b and C5b-9 showed that the underlying mechanism of evasion varied in the four strains: B5055 and NTUH-K2044 appeared to bypass recognition by complement entirely, while ATCC 43816 and RH201207 were able to resist killing despite being associated with substantial levels of C5b-9. All rfaH and lpp mutants bound C3b and C5b-9 in large quantities. Our findings show that, even among this small selection of isolates, K. pneumoniae adopts differing mechanisms and utilizes distinct gene sets to avoid complement attack.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Carboxy-Lyases/immunology , Gene Expression Regulation, Bacterial/immunology , Genes, Bacterial , Immune Evasion , Klebsiella pneumoniae/immunology , Peptide Elongation Factors/immunology , Bacterial Outer Membrane Proteins/genetics , Blood Bactericidal Activity/immunology , Carboxy-Lyases/deficiency , Carboxy-Lyases/genetics , Complement C3b/genetics , Complement C3b/immunology , Complement Membrane Attack Complex/genetics , Complement Membrane Attack Complex/immunology , DNA Transposable Elements , Gene Expression Profiling , Gene Library , Humans , Klebsiella Infections/immunology , Klebsiella Infections/microbiology , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/pathogenicity , Mutation , Peptide Elongation Factors/deficiency , Peptide Elongation Factors/genetics , Sequence Analysis, DNA
17.
J Struct Biol ; 211(2): 107531, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32446810

ABSTRACT

The Membrane Attack Complex-Perforin (MACPF) family is ubiquitously found in all kingdoms. They have diverse cellular roles, however MACPFs with pore-forming toxic function in venoms and poisons are very rare in animals. Here we present the structure of PmPV2, a MACPF toxin from the poisonous apple snail eggs, that can affect the digestive and nervous systems of potential predators. We report the three-dimensional structure of PmPV2, at 17.2 Å resolution determined by negative-stain electron microscopy and its solution structure by small angle X-ray scattering (SAXS). We found that PV2s differ from nearly all MACPFs in two respects: it is a dimer in solution and protomers combine two immune proteins into an AB toxin. The MACPF chain is linked by a single disulfide bond to a tachylectin chain, and two heterodimers are arranged head-to-tail by non-covalent forces in the native protein. MACPF domain is fused with a putative new Ct-accessory domain exclusive to invertebrates. The tachylectin is a six-bladed ß-propeller, similar to animal tectonins. We experimentally validated the predicted functions of both subunits and demonstrated for the first time that PV2s are true pore-forming toxins. The tachylectin "B" delivery subunit would bind to target membranes, and then the MACPF "A" toxic subunit would disrupt lipid bilayers forming large pores altering the plasma membrane conductance. These results indicate that PV2s toxicity evolved by linking two immune proteins where their combined preexisting functions gave rise to a new toxic entity with a novel role in defense against predation. This structure is an unparalleled example of protein exaptation.


Subject(s)
Complement Membrane Attack Complex/ultrastructure , Lectins/ultrastructure , Perforin/ultrastructure , Protein Conformation , Amino Acid Sequence/genetics , Animals , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Complement Membrane Attack Complex/chemistry , Complement Membrane Attack Complex/immunology , Crystallography, X-Ray , Dimerization , Lectins/chemistry , Lectins/immunology , Models, Molecular , Perforin/chemistry , Perforin/immunology , Protein Subunits/genetics , Scattering, Small Angle , Snails/ultrastructure , X-Ray Diffraction
18.
Nephron ; 144(4): 195-203, 2020.
Article in English | MEDLINE | ID: mdl-32050203

ABSTRACT

A membranoproliferative pattern of glomerular injury is frequently observed in patients with complement-mediated disorders, such as C3 glomerulopathies (C3G) and primary immune complex-mediated membranoproliferative glomerulonephritis (IC-MPGN). The outcomes of C3G and -IC-MPGN are poor, independently of immunosuppressive therapy. However, two 48-week treatment periods with the anti-C5 monoclonal antibody eculizumab, divided by a -12-week washout period, achieved remission of proteinuria and stabilization/improvement of the glomerular filtration rate (GFR), measured through iohexol plasma clearance, in 3 of 10 patients with biopsy-proven MPGN, nephrotic syndrome and terminal complement complex sC5b-9 plasma levels >1,000 mg/mL, at inclusion. Baseline and end-of-study kidney biopsies were available for 2 patients with IC-MPGN, and their baseline characteristics were similar. However, in 1 patient proteinuria and GFR did not improve during the study, whereas in the other proteinuria decreased from 4.84 to 2.12 g/24-h and GFR increased from 91.5 to 142.7 mL/min/1.73 m2. Glomerular inflammation improved and median (interquartile range) glomerular staining for C5b-9 decreased in both cases: from 23.6 to 18.2% (p = 0.021) in the patient who achieved remission and from 15.8 to 10.7% (p = 0.019) in the patient with persistent proteinuria. Chronic glomerular lesions progressed and C3 glomerular staining and electron-dense deposits did not change appreciably in either case. However, in the patient who achieved remission, ultrastructural evaluation revealed features of glomerular microangiopathy at inclusion, which fully recovered posttreatment. Podocyte foot process effacement was observed in both patients at inclusion, but recovered only in the patient with microangiopathy. Thus, in 2 patients with -IC-MPGN, chronic glomerular changes progressed despite eculizumab-induced amelioration of glomerular inflammation and inhibition of sC5b-9 deposition, and independently of treatment effects on proteinuria and podocytes. The finding that the regression of microangiopathic changes was associated with improved clinical outcomes suggests that C5 blockade might have a therapeutic role in patients with IC-MPGN displaying microangiopathic endothelial injury.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antigen-Antibody Complex/immunology , Complement Activation , Complement C3-C5 Convertases/antagonists & inhibitors , Complement Membrane Attack Complex/immunology , Glomerulonephritis, Membranoproliferative/drug therapy , Adolescent , Complement C3-C5 Convertases/analysis , Female , Glomerular Filtration Rate , Glomerulonephritis, Membranoproliferative/immunology , Glomerulonephritis, Membranoproliferative/pathology , Humans , Male
19.
Pediatr Nephrol ; 35(5): 843-850, 2020 05.
Article in English | MEDLINE | ID: mdl-31932958

ABSTRACT

BACKGROUND: IgA nephropathy (IgAN) and Henoch-Schönlein purpura are common glomerular disorders in children sharing the same histopathologic pattern of IgA deposits within the mesangium, even if their physiopathology may be different. Repeated exposure to pathogens induces the production of abnormal IgA1. The immune complex deposition in the renal mesangium in IgAN or potentially in small vessels in Henoch-Schönlein purpura induces complement activation via the alternative and lectin pathways. Recent studies suggest that levels of membrane attack complex (MAC) in the urine might be a useful indicator of renal injury. Because of the emerging availability of therapies that selectively block complement activation, the aim of the present study is to investigate whether MAC immunostaining might be a useful marker of IgA-mediated renal injury. METHODS: We conducted immunohistochemistry analysis of the MAC on renal biopsies from 67 pediatric patients with IgAN and Henoch-Schönlein purpura. We classified their renal biopsies according to the Oxford classification, retrieved symptoms, biological parameters, treatment, and follow-up. RESULTS: We found MAC expression was significantly related to impaired renal function and patients whose clinical course required therapy. MAC deposits tend to be more abundant in patients with decreased glomerular filtration rate (p = 0.02), patients with proteinuria > 0.750 g/day/1.73 m2, and with nephrotic syndrome. No correlation with histological alterations was observed. CONCLUSIONS: We conclude that MAC deposition could be a useful additional indicator of renal injury in patients with IgAN and Henoch-Schönlein purpura, independent of other indicators.


Subject(s)
Complement Membrane Attack Complex/analysis , Glomerular Mesangium/pathology , Glomerulonephritis, IGA/diagnosis , IgA Vasculitis/diagnosis , Immunosuppressive Agents/therapeutic use , Adolescent , Biomarkers/analysis , Biopsy , Child , Child, Preschool , Complement Membrane Attack Complex/immunology , Complement Pathway, Alternative/drug effects , Complement Pathway, Alternative/immunology , Complement Pathway, Mannose-Binding Lectin/drug effects , Complement Pathway, Mannose-Binding Lectin/immunology , Feasibility Studies , Female , Follow-Up Studies , Glomerular Mesangium/immunology , Glomerulonephritis, IGA/drug therapy , Glomerulonephritis, IGA/immunology , Glomerulonephritis, IGA/pathology , Humans , IgA Vasculitis/drug therapy , IgA Vasculitis/immunology , IgA Vasculitis/pathology , Immunoglobulin A/immunology , Immunosuppressive Agents/pharmacology , Male , Prognosis , Retrospective Studies , Treatment Outcome
20.
Front Immunol ; 11: 599974, 2020.
Article in English | MEDLINE | ID: mdl-33643288

ABSTRACT

The membrane attack complex-also known as C5b-9-is the end-product of the classical, lectin, and alternative complement pathways. It is thought to play an important role in the pathogenesis of various kidney diseases by causing cellular injury and tissue inflammation, resulting in sclerosis and fibrosis. These deleterious effects are, consequently, targeted in the development of novel therapies that inhibit the formation of C5b-9, such as eculizumab. To clarify how C5b-9 contributes to kidney disease and to predict which patients benefit from such therapy, knowledge on deposition of C5b-9 in the kidney is essential. Because immunohistochemical staining of C5b-9 has not been routinely conducted and never been compared across studies, we provide a review of studies on deposition of C5b-9 in healthy and diseased human kidneys. We describe techniques to stain deposits and compare the occurrence of deposits in healthy kidneys and in a wide spectrum of kidney diseases, including hypertensive nephropathy, diabetic nephropathy, membranous nephropathy, IgA nephropathy, lupus nephritis, C3 glomerulopathy, and thrombotic microangiopathies such as the atypical hemolytic uremic syndrome, vasculitis, interstitial nephritis, acute tubular necrosis, kidney tumors, and rejection of kidney transplants. We summarize how these deposits are related with other histological lesions and clinical characteristics. We evaluate the prognostic relevance of these deposits in the light of possible treatment with complement inhibitors.


Subject(s)
Complement Inactivating Agents/therapeutic use , Complement Membrane Attack Complex/immunology , Kidney Diseases/drug therapy , Kidney Diseases/immunology , Kidney/immunology , Humans , Kidney/pathology , Kidney Diseases/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...