Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Viruses ; 15(6)2023 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-37376569

RESUMEN

The complement system is a key component of the innate immune response to viruses and proinflammatory events. Exaggerated complement activation has been attributed to the induction of a cytokine storm in severe SARS-CoV-2 infection. However, there is also an argument for the protective role of complement proteins, given their local synthesis or activation at the site of viral infection. This study investigated the complement activation-independent role of C1q and C4b-binding protein (C4BP) against SARS-CoV-2 infection. The interactions of C1q, its recombinant globular heads, and C4BP with the SARS-CoV-2 spike and receptor binding domain (RBD) were examined using direct ELISA. In addition, RT-qPCR was used to evaluate the modulatory effect of these complement proteins on the SARS-CoV-2-mediated immune response. Cell binding and luciferase-based viral entry assays were utilised to assess the effects of C1q, its recombinant globular heads, and C4BP on SARS-CoV-2 cell entry. C1q and C4BP bound directly to SARS-CoV-2 pseudotype particles via the RBD domain of the spike protein. C1q via its globular heads and C4BP were found to reduce binding as well as viral transduction of SARS-CoV-2 spike protein expressing lentiviral pseudotypes into transfected A549 cells expressing human ACE2 and TMPRSS2. Furthermore, the treatment of the SARS-CoV-2 spike, envelope, nucleoprotein, and membrane protein expressing alphaviral pseudotypes with C1q, its recombinant globular heads, or C4BP triggered a reduction in mRNA levels of proinflammatory cytokines and chemokines such as IL-1ß, IL-8, IL-6, TNF-α, IFN-α, and RANTES (as well as NF-κB) in A549 cells expressing human ACE2 and TMPRSS2. In addition, C1q and C4BP treatment also reduced SARS-CoV-2 pseudotype infection-mediated NF-κB activation in A549 cells expressing human ACE2 and TMPRSS2. C1q and C4BP are synthesised primarily by hepatocytes; however, they are also produced by macrophages, and alveolar type II cells, respectively, locally at the pulmonary site. These findings support the notion that the locally produced C1q and C4BP can be protective against SARS-CoV-2 infection in a complement activation-independent manner, offering immune resistance by inhibiting virus binding to target host cells and attenuating the infection-associated inflammatory response.


Asunto(s)
COVID-19 , Proteína de Unión al Complemento C4b , Humanos , Proteína de Unión al Complemento C4b/química , Proteína de Unión al Complemento C4b/metabolismo , Complemento C1q/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , FN-kappa B/metabolismo , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Activación de Complemento , Proteínas del Sistema Complemento/metabolismo , Unión Proteica
2.
Acta Crystallogr D Struct Biol ; 76(Pt 8): 759-770, 2020 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-32744258

RESUMEN

BibA, a group B streptococcus (GBS) surface protein, has been shown to protect the pathogen from phagocytic killing by sequestering a complement inhibitor: C4b-binding protein (C4BP). Here, the X-ray crystallographic structure of a GBS BibA fragment (BibA126-398) and a low-resolution small-angle X-ray scattering (SAXS) structure of the full-length N-terminal domain (BibA34-400) are described. The BibA126-398 fragment crystal structure displayed a novel and predominantly helical structure. The tertiary arrangement of helices forms four antiparallel three-helix-bundle-motif repeats, with one long helix from a bundle extending into the next. Multiple mutations on recombinant BibA34-400 delayed the degradation of the protein, and circular dichroism spectroscopy of BibA34-400 suggested a similar secondary-structure composition to that observed in the crystallized BibA126-398 fragment. A model was generated for the 92 N-terminal residues (BibA34-125) using structural similarity prediction programs, and a BibA34-400 model was generated by combining the coordinates of BibA34-126 and BibA126-398. The X-ray structure of BibA126-398 and the model of BibA34-400 fitted well into the calculated SAXS envelope. One possible binding site for the BibA N-terminal domain was localized to the N-terminal CCP (complement-control protein) domains of the C4BP α-chain, as indicated by the decreased binding of BibA to a ΔCCP1 C4BP α-chain mutant. In summary, it is suggested that the GBS surface protein BibA, which consists of three antiparallel α-helical-bundle motifs, is unique and belongs to a new class of Gram-positive surface adhesins.


Asunto(s)
Adhesinas Bacterianas/química , Streptococcus agalactiae/metabolismo , Sitios de Unión , Proteína de Unión al Complemento C4b/química , Cristalografía por Rayos X , Conformación Proteica en Hélice alfa
3.
Sheng Wu Gong Cheng Xue Bao ; 36(4): 693-699, 2020 Apr 25.
Artículo en Chino | MEDLINE | ID: mdl-32347063

RESUMEN

To study the interaction between C4b-binding protein (C4BP) and Riemerella anatipestifer (RA), we cloned duck C4BPα, conducted prokaryotic expression and prepared the polyclonal antibody by immunizing mice. Then indirect immunofluorescence assay and dot blotting hybridization assay were used to verify the interaction between C4BP and RA. The full length of duck C4BPα nucleotide sequence was 1 230 bp, with the highest similarity to chicken C4BPα (82.1%). Phylogenetic tree analysis showed that duck C4BPα and chicken C4BPα were on the same phylogenetic tree branch and the genetic evolution relationship between them was the closest. C4BPα was efficiently expressed in Escherichia coli BL21 (DE3). The recombinant proteins existed in intracellular soluble form. The titer of polyclonal antibody was more than 1:10 000 and polyclonal antibodies could specifically recognize the recombinant proteins. The results of indirect immunofluorescence assay and dot blot hybridization assay showed that RA could interact with duck C4BP. The results provide a basis to further reveal the pathogenesis of RA.


Asunto(s)
Proteína de Unión al Complemento C4b , Patos , Regulación de la Expresión Génica , Riemerella , Animales , Clonación Molecular , Proteína de Unión al Complemento C4b/química , Proteína de Unión al Complemento C4b/genética , Proteína de Unión al Complemento C4b/metabolismo , Patos/clasificación , Patos/genética , Patos/microbiología , Ratones , Filogenia , Riemerella/metabolismo
4.
Sci Rep ; 9(1): 5519, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30940885

RESUMEN

The non-canonical NF-κB signaling may be a central integrator of a placental clock that governs the length of human pregnancy. We sought to identify fetal signals that could activate this NF-κB pathway in the placenta, and in turn, contribute to the onset of labor. Proteomics analysis of exosomes purified from fetal cord arterial blood revealed a total of 328 proteins, among which 48 were more significantly abundant (p < 0.01) in samples from women who delivered following elective Cesarean-section at term (39 to 40 weeks of estimated gestational age, EGA) compared to those who had elective Cesarean deliveries near term (35 to 36 weeks of EGA). Computational, crystal structural, and gene functional analyses showed that one of these 48 proteins, C4BPA, binds to CD40 of placental villous trophoblast to activate p100 processing to p52, and in turn, pro-labor genes. These results suggest that fetal C4BPA-induced activation of non-canonical NF-κB in human placenta may play a critical role in processes of term or preterm labor.


Asunto(s)
Proteína de Unión al Complemento C4b/metabolismo , Exosomas/metabolismo , Pulmón/embriología , Subunidad p52 de NF-kappa B/metabolismo , Placenta/metabolismo , Antígenos CD40/metabolismo , Proteína de Unión al Complemento C4b/química , Femenino , Sangre Fetal/metabolismo , Humanos , Trabajo de Parto/metabolismo , Pulmón/metabolismo , Modelos Moleculares , FN-kappa B/metabolismo , Embarazo , Proteómica/métodos , Transducción de Señal
5.
Front Immunol ; 9: 1945, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30210498

RESUMEN

The complement system is a tightly regulated network of proteins involved in defense against pathogens, inflammatory processes, and coordination of the innate and adaptive immune responses. Dysregulation of the complement cascade is associated with many inflammatory disorders. Thus, inhibition of the complement system has emerged as an option for treatment of a range of different inflammatory diseases. MAP-1 is a pattern recognition molecule (PRM)-associated inhibitor of the lectin pathway of the complement system, whereas C4b-binding protein (C4BP) regulates both the classical and lectin pathways. In this study we generated chimeric proteins consisting of MAP-1 and the first five domains of human C4BP (C4BP1-5) in order to develop a targeted inhibitor acting at different levels of the complement cascade. Two different constructs were designed and expressed in CHO cells where MAP-1 was fused with C4BP1-5 in either the C- or N-terminus. The functionality of the chimeric proteins was assessed using different in vitro complement activation assays. Both chimeric proteins displayed the characteristic Ca2+-dependent dimerization and binding to PRMs of native MAP-1, as well as the co-factor activity of native C4BP. In ELISA-based complement activation assays they could effectively inhibit the lectin and classical pathways. Notably, MAP-1:C4BP1-5 was five times more effective than rMAP-1 and rC4BP1-5 applied at the same time, emphasizing the advantage of a single inhibitor containing both functional domains. The MAP-1/C4BP chimeras exert unique complement inhibitory properties and represent a novel therapeutic approach targeting both upstream and central complement activation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Reguladoras de la Apoptosis , Proteína de Unión al Complemento C4b , Lectina de Unión a Manosa de la Vía del Complemento/inmunología , Proteínas Recombinantes de Fusión , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Células CHO , Proteína de Unión al Complemento C4b/química , Proteína de Unión al Complemento C4b/genética , Proteína de Unión al Complemento C4b/inmunología , Cricetulus , Ensayo de Inmunoadsorción Enzimática , Humanos
6.
Biomed Chromatogr ; 32(5): e4180, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29265394

RESUMEN

Serum levels of fully sialylated C4-binding protein (FS-C4BP) are remarkably elevated in patients with epithelial ovarian cancer (EOC) and can be used as a marker to distinguish ovarian clear cell carcinoma from endometrioma. This study aimed to develop a stable, robust and reliable liquid chromatography-hybrid mass spectrometry (UPLC-MS/MS) based diagnostic method that would generalize FS-C4BP as a clinical EOC biomarker. Glycopeptides derived from 20 µL of trypsin-digested serum glycoprotein were analyzed via UPLC equipped with an electrospray ionization time-of-flight mass spectrometer. This UPLC-MS/MS-based diagnostic method was optimized for FS-C4BP and validated using sera from 119 patients with EOC and 127 women without cancer. A1958 (C4BP peptide with two fully sialylated biantennary glycans) was selected as a marker of FS-C4BP because its level in serum was highest among FS-C4BP family members. Preparation and UPLC-MS/MS were optimized for A1958, and performance and robustness were significantly improved relative to our previous method. An area under the curve analysis of the FS-C4BP index receiver operating characteristic curve revealed that the ratio between A1958 and A1813 (C4BP peptide with two partially sialylated biantennary glycans) reached 85%. A combination of the FS-C4BP index and carbohydrate antigen-125 levels further enhanced the sensitivity and specificity.


Asunto(s)
Biomarcadores de Tumor/sangre , Cromatografía Líquida de Alta Presión/métodos , Proteína de Unión al Complemento C4b/análisis , Neoplasias Glandulares y Epiteliales/sangre , Neoplasias Glandulares y Epiteliales/diagnóstico , Neoplasias Ováricas/sangre , Neoplasias Ováricas/diagnóstico , Espectrometría de Masas en Tándem/métodos , Anciano , Carcinoma Epitelial de Ovario , Proteína de Unión al Complemento C4b/química , Femenino , Humanos , Persona de Mediana Edad , Ácido N-Acetilneuramínico/química , Reproducibilidad de los Resultados
7.
Bioconjug Chem ; 28(5): 1544-1551, 2017 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-28437083

RESUMEN

Engineering modular platforms to control biomolecular architecture can advance both the understanding and the manipulation of biological systems. Icosahedral particles uniformly displaying single antigens stimulate potent immune activation and have been successful in various licensed vaccines. However, it remains challenging to display multiple antigens on a single particle and to induce broader immunity protective across strains or even against distinct diseases. Here, we design a dually addressable synthetic nanoparticle by engineering the multimerizing coiled-coil IMX313 and two orthogonally reactive split proteins. SpyCatcher protein forms an isopeptide bond with SpyTag peptide through spontaneous amidation. SnoopCatcher forms an isopeptide bond with SnoopTag peptide through transamidation. SpyCatcher-IMX-SnoopCatcher provides a modular platform, whereby SpyTag-antigen and SnoopTag-antigen can be multimerized on opposite faces of the particle simply upon mixing. We demonstrate efficient derivatization of the platform with model proteins and complex pathogen-derived antigens. SpyCatcher-IMX-SnoopCatcher was expressed in Escherichia coli and was resilient to lyophilization or extreme temperatures. For the next generation of malaria vaccines, blocking the transmission of the parasite from human to mosquito is an important goal. SpyCatcher-IMX-SnoopCatcher multimerization of the leading transmission-blocking antigens Pfs25 and Pfs28 greatly enhanced the antibody response to both antigens in comparison to the monomeric proteins. This dual plug-and-display architecture should help to accelerate vaccine development for malaria and other diseases.


Asunto(s)
Proteína de Unión al Complemento C4b/inmunología , Proteínas de Escherichia coli/inmunología , Proteínas Protozoarias/inmunología , Animales , Proteína de Unión al Complemento C4b/química , Proteínas de Escherichia coli/química , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Proteínas Protozoarias/química , Vacunación , Vacunas Sintéticas/química , Vacunas Sintéticas/inmunología
8.
PLoS One ; 12(1): e0168814, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28125581

RESUMEN

The surface protein SdrE, a microbial surface components recognizing adhesive matrix molecule (MSCRAMM) family protein expressed on the surface of Staphylococcus aureus (S. aureus), can recognize human complement regulator Factor H and C4BP, thus making it a potentially promising vaccine candidate. In this study, SdrE278-591 was found to directly affect S. aureus host cell invasion. Additionally, the crystal structure of SdrE278-591 at a resolution of 1.25 Å was established, with the three-dimensional structure revealing N2-N3 domains which fold in a manner similar to an IgG fold. Furthermore, a putative ligand binding site located at a conserved charged groove formed by the interface between N2 and N3 domains was identified, with ß2 suspected to occupy the ligand recognizing site and undergo a structural rearrangement to allow ligand binding. Overall, these findings have further contributed to the understanding of SdrE as a key factor for S. aureus invasivity and will enable a better understanding of bacterial infection processes.


Asunto(s)
Proteínas Bacterianas/química , Proteína de Unión al Complemento C4b/química , Factor H de Complemento/química , Mutación , Staphylococcus aureus/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Sitios de Unión , Línea Celular Tumoral , Clonación Molecular , Proteína de Unión al Complemento C4b/genética , Proteína de Unión al Complemento C4b/inmunología , Factor H de Complemento/genética , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/genética , Ligandos , Modelos Moleculares , Osteoblastos/inmunología , Osteoblastos/microbiología , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Staphylococcus aureus/patogenicidad
9.
Nat Microbiol ; 1(11): 16155, 2016 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-27595425

RESUMEN

No vaccine exists against group A Streptococcus (GAS), a leading cause of worldwide morbidity and mortality. A severe hurdle is the hypervariability of its major antigen, the M protein, with >200 different M types known. Neutralizing antibodies typically recognize M protein hypervariable regions (HVRs) and confer narrow protection. In stark contrast, human C4b-binding protein (C4BP), which is recruited to the GAS surface to block phagocytic killing, interacts with a remarkably large number of M protein HVRs (apparently ∼90%). Such broad recognition is rare, and we discovered a unique mechanism for this through the structure determination of four sequence-diverse M proteins in complexes with C4BP. The structures revealed a uniform and tolerant 'reading head' in C4BP, which detected conserved sequence patterns hidden within hypervariability. Our results open up possibilities for rational therapies that target the M-C4BP interaction, and also inform a path towards vaccine design.


Asunto(s)
Antígenos Bacterianos/química , Proteínas de la Membrana Bacteriana Externa/química , Proteínas Portadoras/química , Proteína de Unión al Complemento C4b/química , Secuencia de Aminoácidos , Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Sitios de Unión , Proteínas Portadoras/metabolismo , Proteína de Unión al Complemento C4b/metabolismo , Proteínas Inactivadoras de Complemento , Secuencia Conservada , Interacciones Huésped-Patógeno , Humanos
10.
Gynecol Oncol ; 139(3): 520-8, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26477941

RESUMEN

OBJECTIVE: While a certain fraction of endometriomas can develop de novo epithelial ovarian cancer (EOC) such as clear cell carcinoma (OCCC), there is currently no useful biomarker available for early detection of OCCC from endometriomas. The aim of this study was to describe the diagnostic utility of a novel biomarker for EOC especially for OCCC to distinguish from endometrioma. METHODS: More than 100,000 glycan structures of serum glycoproteins obtained from 134 pretreatment all stage EOC patients (including 45 OCCCs) and 159 non-cancer control women (including 36 endometriomas) were explored for a mass spectrum approach. Diagnostic accuracy of identified biomarker was compared to the one of CA-125 by comparing area under curve (AUC) and positive/negative predictive values (PPV and NPV). RESULTS: A2160, a fully-sialylated alpha-chain of complement 4-binding protein, was identified as a candidate target marker. A2160 was significantly elevated in all stages of OCCC compared to with endometriomas. Diagnostic accuracy of A2160 (cutoff 1.6U/mL) to distinguish early stage OCCC from endometrioma is significantly higher than that of CA-125 (cutoff 35IU/L): AUC for A2160 versus CA-125, 0.92 versus 0.67; PPV 95% versus 64%; and NPV 85% versus 58%. In addition, fully-sialylated glycans had a higher accuracy for diagnosing EOC as compared to partially-sialylated glycans of alpha-chain of complement 4-binding protein. CONCLUSION: Our study suggested that A2160 may be a useful biomarker to distinguish early-stage OCCC from endometrioma. This new biomarker can be potentially applied for the monitoring of endometrioma patients, making possible the early diagnosis of OCCC.


Asunto(s)
Adenocarcinoma de Células Claras/sangre , Adenocarcinoma de Células Claras/patología , Biomarcadores de Tumor/metabolismo , Proteína de Unión al Complemento C4b/metabolismo , Glicopéptidos/sangre , Neoplasias Ováricas/sangre , Neoplasias Ováricas/patología , Adulto , Anciano , Área Bajo la Curva , Biomarcadores de Tumor/química , Antígeno Ca-125/sangre , Estudios de Casos y Controles , Cromatografía Liquida , Proteína de Unión al Complemento C4b/química , Endometriosis/sangre , Femenino , Humanos , Espectrometría de Masas , Persona de Mediana Edad , Estadificación de Neoplasias , Valor Predictivo de las Pruebas , Curva ROC
11.
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
12.
Eur J Immunol ; 44(3): 742-51, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24765656

RESUMEN

The outer membrane protein Ail of Yersinia pestis mediates several virulence functions, including serum resistance. Here, we demonstrate that Ail binds C4b-binding protein (C4BP), the primary fluid-phase regulator of the classical and lectin pathways. Noncovalent binding of C4 and C4b to Ail was also observed. C4BP bound to Ail can act as a cofactor to the serine protease factor I (fI) in the cleavage of fluid-phase C4b. Employing a panel of C4BP alpha-chain mutants, we observed that the absence of complement control protein domain 6 and 8 reduced binding to Ail. Immunoblot analysis of normal human serum (NHS)-treated bacteria revealed minimal C4b alpha'-chain complexes with bacterial outer membrane targets. Addition of the anti-C4BP monoclonal antibody MK104 to NHS restored C4b-alpha' chain target complexes, suggesting that C4b binds covalently to targets on the Y. pestis surface. C4b bound to Ail noncovalently was also cleaved in a C4BP and fI-dependent manner, leaving the C4c fragment bound to Ail. MK104 also prevented the cleavage of noncovalently bound C4b. Collectively, these data suggest that when C4BP is bound to Ail, fI can cleave and inactivate C4b that has bound covalently to bacterial surface structures as well as C4b bound noncovalently to Ail.


Asunto(s)
Activación de Complemento/inmunología , Proteína de Unión al Complemento C4b/metabolismo , Complemento C4b/inmunología , Complemento C4b/metabolismo , Fibrinógeno/metabolismo , Peste/inmunología , Peste/metabolismo , Yersinia pestis/inmunología , Sitios de Unión , Complemento C4b/química , Proteína de Unión al Complemento C4b/química , Vía Clásica del Complemento/inmunología , Humanos , Cinética , Unión Proteica/inmunología
13.
Biomol NMR Assign ; 8(1): 1-6, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23138856

RESUMEN

The complement 4 binding protein (C4bp) plays a crucial role in the inhibition of the complement cascade. It has an extraordinary seven-arm octopus-like structure with 7 tentacle-like identical chains, held together at their C-terminal end. The C-terminal domain does oligomerize in isolation, and is necessary and sufficient to oligomerize full-length C4bp. It is predicted to form a seven-helix coiled coil, and its multimerization properties make it a promising vaccine adjuvant, probably by enhancing the structural stability and binding affinity of the presented antigen. Here, we present the solid-state NMR resonance assignment of the human C4bp C-terminal oligomerization Domain, hC4pbOD, and the corresponding secondary chemical shifts.


Asunto(s)
Proteína de Unión al Complemento C4b/química , Resonancia Magnética Nuclear Biomolecular , Multimerización de Proteína , Secuencia de Aminoácidos , Humanos , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
14.
Biologicals ; 41(3): 176-83, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23410583

RESUMEN

Nanofiltration assures that protein therapeutics are free of adventitious agents such as viruses. Nanofilter pores must allow passage of protein drugs but be small enough to retain viruses. Five nanofilters have been evaluated to identify those that can be used interchangeably to yield a high purity Coagulation Factor IX product. When product preparations prior to nanofiltration were analyzed using electrophoresis, Western blot, liquid chromatography - tandem mass spectrometry and size exclusion HPLC, factor IX, inter - α - trypsin inhibitor and C4b binding protein (C4BP) were observed. C4BP was removed from product by all five nanofilters when nanofiltration was performed at physiological ionic strength. However, at high ionic strength, C4BP was removed by only two nanofilters. HPLC indicated that the Stokes radius of C4BP was larger at low ionic strength than at high ionic strength. The results suggest that C4BP exists in an open conformation at physiological ionic strength and is removed by nanofiltration whereas, at high ionic strength, the protein collapses to an extent that allows passage through some nanofilters. Manufacturers should be aware that protein contaminants in other nanofiltered protein drugs could behave similarly and conditions of nanofiltration must be evaluated to ensure consistent product purity.


Asunto(s)
Factor IX/química , Factor IX/aislamiento & purificación , Ultrafiltración/instrumentación , Ultrafiltración/métodos , Western Blotting , Cromatografía en Gel , Cromatografía Liquida , Proteína de Unión al Complemento C4b/química , Proteína de Unión al Complemento C4b/aislamiento & purificación , Proteína de Unión al Complemento C4b/metabolismo , Factor IX/metabolismo , Humanos , Concentración Osmolar , Tamaño de la Partícula , Porosidad , Espectrometría de Masas en Tándem , Inhibidores de Tripsina/química , Inhibidores de Tripsina/aislamiento & purificación , Inhibidores de Tripsina/metabolismo
15.
J Immunol ; 190(6): 2857-72, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23390292

RESUMEN

The classical pathway complement regulator C4b-binding protein (C4BP) is composed of two polypeptides (α- and ß-chains), which form three plasma oligomers with different subunit compositions (α7ß1, α7ß0, and α6ß1). We show in this article that the C4BP α7ß0 isoform (hereafter called C4BP[ß(-)] [C4BP lacking the ß-chain]), overexpressed under acute-phase conditions, induces a semimature, tolerogenic state on human monocyte-derived dendritic cells (DCs) activated by a proinflammatory stimulus. C4BP isoforms containing ß-chain (α7ß1 and α6ß1; C4BP[ß(+)]) neither interfered with the normal maturation of DCs nor competed with C4BP(ß(-)) activity on these cells. Immature DCs (iDCs) treated with C4BP(ß(-)) retained high endocytic activity, but, upon LPS treatment, they did not upregulate surface expression of CD83, CD80, and CD86. Transcriptional profiling of these semimature DCs revealed that treatment with C4BP(ß(-)) prevented the induction of IDO and BIC-1, whereas TGF-ß1 expression was maintained to the level of iDCs. C4BP(ß(-))-treated DCs were also unable to release proinflammatory Th1 cytokines (IL-12, TNF-α, IFN-γ, IL-6, IL-8) and, conversely, increased IL-10 secretion. They prevented surface CCR7 overexpression and, accordingly, displayed reduced chemotaxis, being morphologically indistinguishable from iDCs. Moreover, C4BP(ß(-))-treated DCs failed to enhance allogeneic T cell proliferation, impairing IFN-γ production in these cells and, conversely, promoting CD4(+)CD127(low/neg)CD25(high)Foxp3(+) T cells. Deletion mutant analysis revealed that the complement control protein-6 domain of the α-chain is necessary for the tolerogenic activity of C4BP(ß(-)). Our data demonstrate a novel anti-inflammatory and immunomodulatory function of the complement regulator C4BP, suggesting a relevant role of the acute-phase C4BP(ß(-)) isoform in a number of pathophysiological conditions and potential applications in autoimmunity and transplantation.


Asunto(s)
Antiinflamatorios no Esteroideos/química , Diferenciación Celular/inmunología , Proteína de Unión al Complemento C4b/fisiología , Células Dendríticas/química , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad/fisiología , Diferenciación Celular/genética , Proteína de Unión al Complemento C4b/química , Proteína de Unión al Complemento C4b/genética , Células Dendríticas/patología , Células HEK293 , Antígenos de Histocompatibilidad/química , Antígenos de Histocompatibilidad/genética , Humanos , Tolerancia Inmunológica/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/prevención & control , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología
16.
J Mol Biol ; 425(8): 1302-17, 2013 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-23274142

RESUMEN

The complement system as a major part of innate immunity is the first line of defense against invading microorganisms. Orchestrated by more than 60 proteins, its major task is to discriminate between host cells and pathogens and to initiate immune response. Additional recognition of necrotic or apoptotic cells demands a fine-tune regulation of this powerful system. C4b-binding protein (C4BP) is the major inhibitor of the classical complement and lectin pathway. The crystal structure of the human C4BP oligomerization domain in its 7α isoform and molecular simulations provide first structural insights of C4BP oligomerization. The heptameric core structure is stabilized by intermolecular disulfide bonds. In addition, thermal shift assays indicate that layers of electrostatic interactions mainly contribute to the extraordinary thermodynamic stability of the complex. These findings make C4BP a promising scaffold for multivalent ligand display with applications in immunology and biological chemistry.


Asunto(s)
Proteína de Unión al Complemento C4b/química , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Conformación Proteica , Multimerización de Proteína , Electricidad Estática , Termodinámica
17.
J Biol Chem ; 287(14): 10824-33, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22334700

RESUMEN

Islet amyloid polypeptide (IAPP) is synthesized in pancreatic ß-cells and co-secreted with insulin. Aggregation and formation of IAPP-amyloid play a critical role in ß-cell death in type 2 diabetic patients. Because Aß-fibrils in Alzheimer disease activate the complement system, we have here investigated specific interactions between IAPP and complement factors. IAPP fibrils triggered limited activation of complement in vitro, involving both the classical and the alternative pathways. Direct binding assays confirmed that IAPP fibrils interact with globular head domains of complement initiator C1q. Furthermore, IAPP also bound complement inhibitors factor H and C4b-binding protein (C4BP). Recombinant C4BP mutants were used to show that complement control protein (CCP) domains 8 and 2 of the α-chain were responsible for the strong, hydrophobic binding of C4BP to IAPP. Immunostaining of pancreatic sections from type 2 diabetic patients revealed the presence of complement factors in the islets and varying degree of co-localization between IAPP fibrils and C1q, C3d, as well as C4BP and factor H but not membrane attack complex. Furthermore, C4BP enhanced formation of IAPP fibrils in vitro. We conclude that C4BP binds to IAPP thereby limiting complement activation and may be enhancing formation of IAPP fibrils from cytotoxic oligomers.


Asunto(s)
Proteína de Unión al Complemento C4b/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/química , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Multimerización de Proteína , Amiloide/química , Amiloide/metabolismo , Animales , Proteína de Unión al Complemento C4b/química , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Páncreas/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Estructura Secundaria de Proteína , Transporte de Proteínas , Ratas
18.
J Proteomics ; 75(6): 1924-32, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22266103

RESUMEN

Cationic liposomes (CLs) are one of the most employed nonviral nanovector systems in gene therapy. However, their transfection efficiency is strongly affected by interactions with plasma components, that lead to the formation of a "protein corona" onto CL surface. The interactions between nanoparticles entering the body and biomolecules have an essential role for their biodistribution. Because the knowledge of proteins adsorbed onto vector surface could be useful in the screening of new, more efficient and more biocompatible liposomal formulations, the behavior of three CLs with different membrane charge densities was investigated. The proteins of the three coronas were identified by nano-liquid chromatography-tandem mass spectrometry, and quantified with label-free spectral counting strategy. Fibrinogen displayed higher association with CLs with high membrane charge density, while apolipoproteins and C4b-binding protein with CLs with low membrane charge density. These results are discussed in terms of the different lipid compositions of CLs and may have a deep biological impact for in vivo applications. Surface charge of nanoparticles is emerging as a relevant factor determining the corona composition after interaction with plasma proteins. Remarkably, it is also shown that the charge of the protein corona formed around CLs is strongly related to their membrane charge density.


Asunto(s)
Proteínas Sanguíneas/química , Cationes/química , Liposomas/química , Nanopartículas/administración & dosificación , Propiedades de Superficie , Adsorción , Adulto , Apolipoproteínas/química , Proteína de Unión al Complemento C4b/química , Fibrinógeno/química , Humanos
19.
J Biol Chem ; 287(11): 8092-100, 2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22267731

RESUMEN

PRELP is a 58-kDa proteoglycan found in a variety of extracellular matrices, including cartilage and at several basement membranes. In rheumatoid arthritis (RA), the cartilage tissue is destroyed and fragmented molecules, including PRELP, are released into the synovial fluid where they may interact with components of the complement system. In a previous study, PRELP was found to interact with the complement inhibitor C4b-binding protein, which was suggested to locally down-regulate complement activation in joints during RA. Here we show that PRELP directly inhibits all pathways of complement by binding C9 and thereby prevents the formation of the membrane attack complex (MAC). PRELP does not interfere with the interaction between C9 and already formed C5b-8, but inhibits C9 polymerization thereby preventing formation of the lytic pore. The alternative pathway is moreover inhibited already at the level of C3-convertase formation due to an interaction between PRELP and C3. This suggests that PRELP may down-regulate complement attack at basement membranes and on damaged cartilage and therefore limit pathological complement activation in inflammatory disease such as RA. The net outcome of PRELP-mediated complement inhibition will highly depend on the local concentration of other complement modulating molecules as well as on the local concentration of available complement proteins.


Asunto(s)
Artritis Reumatoide/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Membrana Basal/química , Membrana Basal/metabolismo , Membrana Basal/patología , Activación de Complemento/genética , Convertasas de Complemento C3-C5/química , Convertasas de Complemento C3-C5/genética , Convertasas de Complemento C3-C5/metabolismo , Proteína de Unión al Complemento C4b/química , Proteína de Unión al Complemento C4b/genética , Proteína de Unión al Complemento C4b/metabolismo , Complemento C9/química , Complemento C9/genética , Complemento C9/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/química , Complejo de Ataque a Membrana del Sistema Complemento/genética , Proteínas del Sistema Complemento/química , Proteínas del Sistema Complemento/genética , Proteínas del Sistema Complemento/metabolismo , Regulación hacia Abajo/genética , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/genética , Femenino , Glicoproteínas/química , Glicoproteínas/genética , Células HEK293 , Humanos , Masculino
20.
PLoS One ; 6(8): e23991, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21915248

RESUMEN

The long pentraxin 3 (PTX3) is a soluble recognition molecule with multiple functions including innate immune defense against certain microbes and the clearance of apoptotic cells. PTX3 interacts with recognition molecules of the classical and lectin complement pathways and thus initiates complement activation. In addition, binding of PTX3 to the alternative complement pathway regulator factor H was shown. Here, we show that PTX3 binds to the classical and lectin pathway regulator C4b-binding protein (C4BP). A PTX3-binding site was identified within short consensus repeats 1-3 of the C4BP α-chain. PTX3 did not interfere with the cofactor activity of C4BP in the fluid phase and C4BP maintained its complement regulatory activity when bound to PTX3 on surfaces. While C4BP and factor H did not compete for PTX3 binding, the interaction of C4BP with PTX3 was inhibited by C1q and by L-ficolin. PTX3 bound to human fibroblast- and endothelial cell-derived extracellular matrices and recruited functionally active C4BP to these surfaces. Whereas PTX3 enhanced the activation of the classical/lectin pathway and caused enhanced C3 deposition on extracellular matrix, deposition of terminal pathway components and the generation of the inflammatory mediator C5a were not increased. Furthermore, PTX3 enhanced the binding of C4BP to late apoptotic cells, which resulted in an increased rate of inactivation of cell surface bound C4b and a reduction in the deposition of C5b-9. Thus, in addition to complement activators, PTX3 interacts with complement inhibitors including C4BP. This balanced interaction on extracellular matrix and on apoptotic cells may prevent excessive local complement activation that would otherwise lead to inflammation and host tissue damage.


Asunto(s)
Proteína C-Reactiva/metabolismo , Proteína de Unión al Complemento C4b/metabolismo , Componente Amiloide P Sérico/metabolismo , Apoptosis/genética , Apoptosis/fisiología , Sitios de Unión , Proteína C-Reactiva/química , Calcio/metabolismo , Células Cultivadas , Proteína de Unión al Complemento C4b/química , Matriz Extracelular/metabolismo , Humanos , Células Jurkat/metabolismo , Unión Proteica , Componente Amiloide P Sérico/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...