Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 179
Filtrar
1.
Nat Commun ; 12(1): 6086, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34667172

RESUMEN

Unregulated complement activation causes inflammatory and immunological pathologies with consequences for human disease. To prevent bystander damage during an immune response, extracellular chaperones (clusterin and vitronectin) capture and clear soluble precursors to the membrane attack complex (sMAC). However, how these chaperones block further polymerization of MAC and prevent the complex from binding target membranes remains unclear. Here, we address that question by combining cryo electron microscopy (cryoEM) and cross-linking mass spectrometry (XL-MS) to solve the structure of sMAC. Together our data reveal how clusterin recognizes and inhibits polymerizing complement proteins by binding a negatively charged surface of sMAC. Furthermore, we show that the pore-forming C9 protein is trapped in an intermediate conformation whereby only one of its two transmembrane ß-hairpins has unfurled. This structure provides molecular details for immune pore formation and helps explain a complement control mechanism that has potential implications for how cell clearance pathways mediate immune homeostasis.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/química , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Complemento C8/química , Complemento C8/metabolismo , Complemento C9/química , Complemento C9/inmunología , Microscopía por Crioelectrón , Humanos , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios Proteicos
2.
Front Immunol ; 11: 569549, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193347

RESUMEN

The complement system has developed different strategies to clear infections by several effector mechanisms, such as opsonization, which supports phagocytosis, attracting immune cells by C3 and C5 cleavage products, or direct killing of pathogens by the formation of the membrane attack complex (MAC). As the Zika virus (ZIKV) activates the classical complement pathway and thus has to avoid clearance by the complement system, we analyzed putative viral escape mechanisms, which limit virolysis. We identified binding of the recombinant viral envelope E protein to components of the terminal pathway complement (C5b6, C7, C8, and C9) by ELISA. Western blot analyses revealed that ZIKV E protein interfered with the polymerization of C9, induced on cellular surfaces, either by purified terminal complement proteins or by normal human serum (NHS) as a source of the complement. Further, the hemolytic activity of NHS was significantly reduced in the presence of the recombinant E protein or entire viral particles. This data indicates that ZIKV reduces MAC formation and complement-mediated lysis by binding terminal complement proteins to the viral E protein.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/metabolismo , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología , Virus Zika/inmunología , Línea Celular , Membrana Celular/inmunología , Membrana Celular/metabolismo , Activación de Complemento/inmunología , Complemento C9/inmunología , Complemento C9/metabolismo , Vía Clásica del Complemento , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Unión Proteica , Multimerización de Proteína
3.
Ecotoxicology ; 29(7): 837-845, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32656652

RESUMEN

Vibrio alginolyticus is posting an increasing threat to survival of grouper. Classical complement cascade can trigger initiation of immunity, while complement 9 (C9) is a major complement molecule involved in final step of membrane attack complex (MAC) formation. In this study, full-length EcC9 contained an ORF sequence of 1779 bp, encoding a polypeptide of 592 amino acids. A high-level expression of EcC9 mRNA was observed in liver. Following vibrio challenge, increased expression levels of EcC1q, EcBf/C2, EcC4, EcC6, EcC7 and EcC9 mRNA were detected in liver and kidney. These results implied that elevated expression level of classical complement pathway (CCP) and terminal complement components (TCCs) may assess toxicological effect of V. alginolyticus.


Asunto(s)
Lubina/genética , Lubina/inmunología , Complemento C9/genética , Complemento C9/inmunología , Enfermedades de los Peces/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Complemento C9/química , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Perfilación de la Expresión Génica/veterinaria , Filogenia , Alineación de Secuencia/veterinaria , Vibrio alginolyticus/fisiología
4.
Fish Shellfish Immunol ; 104: 101-110, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32464273

RESUMEN

The ninth complement component (C9) is a terminal complement component (TCC) that is involved in creating the membrane attack complex (MAC) on the target cell surface. In this study, the CsC9 (C9 of Cynoglossus semilaevis) cDNA sequence was cloned and characterized. The full-length CsC9 cDNA measured 2,150 bp, containing an open reading frame (ORF) of 1,803 bp, a 5'-untranslated region (UTR) of 24 bp and a 3'-UTR of 323 bp. A domain search revealed that the CsC9 protein contains five domains, including two TSP1s, an LDLRA, an EGF, and a MACPF. Quantitative real-time PCR analysis showed that CsC9 at the mRNA level was expressed in all the tested tissues, with the highest expression being observed in the liver. CsC9 expression is significantly upregulated in the tested tissues after challenge with Vibrio anguillarum. To further characterize the role of CsC9, peripheral blood mononuclear cells of C. semilaevis were used for transcriptome analysis after incubation with recombinant CsC9 (rCsC9) protein. A total of 3,775 significant differentially expressed genes (DEGs) were identified between the control and the rCsC9-treated group, including 2,063 upregulated genes and 1,712 downregulated genes. KEGG analyses revealed that the DEGs were enriched in cell adhesion molecules, cytokine-cytokine receptor interactions, T cell receptor signaling pathways, B cell receptor signaling pathways and Toll-like receptor signaling pathways. The results of this study indicate that in addition to participating in MAC formation, CsC9 might play multiple roles in the innate and adaptive immunity of C. semilaevis.


Asunto(s)
Complemento C9/genética , Complemento C9/inmunología , Enfermedades de los Peces/inmunología , Peces Planos/genética , Peces Planos/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Inmunidad Adaptativa , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Complemento C9/química , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Perfilación de la Expresión Génica/veterinaria , Leucocitos/metabolismo , Filogenia , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Alineación de Secuencia/veterinaria , Transcriptoma , Vibrio , Vibriosis
5.
J Clin Invest ; 129(3): 1061-1075, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30714990

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is frequently associated in humans with loss-of-function mutations in complement-regulating proteins or gain-of-function mutations in complement-activating proteins. Thus, aHUS provides an archetypal complement-mediated disease with which to model new therapeutic strategies and treatments. Herein, we show that, when transferred to mice, an aHUS-associated gain-of-function change (D1115N) to the complement-activation protein C3 results in aHUS. Homozygous C3 p.D1115N (C3KI) mice developed spontaneous chronic thrombotic microangiopathy together with hematuria, thrombocytopenia, elevated creatinine, and evidence of hemolysis. Mice with active disease had reduced plasma C3 with C3 fragment and C9 deposition within the kidney. Therapeutic blockade or genetic deletion of C5, a protein downstream of C3 in the complement cascade, protected homozygous C3KI mice from thrombotic microangiopathy and aHUS. Thus, our data provide in vivo modeling evidence that gain-of-function changes in complement C3 drive aHUS. They also show that long-term C5 deficiency is not accompanied by development of other renal complications (such as C3 glomerulopathy) despite sustained dysregulation of C3. Our results suggest that this preclinical model will allow testing of novel complement inhibitors with the aim of developing precisely targeted therapeutics that could have application in many complement-mediated diseases.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Activación de Complemento , Complemento C3 , Complemento C5 , Riñón , Mutación Missense , Sustitución de Aminoácidos , Animales , Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/inmunología , Síndrome Hemolítico Urémico Atípico/patología , Complemento C3/genética , Complemento C3/inmunología , Complemento C5/genética , Complemento C5/inmunología , Complemento C9/genética , Complemento C9/inmunología , Modelos Animales de Enfermedad , Glomerulonefritis Membranosa/genética , Glomerulonefritis Membranosa/inmunología , Glomerulonefritis Membranosa/patología , Riñón/inmunología , Riñón/patología , Ratones , Ratones Transgénicos
6.
Fish Shellfish Immunol ; 86: 449-458, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30508672

RESUMEN

The complement system plays an important role in host defense against invading microorganisms. Complement component C9 is the last component that is involved in the formation of the membrane attack complex (MAC) on the surface of target cells. In the present study, the full length C9 cDNA sequence of 1984 bp with an open reading frame (ORF) of 1809 bp was cloned from southern catfish (Silurus meridionalis). The deduced amino acid sequence showed similarity with other teleost fish. The mRNA expression of C9 was detected in the liver, spleen, stomach, intestine, and head kidney, with highest levels detected in the liver. The mRNA of C9 was first detected in the yolk syncytial layer at 34 h post fertilization (hpf) with whole mount in situ hybridization, followed by the liver at 36 h post hatching (hph). The mRNA expression of C9 was upregulated significantly in the liver, spleen, and intestine following the injection with Aeromonas hydrophila, suggesting that C9 played an important role in defense against invading pathogens in southern catfish. Therefore, these results provide important information to understand the functions of C9 during fish early development in fish.


Asunto(s)
Bagres/genética , Bagres/inmunología , Complemento C9/genética , Complemento C9/inmunología , Enfermedades de los Peces/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Aeromonas hydrophila/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Complemento C9/química , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Perfilación de la Expresión Génica/veterinaria , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/veterinaria , Alineación de Secuencia/veterinaria
7.
Mol Vis ; 24: 518-535, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30090015

RESUMEN

Purpose: Variants of complement factor genes, hypoxia and oxidative stress of the outer retina, and systemic hypertension affect the risk of age-related macular degeneration. Hypertension often results from the high intake of dietary salt that increases extracellular osmolarity. We determined the effects of extracellular hyperosmolarity, hypoxia, and oxidative stress on the expression of complement genes in cultured (dedifferentiated) human RPE cells and investigated the effects of C9 siRNA and C9 protein on RPE cells. Methods: Hyperosmolarity was induced by adding 100 mM NaCl or sucrose to the culture medium. Hypoxia was induced by culturing cells in 1% O2 or by adding the hypoxia mimetic CoCl2. Oxidative stress was induced by adding H2O2. Gene and protein expression levels were determined with real-time RT-PCR, western blot, and ELISA analyses. The expression of the nuclear factor of activated T cell 5 (NFAT5) and complement factor (C9) was knocked down with siRNA. Results: Extracellular hyperosmolarity, hypoxia, and oxidative stress strongly increased the transcription of the C9 gene, while the expression of the C3, C5, CFH, and CFB genes was moderately altered or not altered at all. Hyperosmolarity also induced a moderate increase in the cytosolic C9 protein level. The hyperosmotic C9 gene expression was reduced by inhibitors of the p38 MAPK, ERK1/2, JNK, and PI3K signal transduction pathways and of the transcription factors STAT3 and NFAT5. The hypoxic C9 gene expression was reduced by a STAT3 inhibitor. The knockdown of C9 with siRNA decreased the hypoxic vascular endothelial growth factor (VEGF) and NLRP3 gene expression, the hypoxic secretion of VEGF, and the hyperosmotic expression of the NLRP3 gene. Exogenous C9 protein inhibited the hyperosmotic expression of the C9 gene, the hypoxic and hyperosmotic VEGF gene expression, and the hyperosmotic expression of the NLRP3 gene. Both C9 siRNA and C9 protein inhibited inflammasome activation under hyperosmotic conditions, as indicated by the decrease in the cytosolic level of mature IL-1ß. Conclusions: The expression of the C9 gene in cultured RPE cells is highly induced by extracellular hyperosmolarity, hypoxia, and oxidative stress. The data may support the assumption that C9 gene expression may stimulate the expression of inflammatory (NLRP3) and angiogenic growth factors (VEGF) in RPE cells. Extracellular C9 protein may attenuate this effect, in part via negative regulation of the C9 mRNA level.


Asunto(s)
Cobalto/farmacología , Complemento C9/genética , Células Epiteliales/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Cloruro de Sodio/farmacología , Factor A de Crecimiento Endotelial Vascular/genética , Hipoxia de la Célula/efectos de los fármacos , Línea Celular , Complemento C3/genética , Complemento C3/inmunología , Complemento C5/genética , Complemento C5/inmunología , Complemento C9/antagonistas & inhibidores , Complemento C9/inmunología , Factor B del Complemento/genética , Factor B del Complemento/inmunología , Factor H de Complemento/genética , Factor H de Complemento/inmunología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Humanos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Concentración Osmolar , Presión Osmótica/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/inmunología , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Factor A de Crecimiento Endotelial Vascular/inmunología
8.
Fish Shellfish Immunol ; 81: 1-9, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29981471

RESUMEN

The redlip mullet (Liza haematocheila) is one of the most economically important fish in Korea and other East Asian countries; it is susceptible to infections by pathogens such as Lactococcus garvieae, Argulus spp., Trichodina spp., and Vibrio spp. Learning about the mechanisms of the complement system of the innate immunity of redlip mullet is important for efforts towards eradicating pathogens. Here, we report a comprehensive study of the terminal complement complex (TCC) components that form the membrane attack complex (MAC) through in-silico characterization and comparative spatial and temporal expression profiling. Five conserved domains (TSP1, LDLa, MACPF, CCP, and FIMAC) were detected in the TCC components, but the CCP and FIMAC domains were absent in MuC8ß and MuC9. Expression analysis of four TCC genes from healthy redlip mullets showed the highest expression levels in the liver, whereas limited expression was observed in other tissues; immune-induced expression in the head kidney and spleen revealed significant responses against Lactococcus garvieae and poly I:C injection, suggesting their involvement in MAC formation in response to harmful pathogenic infections. Furthermore, the response to poly I:C may suggest the role of TCC components in the breakdown of the membrane of enveloped viruses. These findings may help to elucidate the mechanisms behind the complement system of the teleosts innate immunity.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/genética , Inmunidad Innata , Smegmamorpha/inmunología , Animales , Complemento C6/genética , Complemento C6/inmunología , Complemento C7/genética , Complemento C7/inmunología , Complemento C8/genética , Complemento C8/inmunología , Complemento C9/genética , Complemento C9/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Perfilación de la Expresión Génica , Lactococcus , Lipopolisacáridos , Hígado/inmunología , Poli I-C/farmacología , Smegmamorpha/genética , Bazo/inmunología
9.
PLoS One ; 13(6): e0198472, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29894483

RESUMEN

Immunoproteomic analysis was performed to identify unknown, pathology-related molecules in patients with seronegative (SN) obstetric antiphospholipid syndrome (APS) who clinically satisfied the diagnostic criteria for APS, but not the serological criteria. We collected peripheral blood from 13 SN-APS outpatients with known thrombotic predisposition, 13 with no known thrombotic predisposition, and four multiparous women with no history of miscarriage (control). Plasma proteins from volunteers were purified and used as plasma protein antigens. Two-dimensional immunoblotting was performed using pooled control or SN-APS serum samples as the primary antibodies. Mass spectrometry of reactive spots specific to SN-APS serum led to the identification of complement molecule C9. Western blotting using commercial purified alkylated C9 was performed to detect autoantibodies. Examination of individual patient serum identified reactivity in one patient with, and in two patients without known thrombotic predisposition. This study suggests that SN-APS pathologies were associated with autoantibodies that react to specific C9 epitopes.


Asunto(s)
Síndrome Antifosfolípido/inmunología , Autoanticuerpos/sangre , Complemento C9/inmunología , Proteómica/métodos , Adulto , Síndrome Antifosfolípido/sangre , Biomarcadores/sangre , Electroforesis en Gel Bidimensional , Epítopos/inmunología , Femenino , Humanos , Masculino , Espectrometría de Masas
10.
Mol Vis ; 24: 75-82, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29410599

RESUMEN

Purpose: A recent genome-wide association study by the International Age-related Macular Degeneration Genomics Consortium (IAMDGC) identified seven rare variants that are individually associated with age-related macular degeneration (AMD), the most common cause of vision loss in the elderly. In literature, several of these rare variants have been reported with different frequencies and odds ratios across populations of Europe and North America. Here, we aim to describe the representation of these seven AMD-associated rare variants in different geographic regions based on 24 AMD studies. Methods: We explored the occurrence of seven rare variants independently associated with AMD (CFH rs121913059 (p.Arg1210Cys), CFI rs141853578 (p.Gly119Arg), C3 rs147859257 (p.Lys155Gln), and C9 rs34882957 (p.Pro167Ser)) and three non-coding variants in or near the CFH gene (rs148553336, rs35292876, and rs191281603) in 24 AMD case-control studies. We studied the difference in distribution, interaction, and effect size for each of the rare variants based on the minor allele frequency within the different geographic regions. Results: We demonstrate that two rare AMD-associated variants in the CFH gene (rs121913059 [p.Arg1210Cys] and rs35292876) deviate in frequency among different geographic regions (p=0.004 and p=0.001, respectively). The risk estimates of each of the seven rare variants were comparable across the geographic regions. Conclusions: The results emphasize the importance of identifying population-specific rare variants, for example, by performing sequencing studies in case-control studies of various populations, because their identification may have implications for diagnostic screening and personalized treatment.


Asunto(s)
Factor H de Complemento/genética , Predisposición Genética a la Enfermedad , Degeneración Macular/genética , Polimorfismo de Nucleótido Simple , Anciano , Alelos , Estudios de Casos y Controles , Complemento C3/genética , Complemento C3/inmunología , Complemento C9/genética , Complemento C9/inmunología , Factor H de Complemento/inmunología , Factor I de Complemento/genética , Factor I de Complemento/inmunología , Europa (Continente) , Femenino , Expresión Génica , Frecuencia de los Genes , Estudio de Asociación del Genoma Completo , Geografía , Humanos , Degeneración Macular/diagnóstico , Degeneración Macular/inmunología , Degeneración Macular/patología , Masculino
11.
Clin Immunol ; 181: 24-28, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28578024

RESUMEN

We examined complement-dependent cytotoxicity (CDC) by hexamer formation-enhanced CD20 mAb Hx-7D8 of patient-derived chronic lymphocytic leukemia (CLL) cells that are relatively resistant to CDC. CDC was analyzed in normal human serum (NHS) and serum from an individual genetically deficient for C9. Hx-7D8 was able to kill up to 80% of CLL cells in complete absence of C9. We conclude that the narrow C5b-8 pores formed without C9 are sufficient for CDC due to efficient antibody-mediated hexamer formation. In the absence of C9, we observed transient intracellular increases of Ca2+ during CDC (as assessed with FLUO-4) that were extended in time. This suggests that small C5b-8 pores allow Ca2+ to enter the cell, while dissipation of the fluorescent signal accompanying cell disintegration is delayed. The Ca2+ signal is retained concomitantly with TOPRO-3 (viability dye) staining, thereby confirming that Ca2+ influx represents the most proximate mediator of cell death by CDC.


Asunto(s)
Complemento C9/deficiencia , Proteínas del Sistema Complemento/inmunología , Síndromes de Inmunodeficiencia/inmunología , Leucemia Linfocítica Crónica de Células B , Rituximab/farmacología , Calcio/metabolismo , Supervivencia Celular/efectos de los fármacos , Complemento C9/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Proteínas del Sistema Complemento/metabolismo , Enfermedades por Deficiencia de Complemento Hereditario , Humanos , Inmunoterapia , Polimerizacion
12.
Microbiol Immunol ; 61(2): 75-84, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28150868

RESUMEN

Vitronectin (Vn), a multifunctional protein of blood and extracellular matrix, interacts with complement C9. This interaction may modulate innate immunity. Details of Vn-C9 interactions are limited. Vn-C9 interactions were assessed by employing a goat homologous system and observing Vn binding to C9 in three different assays. Using recombinant fragments, C9 binding was mapped to the N-terminus of Vn. Site directed mutagenesis was performed to alter the second arginine glycine aspartic acid (RGD) sequence (RGD-2) of Vn. Changing R to G or D to A in RGD-2 caused significant decrease in Vn binding to C9 whereas changing of R to G in the first RGD motif (RGD-1) had no effect on Vn binding to C9. These results imply that the RGD-2 of goat Vn is involved in C9 binding. In a competitive binding assay, the presence of soluble RGD peptide inhibited Vn binding to C9 whereas heparin had no effect. Vn binding to C9 was also evaluated in terms of bacterial pathogenesis. Serum dependent inhibition of Escherichia coli growth was significantly reverted when Vn or its N-fragment were included in the assay. The C-fragment, which did not support C9 binding, also partly nullified serum-dependent inhibition of bacterial growth, probably through other serum component(s).


Asunto(s)
Secuencias de Aminoácidos , Complemento C9/metabolismo , Factores Inmunológicos/metabolismo , Vitronectina/metabolismo , Animales , Sitios de Unión , Actividad Bactericida de la Sangre , Complemento C9/inmunología , Análisis Mutacional de ADN , Escherichia coli/inmunología , Cabras , Factores Inmunológicos/inmunología , Mutagénesis Sitio-Dirigida , Unión Proteica , Vitronectina/genética , Vitronectina/inmunología
13.
J Immunol ; 197(5): 1762-75, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27474078

RESUMEN

Recently, we demonstrated that IgG Abs can organize into ordered hexamers after binding their cognate Ags expressed on cell surfaces. This process is dependent on Fc:Fc interactions, which promote C1q binding, the first step in classical pathway complement activation. We went on to engineer point mutations that stimulated IgG hexamer formation and complement-dependent cytotoxicity (CDC). The hexamer formation-enhanced (HexaBody) CD20 and CD38 mAbs support faster, more robust CDC than their wild-type counterparts. To further investigate the CDC potential of these mAbs, we used flow cytometry, high-resolution digital imaging, and four-color confocal microscopy to examine their activity against B cell lines and primary chronic lymphocytic leukemia cells in sera depleted of single complement components. We also examined the CDC activity of alemtuzumab (anti-CD52) and mAb W6/32 (anti-HLA), which bind at high density to cells and promote substantial complement activation. Although we observed little CDC for mAb-opsonized cells reacted with sera depleted of early complement components, we were surprised to discover that the Hexabody mAbs, as well as ALM and W6/32, were all quite effective at promoting CDC in sera depleted of individual complement components C6 to C9. However, neutralization studies conducted with an anti-C9 mAb verified that C9 is required for CDC activity against cell lines. These highly effective complement-activating mAbs efficiently focus activated complement components on the cell, including C3b and C9, and promote CDC with a very low threshold of MAC binding, thus providing additional insight into their enhanced efficacy in promoting CDC.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD20/metabolismo , Antígenos/inmunología , Sitios de Unión de Anticuerpos , Complemento C9/metabolismo , Glicoproteínas de Membrana/metabolismo , ADP-Ribosil Ciclasa 1/inmunología , Alemtuzumab , Anticuerpos Monoclonales Humanizados/inmunología , Antígenos CD20/inmunología , Linfocitos B/inmunología , Línea Celular Tumoral , Activación de Complemento , Complemento C3b/metabolismo , Complemento C9/inmunología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Humanos , Glicoproteínas de Membrana/inmunología
14.
Sci Rep ; 6: 30239, 2016 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-27444648

RESUMEN

Terminal complement membrane attack complex (MAC) formation is induced initially by C5b, followed by the sequential condensation of the C6, C7, C8. Polymerization of C9 to the C5b-8 complex forms the C5b-9 (or MAC). The C5b-9 forms lytic or non lytic pores in the cell membrane destroys membrane integrity. The biological functionalities of MAC has been previously investigated by using either the mice deficient in C5 and C6, or MAC's regulator CD59. However, there is no available C9 deficient mice (mC9(-/-)) for directly dissecting the role of C5b-9 in the pathogenesis of human diseases. Further, since C5b-7 and C5b-8 complexes form non lytic pore, it may also plays biological functionality. To better understand the role of terminal complement cascades, here we report a successful generation of mC9(-/-). We demonstrated that lack of C9 attenuates anti-erythrocyte antibody-mediated hemolysis or LPS-induced acute shock. Further, the rescuing effect on the acute shock correlates with the less release of IL-1ß in mC9(-/-), which is associated with suppression of MAC-mediated inflammasome activation in mC9(-/-). Taken together, these results not only confirm the critical role of C5b-9 in complement-mediated hemolysis and but also highlight the critical role of C5b-9 in inflammasome activation.


Asunto(s)
Complemento C5b/genética , Complemento C9/genética , Complejo de Ataque a Membrana del Sistema Complemento/genética , Inflamación/genética , Choque/genética , Animales , Anticuerpos/inmunología , Anticuerpos/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Complemento C5b/inmunología , Complemento C9/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/química , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/genética , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Eritrocitos/inmunología , Eritrocitos/metabolismo , Hemólisis/inmunología , Humanos , Inflamasomas/genética , Inflamasomas/inmunología , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/patología , Lipopolisacáridos/toxicidad , Ratones , Ratones Noqueados , Choque/inducido químicamente , Choque/inmunología , Choque/fisiopatología
15.
Mol Immunol ; 76: 13-21, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27337595

RESUMEN

Experimental mouse models have been extensively used to elucidate the role of the complement system in different diseases and injuries. Contribution of gender has revealed an intriguing gender specific difference; female mice often show protection against most complement driven injuries such as ischemia/reperfusion injury, graft rejection and sepsis. Interestingly, early studies to the mouse complement system revealed that female mice have very low total complement activity (CH50), which is related to androgen regulation of hepatic complement synthesis. Here, our aim was to understand at which level the female specific differences in mouse complement resides. We have used recently developed complement assays to study the functional activities of female and male mice at the level of C3 and C9 activation, and furthermore assayed key complement factor levels in serum of age-matched female and male C57BL/6 mice. Our results show that the female mice have normal complement cascade functionality at the level of C3 activation, which was supported by determinations of early complement factors. However, all pathways are strongly reduced at the level of C9 activation, suggesting a terminal pathway specific difference. This was in line with C6 and C9 measurements, showing strongly decreased levels in females. Furthermore, similar gender differences were also found in BALB/cJ mice, but not in CD-1 mice. Our results clearly demonstrate that the complement system in females of frequently used mouse strains is restricted by the terminal pathway components and that the perceived female specific protection against experimental disease and injury might be in part explained by the inability promote inflammation through C5b-9.


Asunto(s)
Activación de Complemento/inmunología , Complemento C3/inmunología , Complemento C6/inmunología , Complemento C9/inmunología , Caracteres Sexuales , Animales , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
16.
Fish Shellfish Immunol ; 51: 211-219, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26902705

RESUMEN

The large yellow croaker Larimichthys crocea, as one of the most economically important marine fish in China and East Asian countries, are facing the fatal attraction of various pathogens in recent years. Elucidation of the organism immunomodulatory mechanism of croaker response to pathogen infection is essential for the disease control. In present study, we reported for the first time the molecular characterization and expression analysis of two terminal complement components (TCCs) of croaker, Lc-C7 and Lc-C9. These two structural conserved TCCs were detected in many tissues in adult healthy fish, with highest levels detected in liver. The transcriptional expression analysis of Lc-C7 and Lc-C9 at different developmental stages showed a continuous increase towards hatch, however the two TCCs mRNA were not detected at the unfertilized stage, hinting the origination of these two TCCs after fertilization. Rapid and drastic responses to Vibrio alginolyticus challenge were observed for Lc-C7 and Lc-C9, suggesting the involvement of component C7 and C9 in innate immune responses to pathogenic invasion in teleost fish. These findings could deepen our understanding about immunomodulatory mechanisms of croaker and shed a new light to the role of component system in teleostean immunomodulation.


Asunto(s)
Complemento C7/inmunología , Complemento C9/inmunología , Enfermedades de los Peces/inmunología , Proteínas de Peces/inmunología , Perciformes/inmunología , Vibriosis/inmunología , Vibrio alginolyticus , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Complemento C7/genética , Complemento C9/genética , ADN Complementario/genética , Proteínas de Peces/genética , Riñón Cefálico/inmunología , Hígado/inmunología , Perciformes/genética , Filogenia , ARN Mensajero/genética , Vibriosis/veterinaria
17.
J Immunol Methods ; 419: 25-34, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25733354

RESUMEN

The complement system is an essential component of our innate immunity, both for the protection against infections and for proper handling of dying cells. However, the complement system can also contribute to tissue injury and inflammatory responses. In view of novel therapeutic possibilities, there is an increasing interest in measurement of the complement system activation in the systemic compartment, both in the clinical setting as well as in experimental models. Here we describe in parallel a sensitive and specific sandwich ELISA detecting mouse C3 activation fragments C3b/C3c/iC3b, as well as functional complement ELISAs detecting specific activities of the three complement pathways at the level of C3 and at the level of C9 activation. In a murine model of renal ischaemia/reperfusion injury (IRI) we found transient complement activation as shown by generation of C3b/C3c/iC3b fragments at 24 h following reperfusion, which returned to base-line at 3 and 7 days post reperfusion. When the pathway specific complement activities were measured at the level of C3 activation, we found no significant reduction in any of the pathways. However, the functional complement activity of all three pathways was significantly reduced when measured at the level of C9, with the strongest reduction being observed in the alternative pathway. For all three pathways there was a strong correlation between the amount of C3 fragments and the reduction in functional complement activity. Moreover, at 24 h both C3 fragments and the functional complement activities showed a correlation with the rise in serum creatinine. Together our results show that determination of the systemic pathway specific complement activity is feasible in experimental mouse models and that they are useful in understanding complement activation and inhibition in vivo.


Asunto(s)
Activación de Complemento/inmunología , Complemento C3b/inmunología , Complemento C3c/inmunología , Riñón/inmunología , Daño por Reperfusión/inmunología , Animales , Activación de Complemento/genética , Complemento C3b/genética , Complemento C3b/metabolismo , Complemento C3c/genética , Complemento C3c/metabolismo , Complemento C9/inmunología , Complemento C9/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Estudios de Factibilidad , Riñón/irrigación sanguínea , Riñón/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Reproducibilidad de los Resultados , Factores de Tiempo
18.
Infect Immun ; 83(3): 888-97, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25534939

RESUMEN

Upon infection, pathogenic Leptospira species bind several complement regulators in order to overcome host innate immunity. We previously characterized a 20-kDa leptospiral surface protein which interacts with C4b binding protein (C4BP): leptospiral complement regulator-acquiring protein A (LcpA). Here we show that LcpA also interacts with human factor H (FH), which remains functionally active once bound to the protein. Antibodies directed against short consensus repeat 20 (SCR20) inhibited binding of FH to LcpA by approximately 90%, thus confirming that this particular domain is involved in the interaction. We have also shown for the first time that leptospires bind human vitronectin and that the interaction is mediated by LcpA. Coincubation with heparin blocked LcpA-vitronectin interaction in a dose-dependent manner, strongly suggesting that binding may occur through the heparin binding domains of vitronectin. LcpA also bound to the terminal pathway component C9 and inhibited Zn(2+)-induced polymerization and membrane attack complex (MAC) formation. Competitive binding assays indicated that LcpA interacts with C4BP, FH, and vitronectin through distinct sites. Taken together, our findings indicate that LcpA may play a role in leptospiral immune evasion.


Asunto(s)
Proteínas Bacterianas/química , Leptospira interrogans/química , Leptospira/química , Fragmentos de Péptidos/química , Vitronectina/química , Anticuerpos Monoclonales/química , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/inmunología , Sitios de Unión , Unión Competitiva , Activación de Complemento , Proteína de Unión al Complemento C4b/química , Proteína de Unión al Complemento C4b/inmunología , Complemento C9/química , Complemento C9/inmunología , Factor H de Complemento/química , Factor H de Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/química , Heparina/química , Humanos , Evasión Inmune , Leptospira/inmunología , Leptospira/patogenicidad , Leptospira interrogans/inmunología , Leptospira interrogans/patogenicidad , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/inmunología , Unión Proteica , Vitronectina/inmunología , Zinc/química
19.
PLoS Negl Trop Dis ; 9(12): e0004310, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26720603

RESUMEN

BACKGROUND: Trichinella spiralis expresses paramyosin (Ts-Pmy) as a defense mechanism. Ts-Pmy is a functional protein with binding activity to human complement C8 and C9 and thus plays a role in evading the attack of the host's immune system. In the present study, the binding activity of Ts-Pmy to human complement C1q and its ability to inhibit classical complement activation were investigated. METHODS AND FINDINGS: The binding of recombinant and natural Ts-Pmy to human C1q were determined by ELISA, Far Western blotting and immunoprecipitation, respectively. Binding of recombinant Ts-Pmy (rTs-Pmy) to C1q inhibited C1q binding to IgM and consequently inhibited C3 deposition. The lysis of antibody-sensitized erythrocytes (EAs) elicited by the classical complement pathway was also inhibited in the presence of rTs-Pmy. In addition to inhibiting classical complement activation, rTs-Pmy also suppressed C1q binding to THP-1-derived macrophages, thereby reducing C1q-induced macrophages migration. CONCLUSION: Our results suggest that T. spiralis paramyosin plays an important role in immune evasion by interfering with complement activation through binding to C1q in addition to C8 and C9.


Asunto(s)
Complemento C1q/inmunología , Vía Clásica del Complemento/efectos de los fármacos , Evasión Inmune/efectos de los fármacos , Trichinella spiralis/inmunología , Triquinelosis/inmunología , Tropomiosina/farmacología , Animales , Complemento C1q/metabolismo , Complemento C8/antagonistas & inhibidores , Complemento C8/inmunología , Complemento C9/antagonistas & inhibidores , Complemento C9/inmunología , Eritrocitos/efectos de los fármacos , Femenino , Hemólisis , Humanos , Factores Inmunológicos/inmunología , Factores Inmunológicos/metabolismo , Factores Inmunológicos/farmacología , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Proteínas Recombinantes , Tropomiosina/inmunología , Tropomiosina/metabolismo
20.
J Biol Chem ; 289(21): 15014-22, 2014 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-24719326

RESUMEN

Mortalin/GRP75, the mitochondrial heat shock protein 70, plays a role in cell protection from complement-dependent cytotoxicity (CDC). As shown here, interference with mortalin synthesis enhances sensitivity of K562 erythroleukemia cells to CDC, whereas overexpression of mortalin leads to their resistance to CDC. Quantification of the binding of the C5b-9 membrane attack complex to cells during complement activation shows an inverse correlation between C5b-9 deposition and the level of mortalin in the cell. Following transfection, mortalin-enhanced GFP (EGFP) is located primarily in mitochondria, whereas mortalinΔ51-EGFP lacking the mitochondrial targeting sequence is distributed throughout the cytoplasm. Overexpressed cytosolic mortalinΔ51-EGFP has a reduced protective capacity against CDC relative to mitochondrial mortalin-EGFP. Mortalin was previously shown by us to bind to components of the C5b-9 complex. Two functional domains of mortalin, the N-terminal ATPase domain and the C-terminal substrate-binding domain, were purified after expression in bacteria. Similar to intact mortalin, the ATPase domain, but not the substrate-binding domain, was found to bind to complement proteins C8 and C9 and to inhibit zinc-induced polymerization of C9. Binding of mortalin to complement C9 and C8 occurs through an ionic interaction that is nucleotide-sensitive. We suggest that to express its full protective effect from CDC, mortalin must first reach the mitochondria. In addition, mortalin can potentially target the C8 and C9 complement components through its ATPase domain and inhibit C5b-9 assembly and stability.


Asunto(s)
Complemento C9/inmunología , Proteínas del Sistema Complemento/inmunología , Citotoxicidad Inmunológica/inmunología , Proteínas HSP70 de Choque Térmico/inmunología , Adenosina Difosfato/inmunología , Adenosina Difosfato/farmacología , Adenosina Trifosfatasas/inmunología , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/inmunología , Adenosina Trifosfato/farmacología , Sitios de Unión/genética , Sitios de Unión/inmunología , Western Blotting , Complemento C9/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Proteínas del Sistema Complemento/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Células K562 , Microscopía Confocal , Unión Proteica/efectos de los fármacos , Unión Proteica/inmunología , Interferencia de ARN , Cloruro de Sodio/inmunología , Cloruro de Sodio/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...