Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Annu Rev Immunol ; 28: 131-55, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19947883

RESUMO

Complement is an innate immune system that is a first line of defense against pathogens and facilitates elimination of apoptotic and injured cells. During complement activation, the complement convertases are assembled on target surfaces and initiate their proteolytic activities, a process that marks targets for phagocytosis and/or lysis. The complement alternative activation pathway has been implicated in a number of autoimmune conditions including arthritis and age-related macular degeneration. Properdin, a plasma component that is also released by activated neutrophils, is critical in the stabilization of alternative pathway convertases. Recently, it has been shown that properdin is also a pattern-recognition molecule that binds to certain microbial surfaces, apoptotic cells, and necrotic cells. Once bound to a surface, properdin can direct convertase formation and target uptake. New studies are now focusing on a role for properdin in inflammatory and autoimmune diseases. This review examines the new properdin findings and their implications.


Assuntos
Properdina/imunologia , Animais , Doenças Autoimunes/imunologia , Ativação do Complemento , Proteínas do Sistema Complemento/imunologia , Humanos , Inflamação/imunologia , Neutrófilos/imunologia , Properdina/química
2.
Allergy ; 2024 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-39033312

RESUMO

BACKGROUND: During the COVID-19 pandemic, novel nanoparticle-based mRNA vaccines were developed. A small number of individuals developed allergic reactions to these vaccines although the mechanisms remain undefined. METHODS: To understand COVID-19 vaccine-mediated allergic reactions, we enrolled 19 participants who developed allergic events within 2 h of vaccination and 13 controls, nonreactors. Using standard hemolysis assays, we demonstrated that sera from allergic participants induced stronger complement activation compared to nonallergic subjects following ex vivo vaccine exposure. RESULTS: Vaccine-mediated complement activation correlated with anti-polyethelyne glycol (PEG) IgG (but not IgM) levels while anti-PEG IgE was undetectable in all subjects. Depletion of total IgG suppressed complement activation in select individuals. To investigate the effects of vaccine excipients on basophil function, we employed a validated indirect basophil activation test that stratified the allergic populations into high and low responders. Complement C3a and C5a receptor blockade in this system suppressed basophil response, providing strong evidence for complement involvement in vaccine-mediated basophil activation. Single-cell multiome analysis revealed differential expression of genes encoding the cytokine response and Toll-like receptor (TLR) pathways within the monocyte compartment. Differential chromatin accessibility for IL-13 and IL-1B genes was found in allergic and nonallergic participants, suggesting that in vivo, epigenetic modulation of mononuclear phagocyte immunophenotypes determines their subsequent functional responsiveness, contributing to the overall physiologic manifestation of vaccine reactions. CONCLUSION: These findings provide insights into the mechanisms underlying allergic reactions to COVID-19 mRNA vaccines, which may be used for future vaccine strategies in individuals with prior history of allergies or reactions and reduce vaccine hesitancy.

3.
J Immunol ; 200(8): 2786-2797, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29531168

RESUMO

Factor D (FD) is an essential component of the complement alternative pathway (AP). It is an attractive pharmaceutical target because it is an AP-specific protease circulating in blood. Most components of the complement activation pathways are produced by the liver, but FD is highly expressed by adipose tissue. Two critical questions are: 1) to what degree does adipose tissue contribute to circulating FD levels and 2) what quantity of FD is sufficient to maintain a functional AP? To address these issues, we studied a novel mouse strain with complete lipodystrophy (LD), the fld mouse with partial LD, an FD-deficient mouse, and samples from lipodystrophic patients. FD was undetectable in the serum of LD mice, which also showed minimal AP function. Reconstitution with purified FD, serum mixing experiments, and studies of partial LD mice all demonstrated that a low level of serum FD is sufficient for normal AP activity in the mouse system. This conclusion was further supported by experiments in which wild-type adipose precursors were transplanted into LD mice. Our results indicate that almost all FD in mouse serum is derived from adipose tissue. In contrast, FD levels were reduced ∼50% in the sera of patients with congenital generalized LD. Our studies further demonstrate that a relatively small amount of serum FD is sufficient to facilitate significant time-dependent AP activity in humans and in mice. Furthermore, this observation highlights the potential importance of obtaining nearly complete inhibition of FD in treating alternative complement activation in various autoimmune and inflammatory human diseases.


Assuntos
Tecido Adiposo/metabolismo , Fator D do Complemento/metabolismo , Lipodistrofia/sangue , Animais , Fator D do Complemento/análise , Humanos , Camundongos
4.
J Immunol ; 196(3): 1355-65, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26729803

RESUMO

Factor H is a circulating protein that regulates activation of the alternative pathway (AP) of complement. Mutations and genetic variations of factor H are associated with several AP-mediated diseases, highlighting the critical role of factor H in AP regulation. AP-mediated inflammation is typically triggered by illness or tissue injury, however, and tissue injury can trigger AP activation in individuals with fully functional factor H. This suggests that factor H function is affected by local conditions within tissues. We hypothesized that inducible proteins impair the ability of factor H to locally control the AP, thereby increasing AP activation. We used purified murine factor H to immunoprecipitate binding partners from mouse kidneys. Using immunoaffinity liquid chromatography-mass spectrometry, we identified annexin A2 as a factor H binding partner. Further experiments showed that annexin A2 reduces the binding of factor H to cell surfaces. Recombinant annexin A2 impaired complement regulation by factor H and increased complement activation on renal cell surfaces in vitro and in vivo. In a murine model of acute pneumococcal otitis media, the administration of annexin A2 increased AP-mediated bacterial opsonization and clearance. In conclusion, the local production of annexin A2 within tissues suppresses regulation of the AP by factor H. Annexin A2 can contribute to AP-mediated tissue inflammation by locally impairing factor H function, but it can also improve complement-mediated bacterial clearance.


Assuntos
Anexina A2/imunologia , Ativação do Complemento/imunologia , Fator H do Complemento/imunologia , Injúria Renal Aguda/imunologia , Animais , Western Blotting , Cromatografia Líquida , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunofluorescência , Imuno-Histoquímica , Imunoprecipitação , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Otite Média/imunologia , Traumatismo por Reperfusão/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Proc Natl Acad Sci U S A ; 110(46): E4335-44, 2013 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-24167262

RESUMO

Abdominal aortic aneurysm (AAA) is a common vascular disease associated with high mortality rate due to progressive enlargement and eventual rupture. There is currently no established therapy known to alter the rate of aneurysmal expansion. Thus, understanding the processes that initiate and sustain aneurysmal growth is pivotal for the development of medical therapies aimed at halting disease progression. Using an elastase-induced AAA mouse model that recapitulates key features of human AAA, we previously reported that a natural IgG antibody directs alternative pathway complement activation and initiates the inflammatory process that culminates in aneurysmal development. The target of this natural antibody, however, was unknown. Herein we identify a natural IgG that binds to fibrinogen deposited in elastase-perfused aortic tissues, activates the complement lectin pathway (LP), and induces AAA. Moreover, we establish that alterations in the glycosylation patterns of this antibody critically affect its ability to activate the LP in vivo. We find that LP activation precedes the alternative pathway and absence of the LP complement protein mannan-binding lectin abrogates elastase-induced AAA. In human AAA tissues the mouse anti-fibrinogen antibody recognizes epitopes that localize to the same areas that stain positively for mannan-binding lectin, which suggests that the complement LP is engaged in humans as well. Lastly, we demonstrate that circulating antibodies in a subset of AAA patients react against fibrinogen or fibrinogen-associated epitopes in human aneurysmal tissues. Our findings support the concept that an autoimmune process directed at aortic wall self-antigens may play a central role in the immunopathogenesis of AAA.


Assuntos
Aneurisma da Aorta Abdominal/etiologia , Ativação do Complemento/imunologia , Lectina de Ligação a Manose da Via do Complemento/imunologia , Fibrinogênio/imunologia , Imunoglobulina G/imunologia , Análise de Variância , Animais , Aneurisma da Aorta Abdominal/imunologia , Western Blotting , Ensaio de Imunoadsorção Enzimática , Proteínas da Matriz Extracelular/metabolismo , Imunofluorescência , Humanos , Imunoglobulina G/metabolismo , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Elastase Pancreática
6.
J Biol Chem ; 289(1): 450-63, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24214979

RESUMO

To survive and replicate within the human host, malaria parasites must invade erythrocytes. Invasion can be mediated by the P. falciparum reticulocyte-binding homologue protein 4 (PfRh4) on the merozoite surface interacting with complement receptor type 1 (CR1, CD35) on the erythrocyte membrane. The PfRh4 attachment site lies within the three N-terminal complement control protein modules (CCPs 1-3) of CR1, which intriguingly also accommodate binding and regulatory sites for the key complement activation-specific proteolytic products, C3b and C4b. One of these regulatory activities is decay-accelerating activity. Although PfRh4 does not impact C3b/C4b binding, it does inhibit this convertase disassociating capability. Here, we have employed ELISA, co-immunoprecipitation, and surface plasmon resonance to demonstrate that CCP 1 contains all the critical residues for PfRh4 interaction. We fine mapped by homologous substitution mutagenesis the PfRh4-binding site on CCP 1 and visualized it with a solution structure of CCPs 1-3 derived by NMR and small angle x-ray scattering. We cross-validated these results by creating an artificial PfRh4-binding site through substitution of putative PfRh4-interacting residues from CCP 1 into their homologous positions within CCP 8; strikingly, this engineered binding site had an ∼30-fold higher affinity for PfRh4 than the native one in CCP 1. These experiments define a candidate site on CR1 by which P. falciparum merozoites gain access to human erythrocytes in a non-sialic acid-dependent pathway of merozoite invasion.


Assuntos
Proteínas de Membrana/metabolismo , Merozoítos/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Receptores de Complemento 3b/metabolismo , Sítios de Ligação , Complemento C3b/química , Complemento C3b/genética , Complemento C3b/metabolismo , Complemento C4b/química , Complemento C4b/genética , Complemento C4b/metabolismo , Eritrócitos/química , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Células HEK293 , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Merozoítos/química , Mutagênese , Ressonância Magnética Nuclear Biomolecular , Plasmodium falciparum/química , Plasmodium falciparum/genética , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Receptores de Complemento 3b/química , Receptores de Complemento 3b/genética , Espalhamento a Baixo Ângulo , Ressonância de Plasmônio de Superfície , Difração de Raios X
7.
Nanomedicine ; 11(3): 601-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25652900

RESUMO

High-relaxivity T1-weighted (T1w) MR molecular imaging nanoparticles typically present high surface gadolinium payloads that can elicit significant acute complement activation (CA). The objective of this research was to develop a high T1w contrast nanoparticle with improved safety. We report the development, optimization, and characterization of a gadolinium-manganese hybrid nanocolloid (MnOL-Gd NC; 138±10 (Dav)/nm; PDI: 0.06; zeta: -27±2 mV). High r1 particulate relaxivity with minute additions of Gd-DOTA-lipid conjugate to the MnOL nanocolloid surface achieved an unexpected paramagnetic synergism. This hybrid MnOL-Gd NC provided optimal MR TSE signal intensity at 5 nM/voxel and lower levels consistent with the level expression anticipated for sparse biomarkers, such as neovascular integrins. MnOL NC produced optimal MR TSE signal intensity at 10 nM/voxel concentrations and above. Importantly, MnOL-Gd NC avoided acute CA in vitro and in vivo while retaining minimal transmetallation risk. From the clinical editor: The authors developed a gadolinium-manganese hybrid nanocolloid (MnOL-Gd NC) in this study. These were used as a high-relaxivity paramagnetic MR molecular imaging agent in experimental models. It was shown that MnOL-Gd NC could provide high T1w MR contrast for targeted imaging. As the level of gadolinium used was reduced, there was also reduced risk of systemic side effects from complement activation.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Meios de Contraste , Gadolínio , Imageamento por Ressonância Magnética , Manganês , Nanopartículas , Animais , Biomarcadores/sangue , Coloides , Meios de Contraste/efeitos adversos , Meios de Contraste/química , Meios de Contraste/farmacologia , Avaliação Pré-Clínica de Medicamentos , Gadolínio/efeitos adversos , Gadolínio/química , Gadolínio/farmacologia , Manganês/efeitos adversos , Manganês/química , Manganês/farmacologia , Camundongos , Nanopartículas/efeitos adversos , Nanopartículas/química
8.
Proc Natl Acad Sci U S A ; 109(7): E415-22, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22308431

RESUMO

Abdominal aortic aneurysm (AAA) is a complex inflammatory vascular disease. There are currently limited treatment options for AAA when surgery is inapplicable. Therefore, insights into molecular mechanisms underlying AAA pathogenesis may reveal therapeutic targets that could be manipulated pharmacologically or biologically to halt disease progression. Using an elastase-induced AAA mouse model, we previously established that the complement alternative pathway (AP) plays a critical role in the development of AAA. However, the mechanism by which complement AP is initiated remains undefined. The complement protein properdin, traditionally viewed as a positive regulator of the AP, may also initiate complement activation by binding directly to target surfaces. In this study, we sought to determine whether properdin serves as a focal point for the initiation of the AP complement activation in AAA. Using a properdin loss of function mutation in mice and a mutant form of the complement factor B protein that produces a stable, properdin-free AP C3 convertase, we show that properdin is required for the development of elastase-induced AAA in its primary role as a convertase stabilizer. Unexpectedly, we find that, in AAA, natural IgG antibodies direct AP-mediated complement activation. The absence of IgG abrogates C3 deposition in elastase-perfused aortic wall and protects animals from AAA development. We also determine that blockade of properdin activity prevents aneurysm formation. These results indicate that an innate immune response to self-antigens activates the complement system and initiates the inflammatory cascade in AAA. Moreover, the study suggests that properdin-targeting strategies may halt aneurysmal growth.


Assuntos
Aneurisma da Aorta Abdominal/metabolismo , Proteínas do Sistema Complemento/metabolismo , Modelos Animais de Doenças , Properdina/metabolismo , Animais , Antibacterianos/farmacologia , Ativação do Complemento/efeitos dos fármacos , Camundongos
9.
Nanomedicine ; 10(3): 651-60, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24211337

RESUMO

Nanoparticles offer new options for medical diagnosis and therapeutics with their capacity to specifically target cells and tissues with imaging agents and/or drug payloads. The unique physical aspects of nanoparticles present new challenges for this promising technology. Studies indicate that nanoparticles often elicit moderate to severe complement activation. Using human in vitro assays that corroborated the mouse in vivo results we previously presented mechanistic studies that define the pathway and key components involved in modulating complement interactions with several gadolinium-functionalized perfluorocarbon nanoparticles (PFOB). Here we employ a modified in vitro hemolysis-based assay developed in conjunction with the mouse in vivo model to broaden our analysis to include PFOBs of varying size, charge and surface chemistry and examine the variations in nanoparticle-mediated complement activity between individuals. This approach may provide the tools for an in-depth structure-activity relationship study that will guide the eventual development of biocompatible nanoparticles. FROM THE CLINICAL EDITOR: Unique physical aspects of nanoparticles may lead to moderate to severe complement activation in vivo, which represents a challenge to clinical applicability. In order to guide the eventual development of biocompatible nanoparticles, this team of authors report a modified in vitro hemolysis-based assay developed in conjunction with their previously presented mouse model to enable in-depth structure-activity relationship studies.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Fluorocarbonos/imunologia , Hemólise/efeitos dos fármacos , Nanopartículas/metabolismo , Animais , Fluorocarbonos/química , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/química , Tamanho da Partícula
10.
Artigo em Inglês | MEDLINE | ID: mdl-36853837

RESUMO

Complement is a major innate defense system that protects the intravascular space from microbial invasion. Complement activation results in the assembly of C3 convertases, serine proteases that cleave complement protein C3, generating bioactive fragments C3a and C3b. The complement response is rapid and robust, largely due to a positive feedback regulatory loop mediated by alternative pathway (AP) C3 convertase. C3 nephritic factors (C3NEFs) are autoantibodies that stabilize AP convertase, resulting in uncontrolled C3 cleavage, which, in principle, can promote critical tissue injury similar to that seen in certain renal conditions. Investigations of C3NEFs are hampered by a challenging issue: each C3NEF is derived from a different donor source, and there is no method to compare one C3NEF to another. We have identified a widely available mouse anti-C3 mAb that, similar to many C3NEFs, can stabilize functional AP convertase in a form resistant to decay acceleration by multiple complement regulators. The antibody requires the presence of properdin to confer convertase stability, and hampers the activity of Salp20, a tic salivary protein that accelerates convertase dissociation by displacing properdin from the convertase complex. This mAb can serve as an urgently needed standard for the investigation of C3NEFs. This study also provides novel insights into the dynamics of AP convertase.


Assuntos
Anticorpos Monoclonais , Fator Nefrítico do Complemento 3 , Animais , Camundongos , Properdina , Autoanticorpos
11.
J Biol Chem ; 286(41): 35725-35732, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-21862585

RESUMO

Factor B is a zymogen that carries the catalytic site of the complement alternative pathway C3 convertase. During convertase assembly, factor B associates with C3b and Mg(2+) forming a pro-convertase C3bB(Mg(2+)) that is cleaved at a single factor B site by factor D. In free factor B, a pair of salt bridges binds the Arg(234) side chain to Glu(446) and to Glu(207), forming a double latch structure that sequesters the scissile bond (between Arg(234) and Lys(235)) and minimizes its unproductive cleavage. It is unknown how the double latch is released in the pro-convertase. Here, we introduce single amino acid substitutions into factor B that preclude one or both of the Arg(234) salt bridges, and we examine their impact on several different pro-convertase complexes. Our results indicate that loss of the Arg(234)-Glu(446) salt bridge partially stabilizes C3bB(Mg(2+)). Loss of the Arg(234)-Glu(207) salt bridge has lesser effects. We propose that when factor B first associates with C3b, it bears two intact Arg(234) salt bridges. The complex rapidly dissociates unless the Arg(234)-Glu(446) salt bridge is released whereupon conformational changes occur that activate the metal ion-dependent adhesion site and partially stabilize the complex. The remaining salt bridge is then released, exposing the scissile bond and permitting factor D cleavage.


Assuntos
Complemento C3b/química , Fator B do Complemento/química , Fator D do Complemento/química , Complexos Multienzimáticos/química , Substituição de Aminoácidos , Complemento C3b/genética , Complemento C3b/metabolismo , Fator B do Complemento/genética , Fator B do Complemento/metabolismo , Fator D do Complemento/genética , Fator D do Complemento/metabolismo , Humanos , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Mutação de Sentido Incorreto , Ligação Proteica
12.
J Biol Chem ; 286(1): 123-30, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21047788

RESUMO

A wide variety of nanomaterials are currently being developed for use in the detection and treatment of human diseases. However, there is no systematic way to measure and predict the action of such materials in biological contexts. Lipid-encapsulated nanoparticles (NPs) are a class of nanomaterials that includes the liposomes, the most widely used and clinically proven type of NPs. Liposomes can, however, activate the complement system, an important branch of innate immunity, resulting in undesirable consequences. Here, we describe the complement response to lipid-encapsulated NPs that are functionalized on the surface with various lipid-anchored gadolinium chelates. We developed a quantitative approach to examine the interaction of NPs with the complement system using in vitro assays and correlating these results with those obtained in an in vivo mouse model. Our results indicate that surface functionalization of NPs with certain chemical structures elicits swift complement activation that is initiated by a natural IgM antibody and propagated via the classical pathway. The intensity of the response is dependent on the chemical structures of the lipid-anchored chelates and not zeta potential effects alone. Moreover, the extent of complement activation may be tempered by complement inhibiting regulatory proteins that bind to the surface of NPs. These findings represent a step forward in the understanding of the interactions between nanomaterials and the host innate immune response and provide the basis for a systematic structure-activity relationship study to establish guidelines that are critical to the future development of biocompatible nanotherapeutics.


Assuntos
Anticorpos/imunologia , Proteínas do Sistema Complemento/imunologia , Nanocápsulas/química , Fosfolipídeos , Animais , Desenho de Fármacos , Gadolínio/química , Humanos , Imunoglobulina M/imunologia , Camundongos , Nanocápsulas/efeitos adversos , Propriedades de Superfície
13.
Proc Natl Acad Sci U S A ; 105(26): 9023-8, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18579773

RESUMO

Apoptotic cells must be rapidly eliminated to avoid harmful inflammatory and autoimmune reactions. Innate immunity is designed/poised to identify dying cells by their unique surface-associated molecular patterns. Here we demonstrate for the first time, to our knowledge, that the human complement protein properdin binds to early apoptotic T cells and initiates complement activation, leading to C3b opsonization and ingestion by phagocytic cells. Properdin binding was facilitated by the glycosaminoglycan chains of surface proteoglycans. Properdin released by activated neutrophils was particularly effective at recognition of apoptotic T cells, whereas the binding activity of properdin in the serum appeared to be inhibited. "Properdin tagging" of apoptotic T cells also induced their uptake by phagocytes independent of complement activation or other complement proteins. Although our findings were made primarily with apoptotic T cells, they suggest that properdin could play a similar role during apoptosis of other cell types.


Assuntos
Apoptose , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Ativação do Complemento/imunologia , Fagocitose/imunologia , Properdina/imunologia , Linfócitos T CD4-Positivos/patologia , Complemento C3b/imunologia , Células Dendríticas/imunologia , Glicosaminoglicanos/imunologia , Humanos , Macrófagos/imunologia , Neutrófilos/imunologia , Fagócitos/citologia , Fagócitos/imunologia , Ligação Proteica , Proteoglicanas/imunologia
14.
Circulation ; 119(13): 1805-13, 2009 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-19307471

RESUMO

BACKGROUND: We previously established that neutrophils play a critical role in the development of experimental abdominal aortic aneurysm (AAA). The signal that initiates the influx of neutrophils to the aortic wall, however, remains unknown. In this study, we tested the hypothesis that complement participates in the development of AAA by providing the necessary chemotactic signal that recruits neutrophils to the aortic wall. METHODS AND RESULTS: Using an elastase-induced model of AAA, we showed that pretreatment of C57BL/6 mice with cobra venom factor, which depleted serum of complement activity, protected mice from AAA development. Whereas control mice exhibited a mean aortic diameter of 156+/-2% on day 14 after elastase perfusion, mice treated with cobra venom factor exhibited a mean aortic diameter of 90+/-4% (P<0.001). Examination of mice deficient in factor B further indicated that the alternative pathway of complement played a major role in this process (mean aortic diameter of 105+/-4% in factor B-deficient mice, P<0.001 compared with controls). Activation of the alternative pathway led to generation of the anaphylatoxins C3a and C5a, which recruited neutrophils to the aortic wall. Moreover, antagonism of both C3a and C5a activity was required to block AAA, which suggests that each can independently promote the aneurysmal phenotype. In addition, we demonstrated that complement alternative-pathway involvement was not restricted to this experimental model but was also evident in human AAAs. CONCLUSIONS: The identification of involvement of the complement system in the pathophysiology of AAA provides a new target for therapeutic intervention in this common disease.


Assuntos
Aneurisma da Aorta Abdominal/imunologia , Aneurisma da Aorta Abdominal/metabolismo , Proteínas do Sistema Complemento/metabolismo , Neutrófilos/imunologia , Elastase Pancreática/metabolismo , Animais , Aorta/imunologia , Aorta/metabolismo , Complemento C3a/imunologia , Complemento C3a/metabolismo , Complemento C4/imunologia , Complemento C4/metabolismo , Complemento C5a/imunologia , Complemento C5a/metabolismo , Fator B do Complemento/genética , Fator B do Complemento/imunologia , Fator B do Complemento/metabolismo , Proteínas do Sistema Complemento/imunologia , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Neutrófilos/metabolismo
15.
Neurology ; 95(14): e1918-e1931, 2020 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-32887784

RESUMO

OBJECTIVE: To characterize lesion evolution and neurodegeneration in retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations (RVCL-S) using multimodal MRI. METHODS: We prospectively performed MRI and cognitive testing in RVCL-S and healthy control cohorts. Gray and white matter volume and disruption of white matter microstructure were quantified. Asymmetric spin echo acquisition permitted voxel-wise oxygen extraction fraction (OEF) calculation as an in vivo marker of microvascular ischemia. The RVCL-S cohort was included in a longitudinal analysis of lesion subtypes in which hyperintense lesions on fluid-attenuated inversion recovery (FLAIR), T1-postgadolinium, and diffusion-weighted imaging were delineated and quantified volumetrically. RESULTS: Twenty individuals with RVCL-S and 26 controls were enrolled. White matter volume and microstructure declined faster in those with RVCL-S compared to controls. White matter atrophy in RVCL-S was highly linear (ρ = -0.908, p < 0.0001). Normalized OEF was elevated in RVCL-S and increased with disease duration. Multiple cognitive domains, specifically those measuring working memory and processing speed, were impaired in RVCL-S. Lesion volumes, regardless of subtype, progressed/regressed with high variability as a function of age, while FLAIR lesion burden increased near time to death (p < 0.001). CONCLUSION: RVCL-S is a monogenic microvasculopathy affecting predominantly the white matter with regard to atrophy and cognitive impairment. White matter volumes in RVCL-S declined linearly, providing a potential metric against which to test the efficacy of future therapies. Progressive elevation of white matter OEF suggests that microvascular ischemia may underlie neurodegeneration in RVCL-S.


Assuntos
Disfunção Cognitiva/patologia , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/patologia , Degeneração Neural/patologia , Doenças Retinianas/patologia , Doenças Vasculares/patologia , Substância Branca/patologia , Adulto , Disfunção Cognitiva/diagnóstico por imagem , Feminino , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/diagnóstico por imagem , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Degeneração Neural/diagnóstico por imagem , Neuroimagem/métodos , Doenças Retinianas/diagnóstico por imagem , Doenças Vasculares/diagnóstico por imagem , Substância Branca/diagnóstico por imagem
16.
Mol Immunol ; 45(16): 4048-56, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18692243

RESUMO

Properdin was first described over 50 years ago by Louis Pillemer and his collaborators as a vital component of an antibody-independent complement activation pathway. In the 1970s properdin was shown to be a stabilizing component of the alternative pathway convertases, the central enzymes of the complement cascade. Recently we have reported that properdin can also bind to target cells and microbes, provide a platform for convertase assembly and function, and promote target phagocytosis. Evidence is emerging that suggests that properdin interacts with a network of target ligands, phagocyte receptors, and serum regulators. Here we review the new findings and their possible implications.


Assuntos
Via Alternativa do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Properdina/imunologia , Animais , Proteínas do Sistema Complemento/metabolismo , Humanos , Modelos Imunológicos , Properdina/metabolismo
17.
Mol Immunol ; 45(11): 3125-32, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18452991

RESUMO

UNLABELLED: Complement signaling has been implicated as important for normal hepatic regeneration. However, the specific mechanism by which complement is activated during liver regeneration remains undefined. To address this question, we investigated the hepatic regenerative response to partial hepatectomy in wildtype mice, C3-, C4-, and factor B-null mice, and C4-null mice treated with a factor B neutralizing antibody (mAb 1379). The results showed that following partial hepatectomy, C3-null mice exhibit reduced hepatic regeneration compared to wildtype mice as assessed by quantification of hepatic cyclin D1 expression and hepatocellular DNA synthesis and mitosis. In contrast, C4-null mice and factor B-null mice demonstrated normal liver regeneration. Moreover, animals in which all of the traditional upstream C3 activation pathways were disrupted, i.e. C4-null mice treated with mAb 1379, exhibited normal C3 activation and hepatocellular proliferation following partial hepatectomy. In order to define candidate non-traditional mechanisms of C3 activation during liver regeneration, plasmin and thrombin were investigated for their abilities to activate C3 in mouse plasma in vitro. The results showed that both proteases are capable of initiating C3 activation, and that plasmin can do so independent of the classical and alternative pathways. CONCLUSIONS: These results show that C3 is required for a normal hepatic regenerative response, but that disruption of the classical- or lectin-dependent pathways (C4-dependent), the alternative pathway (factor B-dependent), or all of these pathways does not impair the hepatic regenerative response, and indicate that non-traditional mechanisms by which C3 is activated during hepatic regeneration must exist. In vitro analysis raises the possibility that plasmin may contribute to non-traditional complement activation during liver regeneration in vivo.


Assuntos
Ativação do Complemento , Complemento C3/imunologia , Regeneração Hepática/imunologia , Animais , Anticorpos Monoclonais/imunologia , Complemento C3/deficiência , Complemento C4/deficiência , Complemento C4/imunologia , Fator B do Complemento/deficiência , Fator B do Complemento/imunologia , Immunoblotting , Fígado/citologia , Fígado/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Testes de Neutralização , Processamento de Proteína Pós-Traducional
18.
Mol Immunol ; 114: 629-642, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31542608

RESUMO

Neutrophils are essential to the pathogenesis of many inflammatory diseases. In the autoantibody-mediated K/BxN model of inflammatory arthritis, the alternative pathway (AP) of complement and Fc gamma receptors (FcγRs) are required for disease development while the classical pathway is dispensable. The reason for this differential requirement is unknown. We show that within minutes of K/BxN serum injection complement activation (CA) is detected on circulating neutrophils, as evidenced by cell surface C3 fragment deposition. CA requires the AP factor B and FcγRs but not C4, implying that engagement of FcγRs by autoantibody or immune complexes directly triggers AP C3 convertase assembly. The absence of C5 does not prevent CA on neutrophils but diminishes the upregulation of adhesion molecules. In vivo two-photon microscopy reveals that CA on neutrophils is critical for neutrophil extravasation and generation of C5a at the site of inflammation. C5a stimulates the release of neutrophil proteases, which contribute to the degradation of VE-cadherin, an adherens junction protein that regulates endothelial barrier integrity. C5a receptor antagonism blocks the extracellular release of neutrophil proteases, suppressing VE-cadherin degradation and neutrophil transendothelial migration in vivo. These results elucidate the AP-dependent intravascular neutrophil-endothelial interactions that initiate the inflammatory cascade in this disease model but may be generalizable to neutrophil extravasation in other inflammatory processes.


Assuntos
Adesão Celular/imunologia , Ativação do Complemento/imunologia , Complemento C5a/imunologia , Células Endoteliais da Veia Umbilical Humana/imunologia , Neutrófilos/imunologia , Animais , Complexo Antígeno-Anticorpo/imunologia , Autoanticorpos/imunologia , Células Cultivadas , Feminino , Humanos , Masculino , Camundongos Endogâmicos C57BL , Receptores de IgG/imunologia
19.
Am J Reprod Immunol ; 80(4): e12997, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29924462

RESUMO

PROBLEM: Crry is a widely expressed type 1 transmembrane complement regulatory protein in rodents which protects self-tissue by downregulating C3 activation. Crry-/- concepti produced by Crry+/-  × Crry+/- matings are attacked by maternal complement system leading to loss before day 10. The membrane attack complex is not the mediator of this death. We hypothesized that the ability of C3b to engage the alternative pathway's feedback loop relatively unchecked on placental membranes induces the lesion yielding the demise of the Crry-/- mouse. METHOD OF STUDY: We investigated the basis of Crry-/- conceptus demise by depleting maternal complement with cobra venom factor and blocking antibodies. We monitored their effects primarily by genotyping and histologic analyses. RESULTS: We narrowed the critical period of the complement effect from 6.5 to 8.5 days post-coitus (dpc), which is immediately after the conceptus is exposed to maternal blood. Deposition by 5.5 dpc of maternal C3b on the placental vasculature lacking Crry-/- yielded loss of the conceptus by 8.5 dpc. Fusion of the allantois to the chorion during placental assembly did not occur, fetal vessels originating in the allantois did not infiltrate the chorioallantoic placenta, the chorionic plate failed to develop, and the labyrinthine component of the placenta did not mature. CONCLUSION: Our data are most consistent with the deposition of C3b being responsible for the failure of the allantois to fuse to the chorion leading to subsequent conceptus demise.


Assuntos
Aborto Espontâneo/genética , Ativação do Complemento/imunologia , Complemento C3b/imunologia , Via Alternativa do Complemento/imunologia , Embrião de Mamíferos/patologia , Receptores de Complemento/genética , Aborto Espontâneo/imunologia , Animais , Convertases de Complemento C3-C5 da Via Alternativa/metabolismo , Embrião de Mamíferos/imunologia , Feminino , Camundongos , Camundongos Knockout , Placenta/imunologia , Placenta/patologia , Gravidez , Receptores de Complemento 3b
20.
Mol Immunol ; 42(1): 87-97, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15488947

RESUMO

Studies in gene-targeted mice have demonstrated that factor B of the alternative complement pathway plays an important role in several disease models, but an exogenous inhibitor of factor B has not previously been available. We have developed an inhibitory monoclonal antibody directed against a critical epitope on mouse factor B and have tested it in a model of antiphospholipid (aPL) antibody (Ab)-induced fetal loss. Gene-targeted factor B-deficient mice (fB-/-) were injected with a fusion protein comprised of the second and third short consensus repeat (SCR) domains of mouse factor B linked to a mouse IgG1 Fc domain. Hybridomas were made from splenocytes of the immunized mouse. One mAb, designated 1379, produced an IgG1 antibody that inhibited alternative pathway activation in vitro and in vivo by preventing formation of the C3bBb complex. Strikingly, this mAb inhibited alternative pathway activation in serum from mice, rats, humans, monkeys, pigs and horses. Fab fragments made from this mAb also inhibited alternative pathway activation. Epitope mapping demonstrated that this antibody binds to factor B within the third SCR domain. When mAb 1379 was administered to mice that also received human IgG containing antiphospholipid antibodies, it provided significant protection from antiphospholipid antibody-induced complement activation and fetal loss. Thus, this mAb to factor B has broad species reactivity and effectively inhibits alternative pathway activation. The mAb protects mice in an in vivo model of antiphospholipid antibody syndrome, demonstrating the therapeutic potential for the inhibition of factor B in this disease.


Assuntos
Anticorpos Antifosfolipídeos/efeitos adversos , Ativação do Complemento/efeitos dos fármacos , Morte Fetal/prevenção & controle , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Fator B do Complemento/antagonistas & inibidores , Fator B do Complemento/imunologia , Modelos Animais de Doenças , Mapeamento de Epitopos , Feminino , Morte Fetal/imunologia , Humanos , Camundongos , Camundongos Knockout , Gravidez , Complicações Hematológicas na Gravidez/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa