Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 341
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 212(12): 1922-1931, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38683124

RESUMEN

Although high titers of neutralizing Abs in human serum are associated with protection from reinfection by SARS-CoV-2, there is considerable heterogeneity in human serum-neutralizing Abs against SARS-CoV-2 during convalescence between individuals. Standard human serum live virus neutralization assays require inactivation of serum/plasma prior to testing. In this study, we report that the SARS-CoV-2 neutralization titers of human convalescent sera were relatively consistent across all disease states except for severe COVID-19, which yielded significantly higher neutralization titers. Furthermore, we show that heat inactivation of human serum significantly lowered neutralization activity in a live virus SARS-CoV-2 neutralization assay. Heat inactivation of human convalescent serum was shown to inactivate complement proteins, and the contribution of complement in SARS-CoV-2 neutralization was often >50% of the neutralizing activity of human sera without heat inactivation and could account for neutralizing activity when standard titers were zero after heat inactivation. This effect was also observed in COVID-19 vaccinees and could be abolished in individuals who were undergoing treatment with therapeutic anti-complement Abs. Complement activity was mainly dependent on the classical pathway with little contributions from mannose-binding lectin and alternative pathways. Our study demonstrates the importance of the complement pathway in significantly increasing viral neutralization activity against SARS-CoV-2 in spike seropositive individuals.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Vacunas contra la COVID-19 , COVID-19 , Vía Clásica del Complemento , Pruebas de Neutralización , SARS-CoV-2 , Humanos , SARS-CoV-2/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , COVID-19/inmunología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Vía Clásica del Complemento/inmunología , Vacunas contra la COVID-19/inmunología , Masculino , Femenino , Persona de Mediana Edad , Adulto , Convalecencia , Anciano , Proteínas del Sistema Complemento/inmunología
2.
J Biol Chem ; 300(5): 107236, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38552741

RESUMEN

The complement system serves as the first line of defense against invading pathogens by promoting opsonophagocytosis and bacteriolysis. Antibody-dependent activation of complement occurs through the classical pathway and relies on the activity of initiating complement proteases of the C1 complex, C1r and C1s. The causative agent of Lyme disease, Borrelia burgdorferi, expresses two paralogous outer surface lipoproteins of the OspEF-related protein family, ElpB and ElpQ, that act as specific inhibitors of classical pathway activation. We have previously shown that ElpB and ElpQ bind directly to C1r and C1s with high affinity and specifically inhibit C2 and C4 cleavage by C1s. To further understand how these novel protease inhibitors function, we carried out a series of hydrogen-deuterium exchange mass spectrometry (HDX-MS) experiments using ElpQ and full-length activated C1s as a model of Elp-protease interaction. Comparison of HDX-MS profiles between unbound ElpQ and the ElpQ/C1s complex revealed a putative C1s-binding site on ElpQ. HDX-MS-guided, site-directed ElpQ mutants were generated and tested for direct binding to C1r and C1s using surface plasmon resonance. Several residues within the C-terminal region of ElpQ were identified as important for protease binding, including a single conserved tyrosine residue that was required for ElpQ- and ElpB-mediated complement inhibition. Collectively, our study identifies key molecular determinants for classical pathway protease recognition by Elp proteins. This investigation improves our understanding of the unique complement inhibitory mechanism employed by Elp proteins which serve as part of a sophisticated complement evasion system present in Lyme disease spirochetes.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa , Borrelia burgdorferi , Vía Clásica del Complemento , Humanos , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Borrelia burgdorferi/inmunología , Borrelia burgdorferi/metabolismo , Borrelia burgdorferi/genética , Complemento C1r/metabolismo , Complemento C1r/genética , Complemento C1s/metabolismo , Complemento C1s/genética , Complemento C1s/química , Vía Clásica del Complemento/inmunología , Lipoproteínas/metabolismo , Lipoproteínas/genética , Lipoproteínas/química , Lipoproteínas/inmunología , Enfermedad de Lyme/genética , Enfermedad de Lyme/inmunología , Enfermedad de Lyme/microbiología , Unión Proteica
3.
Eur J Immunol ; 54(7): e2350918, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38629181

RESUMEN

For many years complement activation in systemic lupus erythematosus (SLE) was viewed as a major cause of tissue injury. However, human and murine studies showed that complement plays a protective as well as a proinflammatory role in tissue damage. A hierarchy is apparent with early classical pathway components, particularly C1q, exerting the greatest influence. Understanding the mechanisms underlying the protective function(s) of complement remains an important challenge for the future and has implications for the use of complement therapy in SLE. We review recent advances in the field and give a new perspective on the complement conundrum in SLE.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento , Lupus Eritematoso Sistémico , Lupus Eritematoso Sistémico/inmunología , Humanos , Animales , Activación de Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Complemento C1q/inmunología , Complemento C1q/metabolismo , Ratones , Vía Clásica del Complemento/inmunología
4.
J Biol Chem ; 297(3): 101085, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34411562

RESUMEN

The complement cascade is a key component of the innate immune system that is rapidly recruited through a cascade of enzymatic reactions to enable the recognition and clearance of pathogens and promote tissue repair. Despite its well-understood role in immunology, recent studies have highlighted new and unexpected roles of the complement cascade in neuroimmune interaction and in the regulation of neuronal processes during development, aging, and in disease states. Complement signaling is particularly important in directing neuronal responses to tissue injury, neurotrauma, and nerve lesions. Under physiological conditions, complement-dependent changes in neuronal excitability, synaptic strength, and neurite remodeling promote nerve regeneration, tissue repair, and healing. However, in a variety of pathologies, dysregulation of the complement cascade leads to chronic inflammation, persistent pain, and neural dysfunction. This review describes recent advances in our understanding of the multifaceted cross-communication that takes place between the complement system and neurons. In particular, we focus on the molecular and cellular mechanisms through which complement signaling regulates neuronal excitability and synaptic plasticity in the nociceptive pathways involved in pain processing in both health and disease. Finally, we discuss the future of this rapidly growing field and what we believe to be the significant knowledge gaps that need to be addressed.


Asunto(s)
Vía Clásica del Complemento/inmunología , Neuroinmunomodulación/fisiología , Dolor Nociceptivo/fisiopatología , Animales , Activación de Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Humanos , Inmunidad Innata/fisiología , Neuroinmunomodulación/inmunología , Plasticidad Neuronal/fisiología , Neuronas , Nocicepción , Dolor Nociceptivo/inmunología , Dolor/inmunología , Dolor/fisiopatología , Transducción de Señal
5.
Nature ; 534(7608): 538-43, 2016 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-27337340

RESUMEN

Over 50% of patients who survive neuroinvasive infection with West Nile virus (WNV) exhibit chronic cognitive sequelae. Although thousands of cases of WNV-mediated memory dysfunction accrue annually, the mechanisms responsible for these impairments are unknown. The classical complement cascade, a key component of innate immune pathogen defence, mediates synaptic pruning by microglia during early postnatal development. Here we show that viral infection of adult hippocampal neurons induces complement-mediated elimination of presynaptic terminals in a murine WNV neuroinvasive disease model. Inoculation of WNV-NS5-E218A, a WNV with a mutant NS5(E218A) protein leads to survival rates and cognitive dysfunction that mirror human WNV neuroinvasive disease. WNV-NS5-E218A-recovered mice (recovery defined as survival after acute infection) display impaired spatial learning and persistence of phagocytic microglia without loss of hippocampal neurons or volume. Hippocampi from WNV-NS5-E218A-recovered mice with poor spatial learning show increased expression of genes that drive synaptic remodelling by microglia via complement. C1QA was upregulated and localized to microglia, infected neurons and presynaptic terminals during WNV neuroinvasive disease. Murine and human WNV neuroinvasive disease post-mortem samples exhibit loss of hippocampal CA3 presynaptic terminals, and murine studies revealed microglial engulfment of presynaptic terminals during acute infection and after recovery. Mice with fewer microglia (Il34(-/-) mice with a deficiency in IL-34 production) or deficiency in complement C3 or C3a receptor were protected from WNV-induced synaptic terminal loss. Our study provides a new murine model of WNV-induced spatial memory impairment, and identifies a potential mechanism underlying neurocognitive impairment in patients recovering from WNV neuroinvasive disease.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Trastornos de la Memoria/patología , Trastornos de la Memoria/virología , Microglía/inmunología , Plasticidad Neuronal , Terminales Presinápticos/patología , Virus del Nilo Occidental/patogenicidad , Animales , Región CA3 Hipocampal/inmunología , Región CA3 Hipocampal/patología , Región CA3 Hipocampal/virología , Activación de Complemento , Vía Clásica del Complemento/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Trastornos de la Memoria/inmunología , Trastornos de la Memoria/fisiopatología , Ratones , Neuronas/inmunología , Neuronas/patología , Neuronas/virología , Terminales Presinápticos/inmunología , Memoria Espacial , Fiebre del Nilo Occidental/patología , Fiebre del Nilo Occidental/fisiopatología , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/inmunología
6.
Blood ; 132(23): 2431-2440, 2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30309891

