Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Neurochem ; 157(3): 479-493, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33190236

RESUMEN

Immune system components also regulate synapse formation and refinement in neurodevelopment. The complement pathway, associated with cell lysis and phagocytosis, is implicated in synaptic elimination. Aberrant adolescent synaptic pruning may underpin schizophrenia onset; thus, changes in cortical complement activity during human development are of major interest. Complement is genetically linked to schizophrenia via increased C4 copy number variants, but the developmental trajectory of complement expression in the human brain is undetermined. As complement increases during periods of active synaptic engulfment in rodents, we hypothesized that complement expression would increase during postnatal development in humans, particularly during adolescence. Using human postmortem prefrontal cortex, we observed that complement activator (C1QB and C3) transcripts peaked in early neurodevelopment, and were highest in toddlers, declining in teenagers (all ANCOVAs between F = 2.41 -3.325, p = .01-0.05). We found that C4 protein was higher at 1-5 years (H = 16.378, p = .012), whereas C3 protein levels were unchanged with age. The microglial complement receptor subunit CD11b increased in mRNA early in life and peaked in the toddler brain (ANCOVA: pH, F = 4.186, p = .003). Complement inhibitors (CD46 and CD55) increased at school age, but failed to decrease like complement activators (both ANCOVAs, F > 4.4, p < .01). These data suggest the activation of complement in the human prefrontal cortex occurs between 1 and 5 years. We did not find evidence of induction of complement factors during adolescence and instead found increased or sustained levels of complement inhibitor mRNA at maturation. Dysregulation of these typical patterns of complement may predispose the brain to neurodevelopmental disorders such as autism or schizophrenia.


Asunto(s)
Envejecimiento/metabolismo , Química Encefálica/fisiología , Encéfalo/crecimiento & desarrollo , Proteínas del Sistema Complemento/metabolismo , Adolescente , Adulto , Antígeno CD11b/biosíntesis , Antígeno CD11b/genética , Antígeno CD56/genética , Antígeno CD56/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Niño , Preescolar , Vía Clásica del Complemento/genética , ADN/biosíntesis , ADN/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Recién Nacido , Masculino , Proteína Cofactora de Membrana/genética , Proteína Cofactora de Membrana/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Corteza Prefrontal/crecimiento & desarrollo , Corteza Prefrontal/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Adulto Joven
2.
PLoS Pathog ; 15(3): e1007659, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30897158

RESUMEN

The carboxy-terminal domain of the BBK32 protein from Borrelia burgdorferi sensu stricto, termed BBK32-C, binds and inhibits the initiating serine protease of the human classical complement pathway, C1r. In this study we investigated the function of BBK32 orthologues of the Lyme-associated Borrelia burgdorferi sensu lato complex, designated BAD16 from B. afzelii strain PGau and BGD19 from B. garinii strain IP90. Our data show that B. afzelii BAD16-C exhibits BBK32-C-like activities in all assays tested, including high-affinity binding to purified C1r protease and C1 complex, and potent inhibition of the classical complement pathway. Recombinant B. garinii BGD19-C also bound C1 and C1r with high-affinity yet exhibited significantly reduced in vitro complement inhibitory activities relative to BBK32-C or BAD16-C. Interestingly, natively produced BGD19 weakly recognized C1r relative to BBK32 and BAD16 and, unlike these proteins, BGD19 did not confer significant protection from serum killing. Site-directed mutagenesis was performed to convert BBK32-C to resemble BGD19-C at three residue positions that are identical between BBK32 and BAD16 but different in BGD19. The resulting chimeric protein was designated BXK32-C and this BBK32-C variant mimicked the properties observed for BGD19-C. To query the disparate complement inhibitory activities of BBK32 orthologues, the crystal structure of BBK32-C was solved to 1.7Å limiting resolution. BBK32-C adopts an anti-parallel four-helix bundle fold with a fifth alpha-helix protruding from the helical core. The structure revealed that the three residues targeted in the BXK32-C chimera are surface-exposed, further supporting their potential relevance in C1r binding and inhibition. Additional binding assays showed that BBK32-C only recognized C1r fragments containing the serine protease domain. The structure-function studies reported here improve our understanding of how BBK32 recognizes and inhibits C1r and provide new insight into complement evasion mechanisms of Lyme-associated spirochetes of the B. burgdorferi sensu lato complex.


Asunto(s)
Proteínas Bacterianas/genética , Borrelia burgdorferi/genética , Vía Clásica del Complemento/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/ultraestructura , Borrelia burgdorferi/inmunología , Grupo Borrelia Burgdorferi , Complemento C1r/metabolismo , Vía Clásica del Complemento/fisiología , Proteínas del Sistema Complemento/metabolismo , Humanos , Enfermedad de Lyme/fisiopatología , Dominios Proteicos/fisiología , Proteínas Recombinantes , Análisis de Secuencia de Proteína
3.
J Autoimmun ; 124: 102728, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34592707

RESUMEN

Extremely rare reactions characterized by thrombosis and thrombocytopenia have been described in subjects that received ChAdOx1 nCoV-19 vaccination 5-16 days earlier. Although patients with vaccine-induced thrombotic thrombocytopenia (VITT) have high levels of antibodies to platelet factor 4 (PF4)-polyanion complexes, the exact mechanism of the development of thrombosis is still unknown. Here we reported serum studies as well as proteomics and genomics analyses demonstrating a massive complement activation potentially linked to the presence of anti-PF4 antibodies in a patient with severe VITT. At admission, complement activity of the classical and lectin pathways were absent (0% for both) with normal levels of the alternative pathway (73%) in association with elevated levels of the complement activation marker sC5b-9 (630 ng/mL [n.v. 139-462 ng/mL]) and anti-PF4 IgG (1.918 OD [n.v. 0.136-0.300 OD]). The immunoblotting analysis of C2 showed the complete disappearance of its normal band at 110 kDa. Intravenous immunoglobulin treatment allowed to recover complement activity of the classical pathway (91%) and lectin pathway (115%), to reduce levels of sC5b-9 (135 ng/mL) and anti-PF4 IgG (0.681 OD) and to normalize the C2 pattern at immunoblotting. Proteomics and genomics analyses in addition to serum studies showed that the absence of complement activity during VITT was not linked to alterations of the C2 gene but rather to a strong complement activation leading to C2 consumption. Our data in a single patient suggest monitoring complement parameters in other VITT patients considering also the possibility to target complement activation with specific drugs.


Asunto(s)
Vacunas contra la COVID-19/efectos adversos , COVID-19/prevención & control , Complemento C2 , Complejo de Ataque a Membrana del Sistema Complemento , Vía Clásica del Complemento , Lectina de Unión a Manosa de la Vía del Complemento , Púrpura Trombocitopénica Trombótica , SARS-CoV-2 , Adulto , Autoanticuerpos/sangre , Vacunas contra la COVID-19/administración & dosificación , ChAdOx1 nCoV-19 , Complemento C2/genética , Complemento C2/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Vía Clásica del Complemento/efectos de los fármacos , Vía Clásica del Complemento/genética , Lectina de Unión a Manosa de la Vía del Complemento/efectos de los fármacos , Lectina de Unión a Manosa de la Vía del Complemento/genética , Femenino , Humanos , Factor Plaquetario 4/sangre , Púrpura Trombocitopénica Trombótica/sangre , Púrpura Trombocitopénica Trombótica/inducido químicamente , Púrpura Trombocitopénica Trombótica/genética
4.
Hum Mol Genet ; 26(R1): R51-R57, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28482029

RESUMEN

Age-related macular degeneration (AMD) is a major cause of visual impairment that affects the central retina. Genome wide association studies and candidate gene screens have identified members of the complement pathway as contributing to the risk of AMD. In this review, we discuss the complement system, its importance in retinal development and normal physiology, how its dysregulation may contribute to disease, and how it might be targeted to prevent damage to the aging choriocapillaris in AMD.


Asunto(s)
Proteínas del Sistema Complemento/genética , Proteínas del Sistema Complemento/metabolismo , Retina/fisiología , Envejecimiento/metabolismo , Coroides/metabolismo , Vía Clásica del Complemento/genética , Estudio de Asociación del Genoma Completo/métodos , Humanos , Degeneración Macular/genética , Degeneración Macular/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Factores de Riesgo
5.
Mod Pathol ; 31(3): 488-494, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29148534

RESUMEN

A diagnosis of thrombotic microangiopathy on kidney biopsy in a patient presenting with hypertensive emergency has historically elicited the diagnosis of malignant hypertension-associated thrombotic microangiopathy. Recent studies, however, have raised awareness that a number of these patients may actually represent atypical hemolytic uremic syndrome. To further investigate this premise, we performed next-generation sequencing to interrogate the coding regions of 29 complement and coagulation cascade genes associated with atypical hemolytic uremic syndrome in 100 non-elderly patients presenting with severe hypertension, renal failure and a kidney biopsy showing microangiopathic changes limited to the classic accelerated hypertension-associated lesion of arterial intimal edema ('mucoid intimal hyperplasia') in isolation and without accompanying glomerular microthrombi. No pathogenic or likely pathogenic variants were identified in any of the genes analyzed, although 13 patients had rare variants of uncertain significance predicted to be deleterious by all in-silico prediction methods utilized. Accordingly, this large patient cohort showed no definitive burden of disease secondary to genetic variants involving complement or coagulation pathways, which contrasts sharply with the high frequency of similar mutational events reported for atypical hemolytic uremic syndrome. Our results also inform recent data by suggesting that patients who present with severe or malignant hypertension and renal thrombotic microangiopathy may be at higher risk for atypical hemolytic uremic syndrome only if the biopsy shows more active disease that includes glomerular fibrin thrombi.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/diagnóstico , Síndrome Hemolítico Urémico Atípico/genética , Vía Clásica del Complemento/genética , Pruebas Genéticas , Hipertensión/fisiopatología , Riñón/fisiopatología , Microangiopatías Trombóticas/fisiopatología , Adulto , Síndrome Hemolítico Urémico Atípico/etiología , Biopsia , Femenino , Humanos , Hipertensión/complicaciones , Masculino , Persona de Mediana Edad , Mutación , Insuficiencia Renal/complicaciones , Microangiopatías Trombóticas/complicaciones
6.
BMC Nephrol ; 19(1): 355, 2018 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-30541482

RESUMEN

BACKGROUND: Renal thrombotic microangiopathy (TMA) is occasionally seen in biopsies with pauci-immune necrotizing crescentic glomerulonephritis (PCGN). Recent study indicated that the complement activation is more prominent in the ANCA-negative glomerulonephritis. CASE PRESENTATION: We report a case of concurrent TMA and PCGN without ANCA positivity. Interestingly, our patient also had biopsy features supportive of Alport syndrome (AS). Genetic studies identified variants and polymorphisms in alternative complement pathway genes that confer substantial risk of developing atypical hemolytic uremic syndrome (aHUS). CONCLUSIONS: Abnormal activation in complement pathway may represent a common pathogenic link between these three distinct entities.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/complicaciones , Síndrome Hemolítico Urémico Atípico/patología , Glomerulonefritis/complicaciones , Glomerulonefritis/patología , Nefritis Hereditaria/complicaciones , Nefritis Hereditaria/patología , Adulto , Anticuerpos Anticitoplasma de Neutrófilos/sangre , Síndrome Hemolítico Urémico Atípico/genética , Vía Clásica del Complemento/genética , Femenino , Humanos , Riñón/patología
7.
J Immunol ; 188(9): 4421-31, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22442442

RESUMEN

Interactions between spores of Bacillus anthracis and macrophages are critical for the development of anthrax infections, as spores are thought to use macrophages as vehicles to disseminate in the host. In this study, we report a novel mechanism for phagocytosis of B. anthracis spores. Murine macrophage-like cell line RAW264.7, bone marrow-derived macrophages, and primary peritoneal macrophages from mice were used. The results indicated that activation of the classical complement pathway (CCP) was a primary mechanism for spore phagocytosis. Phagocytosis was significantly reduced in the absence of C1q or C3. C3 fragments were found deposited on the spore surface, and the deposition was dependent on C1q and Ca(2+). C1q recruitment to the spore surface was mediated by the spore surface protein BclA, as recombinant BclA bound directly and specifically to C1q and inhibited C1q binding to spores in a dose-dependent manner. C1q binding to spores lacking BclA (ΔbclA) was also significantly reduced compared with wild-type spores. In addition, deposition of both C3 and C4 as well as phagocytosis of spores were significantly reduced when BclA was absent, but were not reduced in the absence of IgG, suggesting that BclA, but not IgG, is important in these processes. Taken together, these results support a model in which spores actively engage CCP primarily through BclA interaction with C1q, leading to CCP activation and opsonophagocytosis of spores in an IgG-independent manner. These findings are likely to have significant implications on B. anthracis pathogenesis and microbial manipulation of complement.


Asunto(s)
Carbunco/inmunología , Bacillus anthracis/fisiología , Vía Clásica del Complemento/inmunología , Macrófagos/inmunología , Glicoproteínas de Membrana/inmunología , Fagocitosis/inmunología , Animales , Carbunco/genética , Anticuerpos Antibacterianos/genética , Anticuerpos Antibacterianos/inmunología , Línea Celular , Complemento C1q/genética , Complemento C1q/inmunología , Complemento C3/genética , Complemento C3/inmunología , Vía Clásica del Complemento/genética , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Esporas Bacterianas/genética , Esporas Bacterianas/inmunología
8.
J Immunol ; 188(9): 4450-9, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22467648

RESUMEN

Ail is a 17-kDa chromosomally encoded outer membrane protein that mediates serum resistance (complement resistance) in the pathogenic Yersiniae (Yersinia pestis, Y. enterocolitica, and Y. pseudotuberculosis). In this article, we demonstrate that Y. pseudotuberculosis Ail from strains PB1, 2812/79, and YPIII/pIB1 (serotypes O:1a, O:1b, and O:3, respectively) can bind the inhibitor of the classical and lectin pathways of complement, C4b-binding protein (C4BP). Binding was observed irrespective of serotype tested and independently of YadA, which is the primary C4BP receptor of Y. enterocolitica. Disruption of the ail gene in Y. pseudotuberculosis resulted in loss of C4BP binding. Cofactor assays revealed that bound C4BP is functional, because bound C4BP in the presence of factor I cleaved C4b. In the absence of YadA, Ail conferred serum resistance to strains PB1 and YPIII, whereas serum resistance was observed in strain 2812/79 in the absence of both YadA and Ail, suggesting additional serum resistance factors. Ail from strain YPIII/pIB1 alone can mediate serum resistance and C4BP binding, because its expression in a serum-sensitive laboratory strain of Escherichia coli conferred both of these phenotypes. Using a panel of C4BP mutants, each deficient in a single complement control protein domain, we observed that complement control protein domains 6-8 are important for binding to Ail. Binding of C4BP was unaffected by increasing heparin or salt concentrations, suggesting primarily nonionic interactions. These results indicate that Y. pseudotuberculosis Ail recruits C4BP in a functional manner, facilitating resistance to attack from complement.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Inactivadoras de Complemento/inmunología , Vía Clásica del Complemento/inmunología , Lectina de Unión a Manosa de la Vía del Complemento/inmunología , Antígenos de Histocompatibilidad/inmunología , Yersinia pseudotuberculosis/inmunología , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/inmunología , Adhesinas Bacterianas/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Actividad Bactericida de la Sangre/genética , Actividad Bactericida de la Sangre/inmunología , Complemento C1/genética , Complemento C1/inmunología , Complemento C1/metabolismo , Proteína de Unión al Complemento C4b , Proteínas Inactivadoras de Complemento/genética , Proteínas Inactivadoras de Complemento/metabolismo , Vía Clásica del Complemento/genética , Lectina de Unión a Manosa de la Vía del Complemento/genética , Escherichia coli/genética , Escherichia coli/inmunología , Escherichia coli/metabolismo , Femenino , Antígenos de Histocompatibilidad/genética , Antígenos de Histocompatibilidad/metabolismo , Humanos , Masculino , Mutación , Unión Proteica/genética , Unión Proteica/inmunología , Especificidad de la Especie , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/metabolismo
9.
Scand J Immunol ; 78(2): 181-93, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23672641

RESUMEN

In the context of immunity, pattern recognition is the art of discriminating friend from foe and innocuous from noxious. The basis of discrimination is the existence of evolutionarily conserved patterns on microorganisms, which are intrinsic to these microorganisms and necessary for their function and existence. Such immutable or slowly evolving patterns are ideal handles for recognition and have been targeted by early cellular immune defence mechanisms such as Toll-like receptors, NOD-like receptors, RIG-I-like receptors, C-type lectin receptors and by humoral defence mechanisms such as the complement system. Complement is a proteolytic cascade system comprising around 35 different soluble and membrane-bound proteins. It constitutes a central part of the innate immune system, mediating several major innate effector functions and modulating adaptive immune responses. The complement cascade proceeds via controlled, limited proteolysis and conformational changes of constituent proteins through three activation pathways: the classical pathway, the alternative pathway and the lectin pathway, which converge in common effector functions. Here, we review the nature of the pattern recognition molecules involved in complement activation, as well as their close relatives with no or unknown capacity for activating complement. We proceed to examine the composition of the pattern recognition complexes involved in complement activation, focusing on those of the lectin pathway, and arrive at a new model for their mechanism of operation, supported by recently emerging evidence.


Asunto(s)
Lectina de Unión a Manosa de la Vía del Complemento/genética , Proteínas del Sistema Complemento/genética , Inmunidad Humoral , Receptores de Reconocimiento de Patrones/inmunología , Complejo Antígeno-Anticuerpo/genética , Complejo Antígeno-Anticuerpo/inmunología , Vía Alternativa del Complemento/genética , Vía Clásica del Complemento/genética , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Modelos Inmunológicos , Multimerización de Proteína , Proteolisis , Receptores de Reconocimiento de Patrones/genética
10.
J Immunol ; 186(5): 3066-75, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21263075

RESUMEN

Complement activation represents a crucial innate defense mechanism to invading microorganisms, but there is an eminent lack of understanding of the separate contribution of the different complement activation pathways to the host response during sepsis. We therefore investigated different innate host immune responses during cecal ligation and puncture (CLP)-induced sepsis in mice lacking either the alternative (fD(-/-)) or classical (C1q(-/-)) complement activation pathway. Both knockout mice strains showed a significantly reduced survival and increased organ dysfunction when compared with control mice. Surprisingly, fD(-/-) mice demonstrated a compensated bacterial clearance capacity as control mice at 6 h post CLP, whereas C1q(-/-) mice were already overwhelmed by bacterial growth at this time point. Interestingly, at 24 h after CLP, fD(-/-) mice failed to clear bacteria in a way comparable to control mice. However, both knockout mice strains showed compromised C3 cleavage during sepsis. Investigating potential causes for this discrepancy, we were able to demonstrate that despite normal bacterial clearance capacity early during the onset of sepsis, fD(-/-) mice displayed increased inflammatory cytokine generation and neutrophil recruitment into lungs and blood when compared with both control- and C1q(-/-) mice, indicating a potential loss of control over these immune responses. Further in vitro experiments revealed a strongly increased Nf-κB activation capacity in isolated neutrophils from fD(-/-) mice, supporting this hypothesis. Our results provide evidence for the new concept that the alternative complement activation pathway exerts a distinctly different contribution to the innate host response during sepsis when compared with the classical pathway.


Asunto(s)
Activación de Complemento/inmunología , Vía Alternativa del Complemento/inmunología , Vía Clásica del Complemento/inmunología , Choque Séptico/inmunología , Animales , Carga Bacteriana/inmunología , Ciego , Activación de Complemento/genética , Complemento C1q/deficiencia , Complemento C1q/genética , Factor D del Complemento/deficiencia , Factor D del Complemento/genética , Vía Alternativa del Complemento/genética , Vía Clásica del Complemento/genética , Inmunidad Innata/genética , Riñón/inmunología , Riñón/microbiología , Riñón/fisiopatología , Ligadura , Hígado/inmunología , Hígado/microbiología , Hígado/fisiopatología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Punciones , Choque Séptico/genética , Choque Séptico/mortalidad , Análisis de Supervivencia
11.
Biochem Biophys Res Commun ; 421(4): 665-70, 2012 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-22542627

RESUMEN

Histone modifying enzymes confer epigenetic marks, directing the changes in gene expression required for diverse cellular processes. Lysine-specific demethylase 1 (LSD1) functions as a transcriptional coregulator by demethylating histone H3 on lysine 4 and lysine 9. Analyzing transcriptomes on microarrays, we identified genes which represent inflammatory-related targets of LSD1. We demonstrate a repressive role of LSD1 in proinflammatory cytokine expression such as IL1α, IL1ß, IL6 and IL8 and classical complement components. Consistently, LSD1 occupies and regulates the promoter of these genes. In addition, we demonstrate that HDAC1 and LSD1 synergistically regulate these inflammatory-related genes. Our data reveal a novel role for LSD1 in suppressing immune responses.


Asunto(s)
Vía Clásica del Complemento/genética , Citocinas/genética , Epigénesis Genética , Histona Desacetilasa 1/metabolismo , Histona Demetilasas/metabolismo , Inmunidad/genética , Inflamación/genética , Línea Celular Tumoral , Citocinas/antagonistas & inhibidores , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Células Hep G2 , Histona Desacetilasa 1/genética , Histona Demetilasas/genética , Humanos
13.
BMC Nephrol ; 13: 161, 2012 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-23199021

RESUMEN

BACKGROUND: Complement activation products are present in atherosclerotic plaques. Recently, binding of complement to elastin and collagen in the aortic wall has been demonstrated, suggesting a role of complement in the development aortic stiffness and atherosclerosis. The definitive role of complement in atherosclerosis and arteriosclerosis, however, remains unclear. CASE PRESENTATION: We here describe a patient with hereditary complete deficiency of complement C4 suffering from Henoch-Schoenlein purpura and on renal replacement therapy for twenty-eight years. The patient had the full range of risk factors for vascular damage such as hypertension, volume overload, hyperphosphatemia and hyperparathyroidism. Despite that, his carotid artery intima media thickness was below the normal range and his pulse wave velocity was normal. In contrast, the patient's coronary and peripheral muscular arteries were heavily calcified. CONCLUSION: This case supports the hypothesis that complement plays an important role in the development of stiffness of elastic arteries. We speculate that inability to activate complement by the classical or lectin pathways protected the patient from atherosclerosis, arteriosclerosis, stiffening and calcification of the aorta and carotid arteries. Inhibition of complement activation may be a potential target for prophylactic and therapeutic interventions.


Asunto(s)
Aterosclerosis/diagnóstico , Aterosclerosis/fisiopatología , Complemento C4/deficiencia , Terapia de Reemplazo Renal/efectos adversos , Rigidez Vascular/genética , Aterosclerosis/genética , Complemento C4/genética , Vía Clásica del Complemento/genética , Lectina de Unión a Manosa de la Vía del Complemento/genética , Elasticidad/fisiología , Humanos , Masculino , Persona de Mediana Edad , Terapia de Reemplazo Renal/tendencias , Factores de Tiempo
14.
Clin Immunol ; 139(1): 85-93, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21300570

RESUMEN

Functional C1-inhibitor (C1-inh) and C4 are potential severity markers of hereditary angioedema due to deficiency of C1-inh (HAE-C1-inh), and the complexes generated through complement activation may be relevant. We studied the association between disease severity and complement parameters in 105 HAE-C1-inh patients. Disease severity was characterized by the number of angioedema attacks or alternatively, by the number of C1-inh concentrate ampoules (C1-inh-amp) used for the treatment of attacks. Median C1rC1sC1-inh level was higher (32.8 U/ml vs. 3.4 U/ml; p<0.0001) in patients, compared to controls. C1rC1sC1-inh and C1-inh strongly correlated with attack number and C1-inh-amp, both in the whole patient population and in the subgroup on danazol prophylaxis. Both C1rC1sC1-inh and C1-inh are suitable for predicting disease severity based on attack frequency and C1-inh-amp (OR=4.38[1.43-13.43], p=0.010 and 11.78[2.54-54.67], p=0.002, respectively). We presume that both C1rC1sC1-inh and C1-inh might prove sensitive predictors of the severity of HAE-C1-inh.


Asunto(s)
Angioedemas Hereditarios/metabolismo , Vía Clásica del Complemento/genética , Vía Clásica del Complemento/fisiología , Adolescente , Adulto , Anciano , Angioedemas Hereditarios/tratamiento farmacológico , Niño , Preescolar , Danazol/uso terapéutico , Antagonistas de Estrógenos/uso terapéutico , Femenino , Marcadores Genéticos , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
15.
Methods Mol Biol ; 2227: 159-178, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33847941

RESUMEN

The regulators of complement activation (RCA) gene cluster in 1q31-1q32 includes most of the genes encoding complement regulatory proteins. Genetic variability in the RCA gene cluster frequently involve copy number variations (CNVs), a type of chromosome structural variation causing alterations in the number of copies of specific regions of DNA. CNVs in the RCA gene cluster often relate with gene rearrangements that result in the generation of novel genes, carrying internal duplications or deletions, and hybrid genes, resulting from the fusion or exchange of genetic material between two different genes. These gene rearrangements are strongly associated with a number of rare and common diseases characterized by complement dysregulation. Identification of CNVs in the RCA gene cluster is critical in the molecular diagnostic of these diseases. It can be done by bioinformatics analysis of DNA sequence data generated by massive parallel sequencing techniques (NGS, next generation sequencing) but often requires special techniques like multiplex ligation-dependent probe amplification (MLPA). This is because the currently used massive parallel DNA sequencing approaches do not easily identify all the structural variations in the RCA gene cluster. We will describe here how to use the MLPA assays and two computational tools to analyze NGS data, NextGENe and ONCOCNV, to detect CNVs and gene rearrangements in the RCA gene cluster.


Asunto(s)
Activación de Complemento/genética , Análisis Mutacional de ADN/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Reacción en Cadena de la Polimerasa Multiplex/métodos , Aberraciones Cromosómicas , Enzimas Activadoras de Complemento/genética , Vía Alternativa del Complemento/genética , Vía Clásica del Complemento/genética , Proteínas del Sistema Complemento/genética , Variaciones en el Número de Copia de ADN , Reordenamiento Génico , Pruebas Genéticas/métodos , Humanos , Familia de Multigenes/genética , Análisis de Secuencia de ADN
16.
Sci Rep ; 11(1): 13114, 2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34162900

RESUMEN

The currently largest genome-wide association study (GWAS) for age-related macular degeneration (AMD) defines disease association with genome-wide significance for 52 independent common and rare genetic variants across 34 chromosomal loci. Overall, these loci contain over 7200 variants and are enriched for genes with functions indicating several shared cellular processes. Still, the precise mechanisms leading to AMD pathology are largely unknown. Here, we exploit the phenomenon of epistatic interaction to identify seemingly independent AMD-associated variants that reveal joint effects on gene expression. We focus on genetic variants associated with lipid metabolism, organization of extracellular structures, and innate immunity, specifically the complement cascade. Multiple combinations of independent variants were used to generate genetic risk scores allowing gene expression in liver to be compared between low and high-risk AMD. We identified genetic variant combinations correlating significantly with expression of 26 genes, of which 19 have not been associated with AMD before. This study defines novel targets and allows prioritizing further functional work into AMD pathobiology.


Asunto(s)
Epistasis Genética/genética , Sitios Genéticos/genética , Degeneración Macular/genética , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Ligasas de Carbono-Nitrógeno con Glutamina como Donante de Amida-N/genética , Ligasas de Carbono-Nitrógeno con Glutamina como Donante de Amida-N/metabolismo , Vía Clásica del Complemento/genética , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Variación Genética/genética , Humanos , Metabolismo de los Lípidos/genética , Hígado/metabolismo
17.
Genes Immun ; 11(1): 1-10, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19675582

RESUMEN

Complement activation in myasthenia gravis (MG) may damage muscle endplate and complement regulatory proteins such as decay-accelerating factor (DAF) or CD55 may be protective. We hypothesize that the increased prevalence of severe extraocular muscle (EOM) dysfunction among African MG subjects reported earlier may result from altered DAF expression. To test this hypothesis, we screened the DAF gene sequences relevant to the classical complement pathway and found an association between myasthenics with EOM paresis and the DAF regulatory region c.-198C>G SNP (odds ratio=8.6; P=0.0003). This single nucleotide polymorphism (SNP) results in a twofold activation of a DAF 5'-flanking region luciferase reporter transfected into three different cell lines. Direct matching of the surrounding SNP sequence within the DAF regulatory region with the known transcription factor-binding sites suggests a loss of an Sp1-binding site. This was supported by the observation that the c.-198C>G SNP did not show the normal lipopolysaccharide-induced DAF transcriptional upregulation in lymphoblasts from four patients. Our findings suggest that at critical periods during autoimmune MG, this SNP may result in inadequate DAF upregulation with consequent complement-mediated EOM damage. Susceptible individuals may benefit from anti-complement therapy in addition to immunosuppression.


Asunto(s)
Antígenos CD55/genética , Vía Clásica del Complemento/genética , Miastenia Gravis/genética , Polimorfismo de Nucleótido Simple , Elementos de Respuesta/genética , Animales , Población Negra/genética , Antígenos CD55/biosíntesis , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Femenino , Humanos , Terapia de Inmunosupresión , Lipopolisacáridos/farmacología , Masculino , Ratones , Miastenia Gravis/metabolismo , Miastenia Gravis/terapia , Paresia , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética
19.
Artículo en Inglés | MEDLINE | ID: mdl-20635792

RESUMEN

Hereditary complete deficiency of complement component C1q is associated with a high prevalence of systemic lupus erythematosus and increased susceptibility to severe recurrent infections. An 11-year-old girl was screened for immunodeficiency due to a history of recurrent meningitis and pneumonia. Immunologic studies revealed absence of classic pathway hemolytic activity and undetectable levels of Clq. Exon-specific amplification of genomic DNA by polymerase chain reaction followed by direct sequence analysis revealed a novel homozygous missense mutation at codon 48 in the C1q C gene causing a glycine-to-arginine substitution affecting the collagen-like region of C1q. No changes were seen in the exons of the A and B chains. The mutation affected both the formation and the secretion of C1q variant molecules. We describe a novel mutation in the C1q C chain gene that leads to an interchange in amino acids resulting in absence of C1q in serum.


Asunto(s)
Complemento C1q/deficiencia , Complemento C1q/genética , Niño , Complemento C1q/inmunología , Vía Clásica del Complemento/genética , Vía Clásica del Complemento/inmunología , ADN/química , ADN/genética , Femenino , Homocigoto , Humanos , Masculino , Mutación Missense/inmunología , Linaje , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Turquía
20.
J Trauma ; 66(5): 1265-70; discussion 1270-2, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19430225

RESUMEN

BACKGROUND: Trauma is a disease of inflammation. Complement Component 2 (C2) is a protease involved in activation of complement through the classical pathway and has been implicated in a variety of chronic inflammatory diseases. We hypothesized that genetic variation in C2 (E318D) identifies a high-risk subgroup of patients with trauma reflecting increased mortality and infection (ventilator-associated pneumonia [VAP]). Consequently, genetic variation in C2 may stratify patient risk and illuminate underlying mechanisms for therapeutic intervention. METHODS: DNA samples from 702 patients with trauma were genotyped for C2 E318D and linked with covariates (age: mean 42.8 years, gender: 74% male, ethnicity: 80% white, mechanism: 84% blunt, injury severity score: mean 25.0, admission lactate: mean 3.13 mEq/L) and outcomes: mortality 9.9% and VAP: 18.5%. VAP was defined by quantitative bronchoalveolar lavage (> 10). Multivariate regression analysis determined the relationship of genotype and covariates to risk of death and VAP. However, patients with injury severity score > or = 45 were excluded from the multivariate analysis, as magnitude of injury overwhelms genetics and covariates in determining outcome. RESULTS: Fifty-two patients (8.3%) had the high-risk heterozygous genotype, associated with a significant increase in mortality and VAP. CONCLUSION: In 702 patients with trauma, 8.3% had a high-risk genetic variation in C2 associated with increased mortality (odds ratio = 2.65) and infection (odds ratio = 2.00). This variation: (1) identifies a previously unknown high-risk group for infection and mortality; (2) can be determined at admission; (3) may provide opportunity for early therapeutic intervention; and (4) requires validation in a distinct cohort of patients.


Asunto(s)
Causas de Muerte , Complemento C2/genética , Vía Clásica del Complemento/genética , Variación Genética , Mortalidad Hospitalaria/tendencias , Neumonía Asociada al Ventilador/genética , Heridas y Lesiones/genética , Adulto , Distribución por Edad , Análisis de Varianza , Estudios de Cohortes , Complemento C2/análisis , Infección Hospitalaria/diagnóstico , Infección Hospitalaria/genética , Infección Hospitalaria/mortalidad , Femenino , Predisposición Genética a la Enfermedad/epidemiología , Hospitales Universitarios , Humanos , Incidencia , Puntaje de Gravedad del Traumatismo , Modelos Logísticos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Oportunidad Relativa , Neumonía Asociada al Ventilador/mortalidad , Pronóstico , Medición de Riesgo , Sensibilidad y Especificidad , Distribución por Sexo , Centros Traumatológicos , Heridas y Lesiones/diagnóstico , Heridas y Lesiones/mortalidad , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA