Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Front Immunol ; 13: 815833, 2022.
Article in English | MEDLINE | ID: mdl-35250994

ABSTRACT

The coronavirus disease-2019 (COVID-19) caused by the SARS-CoV-2 virus may vary from asymptomatic to severe infection with multi-organ failure and death. Increased levels of circulating complement biomarkers have been implicated in COVID-19-related hyperinflammation and coagulopathy. We characterized systemic complement activation at a cellular level in 49-patients with COVID-19. We found increases of the classical complement sentinel C1q and the downstream C3 component on circulating blood monocytes from COVID-19 patients when compared to healthy controls (HCs). Interestingly, the cell surface-bound complement inhibitor CD55 was also upregulated in COVID-19 patient monocytes in comparison with HC cells. Monocyte membrane-bound C1q, C3 and CD55 levels were associated with plasma inflammatory markers such as CRP and serum amyloid A during acute infection. Membrane-bounds C1q and C3 remained elevated even after a short recovery period. These results highlight systemic monocyte-associated complement activation over a broad range of COVID-19 disease severities, with a compensatory upregulation of CD55. Further evaluation of complement and its interaction with myeloid cells at the membrane level could improve understanding of its role in COVID-19 pathogenesis.


Subject(s)
COVID-19/immunology , Complement Activation/immunology , Complement System Proteins/immunology , Monocytes/immunology , Adult , Biomarkers/blood , COVID-19/blood , COVID-19/virology , Complement Inactivating Agents/immunology , Cytokines/immunology , Female , Humans , Immunologic Factors/immunology , Male , Middle Aged , Monocytes/virology , SARS-CoV-2/immunology
2.
Nat Commun ; 13(1): 317, 2022 01 14.
Article in English | MEDLINE | ID: mdl-35031611

ABSTRACT

Activation of the serum-resident complement system begins a cascade that leads to activation of membrane-resident complement receptors on immune cells, thus coordinating serum and cellular immune responses. Whilst many molecules act to control inappropriate activation, Properdin is the only known positive regulator of the human complement system. By stabilising the alternative pathway C3 convertase it promotes complement self-amplification and persistent activation boosting the magnitude of the serum complement response by all triggers. In this work, we identify a family of tick-derived alternative pathway complement inhibitors, hereafter termed CirpA. Functional and structural characterisation reveals that members of the CirpA family directly bind to properdin, inhibiting its ability to promote complement activation, and leading to potent inhibition of the complement response in a species specific manner. We provide a full functional and structural characterisation of a properdin inhibitor, opening avenues for future therapeutic approaches.


Subject(s)
Arthropod Proteins/chemistry , Arthropod Proteins/immunology , Complement Inactivating Agents/chemistry , Complement Inactivating Agents/immunology , Properdin/immunology , Rhipicephalus/immunology , Amino Acid Sequence , Animals , Arthropod Proteins/genetics , Complement Activation , Complement C3/chemistry , Complement C3/immunology , Complement Pathway, Alternative , Humans , Kinetics , Properdin/chemistry , Properdin/genetics , Rhipicephalus/chemistry , Rhipicephalus/genetics , Sequence Alignment
3.
Mol Immunol ; 128: 175-187, 2020 12.
Article in English | MEDLINE | ID: mdl-33137606

ABSTRACT

It has long been known that the complement cascade is activated in various forms of glomerulonephritis. In many of these diseases, immune-complexes deposit in the glomeruli and activate the classical pathway. Researchers have also identified additional mechanisms by which complement is activated in the kidney, including diseases in which the alternative and lectin pathways are activated. The kidney appears to be particularly susceptible to activation of the alternative pathway, and this pathway has been implicated as a primary driver of atypical hemolytic uremic syndrome, C3 glomerulopathy, anti-neutrophil cytoplasmic antibody-associated vasculitis, as well as some forms of immune-complex glomerulonephritis. In this paper we review the shared and distinct mechanisms by which complement is activated in these different diseases. We also review the opportunities for using therapeutic complement inhibitors to treat kidney diseases.


Subject(s)
Complement System Proteins/immunology , Kidney Diseases/immunology , Animals , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/immunology , Antigen-Antibody Complex/immunology , Atypical Hemolytic Uremic Syndrome/immunology , Complement Activation/immunology , Complement Inactivating Agents/immunology , Glomerulonephritis/immunology , Humans , Kidney/immunology
4.
Expert Opin Biol Ther ; 20(9): 991-998, 2020 09.
Article in English | MEDLINE | ID: mdl-32602752

ABSTRACT

INTRODUCTION: Acetylcholine receptor antibody-positive generalized myasthenia gravis (gMG) is effectively treated with symptomatic and immunosuppressive drugs but a proportion of patients has a persistent disease and severe adverse events (AEs). The unmet medical needs are specific immunosuppression and AE lowering. Eculizumab blocks C5 protecting neuromuscular junction from the destructive autoantibody effects. Phase II (Study C08-001) and III (ECU-MG-301) studies, with the open-label extension (ECU-MG-302), demonstrated eculizumab efficacy and safety in refractory gMG patients. AREAS COVERED: We provide an overview of eculizumab biological features, clinical efficacy, and safety in gMG patients, highlighting our perspective on the drug positioning in the MG treatment algorithm. EXPERT OPINION: Eculizumab has the potential to significantly change the immunosuppressive approach in gMG offering the opportunity to avoid or delay corticosteroids' use due to its speed and selective mechanism of action. Eculizumab prescription will depend on: 1. ability to modify the natural disease course; 2. sustainability in the clinical practice (cost/effectiveness ratio); 3. drug-induced AE reduction. At present we are missing a controlled study on its use as a first-line treatment. We think that immunosuppression in MG will change significantly in the next years by adopting more focused 'Precision Medicine' approaches, and Eculizumab seems to satisfy such a promise.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Complement Inactivating Agents/therapeutic use , Myasthenia Gravis/drug therapy , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/metabolism , Clinical Trials as Topic , Complement C5/immunology , Complement Inactivating Agents/immunology , Complement Inactivating Agents/metabolism , Half-Life , Humans , Myasthenia Gravis/pathology , Treatment Outcome
5.
Adv Chronic Kidney Dis ; 27(2): 104-110, 2020 03.
Article in English | MEDLINE | ID: mdl-32553242

ABSTRACT

C3 glomerulopathy (C3G) is a rare set of kidney diseases with 2 patterns: C3 glomerulonephritis (C3GN) and dense deposit disease. Pathogenesis of both diseases is due to complement dysregulation in the alternative pathway. Acquired or genetic alterations of the regulatory proteins of the complement pathway result in C3G. Although the disease is characterized by low C3 levels in serum and C3-dominant staining by immunofluorescence on biopsy, other disease entities such as infection-related glomerulonephritis and masked monoclonal deposits can present similarly. Both the C3GN and dense deposit disease variants of C3G are progressive and recur in transplanted kidneys. Although no direct treatment is available, complement blockers are either available or in the clinical trial phase. This review will survey the pathogenesis of C3GN and current treatment options.


Subject(s)
Complement C3/immunology , Complement Inactivating Agents , Complement Pathway, Alternative , Glomerulonephritis, Membranoproliferative , Complement Inactivating Agents/immunology , Complement Inactivating Agents/pharmacology , Complement Pathway, Alternative/drug effects , Complement Pathway, Alternative/immunology , Glomerulonephritis, Membranoproliferative/immunology , Glomerulonephritis, Membranoproliferative/physiopathology , Glomerulonephritis, Membranoproliferative/therapy , Humans , Prognosis
6.
Adv Chronic Kidney Dis ; 27(2): 111-119, 2020 03.
Article in English | MEDLINE | ID: mdl-32553243

ABSTRACT

Immunoglobulin A (IgA) nephropathy (IgAN) is an important cause of chronic and end-stage kidney disease. IgAN pathogenesis is incompletely understood. In particular, we cannot adequately explain the heterogeneity in clinical and histologic features and severities that characterizes IgAN. This limits patient stratification to appropriate and effective treatments and the development of disease-targeted therapies. Studies of the role of the alternative, lectin, and terminal complement pathways in IgAN have enhanced our understanding of disease pathogenesis and inform the development of novel diagnostic and therapeutic strategies. For example, recent genetic, serologic, and immunohistologic evidence suggests that imbalances between the main alternative complement pathway regulator protein (factor H) and competitor proteins that deregulate complement activity (factor H-related proteins 1 and 5, FHR1, and FHR5) associate with IgAN severity: a relative abundance of FHR1 and FHR5 amplifies complement-dependent inflammation and exacerbates kidney injury. Ongoing characterization of the mechanisms by which complement activity contributes to IgAN pathogenesis will facilitate the development of complement-based diagnostic techniques, biomarkers of disease activity and severity, and novel targeted therapies.


Subject(s)
Complement Inactivating Agents , Complement Pathway, Alternative , Glomerulonephritis, IGA , Complement Activation/drug effects , Complement Activation/immunology , Complement Inactivating Agents/immunology , Complement Inactivating Agents/pharmacology , Complement Pathway, Alternative/genetics , Complement Pathway, Alternative/immunology , Glomerulonephritis, IGA/immunology , Glomerulonephritis, IGA/physiopathology , Glomerulonephritis, IGA/therapy , Humans , Prognosis , Severity of Illness Index
7.
Adv Chronic Kidney Dis ; 27(2): 128-137, 2020 03.
Article in English | MEDLINE | ID: mdl-32553245

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) is a severe thrombotic microangiopathy characterized by over-activation of the alternative complement pathway. The etiology of the dysregulated complement system is commonly a genetic variant in one or more complement proteins as identified in ∼ 60%-70% patients. The risk of recurrence after a kidney transplantation is high and depends on the underlying complement abnormality. For a long time, kidney transplantation was contraindicated in these patients because of the high rate of recurrence and subsequent allograft loss. Over the past decade, advancements in the understanding of etiopathogenesis of aHUS and approval of the anti-complement drug, eculizumab, have allowed for successful kidney transplantation in these patients. All patients with ESRD due to aHUS should undergo screening for complement genetic variants. Patients in whom a genetic variant is not identified or in whom a genetic variant of uncertain significance is identified should undergo further testing to determine etiology of disease. This review aims to shed light on the diagnostic and therapeutic considerations in patients with aHUS preceding and following kidney transplantation.


Subject(s)
Antibodies, Monoclonal, Humanized , Atypical Hemolytic Uremic Syndrome , Complement Pathway, Alternative , Graft Rejection , Kidney Transplantation/adverse effects , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/physiopathology , Atypical Hemolytic Uremic Syndrome/surgery , Complement C5/antagonists & inhibitors , Complement Inactivating Agents/immunology , Complement Inactivating Agents/pharmacology , Complement Pathway, Alternative/drug effects , Complement Pathway, Alternative/genetics , Graft Rejection/immunology , Graft Rejection/prevention & control , Humans , Kidney Transplantation/methods , Secondary Prevention/methods
8.
Adv Chronic Kidney Dis ; 27(2): 138-148, 2020 03.
Article in English | MEDLINE | ID: mdl-32553246

ABSTRACT

Antibody-mediated rejection (AMR) is one of the leading causes of kidney allograft failure and is usually mediated by anti-human leukocyte antigen donor-specific antibodies (DSAs). Activation of classical pathway of the complement system is responsible for downstream effects of DSA and account for significant manifestations of AMR. Currently, the treatment of AMR is based on strategies to remove preformed antibodies or to prevent their production; however, these strategies are often unsuccessful. It is theoretically possible to inhibit complement activity to prevent the effect of DSA on kidney allograft function. Complement inhibitors such as eculizumab, a complement 5 monoclonal antibody, and complement 1 esterase inhibitors (C1 INHs) have been used in prevention and treatment of AMR with variable success. Eculizumab and C1 INH seem to reduce the incidence of early AMR and allow transplantation in highly sensitized kidney transplant recipients, but data on their long-term effect on kidney allograft function are limited. Several case reports described the successful use of eculizumab in the treatment of AMR, but there are no randomized controlled studies that showed efficacy. Treatment of AMR with C1 INH, in addition to standard of care, did not change short-term outcome but long-term studies are underway.


Subject(s)
Antibodies, Monoclonal, Humanized , Complement C1 Inhibitor Protein , Complement Pathway, Classical , Graft Rejection , HLA Antigens/immunology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Complement C1 Inhibitor Protein/immunology , Complement C1 Inhibitor Protein/pharmacology , Complement Inactivating Agents/immunology , Complement Inactivating Agents/pharmacology , Complement Pathway, Classical/drug effects , Complement Pathway, Classical/immunology , Graft Rejection/immunology , Graft Rejection/prevention & control , Humans , Kidney Failure, Chronic/surgery , Kidney Transplantation/adverse effects
9.
Adv Chronic Kidney Dis ; 27(2): 155-164, 2020 03.
Article in English | MEDLINE | ID: mdl-32553248

ABSTRACT

Complement-mediated disorders in pregnancy span a large spectrum and have been implicated in all three complement pathways: classical, lectin, and alternative. Our understanding of these disorders in recent years has advanced due to a better understanding of complement regulatory proteins, such as complement factor H, complement factor I, membrane cofactor protein, and thrombomodulin that particularly affect the alternative complement pathway. Enthusiasm in genotyping for mutations that encode these proteins has allowed us to study the presence of genetic variants which may predispose women to develop conditions such as pregnancy-associated hemolytic uremic syndrome (P-aHUS), thrombotic thrombocytopenic purpura, preeclampsia/hemolysis, elevated liver enzymes, low platelets (HELLP), systemic lupus erythematosus/antiphospholipid syndrome, and peripartum cardiomyopathy. The advent of the anti-C5-antibody eculizumab to quench the complement cascade has already proven in small case series to improve maternal kidney outcomes in complement-mediated obstetric catastrophes such as P-aHUS and HELLP. In this review, we will detail the pathogenesis behind these complement-mediated pregnancy disorders, the role of complement variants in disease phenotype, and the most up-to-date experience with eculizumab in this population.


Subject(s)
Complement Activation/immunology , Complement Inactivating Agents , Complement System Proteins , Hemolytic-Uremic Syndrome , Pregnancy Complications , Complement Inactivating Agents/immunology , Complement Inactivating Agents/pharmacology , Complement System Proteins/genetics , Complement System Proteins/immunology , Female , HELLP Syndrome/immunology , HELLP Syndrome/prevention & control , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/prevention & control , Humans , Pregnancy , Pregnancy Complications/immunology , Pregnancy Complications/physiopathology , Pregnancy Complications/prevention & control
11.
Adv Chronic Kidney Dis ; 27(2): 95-103, 2020 03.
Article in English | MEDLINE | ID: mdl-32553251

ABSTRACT

The complement system is an evolutionarily ancient arm of the innate immune system. It remains, however, one of the last major pathways in immunology for which specific pharmaceutical antagonists have been developed. In recent years, a fundamental role for complement has been described in many different renal diseases, including both pauci-immune as well as immune-complex diseases. Since the 2011 FDA approval of eculizumab, the only marketed complement antagonist, no new therapeutics have entered clinical practice. There are now multiple new agents in clinical trials, from oral molecules to small inhibitory RNA, that target the classical, lectin, and alternative pathways. Herein we summarize several potential renal diseases in which complement inhibitors may provide a therapeutic benefit, as well as specific complement inhibitors in development.


Subject(s)
Complement Inactivating Agents , Complement System Proteins/immunology , Kidney Diseases , Complement Inactivating Agents/immunology , Complement Inactivating Agents/pharmacology , Humans , Kidney Diseases/drug therapy , Kidney Diseases/immunology , Molecular Targeted Therapy/trends
13.
FASEB J ; 34(5): 6598-6612, 2020 05.
Article in English | MEDLINE | ID: mdl-32219899

ABSTRACT

Inhibition of the complement activation has emerged as an option for treatment of a range of diseases. Activation of the lectin and alternative pathways (LP and AP, respectively) contribute to the deterioration of conditions in certain diseases such as ischemia-reperfusion injuries and age-related macular degeneration (AMD). In the current study, we generated dual complement inhibitors of the pathways MAp44-FH and sMAP-FH by fusing full-length MAp44 or small mannose-binding lectin-associated protein (sMAP), LP regulators, with the N-terminal five short consensus repeat (SCR) domains of complement factor H (SCR1/5-FH), an AP regulator. The murine forms of both fusion proteins formed a complex with endogenous mannose-binding lectin (MBL) or ficolin A in the circulation when administered in mice intraperitoneally. Multiple complement activation assays revealed that sMAP-FH had significantly higher inhibitory effects on activation of the LP and AP in vivo as well as in vitro compared to MAp44-FH. Human form of sMAP-FH also showed dual inhibitory effects on LP and AP activation in human sera. Our results indicate that the novel fusion protein sMAP-FH inhibits both the LP and AP activation in mice and in human sera, and could be an effective therapeutic agent for diseases in which both the LP and AP activation are significantly involved.


Subject(s)
Complement Inactivating Agents/metabolism , Complement Pathway, Alternative/immunology , Lectins/immunology , Mannose-Binding Lectin/metabolism , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Animals , Complement Activation/immunology , Complement Factor H/immunology , Complement Factor H/metabolism , Complement Inactivating Agents/immunology , Female , Humans , Lectins/metabolism , Mannose-Binding Lectin/immunology , Mannose-Binding Protein-Associated Serine Proteases/immunology , Mice , Mice, Inbred C57BL
14.
AAPS J ; 20(6): 106, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30280286

ABSTRACT

Complement protein C5a is recognized as an important component of the alternative complement pathway. Its role is prominent enough to garner interest not only as a biomarker, but also as a potential therapeutic target. Bioanalytical challenges have been posed in proper quantitation of free C5a due to interference from its precursor, C5. Additionally, free therapeutic target quantitation can be difficult due to effects of sample dilution and prolonged sample incubation when therapeutic is used as capture reagent. Gyrolab technology enables quantitation of free target analyte with minimal sample dilution and rapid sample incubations, thus enabling in vitro results that are more representative of in vivo pharmacodynamics. When coupled with strategic sample pretreatment, Gyrolab offers an opportunity to quantitate free C5a in human plasma with an assay that vastly diminishes C5 interference. A Gyrolab assay for the quantitation of free C5a in human plasma was developed and validated. Validation results confirmed that proper sample pretreatment and use of the Gyrolab platform yield accurate and reliable results. Due to the advantages that it provides, Gyrolab has become our default technology of choice for quantitation of free target.


Subject(s)
Analytic Sample Preparation Methods/methods , Biological Assay/methods , Complement C5a/analysis , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/metabolism , Biological Assay/instrumentation , Complement C5a/immunology , Complement C5a/metabolism , Complement Inactivating Agents/immunology , Complement Inactivating Agents/metabolism , Humans , Immunoassay/instrumentation , Immunoassay/methods , Limit of Detection
15.
Molecules ; 23(7)2018 Jul 12.
Article in English | MEDLINE | ID: mdl-30002298

ABSTRACT

Infusion reactions (IRs) are common immune-mediated side effects in patients treated with a variety of drug products, including, but not limited to, nanotechnology formulations. The mechanism of IRs is not fully understood. One of the best studied mechanisms of IRs to nanomedicines is the complement activation. However, it is largely unknown why some patients develop reactions to nanomedicines while others do not, and why some nanoparticles are more reactogenic than others. One of the theories is that the pre-existing anti-polyethylene glycol (PEG) antibodies initiate the complement activation and IRs in patients. In this study, we investigated this hypothesis in the case of PEGylated liposomal doxorubicin (Doxil), which, when used in a clinical setting, is known to induce IRs; referred to as complement activation-related pseudoallergy (CARPA) in sensitive individuals. We conducted the study in vitro using plasma derived from C57BL/6 mice and twenty human donor volunteers. We used mouse plasma to test a library of well-characterized mouse monoclonal antibodies with different specificity and affinity to PEG as it relates to the complement activation by Doxil. We determined the levels of pre-existing polyclonal antibodies that bind to PEG, methoxy-PEG, and PEGylated liposomes in human plasma, and we also assessed complement activation by Doxil and concentrations of complement inhibitory factors H and I in these human plasma specimens. The affinity, specificity, and other characteristics of the human polyclonal antibodies are not known at this time. Our data demonstrate that under in vitro conditions, some anti-PEG antibodies contribute to the complement activation by Doxil. Such contribution, however, needs to be considered in the context of other factors, including, but not limited to, antibody class, type, clonality, epitope specificity, affinity, and titer. In addition, our data contribute to the knowledge base used to understand and improve nanomedicine safety.


Subject(s)
Antibodies, Monoclonal , Complement Activation , Complement Inactivating Agents , Doxorubicin/analogs & derivatives , Polyethylene Glycols , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Complement Activation/drug effects , Complement Activation/immunology , Complement Factor H/immunology , Complement Factor I/immunology , Complement Inactivating Agents/chemistry , Complement Inactivating Agents/immunology , Doxorubicin/pharmacology , Drug Hypersensitivity/immunology , Humans , Mice , Polyethylene Glycols/pharmacology
16.
Emerg Microbes Infect ; 7(1): 77, 2018 Apr 24.
Article in English | MEDLINE | ID: mdl-29691378

ABSTRACT

The pathogenesis of highly pathogenic Middle East respiratory syndrome coronavirus (MERS-CoV) remains poorly understood. In a previous study, we established an hDPP4-transgenic (hDPP4-Tg) mouse model in which MERS-CoV infection causes severe acute respiratory failure and high mortality accompanied by an elevated secretion of cytokines and chemokines. Since excessive complement activation is an important factor that contributes to acute lung injury after viral infection, in this study, we investigated the role of complement in MERS-CoV-induced lung damage. Our study showed that complement was excessively activated in MERS-CoV-infected hDPP4-Tg mice through observations of increased concentrations of the C5a and C5b-9 complement activation products in sera and lung tissues, respectively. Interestingly, blocking C5a production by targeting its receptor, C5aR, alleviated lung and spleen tissue damage and reduced inflammatory responses. More importantly, anti-C5aR antibody treatment led to decreased viral replication in lung tissues. Furthermore, compared with the sham treatment control, apoptosis of splenic cells was less pronounced in the splenic white pulp of treated mice, and greater number of proliferating splenic cells, particularly in the red pulp, was observed. These data indicate that (1) dysregulated host immune responses contribute to the severe outcome of MERS; (2) excessive complement activation, triggered by MERS-CoV infection, promote such dysregulation; and (3) blockade of the C5a-C5aR axis lead to the decreased tissue damage induced by MERS-CoV infection, as manifested by reduced apoptosis and T cell regeneration in the spleen. Therefore, the results of this study suggest a new strategy for clinical intervention and adjunctive treatment in MERS-CoV cases.


Subject(s)
Complement Activation/immunology , Complement C5a/immunology , Dipeptidyl Peptidase 4/genetics , Lung/pathology , Middle East Respiratory Syndrome Coronavirus/immunology , Animals , Apoptosis , Chemokines/immunology , Complement C5a/antagonists & inhibitors , Complement Inactivating Agents/administration & dosage , Complement Inactivating Agents/immunology , Coronavirus Infections/immunology , Coronavirus Infections/therapy , Coronavirus Infections/virology , Cytokines/immunology , Disease Models, Animal , Host-Pathogen Interactions/immunology , Humans , Immunologic Factors/antagonists & inhibitors , Immunologic Factors/immunology , Lung/immunology , Lung/virology , Mice , Mice, Transgenic , Middle East Respiratory Syndrome Coronavirus/pathogenicity , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Spleen/cytology , Spleen/immunology , Spleen/pathology , Spleen/virology , T-Lymphocytes/immunology , Virus Replication/immunology
17.
J Immunol ; 200(10): 3495-3505, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29626087

ABSTRACT

Streptococcus pyogenes is an exclusively human pathogen that can provoke mild skin and throat infections but can also cause fatal septicemia. This gram-positive bacterium has developed several strategies to evade the human immune system, enabling S. pyogenes to survive in the host. These strategies include recruiting several human plasma proteins, such as the complement inhibitor, C4b-binding protein (C4BP), and human (hu)-IgG through its Fc region to the bacterial surface to evade immune recognition. We identified a novel virulence mechanism whereby IgG-enhanced binding of C4BP to five of 12 tested S. pyogenes strains expressed diverse M proteins that are important surface-expressed virulence factors. Importantly, all strains that bound C4BP in the absence of IgG bound more C4BP when IgG was present. Further studies with an M1 strain that additionally expressed protein H, also a member of the M protein family, revealed that binding of hu-IgG Fc to protein H increased the affinity of protein H for C4BP. Increased C4BP binding accentuated complement downregulation, resulting in diminished bacterial killing. Accordingly, mortality from S. pyogenes infection in hu-C4BP transgenic mice was increased when hu-IgG or its Fc portion alone was administered concomitantly. Electron microscopy analysis of human tissue samples with necrotizing fasciitis confirmed increased C4BP binding to S. pyogenes when IgG was present. Our findings provide evidence of a paradoxical function of hu-IgG bound through Fc to diverse S. pyogenes isolates that increases their virulence and may counteract the beneficial effects of IgG opsonization.


Subject(s)
Complement System Proteins/immunology , Immunoglobulin G/immunology , Streptococcus pyogenes/immunology , Virulence/immunology , Animals , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Carrier Proteins/immunology , Complement C4b-Binding Protein/immunology , Complement Inactivating Agents/immunology , Female , Humans , Male , Mice , Mice, Inbred BALB C , Phagocytosis/immunology , Protein Binding/immunology , Streptococcal Infections/immunology , Virulence Factors/immunology
18.
PLoS One ; 11(2): e0148840, 2016.
Article in English | MEDLINE | ID: mdl-26871712

ABSTRACT

Neisseria meningitidis recruits host human complement inhibitors to its surface to down-regulate complement activation and enhance survival in blood. We have investigated whether such complement inhibitor binding occurs after vaccination with native outer membrane vesicles (nOMVs), and limits immunogenicity of such vaccines. To this end, nOMVs reactogenic lipopolysaccharide was detoxified by deletion of the lpxl1 gene (nOMVlpxl1). nOMVs unable to bind human complement factor H (hfH) were generated by additional deletions of the genes encoding factor H binding protein (fHbp) and neisserial surface protein A (NspA) (nOMVdis). Antibody responses elicited in mice with nOMVdis were compared to those elicited with nOMVlpxl1 in the presence of hfH. Results demonstrate that the administration of human fH to mice immunized with fHbp containing OMVlpxl1 decreased immunogenicity against fHbp (but not against the OMV as a whole). The majority of the OMV-induced bactericidal immune response (OMVlpxl1 or OMVdis) was versus PorA. Despite a considerable reduction of hfH binding to nOMVdis, and the absence of the vaccine antigen fHbp, immunogenicity in mice was not different from nOMVlpxl1, in the absence or presence of hfH (serum bactericidal titers of 1:64 vs 1:128 after one dose in the nOMVdis and nOMVlpxl1-immunized groups respectively). Therefore, partial inhibition of fH binding did not enhance immunity in this model.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Complement Inactivating Agents/immunology , Meningitis, Meningococcal/immunology , Meningitis, Meningococcal/prevention & control , Meningococcal Vaccines/immunology , Neisseria meningitidis/immunology , Animals , Antibody Formation , Bacterial Outer Membrane Proteins/therapeutic use , Complement Factor H/immunology , Female , Humans , Immunization , Meningococcal Vaccines/therapeutic use , Mice , Mice, Inbred C57BL
19.
Immunobiology ; 221(4): 503-11, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26792457

ABSTRACT

Paroxysmal nocturnal hemoglobinuria (PNH) is characterized by complement-mediated cell lysis due to deficiency of GPI-anchored complement regulators. Blockage of the lytic pathway by eculizumab is the only available therapy for PNH patients and shows remarkable benefits, but regularly yields PNH erythrocytes opsonized with fragments of complement protein C3, rendering such erythrocytes prone to extravascular hemolysis. This effect is associated with insufficient responsiveness seen in a subgroup of PNH patients. Novel C3-opsonin targeted complement inhibitors act earlier in the cascade, at the level of activated C3 and are engineered from parts of the natural complement regulator Factor H (FH) or complement receptor 2 (CR2). This inhibitor class comprises three variants of "miniFH" and the clinically developed "FH-CR2" fusion-protein (TT30). We show that the approach of FH-CR2 to target C3-opsonins was more efficient in preventing complement activation induced by foreign surfaces, whereas the miniFH variants were substantially more active in controlling complement on PNH erythrocytes. Subtle differences were noted in the ability of each version of miniFH to protect human PNH cells. Importantly, miniFH and FH-CR2 interfered only minimally with complement-mediated serum killing of bacteria when compared to untargeted inhibition of all complement pathways by eculizumab. Thus, the molecular design of each C3-opsonin targeted complement inhibitor determines its potency in respect to the nature of the activator/surface providing potential functionality in PNH.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Complement C3/genetics , Complement Inactivating Agents/pharmacology , Erythrocytes/drug effects , Opsonin Proteins/genetics , Animals , Antibodies, Monoclonal, Humanized/biosynthesis , Antibodies, Monoclonal, Humanized/immunology , Cells, Cultured , Complement Activation/drug effects , Complement Factor H/genetics , Complement Factor H/immunology , Complement Inactivating Agents/immunology , Complement Inactivating Agents/metabolism , Complement Pathway, Alternative , Erythrocytes/immunology , Erythrocytes/pathology , Escherichia coli/drug effects , Escherichia coli/growth & development , Gene Expression , Hemoglobinuria, Paroxysmal/drug therapy , Hemoglobinuria, Paroxysmal/genetics , Hemoglobinuria, Paroxysmal/metabolism , Hemoglobinuria, Paroxysmal/pathology , Hemolysis/drug effects , Humans , Molecular Targeted Therapy , Protein Engineering , Rabbits , Receptors, Complement 3d/genetics , Receptors, Complement 3d/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
20.
J Immunol ; 196(2): 866-76, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26643478

ABSTRACT

The serum proteins factor H (FH), consisting of 20 complement control protein modules (CCPs), and its splice product FH-like protein 1 (FHL-1; consisting of CCPs 1-7) are major regulators of the alternative pathway (AP) of complement activation. The engineered version of FH, miniFH, contains only the N- and C-terminal portions of FH linked by an optimized peptide and shows ∼ 10-fold higher ex vivo potency. We explored the hypothesis that regulatory potency is enhanced by unmasking of a ligand-binding site in the C-terminal CCPs 19-20 that is cryptic in full-length native FH. Therefore, we produced an FH variant lacking the central domains 10-15 (FHΔ10-15). To explore how avidity affects regulatory strength, we generated a duplicated version of miniFH, termed midiFH. We compared activities of FHΔ10-15 and midiFH to miniFH, FH, and FHL-1. Relative to FH, FHΔ10-15 exhibited an altered binding profile toward C3 activation products and a 5-fold-enhanced complement regulation on a paroxysmal nocturnal hemoglobinuria patient's erythrocytes. Contrary to dogma, FHL-1 and FH exhibited equal regulatory activity, suggesting that the role of FHL-1 in AP regulation has been underestimated. Unexpectedly, a substantially increased avidity for complement opsonins, as seen in midiFH, did not potentiate the inhibitory potential on host cells. In conclusion, comparisons of engineered and native FH-based regulators have identified features that determine high AP regulatory activity on host cells. Unrestricted availability of FH CCPs 19-20 and an optimal spatial orientation between the N- and C-terminal FH regions are key.


Subject(s)
Complement C3b Inactivator Proteins/immunology , Complement Factor H/immunology , Complement Inactivating Agents/pharmacology , Complement Pathway, Alternative/immunology , Recombinant Proteins/pharmacology , Amino Acid Sequence , Complement Factor H/chemistry , Complement Inactivating Agents/chemical synthesis , Complement Inactivating Agents/immunology , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Protein Binding , Recombinant Proteins/chemical synthesis , Recombinant Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...