RESUMEN

The mechanisms by which exposure to heparin initiates antibody responses in many, if not most, recipients are poorly understood. We recently demonstrated that antigenic platelet factor 4 (PF4)/heparin complexes activate complement in plasma and bind to B cells. Here, we describe how this process is initiated. We observed wide stable variation in complement activation when PF4/heparin was added to plasma of healthy donors, indicating a responder "phenotype" (high, intermediate, or low). Proteomic analysis of plasma from these healthy donors showed a strong correlation between complement activation and plasma immunoglobulin M (IgM) levels (r = 0.898; P < .005), but not other Ig isotypes. Complement activation response to PF4/heparin in plasma displaying the low donor phenotype was enhanced by adding pooled IgM from healthy donors, but not monoclonal IgM. Depletion of IgM from plasma abrogated C3c generation by PF4/heparin. The complement-activating features of IgM are likely mediated by nonimmune, or natural, IgM, as cord blood and a monoclonal polyreactive IgM generate C3c in the presence of PF4/heparin. IgM facilitates complement and antigen deposition on B cells in vitro and in patients receiving heparin. Anti-C1q antibody prevents IgM-mediated complement activation by PF4/heparin complexes, indicating classical pathway involvement. These studies demonstrate that variability in plasma IgM levels correlates with functional complement responses to PF4/heparin. Polyreactive IgM binds PF4/heparin, triggers activation of the classical complement pathway, and promotes antigen and complement deposition on B cells. These studies provide new insights into the evolution of the heparin-induced thrombocytopenia immune response and may provide a biomarker of risk.


Asunto(s)
Linfocitos B/inmunología , Vía Clásica del Complemento/inmunología , Heparina/inmunología , Inmunoglobulina M/inmunología , Activación de Linfocitos , Factor Plaquetario 4/inmunología , Complemento C3c/inmunología , Vía Clásica del Complemento/efectos de los fármacos , Heparina/farmacología , Humanos , Factor Plaquetario 4/farmacología , Proteómica
7.
Ann Diagn Pathol ; 48: 151564, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32659621

RESUMEN

BACKGROUND: Surveillance transbronchial biopsies are routinely used to assess lung allograft rejection. While the criteria for diagnosing acute cellular rejection have been well-established, the morphological findings associated with antibody mediated rejection are variable. To increase the sensitivity for antibody mediated rejection, a portion of a biopsy can be used for C4d immunofluorescence testing, along with histologic findings and donor specific antibodies. When the number of alveolar pieces in a biopsy is small, the relative utility of sending one piece for C4d testing is unclear. METHODS: Pathology reports of 1400 surveillance transbronchial lung biopsies from 2008 to 2017 were reviewed to obtain the number of pieces of alveolar parenchyma in each case. Based on a standard definition of adequacy as five pieces of well-expanded alveolar parenchyma, reports with five fragments were grouped as "adequate", four pieces as a "marginal" sample, and three or less were considered an "inadequate" sample. RESULTS: Of the 1400 biopsies, 653 specimens had 5 or more pieces of alveolar parenchyma.747 specimens were submitted with less than 5 pieces and 290 of those were considered marginal. In all marginal cases, a piece was withheld for C4d immunofluorescence testing. CONCLUSIONS: About 21% of specimens would have the recommended 5 pieces of alveolar parenchyma if not for the withholding of pieces for C4d IF testing. Over the span of 10 years, 290 such cases were recorded at our institution. Given this nontrivial impact, it is unclear if C4d immunofluorescence testing should be performed on surveillance transbronchial biopsies when the number of pieces in the specimen is marginal.


Asunto(s)
Complemento C4/inmunología , Vía Clásica del Complemento/inmunología , Rechazo de Injerto/inmunología , Trasplante de Pulmón/estadística & datos numéricos , Pulmón/patología , Anticuerpos/inmunología , Biopsia/métodos , Técnica del Anticuerpo Fluorescente/métodos , Rechazo de Injerto/diagnóstico , Rechazo de Injerto/patología , Humanos , Trasplante de Pulmón/efectos adversos , Sensibilidad y Especificidad
8.
PLoS Pathog ; 13(3): e1006248, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28301559

RESUMEN

The pandemic influenza A(H1N1) 2009 virus caused significant morbidity and mortality worldwide thus necessitating the need to understand the host factors that influence its control. Previously, the complement system has been shown to provide protection during the seasonal influenza virus infection, however, the role of individual complement pathways is not yet clear. Here, we have dissected the role of intact complement as well as of its individual activation pathways during the pandemic influenza virus infection using mouse strains deficient in various complement components. We show that the virus infection in C3-/- mice results in increased viral load and 100% mortality, which can be reversed by adoptive transfer of naïve wild-type (WT) splenocytes, purified splenic B cells, or passive transfer of immune sera from WT, but not C3-/- mice. Blocking of C3a and/or C5a receptor signaling in WT mice using receptor antagonists and use of C3aR-/- and C5aR-/- mice showed significant mortality after blocking/ablation of C3aR, with little or no effect after blocking/ablation of C5aR. Intriguingly, deficiency of C4 and FB in mice resulted in only partial mortality (24%-32%) suggesting a necessary cross-talk between the classical/lectin and alternative pathways for providing effective protection. In vitro virus neutralization experiments performed to probe the cross-talk between the various pathways indicated that activation of the classical and alternative pathways in concert, owing to coating of viral surface by antibodies, is needed for its efficient neutralization. Examination of the virus-specific complement-binding antibodies in virus positive subjects showed that their levels vary among individuals. Together these results indicate that cooperation between the classical and alternative pathways not only result in efficient direct neutralization of the pandemic influenza virus, but also lead to the optimum generation of C3a, which when sensed by the immune cells along with the antigen culminates in generation of effective protective immune responses.


Asunto(s)
Vía Alternativa del Complemento/inmunología , Vía Clásica del Complemento/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Traslado Adoptivo , Animales , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Gripe Humana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Pruebas de Neutralización
9.
Haematologica ; 104(2): 403-416, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30262558

RESUMEN

High titers of HLA antibodies are associated with platelet refractoriness, causing poor platelet increments after transfusions in a subset of patients with HLA antibodies. Currently, we do not know the biological mechanisms that explain the variability in clinical responses in HLA alloimmunized patients receiving platelet transfusions. Previously we showed that a subset of anti-HLA IgG-antibodies induces FcγRIIa-dependent platelet activation and enhanced phagocytosis. Here, we investigated whether anti-HLA IgG can induce complement activation on platelets. We found that a subset of anti-HLA IgG induced complement activation via the classical pathway, causing C4b and C3b deposition and formation of the membrane-attack complex. This resulted in permeabilization of platelet membranes and increased calcium influx. Complement activation also caused enhanced α-granule release, as measured by CD62P surface exposure. Blocking studies revealed that platelet activation was caused by FcγRIIa-dependent signaling as well as HLA antibody induced complement activation. Synergistic complement activation employing combinations of monoclonal IgGs suggested that assembly of oligomeric IgG complexes strongly promoted complement activation through binding of IgGs to different antigenic determinants on HLA. In agreement with this, we observed that preventing anti-HLA-IgG hexamer formation using an IgG-Fc:Fc blocking peptide, completely inhibited C3b and C4b deposition. Our results show that HLA antibodies can induce complement activation on platelets including membrane attack complex formation, pore formation and calcium influx. We propose that these events can contribute to fast platelet clearance in vivo in patients refractory to platelet transfusions with HLA alloantibodies, who may benefit from functional-platelet matching and treatment with complement inhibitors.


Asunto(s)
Plaquetas/inmunología , Vía Clásica del Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Antígenos HLA/inmunología , Isoanticuerpos/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Plaquetas/metabolismo , Calcio/metabolismo , Vía Clásica del Complemento/efectos de los fármacos , Proteínas del Sistema Complemento/metabolismo , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/farmacología , Inmunoglobulinas Intravenosas/farmacología , Isoanticuerpos/farmacología , Modelos Biológicos , Activación Plaquetaria/efectos de los fármacos , Unión Proteica , Receptores de IgG/metabolismo
10.
Am J Nephrol ; 49(1): 81-92, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30612132

RESUMEN

Complement activation occurs in many glomerular diseases, the exact pathway(s) of activation has been studied in detail in some diseases but not in all. C4d is generated by the activation of classical and lectin pathways, and its presence can point to the activation of either of these pathways. This review aims to summarize the available data with regard to the deposition of glomerular C4d in native kidney biopsies in different glomerular pathologies that may be useful for future research into the role of complement activation in glomerular diseases. While there is more information on C4d in certain diseases (e.g., Immunoglobulin A (IgA) nephropathy), there is scant data in other diseases (such as focal segmental glomerulosclerosis).


Asunto(s)
Complemento C4b/inmunología , Vía Clásica del Complemento/inmunología , Lectina de Unión a Manosa de la Vía del Complemento/inmunología , Glomerulonefritis por IGA/inmunología , Glomeruloesclerosis Focal y Segmentaria/inmunología , Fragmentos de Péptidos/inmunología , Animales , Modelos Animales de Enfermedad , Glomerulonefritis por IGA/patología , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Glomérulos Renales/inmunología , Glomérulos Renales/patología
11.
J Virol ; 91(19)2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28724763

RESUMEN

Poxviruses display species tropism-variola virus is a human-specific virus, while vaccinia virus causes repeated outbreaks in dairy cattle. Consistent with this, variola virus complement regulator SPICE (smallpox inhibitor of complement enzymes) exhibits selectivity in inhibiting the human alternative complement pathway and vaccinia virus complement regulator VCP (vaccinia virus complement control protein) displays selectivity in inhibiting the bovine alternative complement pathway. In the present study, we examined the species specificity of VCP and SPICE for the classical pathway (CP). We observed that VCP is ∼43-fold superior to SPICE in inhibiting bovine CP. Further, functional assays revealed that increased inhibitory activity of VCP for bovine CP is solely due to its enhanced cofactor activity, with no effect on decay of bovine CP C3-convertase. To probe the structural basis of this specificity, we utilized single- and multi-amino-acid substitution mutants wherein 1 or more of the 11 variant VCP residues were substituted in the SPICE template. Examination of these mutants for their ability to inhibit bovine CP revealed that E108, E120, and E144 are primarily responsible for imparting the specificity and contribute to the enhanced cofactor activity of VCP. Binding and functional assays suggested that these residues interact with bovine factor I but not with bovine C4(H2O) (a moiety conformationally similar to C4b). Mapping of these residues onto the modeled structure of bovine C4b-VCP-bovine factor I supported the mutagenesis data. Taken together, our data help explain why the vaccine strain of vaccinia virus was able to gain a foothold in domesticated animals.IMPORTANCE Vaccinia virus was used for smallpox vaccination. The vaccine-derived virus is now circulating and causing outbreaks in dairy cattle in India and Brazil. However, the reason for this tropism is unknown. It is well recognized that the virus is susceptible to neutralization by the complement classical pathway (CP). Because the virus encodes a soluble complement regulator, VCP, we examined whether this protein displays selectivity in targeting bovine CP. Our data show that it does exhibit selectivity in inhibiting the bovine CP and that this is primarily determined by its amino acids E108, E120, and E144, which interact with bovine serine protease factor I to inactivate bovine C4b-one of the two subunits of CP C3-convertase. Of note, the variola complement regulator SPICE contains positively charged residues at these positions. Thus, these variant residues in VCP help enhance its potency against the bovine CP and thereby the fitness of the virus in cattle.


Asunto(s)
Activación de Complemento/inmunología , Vía Alternativa del Complemento/inmunología , Vía Clásica del Complemento/inmunología , Proteínas de la Matriz Viral/inmunología , Proteínas Virales/inmunología , Tropismo Viral/genética , Secuencia de Aminoácidos , Animales , Bovinos , Proteína de Unión al Complemento C4b/inmunología , Fibrinógeno/metabolismo , Humanos , Alineación de Secuencia , Especificidad de la Especie , Virus Vaccinia/inmunología , Virus Vaccinia/patogenicidad , Proteínas de la Matriz Viral/genética , Proteínas Virales/genética
12.
PLoS Pathog ; 12(1): e1005404, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26808924

RESUMEN

Pathogens that traffic in blood, lymphatics, or interstitial fluids must adopt strategies to evade innate immune defenses, notably the complement system. Through recruitment of host regulators of complement to their surface, many pathogens are able to escape complement-mediated attack. The Lyme disease spirochete, Borrelia burgdorferi, produces a number of surface proteins that bind to factor H related molecules, which function as the dominant negative regulator of the alternative pathway of complement. Relatively less is known about how B. burgdorferi evades the classical pathway of complement despite the observation that some sensu lato strains are sensitive to classical pathway activation. Here we report that the borrelial lipoprotein BBK32 potently and specifically inhibits the classical pathway by binding with high affinity to the initiating C1 complex of complement. In addition, B. burgdorferi cells that produce BBK32 on their surface bind to both C1 and C1r and a serum sensitive derivative of B. burgdorferi is protected from killing via the classical pathway in a BBK32-dependent manner. Subsequent biochemical and biophysical approaches localized the anti-complement activity of BBK32 to its globular C-terminal domain. Mechanistic studies reveal that BBK32 acts by entrapping C1 in its zymogen form by binding and inhibiting the C1 subcomponent, C1r, which serves as the initiating serine protease of the classical pathway. To our knowledge this is the first report of a spirochetal protein acting as a direct inhibitor of the classical pathway and is the only example of a biomolecule capable of specifically and noncovalently inhibiting C1/C1r. By identifying a unique mode of complement evasion this study greatly enhances our understanding of how pathogens subvert and potentially manipulate host innate immune systems.


Asunto(s)
Proteínas Bacterianas/inmunología , Borrelia burgdorferi/inmunología , Activación de Complemento/inmunología , Vía Clásica del Complemento/inmunología , Interacciones Huésped-Parásitos/inmunología , Evasión Inmune/inmunología , Complemento C1/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Immunoblotting , Inmunoprecipitación , Enfermedad de Lyme/inmunología
13.
Mol Psychiatry ; 22(11): 1554-1561, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28761078

RESUMEN

The pathogenesis of schizophrenia is considered to be multi-factorial, with likely gene-environment interactions (GEI). Genetic and environmental risk factors are being identified with increasing frequency, yet their very number vastly increases the scope of possible GEI, making it difficult to identify them with certainty. Accumulating evidence suggests a dysregulated complement pathway among the pathogenic processes of schizophrenia. The complement pathway mediates innate and acquired immunity, and its activation drives the removal of damaged cells, autoantigens and environmentally derived antigens. Abnormalities in complement functions occur in many infectious and autoimmune disorders that have been linked to schizophrenia. Many older reports indicate altered serum complement activity in schizophrenia, though the data are inconclusive. Compellingly, recent genome-wide association studies suggest repeat polymorphisms incorporating the complement 4A (C4A) and 4B (C4B) genes as risk factors for schizophrenia. The C4A/C4B genetic associations have re-ignited interest not only in inflammation-related models for schizophrenia pathogenesis, but also in neurodevelopmental theories, because rodent models indicate a role for complement proteins in synaptic pruning and neurodevelopment. Thus, the complement system could be used as one of the 'staging posts' for a variety of focused studies of schizophrenia pathogenesis. They include GEI studies of the C4A/C4B repeat polymorphisms in relation to inflammation-related or infectious processes, animal model studies and tests of hypotheses linked to autoimmune diseases that can co-segregate with schizophrenia. If they can be replicated, such studies would vastly improve our understanding of pathogenic processes in schizophrenia through GEI analyses and open new avenues for therapy.


Asunto(s)
Activación de Complemento/inmunología , Esquizofrenia/etiología , Esquizofrenia/inmunología , Animales , Encéfalo/inmunología , Activación de Complemento/genética , Complemento C4a/genética , Complemento C4a/metabolismo , Complemento C4b/genética , Complemento C4b/metabolismo , Vía Clásica del Complemento/inmunología , Vía Clásica del Complemento/fisiología , Interacción Gen-Ambiente , Estudio de Asociación del Genoma Completo/métodos , Humanos , Herencia Multifactorial , Polimorfismo Genético/genética , Esquizofrenia/genética
14.
Transpl Int ; 31(7): 761-772, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29537702

RESUMEN

Complement-mediated allograft injury, elicited by donor-specific HLA antibodies (DSA), is a defining pathophysiological characteristic of allograft damage. We aimed to study DSA-induced complement activation as a diagnostic marker of antibody-mediated rejection (AMR) and a risk stratification tool for graft loss in the context of lung transplantation (LT). We identified 38 DSA-positive patients whose serum samples were submitted for C3d deposition testing via the C3d assay. Among these 38 patients, 15 had AMR (DSAPos AMRPos ). Results were reported for each patient as the C3d ratio for each DSA, the immunodominant DSA, and the C3d ratio for all DSA present in a sample (C3d ratioSUM ). DSAPos AMRPos patients had higher C3d ratioSUM values (58.66 (-1.32 to 118.6) vs. 1.52 (0.30 to 2.74), P = 0.0016) and increased immunodominant C3d ratios (41.87 (1.72 to 82.02) vs. 0.69 (0.21 to 1.19), P = 0.001) when compared with DSAPos AMRNeg patients. Specificity and calculated positive predictive value of the immunodominant C3d ratio and BCMsum tests for AMR diagnosis were both 100% (CI = 17.4-100) in this cohort. Worst graft survival was associated with both immunodominant C3d ratio ≥4 or C3d ratioSUM ≥10 or BCMsum >7000, suggesting that the antibody composition and/or strength are the principal determinants of an HLA DSA's capacity to activate complement.


Asunto(s)
Complemento C3d/análisis , Vía Clásica del Complemento/inmunología , Rechazo de Injerto/inmunología , Antígenos HLA/inmunología , Trasplante de Pulmón , Adulto , Estudios de Cohortes , Femenino , Supervivencia de Injerto , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
15.
J Immunol ; 196(1): 385-94, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26608922

RESUMEN

The group B Streptococcus (GBS) is a leading cause of neonatal invasive disease. GBS bacteria are surrounded by a thick capsular polysaccharide that is a potent inhibitor of complement deposition via the alternative pathway. Several of its surface molecules can however activate the classical and lectin complement pathways, rendering this species still vulnerable to phagocytic killing. In this study we have identified a novel secreted protein named complement interfering protein (CIP) that downregulates complement activation via the classical and lectin pathways, but not the alternative pathway. The CIP protein showed high affinity toward C4b and inhibited its interaction with C2, presumably preventing the formation of the C4bC2a convertase. Addition of recombinant CIP to GBS cip-negative bacteria resulted in decreased deposition of C3b on their surface and in diminished phagocytic killing in a whole-blood assay. Our data reveal a novel strategy exploited by GBS to counteract innate immunity and could be valuable for the development of anti-infective agents against this important pathogen.


Asunto(s)
Proteínas Bacterianas/inmunología , Complemento C4b/inmunología , Vía Clásica del Complemento/inmunología , Lectina de Unión a Manosa de la Vía del Complemento/inmunología , Evasión Inmune/inmunología , Streptococcus agalactiae/inmunología , Adulto , Secuencia de Aminoácidos , Activación de Complemento/inmunología , Complemento C3b/biosíntesis , Complemento C3b/inmunología , Vía Alternativa del Complemento/inmunología , Vía Clásica del Complemento/efectos de los fármacos , Lectina de Unión a Manosa de la Vía del Complemento/efectos de los fármacos , Humanos , Inmunidad Innata , Datos de Secuencia Molecular , Fagocitosis/inmunología , Unión Proteica/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Streptococcus agalactiae/genética , Streptococcus agalactiae/metabolismo
16.
Clin Immunol ; 183: 1-7, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28647502

RESUMEN

Eculizumab is a monoclonal anti-C5 antibody used in the treatment of atypical hemolytic uremic syndrome (aHUS). We monitored complement inhibition in 16 eculizumab-treated patients suffering from HUS or transplant rejection (not aHUS patients). Blood samples were obtained one to four weeks after the last eculizumab injection. We observed that eculizumab efficiently blocked the terminal pathway (TP) through classical pathway (CP) activation measured by kinetic hemolytic assay (HA) (<10%) but incompletely blocked the TP through alternative pathway (AP) activation measured by rabbit (APH50>23%) or chicken erythrocytes HA (AP100>15%). Conversely, functional ELISA revealed a complete blockade of TP through AP activation in all patients (<10%). C5a and sC5b9 levels were not correlated with residual APH50 or AP100. Similar results were obtained after in vitro addition of increasing amounts of eculizumab to a control serum (in vitro APH50>60% and AP100>20%). We also showed that ELISA was less sensitive than HA.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Inactivadores del Complemento/uso terapéutico , Vía Alternativa del Complemento/inmunología , Vía Clásica del Complemento/inmunología , Rechazo de Injerto/tratamiento farmacológico , Adolescente , Adulto , Anciano , Síndrome Hemolítico Urémico Atípico/inmunología , Niño , Preescolar , Activación de Complemento/inmunología , Complemento C5a/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Lactante , Trasplante de Riñón , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Adulto Joven
17.
J Immunol ; 194(11): 5488-96, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25911760

RESUMEN

Complement component C4 is a central protein in the classical and lectin pathways within the complement system. During activation of complement, its major fragment C4b becomes covalently attached to the surface of pathogens and altered self-tissue, where it acts as an opsonin marking the surface for removal. Moreover, C4b provides a platform for assembly of the proteolytically active convertases that mediate downstream complement activation by cleavage of C3 and C5. In this article, we present the crystal and solution structures of the 195-kDa C4b. Our results provide the molecular details of the rearrangement accompanying C4 cleavage and suggest intramolecular flexibility of C4b. The conformations of C4b and its paralogue C3b are shown to be remarkably conserved, suggesting that the convertases from the classical and alternative pathways are likely to share their overall architecture and mode of substrate recognition. We propose an overall molecular model for the classical pathway C5 convertase in complex with C5, suggesting that C3b increases the affinity for the substrate by inducing conformational changes in C4b rather than a direct interaction with C5. C4b-specific features revealed by our structural studies are probably involved in the assembly of the classical pathway C3/C5 convertases and C4b binding to regulators.


Asunto(s)
Activación de Complemento/inmunología , Complemento C4b/química , Vía Clásica del Complemento/inmunología , Lectina de Unión a Manosa de la Vía del Complemento/inmunología , Convertasas de Complemento C3-C5/metabolismo , Complemento C3b/genética , Complemento C3b/inmunología , Complemento C4b/inmunología , Complemento C5/genética , Complemento C5/inmunología , Cristalografía por Rayos X , Humanos , Proteínas Opsoninas/inmunología , Unión Proteica/inmunología , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína
18.
J Immunol ; 195(5): 2231-40, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26209620

RESUMEN

Recent reports focusing on virulence factors of periodontal pathogens implicated proteinases as major determinants of remarkable pathogenicity of these species, with special emphasis on their capacity to modulate complement activity. In particular, bacteria-mediated cleavage of C5 and subsequent release of C5a seems to be an important phenomenon in the manipulation of the local inflammatory response in periodontitis. In this study, we present mirolysin, a novel metalloproteinase secreted by Tannerella forsythia, a well-recognized pathogen strongly associated with periodontitis. Mirolysin exhibited a strong effect on all complement pathways. It inhibited the classical and lectin complement pathways due to efficient degradation of mannose-binding lectin, ficolin-2, ficolin-3, and C4, whereas inhibition of the alternative pathway was caused by degradation of C5. This specificity toward complement largely resembled the activity of a previously characterized metalloproteinase of T. forsythia, karilysin. Interestingly, mirolysin released the biologically active C5a peptide in human plasma and induced migration of neutrophils. Importantly, we demonstrated that combination of mirolysin with karilysin, as well as a cysteine proteinase of another periodontal pathogen, Prevotella intermedia, resulted in a strong synergistic effect on complement. Furthermore, mutant strains of T. forsythia, devoid of either mirolysin or karilysin, showed diminished survival in human serum, providing further evidence for the synergistic inactivation of complement by these metalloproteinases. Taken together, our findings on interactions of mirolysin with complement significantly add to the understanding of immune evasion strategies of T. forsythia and expand the knowledge on molecular mechanisms driving pathogenic events in the infected periodontium.


Asunto(s)
Proteínas Bacterianas/inmunología , Infecciones por Bacteroides/inmunología , Bacteroides/inmunología , Activación de Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Metaloproteasas/inmunología , Periodontitis/inmunología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bacteroides/genética , Bacteroides/fisiología , Infecciones por Bacteroides/sangre , Infecciones por Bacteroides/microbiología , Movimiento Celular/inmunología , Vía Alternativa del Complemento/inmunología , Vía Clásica del Complemento/inmunología , Lectina de Unión a Manosa de la Vía del Complemento/inmunología , Hemólisis/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/inmunología , Metaloproteinasas de la Matriz/metabolismo , Metaloproteasas/genética , Metaloproteasas/metabolismo , Viabilidad Microbiana/genética , Viabilidad Microbiana/inmunología , Mutación , Neutrófilos/inmunología , Neutrófilos/metabolismo , Periodontitis/sangre , Periodontitis/microbiología , Ovinos
19.
Lupus ; 25(8): 873-7, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27252264

RESUMEN

C1q is the first component of the classical complement pathway. Both clinically validated in-house ELISA assays as well as commercial ELISA kits are used for detection of anti-C1q antibodies. Anti-C1q autoantibodies can be detected in a wide range of autoimmune diseases and are highly sensitive for hypocomplementemic uticarial vasculitis. In SLE, anti-C1q are strongly associated with proliferative lupus nephritis, and their absence carries a negative predictive value for development of lupus nephritis of close to 100%. Anti-C1q in combination with anti-dsDNA and low complement has the strongest serological association with renal involvement. The anti-C1q titers correlate with global disease activity scores in patients with renal involvement, and higher titers seem to precede renal flares. After the successful treatment of a renal flare, anti-C1q has the tendency to decrease or even become undetectable. The main obstacle to the inclusion of anti-C1q in the classification criteria and clinical management of SLE is the lack of standardized laboratory assays.


Asunto(s)
Anticuerpos Antinucleares/sangre , Complemento C1q/inmunología , Vía Clásica del Complemento/inmunología , Lupus Eritematoso Sistémico/complicaciones , Nefritis Lúpica/inmunología , Biomarcadores/sangre , Humanos , Índice de Severidad de la Enfermedad
20.
J Immunol ; 193(4): 1855-63, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25015832

RESUMEN

Almost all invasive Neisseria meningitidis isolates express capsular polysaccharide. Ab is required for complement-dependent killing of meningococci. Although alternative pathway evasion has received considerable attention, little is known about classical pathway (CP) inhibition by meningococci, which forms the basis of this study. We engineered capsulated and unencapsulated isogenic mutant strains of groups A, B, C, W, and Y meningococci to express similar amounts of the same factor H-binding protein (fHbp; a key component of group B meningococcal vaccines) molecule. Despite similar anti-fHbp mAb binding, significantly less C4b was deposited on all five encapsulated mutants compared with their unencapsulated counterparts (p < 0.01) when purified C1 and C4 were used to deposit C4b. Reduced C4b deposition was the result of capsule-mediated inhibition of C1q engagement by Ab. C4b deposition correlated linearly with C1q engagement by anti-fHbp. Whereas B, C, W, and Y capsules limited CP-mediated killing by anti-fHbp, the unencapsulated group A mutant paradoxically was more resistant than its encapsulated counterpart. Strains varied considerably in their susceptibility to anti-fHbp and complement despite similar Ab binding, which may have implications for the activity of fHbp-based vaccines. Capsule also limited C4b deposition by anti-porin A mAbs. Capsule expression decreased binding of an anti-lipooligosaccharide IgM mAb (∼ 1.2- to 2-fold reduction in fluorescence). Akin to observations with IgG, capsule also decreased IgM-mediated C4b deposition when IgM binding to the mutant strain pairs was normalized. In conclusion, we show that capsular polysaccharide, a critical meningococcal virulence factor, inhibits the CP of complement.


Asunto(s)
Cápsulas Bacterianas/inmunología , Complemento C1q/inmunología , Complemento C4b/inmunología , Vía Clásica del Complemento/inmunología , Evasión Inmune , Neisseria meningitidis/inmunología , Anticuerpos Monoclonales/inmunología , Antígenos Bacterianos/biosíntesis , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/inmunología , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Lipopolisacáridos/inmunología , Neisseria meningitidis/clasificación , Porinas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA