Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Exp Neurol ; 348: 113947, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34902359

RESUMEN

Multiple sclerosis (MS) is a chronic central nervous system demyelinating disease of autoimmune originate. Complement C1q, a complex glycoprotein, mediates a variety of immunoregulatory functions considered important in the prevention of autoimmunity. Although we found that the increased serum C1q level was highly associated with the Fazekas scores and T2 lesion volume of MS patients, the effect and mechanism of C1q on demyelination remains unclear. Cluster analysis and protein array results showed that serum Wnt receptors Frizzled-6 and LRP-6 levels in MS patients were both increased, we proposed that C1q may be involved in demyelination via Wnt signaling. The increased C1q protein levels in the serum and brain tissue were confirmed in a cuprizone (CPZ)-induced demyelination mice model. Moreover, CPZ treatment induced significant increase of LRP-6 and Frizzled-6 protein in mice corpus callosum. LRP-6 extra-cellular domain (LRP-6-ECD) level in the serum and cerebrospinal fluid (CSF) of CPZ mice also significantly increased. Knockdown of the subunit C1s of C1 not only substantially attenuated demyelination, promoted M2 microglia polarization and improved neurological function, but inhibited ß-catenin expression and its nuclear translocation in oligodendrocyte progenitor cells (OPCs). In vitro, C1s silence reversed the increased level of LRP-6-ECD in the medium and ß-catenin expression in OPCs induced by C1q treatment. Meanwhile, inhibition of C1s also markedly lowered the number of EDU positive OPCs, but enhanced the number of CNPase positive oligodendrocyte and the protein of MBP. The present study indicated that C1q was involved in demyelination in response to CPZ in mice by preventing OPC from differentiating into mature oligodendrocyte via Wnt/ß-catenin signaling activation.


Asunto(s)
Diferenciación Celular/fisiología , Complemento C1q/metabolismo , Cuprizona/toxicidad , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Vía de Señalización Wnt/fisiología , Adulto , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Quelantes/toxicidad , Complemento C1q/antagonistas & inhibidores , Complemento C1q/genética , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen/métodos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/genética , Esclerosis Múltiple Recurrente-Remitente/patología , Ratas , Ratas Sprague-Dawley , Vía de Señalización Wnt/efectos de los fármacos
2.
Nature ; 597(7878): 709-714, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34497421

RESUMEN

Multiple sclerosis (MS) lesions that do not resolve in the months after they form harbour ongoing demyelination and axon degeneration, and are identifiable in vivo by their paramagnetic rims on MRI scans1-3. Here, to define mechanisms underlying this disabling, progressive neurodegenerative state4-6 and foster development of new therapeutic agents, we used MRI-informed single-nucleus RNA sequencing to profile the edge of demyelinated white matter lesions at various stages of inflammation. We uncovered notable glial and immune cell diversity, especially at the chronically inflamed lesion edge. We define 'microglia inflamed in MS' (MIMS) and 'astrocytes inflamed in MS', glial phenotypes that demonstrate neurodegenerative programming. The MIMS transcriptional profile overlaps with that of microglia in other neurodegenerative diseases, suggesting that primary and secondary neurodegeneration share common mechanisms and could benefit from similar therapeutic approaches. We identify complement component 1q (C1q) as a critical mediator of MIMS activation, validated immunohistochemically in MS tissue, genetically by microglia-specific C1q ablation in mice with experimental autoimmune encephalomyelitis, and therapeutically by treating chronic experimental autoimmune encephalomyelitis with C1q blockade. C1q inhibition is a potential therapeutic avenue to address chronic white matter inflammation, which could be monitored by longitudinal assessment of its dynamic biomarker, paramagnetic rim lesions, using advanced MRI methods.


Asunto(s)
Astrocitos/patología , Linfocitos/patología , Microglía/patología , Esclerosis Múltiple/patología , Animales , Encéfalo/patología , Complemento C1q/antagonistas & inhibidores , Complemento C1q/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Inflamación/patología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Esclerosis Múltiple/diagnóstico por imagen , RNA-Seq , Transcriptoma , Sustancia Blanca/patología
3.
Clin Exp Immunol ; 206(3): 378-383, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34586637

RESUMEN

Acquired angioedema due to C1-inhibitor (C1-INH) deficiency (AAE-C1-INH) is rare and is associated with underlying lymphoproliferative diseases. C1-INH deficiency may be due to neoplastic over-consumption of C1-INH and the generation of anti-C1-INH autoantibodies. Uncovering an occult malignancy can lead to earlier oncology referral and improvement of angioedema after treatment of the underlying lymphoproliferative disorder. We characterized seven patients with C1-INH-AAE that highlights the importance of recognizing the association between C1-INH-AAE and underlying malignancy. In acute attacks, patients may be resistant to C1-INH therapy due to the presence of anti-C1-INH autoantibodies or rapid complement consumption, and may respond better to icatibant or ecallantide, which directly affect bradykinin. Treatment of the underlying malignancy also improves AAE-C1-INH symptoms and supports the role of lymphoproliferative B cells in AAE-C1-INH pathophysiology. Monitoring levels of C4, C1-INH function and level, and C1q may be predictive of AAE-C1-INH control and be used as surrogates for treatment efficacy. With close monitoring, low-dose danazol can be effective for long-term prophylaxis. Annual evaluation in AAE-C1-INH is recommended if an underlying malignancy is not found, as angioedema may precede the development of malignancy by several years. Our single-center study has aided in standardization of comprehensive AAE-C1-INH diagnosis, treatment, and monitoring strategies towards future therapeutic clinical trials.


Asunto(s)
Angioedema/patología , Proteína Inhibidora del Complemento C1/genética , Angioedema Hereditario Tipos I y II/genética , Trastornos Linfoproliferativos/patología , Anciano , Angioedema/genética , Antiinflamatorios no Esteroideos/uso terapéutico , Autoanticuerpos/inmunología , Bradiquinina/análogos & derivados , Bradiquinina/uso terapéutico , Antagonistas del Receptor de Bradiquinina B2/uso terapéutico , Proteína Inhibidora del Complemento C1/inmunología , Complemento C1q/antagonistas & inhibidores , Complemento C1q/metabolismo , Femenino , Humanos , Trastornos Linfoproliferativos/genética , Masculino , Persona de Mediana Edad , Péptidos/uso terapéutico , Estudios Retrospectivos
4.
PLoS One ; 16(9): e0257960, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34591905

RESUMEN

Perinatal hypoxic ischemic encephalopathy (HIE) remains a major contributor of infant death and long-term disability worldwide. The role played by the complement system in this ischemia-reperfusion injury remains poorly understood. In order to better understand the role of complement activation and other modifiable mechanisms of injury in HIE, we tested the dual-targeting anti-inflammatory peptide, RLS-0071 in an animal model of HIE. Using the well-established HIE rat pup model we measured the effects of RLS-0071 during the acute stages of the brain injury and on long-term neurocognitive outcomes. Rat pups subject to hypoxia-ischemia insult received one of 4 interventions including normothermia, hypothermia and RLS-0071 with and without hypothermia. We measured histopathological effects, brain C1q levels and neuroimaging at day 1 and 21 after the injury. A subset of animals was followed into adolescence and evaluated for neurocognitive function. On histological evaluation, RLS-0071 showed neuronal protection in combination with hypothermia (P = 0.048) in addition to reducing C1q levels in the brain at 1hr (P = 0.01) and at 8 hr in combination with hypothermia (P = 0.005). MRI neuroimaging demonstrated that RLS-0071 in combination with hypothermia reduced lesion volume at 24 hours (P<0.05) as well as decreased T2 signal at day 21 in combination with hypothermia (P<0.01). RLS-0071 alone or in combination with hypothermia improved both short-term and long-term memory. These findings suggest that modulation by RLS-0071 can potentially decrease brain damage resulting from HIE.


Asunto(s)
Encéfalo/efectos de los fármacos , Complemento C1q/antagonistas & inhibidores , Inactivadores del Complemento/farmacología , Vía Clásica del Complemento/efectos de los fármacos , Hipoxia-Isquemia Encefálica/terapia , Animales , Encéfalo/metabolismo , Encéfalo/patología , Terapia Combinada , Complemento C1q/metabolismo , Inactivadores del Complemento/uso terapéutico , Modelos Animales de Enfermedad , Hipotermia Inducida , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/patología , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Destreza Motora/efectos de los fármacos , Destreza Motora/fisiología , Ratas , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Resultado del Tratamiento
5.
Biomed Pharmacother ; 136: 111234, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33454596

RESUMEN

Complement is an important innate immune defence machinery. Once dysregulated, it is often linked to pathogenesis of diverse autoimmune diseases. Artesunate (ART) is a well-known anti-malarial compound. Recently, ART has been highlighted by its potential therapeutic effects on certain complement-related autoimmune diseases. However, the underlying mechanisms are hitherto unknown. In the present study, we found that ART mediated complement interception as validated by analysis of complement haemolytic assay. In cell-based setup using dying Jurkat cells, ART-mediated complement interception was also confirmed. Further, we newly established an ELISA system selectively allowing complement activation via the classical pathway, the lectin pathway and the alternative pathway, respectively. ELISA analysis revealed that ART dose-dependently inhibited C4 activation, C3 activation and terminal complement complex assembly via the effector pathways. ART was found to blockade C1q, C3 and C5 with a lesser extent to properdin. The interaction of ART with C1q was determined to be mediated via C1q globular head region. FACS analysis using ART-conjugated mesoporous silica particles revealed that ART specifically bound the key therapeutic targets of C1q, C3 and C5 on microparticles. In conclusion, we for the first time report the anti-complement bioactivities of ART and suggest a potential therapeutic benefit of ART in the complement-related human diseases.


Asunto(s)
Artesunato/farmacología , Activación de Complemento/efectos de los fármacos , Inactivadores del Complemento/farmacología , Animales , Complemento C1q/antagonistas & inhibidores , Complemento C1q/metabolismo , Complemento C3/antagonistas & inhibidores , Complemento C3/metabolismo , Complemento C4/antagonistas & inhibidores , Complemento C4/metabolismo , Complemento C5/antagonistas & inhibidores , Complemento C5/metabolismo , Hemólisis/efectos de los fármacos , Humanos , Células Jurkat , Necrosis , Oveja Doméstica
6.
Nature ; 588(7838): 459-465, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32866962

RESUMEN

Aberrant aggregation of the RNA-binding protein TDP-43 in neurons is a hallmark of frontotemporal lobar degeneration caused by haploinsufficiency in the gene encoding progranulin1,2. However, the mechanism leading to TDP-43 proteinopathy remains unclear. Here we use single-nucleus RNA sequencing to show that progranulin deficiency promotes microglial transition from a homeostatic to a disease-specific state that causes endolysosomal dysfunction and neurodegeneration in mice. These defects persist even when Grn-/- microglia are cultured ex vivo. In addition, single-nucleus RNA sequencing reveals selective loss of excitatory neurons at disease end-stage, which is characterized by prominent nuclear and cytoplasmic TDP-43 granules and nuclear pore defects. Remarkably, conditioned media from Grn-/- microglia are sufficient to promote TDP-43 granule formation, nuclear pore defects and cell death in excitatory neurons via the complement activation pathway. Consistent with these results, deletion of the genes encoding C1qa and C3 mitigates microglial toxicity and rescues TDP-43 proteinopathy and neurodegeneration. These results uncover previously unappreciated contributions of chronic microglial toxicity to TDP-43 proteinopathy during neurodegeneration.


Asunto(s)
Microglía/metabolismo , Microglía/patología , Neuronas/metabolismo , Neuronas/patología , Progranulinas/deficiencia , Proteinopatías TDP-43/metabolismo , Proteinopatías TDP-43/patología , Envejecimiento/genética , Envejecimiento/patología , Animales , Núcleo Celular/genética , Núcleo Celular/patología , Activación de Complemento/efectos de los fármacos , Activación de Complemento/inmunología , Complemento C1q/antagonistas & inhibidores , Complemento C1q/inmunología , Complemento C3b/antagonistas & inhibidores , Complemento C3b/inmunología , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/farmacología , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Poro Nuclear/metabolismo , Poro Nuclear/patología , Progranulinas/genética , RNA-Seq , Análisis de la Célula Individual , Proteinopatías TDP-43/tratamiento farmacológico , Proteinopatías TDP-43/genética , Tálamo/metabolismo , Tálamo/patología , Transcriptoma
7.
Front Immunol ; 11: 1504, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32849513

RESUMEN

The classical pathway of complement is important for protection against pathogens and in maintaining tissue homeostasis, but excessive or aberrant activation is directly linked to numerous pathologies. We describe the development and in vitro characterization of C1qNb75, a single domain antibody (nanobody) specific for C1q, the pattern recognition molecule of the classical pathway. C1qNb75 binds to the globular head modules of human C1q with sub-nanomolar affinity and impedes classical pathway mediated hemolysis by IgG and IgM. Crystal structure analysis revealed that C1qNb75 recognizes an epitope primarily located in the C1q B-chain that overlaps with the binding sites of IgG and IgM. Thus, C1qNb75 competitively prevents C1q from binding to IgG and IgM causing blockade of complement activation by the classical pathway. Overall, C1qNb75 represents a high-affinity nanobody-based inhibitor of IgG- and IgM-mediated activation of the classical pathway and may serve as a valuable reagent in mechanistic and functional studies of complement, and as an efficient inhibitor of complement under conditions of excessive CP activation.


Asunto(s)
Complemento C1q/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Anticuerpos de Dominio Único/metabolismo , Afinidad de Anticuerpos , Células Cultivadas , Activación de Complemento , Complemento C1q/antagonistas & inhibidores , Vía Clásica del Complemento , Cristalografía por Rayos X , Humanos , Inmunoglobulina G/metabolismo , Inmunoglobulina M/metabolismo , Unión Proteica , Conformación Proteica , Receptores de Reconocimiento de Patrones/genética , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/genética , Relación Estructura-Actividad
8.
Eur J Histochem ; 63(2)2019 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-31060348

RESUMEN

Cognitive impairment in Alzheimer's disease (AD) is usually accompanied by synaptic loss in both the hippocampus and neocortex. In the early stage of AD, amyloid ß-induced synapse changes is the main reason, while in the later stage, the accumulation of Tau protein promotes synapse degeneration as the key factor leading to dementia. MicroRNA (miRNA) is closely related to the expression changes of many AD-related genes. One of the most abundant brain-enriched miRNAs is miR-132, which has been shown to regulate both neuron morphogenesis and plasticity. It has been reported that miR-132 is significantly reduced in the brains of Alzheimer's patients. Genetic deletion of miR-132 in mice promotes Aß deposition, leading to impaired memory and enhanced Tau pathology, but how the miRNA-mediated gene expression dysregulation contributes to AD pathology remains unclear. Here we found the possible downstream target of miR-132 by in silico analysis, namely C1q. C1q is the primary protein of classical complement cascade, which is highly expressed in the synaptic regions of the central nervous system in Alzheimer's patients. However, it is not clear whether miR-132 plays a role in AD through regulating C1q. To address this question, the APP/PS1 transgenic mice were transfected with miR-132 and given C1 inhibitors. Behavior tests were conducted to assess memory and cognitive abilities seven days after administration. In addition, we analyzed the expression of PSD95, Synapsin-1 and phosphorylated (p)-Synapsin. We found that the expression levels of the synaptic proteins treated with miR-132 or C1INH were significantly increased compared with the AD group. Further RT-qPCR result suggested that miR-132 might regulate C1q expression in AD.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Complemento C1q/metabolismo , MicroARNs/metabolismo , Presenilina-1/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Cognición/efectos de los fármacos , Complemento C1q/antagonistas & inhibidores , Complemento C1q/genética , Homólogo 4 de la Proteína Discs Large/genética , Homólogo 4 de la Proteína Discs Large/metabolismo , Reposicionamiento de Medicamentos , Regulación de la Expresión Génica , Masculino , Memoria/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Transgénicos , Sinapsinas/genética , Sinapsinas/metabolismo
9.
Mar Drugs ; 16(12)2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30513754

RESUMEN

Antimicrobial peptides from marine invertebrates are known not only to act like cytotoxic agents, but they also can display some additional activities in mammalian organisms. In particular, these peptides can modulate the complement system as was described for tachyplesin, a peptide from the horseshoe crab. In this work, we investigated the influence on complement activation of the antimicrobial peptide arenicin-1 from the marine polychaete Arenicola marina. To study effects of arenicin on complement activation in human blood serum, we used hemolytic assays of two types, with antibody sensitized sheep erythrocytes and rabbit erythrocytes. Complement activation was also assessed, by the level of C3a production that was measured by ELISA. We found that the effect of arenicin depends on its concentration. At relatively low concentrations the peptide stimulates complement activation and lysis of target erythrocytes, whereas at higher concentrations arenicin acts as a complement inhibitor. A hypothetical mechanism of peptide action is proposed, suggesting its interaction with two complement proteins, C1q and C3. The results lead to the possibility of the development of new approaches for therapy of diseases connected with complement dysregulation, using peptide regulators derived from natural antimicrobial peptides of invertebrates.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Activación de Complemento/efectos de los fármacos , Proteínas del Helminto/farmacología , Poliquetos , Unión Proteica/efectos de los fármacos , Animales , Péptidos Catiónicos Antimicrobianos/aislamiento & purificación , Complemento C1q/agonistas , Complemento C1q/antagonistas & inhibidores , Complemento C1q/inmunología , Complemento C1q/metabolismo , Complemento C3/agonistas , Complemento C3/antagonistas & inhibidores , Complemento C3/inmunología , Complemento C3/metabolismo , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Eritrocitos/efectos de los fármacos , Eritrocitos/inmunología , Eritrocitos/metabolismo , Proteínas del Helminto/aislamiento & purificación , Hemólisis/efectos de los fármacos , Humanos , Conejos , Ovinos
10.
Front Immunol ; 9: 958, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29867943

RESUMEN

IgG antibodies (Abs) mediate their effector functions through the interaction with Fcγ receptors (FcγRs) and the complement factors. The main IgG-mediated complement activation pathway is induced through the binding of complement C1q to IgG Abs. This interaction is dependent on antigen-dependent hexamer formation of human IgG1 and IgG3 to increase the affinity for the six-headed C1q molecule. By contrast, human IgG4 fails to bind to C1q. Instead, it has been suggested that human IgG4 can block IgG1 and IgG3 hexamerization required for their binding to C1q and activating the complement. Here, we show that murine IgG1, which functionally resembles human IgG4 by not interacting with C1q, inhibits the binding of IgG2a, IgG2b, and IgG3 to C1q in vitro, and suppresses IgG2a-mediated complement activation in a hemolytic assay in an antigen-dependent and IgG subclass-specific manner. From this perspective, we discuss the potential of murine IgG1 and human IgG4 to block the complement activation as well as suppressive effects of sialylated IgG subclass Abs on FcγR-mediated immune cell activation. Accumulating evidence suggests that both mechanisms seem to be responsible for preventing uncontrolled IgG (auto)Ab-induced inflammation in mice and humans. Distinct IgG subclass distributions and functionally opposite IgG Fc glycosylation patterns might explain different outcomes of IgG-mediated immune responses and provide new therapeutic options through the induction, enrichment, or application of antigen-specific sialylated human IgG4 to prevent complement and FcγR activation as well.


Asunto(s)
Activación de Complemento/efectos de los fármacos , Complemento C1q/antagonistas & inhibidores , Vía Clásica del Complemento , Inmunoglobulina G/farmacología , Receptores de IgG/antagonistas & inhibidores , Animales , Sitios de Unión de Anticuerpos , Complemento C1q/metabolismo , Glicosilación , Hemólisis , Humanos , Ratones
11.
J Immunol ; 199(3): 1069-1085, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28687659

RESUMEN

Inflammatory processes play a key role in pathophysiology of many neurologic diseases/trauma, but the effect of immune cells and factors on neurotransplantation strategies remains unclear. We hypothesized that cellular and humoral components of innate immunity alter fate and migration of human neural stem cells (hNSC). In these experiments, conditioned media collected from polymorphonuclear leukocytes (PMN) selectively increased hNSC astrogliogenesis and promoted cell migration in vitro. PMN were shown to generate C1q and C3a; exposure of hNSC to PMN-synthesized concentrations of these complement proteins promoted astrogliogenesis and cell migration. Furthermore, in vitro, Abs directed against C1q and C3a reversed the fate and migration effects observed. In a proof-of-concept in vivo experiment, blockade of C1q and C3a transiently altered hNSC migration and reversed astroglial fate after spinal cord injury. Collectively, these data suggest that modulation of the innate/humoral inflammatory microenvironment may impact the potential of cell-based therapies for recovery and repair following CNS pathology.


Asunto(s)
Astrocitos/fisiología , Diferenciación Celular/fisiología , Complemento C1q/biosíntesis , Complemento C3a/biosíntesis , Células-Madre Neurales/fisiología , Neutrófilos/metabolismo , Animales , Astrocitos/efectos de los fármacos , Movimiento Celular , Células Cultivadas , Complemento C1q/antagonistas & inhibidores , Complemento C1q/genética , Complemento C1q/inmunología , Complemento C3a/antagonistas & inhibidores , Complemento C3a/genética , Complemento C3a/inmunología , Medios de Cultivo Condicionados , Humanos , Inmunidad Innata , Ratones , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/inmunología , Neutrófilos/inmunología , Traumatismos de la Médula Espinal/inmunología , Traumatismos de la Médula Espinal/fisiopatología
12.
J Neuroimmunol ; 298: 117-29, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27609284

RESUMEN

Using a previously described retinal explant culture system as an acute injury model, we here explore the role of C1q, the initiator of the classical complement pathway, in neuronal cell survival and retinal homeostasis. Full-thickness adult rat retinal explants were divided into four groups, receiving the following supplementation: C1q (50nM), C1-inhibitor (C1-inh; Berinert; 500mg/l), C1q+C1-inh, and no supplementation (culture controls). Explants were kept for 12h or 2days after which they were examined morphologically and with a panel of immunohistochemical markers. C1q supplementation protects ganglion cells from degeneration within the explant in vitro system. This effect is correlated to an attenuated endogenous production of C1q, and a quiesced gliotic response.


Asunto(s)
Complemento C1q/farmacología , Degeneración Retiniana/patología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Silicatos de Aluminio/farmacología , Animales , Proteínas de Unión al Calcio/metabolismo , Supervivencia Celular/efectos de los fármacos , Complemento C1q/antagonistas & inhibidores , Complemento C1q/uso terapéutico , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Glutamato-Amoníaco Ligasa/metabolismo , Etiquetado Corte-Fin in Situ , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de Microfilamentos/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Ratas , Ratas Sprague-Dawley , Degeneración Retiniana/tratamiento farmacológico , Rodopsina/metabolismo , Factores de Tiempo
13.
Mol Immunol ; 72: 9-18, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26922040

RESUMEN

BACKGROUND: Ethanol feeding in mice activates complement via C1q binding to apoptotic cells in the liver; complement contributes to ethanol-induced inflammation and injury. Despite the critical role of C1q in ethanol-induced injury, the mechanism by which ethanol activates C1q remains poorly understood. Secretory IgM (sIgM), traditionally considered to act as an anti-microbial, also has critical housekeeping functions, facilitating clearance of apoptotic cells, at least in part through activation of C1q. Therefore, we hypothesized that (1) ethanol-induced apoptosis in the liver recruits sIgM, facilitating the activation of C1q and complement and (2) C1INH (C1 esterase inhibitor), which inhibits C1 functional activity, prevents complement activation and decreases ethanol-induced liver injury. METHODS: Female C57BL/6 wild-type, C1qa(-/-), BID(-/-) and sIgM(-/-) mice were fed ethanol containing liquid diets or pair-fed control diets. C1INH or vehicle was given via tail vein injection to ethanol- or pair-fed wild-type mice at 24 and 48h prior to euthanasia. RESULTS: Ethanol exposure increased apoptosis in the liver, as well as the accumulation of IgM in the liver. In the early stages of ethanol feeding, C1q co-localized with IgM in the peri-sinusoidal space of the liver and accumulation of IgM and C3b was dependent on ethanol-induced BID-dependent apoptosis. sIgM(-/-) mice were protected from both ethanol-induced activation of complement and early ethanol-induced liver injury when compared to wild-type mice. Treatment with C1INH also decreased hepatic C3b deposition and ethanol-induced injury. CONCLUSION: These data indicate that sIgM contributes to activation of complement and ethanol-induced increases in inflammatory cytokine expression and hepatocyte injury in the early stages of ethanol-induced liver injury.


Asunto(s)
Apoptosis , Activación de Complemento , Complemento C1q/inmunología , Inmunoglobulina M/inmunología , Hepatopatías Alcohólicas/inmunología , Animales , Proteínas Inactivadoras del Complemento 1/farmacología , Proteína Inhibidora del Complemento C1 , Complemento C1q/antagonistas & inhibidores , Citocinas/biosíntesis , Femenino , Hepatocitos/inmunología , Hepatopatías Alcohólicas/patología , Ratones , Ratones Endogámicos C57BL , Solubilidad
14.
PLoS Negl Trop Dis ; 8(5): e2872, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24854034

RESUMEN

Scabies is an infectious skin disease caused by the mite Sarcoptes scabiei and has been classified as one of the six most prevalent epidermal parasitic skin diseases infecting populations living in poverty by the World Health Organisation. The role of the complement system, a pivotal component of human innate immunity, as an important defence against invading pathogens has been well documented and many parasites have an arsenal of anti-complement defences. We previously reported on a family of scabies mite proteolytically inactive serine protease paralogues (SMIPP-Ss) thought to be implicated in host defence evasion. We have since shown that two family members, SMIPP-S D1 and I1 have the ability to bind the human complement components C1q, mannose binding lectin (MBL) and properdin and are capable of inhibiting all three human complement pathways. This investigation focused on inhibition of the lectin pathway of complement activation as it is likely to be the primary pathway affecting scabies mites. Activation of the lectin pathway relies on the activation of MBL, and as SMIPP-S D1 and I1 have previously been shown to bind MBL, the nature of this interaction was examined using binding and mutagenesis studies. SMIPP-S D1 bound MBL in complex with MBL-associated serine proteases (MASPs) and released the MASP-2 enzyme from the complex. SMIPP-S I1 was also able to bind MBL in complex with MASPs, but MASP-1 and MASP-2 remained in the complex. Despite these differences in mechanism, both molecules inhibited activation of complement components downstream of MBL. Mutagenesis studies revealed that both SMIPP-Ss used an alternative site of the molecule from the residual active site region to inhibit the lectin pathway. We propose that SMIPP-Ss are potent lectin pathway inhibitors and that this mechanism represents an important tool in the immune evasion repertoire of the parasitic mite and a potential target for therapeutics.


Asunto(s)
Lectina de Unión a Manosa de la Vía del Complemento/efectos de los fármacos , Sarcoptes scabiei/enzimología , Serina Proteasas/metabolismo , Serina Proteasas/farmacología , Secuencia de Aminoácidos , Animales , Complemento C1q/antagonistas & inhibidores , Complemento C1q/metabolismo , Interacciones Huésped-Patógeno , Humanos , Lectina de Unión a Manosa/antagonistas & inhibidores , Lectina de Unión a Manosa/metabolismo , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Escabiosis/metabolismo , Alineación de Secuencia , Serina Proteasas/química , Serina Proteasas/genética
15.
Acta Neuropathol ; 125(6): 829-40, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23677375

RESUMEN

Neuromyelitis optica (NMO) is an autoimmune disorder with inflammatory demyelinating lesions in the central nervous system, particularly in the spinal cord and optic nerve. NMO pathogenesis is thought to involve binding of anti-aquaporin-4 (AQP4) autoantibodies to astrocytes, which causes complement-dependent cytotoxicity (CDC) and downstream inflammation leading to oligodendrocyte and neuronal injury. Vasculocentric deposition of activated complement is a prominent feature of NMO pathology. Here, we show that a neutralizing monoclonal antibody against the C1q protein in the classical complement pathway prevents AQP4 autoantibody-dependent CDC in cell cultures and NMO lesions in ex vivo spinal cord slice cultures and in mice. A monoclonal antibody against human C1q with 11 nM binding affinity prevented CDC caused by NMO patient serum in AQP4-transfected cells and primary astrocyte cultures, and prevented complement-dependent cell-mediated cytotoxicity (CDCC) produced by natural killer cells. The anti-C1q antibody prevented astrocyte damage and demyelination in mouse spinal cord slice cultures exposed to AQP4 autoantibody and human complement. In a mouse model of NMO produced by intracerebral injection of AQP4 autoantibody and human complement, the inflammatory demyelinating lesions were greatly reduced by intracerebral administration of the anti-C1q antibody. These results provide proof-of-concept for C1q-targeted monoclonal antibody therapy in NMO. Targeting of C1q inhibits the classical complement pathway directly and causes secondary inhibition of CDCC and the alternative complement pathway. As C1q-targeted therapy leaves the lectin complement activation pathway largely intact, its side-effect profile is predicted to differ from that of therapies targeting downstream complement proteins.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Complemento C1q/antagonistas & inhibidores , Factores Inmunológicos/uso terapéutico , Neuromielitis Óptica/patología , Neuromielitis Óptica/prevención & control , Animales , Acuaporina 4/fisiología , Técnicas de Cultivo de Célula , Activación de Complemento , Cricetulus , Modelos Animales de Enfermedad , Humanos , Ratones , Neuromielitis Óptica/etiología
16.
Mol Biol (Mosk) ; 47(4): 681-8, 2013.
Artículo en Ruso | MEDLINE | ID: mdl-24466758

RESUMEN

Current computational methods have not been able to discover an unknown binding site for low-molecular ligands on a protein receptor and predict parameters of their interaction when this binding site is not distinguished by energy of binding or structural features. Authors propose a method to find an unknown, structurally undefined site for binding low-molecular inhibitors with a protein, as well as to predict kinetic parameters for new compounds using x-ray structure of a protein receptor and experimental interaction constants of a training set of inhibitors. The developed method is applied to discover structural and kinetic parameters of binding C1q, a protein from the first component of complement system, to low-molecular ligands that inhibit its interactions with immune complexes. Authors have suggested that these ligands bind to a region of C1q globular head near residues Arg150 of chain B, and Lys160 and His67 of chain C, supposedly inhibiting the classical pathway of complement activation. Ligands that inhibit interaction of C1q with immune complexes can be used in the therapy of pathological conditions that are related to unwanted complement activation: allergic reactions, xenograft rejection, etc.


Asunto(s)
Sitios de Unión , Simulación del Acoplamiento Molecular/métodos , Proteínas/química , Proteínas/metabolismo , Complemento C1q/antagonistas & inhibidores , Cristalografía por Rayos X , Inmunoglobulina G/metabolismo , Cinética , Ligandos , Modelos Moleculares , Peso Molecular , Conformación Proteica
17.
Immunobiology ; 217(7): 684-91, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22209113

RESUMEN

Lupus nephritis is one of the most severe manifestations of systemic lupus erythematosus. Higher titers of serum anti-C1q autoantibodies correlate with disease activity in patients with lupus nephritis. Anti-C1q autoantibodies have been shown to bind neo-epitopes within the collagen region of human C1q. In a preliminary study, we recently reported that the anti-C1q autoantibodies could also recognize epitopes within the globular domain (gC1q) of the C1q molecule. Here, 38 sera from patients with renal biopsy-proven lupus nephritis were screened for the presence of anti-gC1q autoantibodies, using recombinant globular head regions of individual A (ghA), B (ghB) and C (ghC) chains of human C1q. We isolated anti-gC1q autoantibodies from three selected patients. Human C1q was pre-incubated with increasing concentrations of the isolated anti-ghA, anti-ghB or anti-ghC autoantibodies and its binding to different C1q target molecules such as IgG and CRP was then evaluated. Anti-ghB, but not anti-ghA and anti-ghC autoantibodies, markedly inhibited C1q interaction with IgG as well as CRP. These results appear to suggest that the anti-ghB autoantibodies may partially induce acquired functional C1q deficiency and thus may interfere with the biological function of C1q.


Asunto(s)
Autoanticuerpos/inmunología , Complemento C1q/inmunología , Inmunoglobulina G/inmunología , Nefritis Lúpica/inmunología , Proteínas Recombinantes/inmunología , Adulto , Autoanticuerpos/sangre , Autoanticuerpos/farmacología , Sitios de Unión de Anticuerpos , Proteína C-Reactiva/antagonistas & inhibidores , Proteína C-Reactiva/inmunología , Proteína C-Reactiva/metabolismo , Complemento C1q/antagonistas & inhibidores , Complemento C1q/metabolismo , Epítopos , Femenino , Humanos , Inmunoglobulina G/sangre , Nefritis Lúpica/sangre , Nefritis Lúpica/patología , Masculino , Persona de Mediana Edad , Unión Proteica , Estructura Terciaria de Proteína , Subunidades de Proteína , Proteínas Recombinantes/metabolismo , Índice de Severidad de la Enfermedad
18.
Lupus ; 20(12): 1267-74, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21813587

RESUMEN

OBJECTIVE: Autoantibodies are important in the diagnosis and classification of systemic lupus erythematosus (SLE), but whether they correlate with changes in disease activity within individual patients is controversial. We assessed the association between changes in SLE global and renal activity and changes in several autoantibodies and cell adhesion molecules in patients with SLE. METHODS: Stored sera collected at two or three clinic visits from each of 49 SLE patients (91% female, 59% African-American, 31% Caucasian, 10% other ethnicity, 38% under 30 years, 41% between 30-44 years, and 21% 45-63 years) were analyzed. The visits were chosen to include one visit with proteinuria, and one or two without, for each patient. Global disease activity was measured by the Physician's Global Assessment (PGA), SELENA-SLEDAI (SLE Disease Activity Index modified to exclude anti-dsDNA and complement) and renal activity assessed by urine protein (by urine dipstick) and Renal Activity Score. Sera were assayed for anti-C1q, anti-chromatin, anti-dsDNA, anti-ribosomal P, monocyte chemotactic protein-1 (MCP-1), vascular cell adhesion molecule (VCAM) intercellular adhesion molecule (ICAM) and complement. The associations between changes in disease activity and changes in biomarker levels were assessed. RESULTS: In terms of global disease activity, anti-C1q had the highest association with the PGA (p = 0.09) and was strongly associated with modified SELENA-SLEDAI (p = 0.009). In terms of renal activity, anti-C1q had the highest association with proteinuria (p = 0.079), and was strongly associated with Renal Activity Score (p = 0.006). CONCLUSION: Anti-C1q performed the best of the potential biomarkers, being significantly associated with the modified SELENA-SLEDAI and with the Renal Activity Score. This study indicates the potential superior utility of anti-C1q over anti-dsDNA and other measures to track renal activity.


Asunto(s)
Autoanticuerpos/sangre , Complemento C1q/inmunología , Nefritis Lúpica/inmunología , Adulto , Anticuerpos Antinucleares/sangre , Biomarcadores/sangre , Moléculas de Adhesión Celular/sangre , Quimiocina CCL2/sangre , Estudios de Cohortes , Complemento C1q/antagonistas & inhibidores , Complemento C3/metabolismo , Complemento C4/metabolismo , Femenino , Humanos , Nefritis Lúpica/sangre , Nefritis Lúpica/fisiopatología , Masculino , Persona de Mediana Edad , Proteinuria/sangre , Proteinuria/inmunología , Proteinuria/fisiopatología
19.
Lupus ; 20(12): 1275-84, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21813590

RESUMEN

OBJECTIVE: To evaluate the frequency of primary immunodeficiencies (PID) in juvenile systemic lupus erythematosus (JSLE) patients. METHODS: Some 72 JSLE patients were analyzed for levels of immunoglobulin classes and IgG subclasses and early components of the classical complement pathway. Determination of C4 gene copy number (GCN) and detection of type I C2 deficiency (D) were also performed. RESULTS: PID was identified in 16 patients (22%): C2D in three, C4D in three, C1qD in two, IgG2D (<20 mg/dl) in four, IgAD (<7 mg/dl) in three, and IgMD (<35 mg/dl) in three; one of these patients presented IgA, C2 and C4D. Two patients had low C4 GCN and two had type I C2D. Demographic data, family history of autoimmune disease and PID, JSLE clinical findings, occurrence of infections, disease activity and therapies were similar in patients with and without PID (p > 0.05). Remarkably, the median of Systemic Lupus International Collaborating Clinics/ACR-damage index (SLICC/ACR-DI) was significantly higher in JSLE patients with PID compared with patients without these abnormalities (p = 0.0033), likewise the high frequency of SLICC/ACR-DI > 1 (p = 0.023). CONCLUSIONS: A high frequency of PID was observed in JSLE patients, suggesting that these defects may contribute to lupus development. Our findings indicate that these two groups of PID should be investigated in severe pediatric lupus.


Asunto(s)
Proteínas del Sistema Complemento/deficiencia , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/inmunología , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/inmunología , Adolescente , Anticuerpos Antinucleares/sangre , Autoanticuerpos/sangre , Secuencia de Bases , Niño , Preescolar , Complemento C1q/antagonistas & inhibidores , Complemento C1q/deficiencia , Complemento C1q/inmunología , Complemento C2/deficiencia , Complemento C2/genética , Complemento C4/deficiencia , Complemento C4/genética , Proteínas del Sistema Complemento/genética , Cartilla de ADN/genética , Femenino , Dosificación de Gen , Humanos , Inmunoglobulinas/sangre , Inmunoglobulinas/clasificación , Síndromes de Inmunodeficiencia/genética , Lactante , Lupus Eritematoso Sistémico/genética , Masculino
20.
J Biol Chem ; 286(18): 16459-69, 2011 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-21454703

RESUMEN

C1q is the recognition subunit of the first component of the classical complement pathway. It participates in clearance of immune complexes and apoptotic cells as well as in defense against pathogens. Inappropriate activation of the complement contributes to cellular and tissue damage in different pathologies, urging the need for the development of therapeutic agents that are able to inhibit the complement system. In this study, we report heme as an inhibitor of C1q. Exposure of C1q to heme significantly reduced the activation of the classical complement pathway, mediated by C-reactive protein (CRP) and IgG. Interaction analyses revealed that heme reduces the binding of C1q to CRP and IgG. Furthermore, we demonstrated that the inhibition of C1q interactions results from a direct binding of heme to C1q. Formation of complex of heme with C1q caused changes in the mechanism of recognition of IgG and CRP. Taken together, our data suggest that heme is a natural negative regulator of the classical complement pathway at the level of C1q. Heme may play a role at sites of excessive tissue damage and hemolysis where large amounts of free heme are released.


Asunto(s)
Complemento C1q/metabolismo , Vía Clásica del Complemento/fisiología , Hemo/metabolismo , Proteína C-Reactiva/química , Proteína C-Reactiva/metabolismo , Complemento C1q/antagonistas & inhibidores , Complemento C1q/química , Hemo/química , Hemólisis/fisiología , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/metabolismo , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA