Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 180
Filtrar
1.
J Biol Chem ; 300(5): 107236, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38552741

RESUMEN

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


Asunto(s)
Borrelia burgdorferi , Vía Clásica del Complemento , Borrelia burgdorferi/inmunología , Borrelia burgdorferi/metabolismo , Borrelia burgdorferi/genética , Vía Clásica del Complemento/inmunología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas de la Membrana Bacteriana Externa/química , Humanos , Lipoproteínas/metabolismo , Lipoproteínas/genética , Lipoproteínas/química , Lipoproteínas/inmunología , Complemento C1s/metabolismo , Complemento C1s/genética , Complemento C1s/química , Unión Proteica , Enfermedad de Lyme/inmunología , Enfermedad de Lyme/microbiología , Enfermedad de Lyme/metabolismo , Enfermedad de Lyme/genética , Complemento C1r/metabolismo , Complemento C1r/genética
2.
Mol Immunol ; 166: 29-38, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38218080

RESUMEN

C1s enzyme (active C1s) is a subunit of the complement C1 complex that cleaves low-density lipoprotein receptor-related proteins 5 and 6, leading to Wnt/ß-catenin pathway activation in some cell lines. Macrophages have two major functional polarization states (the classically activated M1 state and the alternatively activated M2 state) and play an essential role in atherosclerosis. An increasing amount of evidence suggests that canonical Wnt signaling is related to macrophage polarization. In this study, we explored the cytoprotective effects of C1s enzyme in macrophages. The results show that C1s enzyme activates canonical Wnt signaling in macrophages, exacerbates macrophage M2 polarization, and inhibits M1 polarization. Moreover, C1s enzyme reduces foam cell formation and simultaneously enhances efferocytosis. This study reveals a novel function of C1s enzyme in macrophages in the context of atherosclerosis.


Asunto(s)
Aterosclerosis , Complemento C1s , Macrófagos , Vía de Señalización Wnt , Humanos , Aterosclerosis/metabolismo , beta Catenina/metabolismo , Células Espumosas/metabolismo , Macrófagos/metabolismo , Complemento C1s/metabolismo
3.
Clin Immunol ; 251: 109629, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37149117

RESUMEN

The objective of this study was to characterize the complement-inhibiting activity of SAR445088, a novel monoclonal antibody specific for the active form of C1s. Wieslab® and hemolytic assays were used to demonstrate that SAR445088 is a potent, selective inhibitor of the classical pathway of complement. Specificity for the active form of C1s was confirmed in a ligand binding assay. Finally, TNT010 (a precursor to SAR445088) was assessed in vitro for its ability to inhibit complement activation associated with cold agglutinin disease (CAD). TNT010 inhibited C3b/iC3b deposition on human red blood cells incubated with CAD patient serum and decreased their subsequent phagocytosis by THP-1 cells. In summary, this study identifies SAR445088 as a potential therapeutic for the treatment of classical pathway-driven diseases and supports its continued assessment in clinical trials.


Asunto(s)
Anemia Hemolítica Autoinmune , Complemento C1s , Humanos , Complemento C1s/metabolismo , Activación de Complemento , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Inactivadores del Complemento/uso terapéutico , Vía Clásica del Complemento
4.
J Med Chem ; 66(9): 6354-6371, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37120845

RESUMEN

A novel series of non-amidine-based C1s inhibitors have been explored. Starting from high-throughput screening hit 3, isoquinoline was replaced with 1-aminophthalazine to enhance C1s inhibitory activity while exhibiting good selectivity against other serine proteases. We first disclose a crystal structure of a complex of C1s and a small-molecule inhibitor (4e), which guided structure-based optimization around the S2 and S3 sites to further enhance C1s inhibitory activity by over 300-fold. Improvement of membrane permeability by incorporation of fluorine at the 8-position of 1-aminophthalazine led to identification of (R)-8 as a potent, selective, orally available, and brain-penetrable C1s inhibitor. (R)-8 significantly inhibited membrane attack complex formation induced by human serum in a dose-dependent manner in an in vitro assay system, proving that selective C1s inhibition blocked the classical complement pathway effectively. As a result, (R)-8 emerged as a valuable tool compound for both in vitro and in vivo assessment.


Asunto(s)
Activación de Complemento , Complemento C1s , Humanos , Complemento C1s/química , Complemento C1s/metabolismo , Serina Endopeptidasas/metabolismo , Encéfalo/metabolismo
5.
Front Immunol ; 13: 1015128, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36275687

RESUMEN

The molecules of the complement system connect the effectors of innate and adaptive immunity and play critical roles in maintaining homeostasis. Among them, the C1 complex, composed of C1q, C1r, and C1s (C1qr2s2), is the initiator of the classical complement activation pathway. While deficiency of C1s is associated with early-onset systemic lupus erythematosus and increased susceptibility to bacteria infections, the gain-of- function variants of C1r and C1s may lead to periodontal Ehlers Danlos syndrome. As C1s is activated under various pathological conditions and associated with inflammation, autoimmunity, and cancer development, it is becoming an informative biomarker for the diagnosis and treatment of a variety of diseases. Thus, more sensitive and convenient methods for assessing the level as well as activity of C1s in clinic samples are highly desirable. Meanwhile, a number of small molecules, peptides, and monoclonal antibodies targeting C1s have been developed. Some of them are being evaluated in clinical trials and one of the antibodies has been approved by US FDA for the treatment of cold agglutinin disease, an autoimmune hemolytic anemia. In this review, we will summarize the biological properties of C1s, its association with development and diagnosis of diseases, and recent progress in developing drugs targeting C1s. These progress illustrate that the C1s molecule is an effective biomarker and promising drug target.


Asunto(s)
Complemento C1r , Complemento C1s , Complemento C1s/metabolismo , Complemento C1q/metabolismo , Activación de Complemento/fisiología , Péptidos , Anticuerpos Monoclonales
6.
J Biol Chem ; 298(11): 102557, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36183830

RESUMEN

Proteolytic cascades comprise several important physiological systems, including a primary arm of innate immunity called the complement cascade. To safeguard against complement-mediated attack, the etiologic agent of Lyme disease, Borreliella burgdorferi, produces numerous outer surface-localized lipoproteins that contribute to successful complement evasion. Recently, we discovered a pair of B. burgdorferi surface lipoproteins of the OspEF-related protein family-termed ElpB and ElpQ-that inhibit antibody-mediated complement activation. In this study, we investigate the molecular mechanism of ElpB and ElpQ complement inhibition using an array of biochemical and biophysical approaches. In vitro assays of complement activation show that an independently folded homologous C-terminal domain of each Elp protein maintains full complement inhibitory activity and selectively inhibits the classical pathway. Using binding assays and complement component C1s enzyme assays, we show that binding of Elp proteins to activated C1s blocks complement component C4 cleavage by competing with C1s-C4 binding without occluding the active site. C1s-mediated C4 cleavage is dependent on activation-induced binding sites, termed exosites. To test whether these exosites are involved in Elp-C1s binding, we performed site-directed mutagenesis, which showed that ElpB and ElpQ binding require C1s residues in the anion-binding exosite located on the serine protease domain of C1s. Based on these results, we propose a model whereby ElpB and ElpQ exploit activation-induced conformational changes that are normally important for C1s-mediated C4 cleavage. Our study expands the known complement evasion mechanisms of microbial pathogens and reveals a novel molecular mechanism for selective C1s inhibition by Lyme disease spirochetes.


Asunto(s)
Borrelia burgdorferi , Enfermedad de Lyme , Humanos , Complemento C1s/química , Complemento C1s/metabolismo , Borrelia burgdorferi/genética , Complemento C4/química , Proteínas del Sistema Complemento/metabolismo , Serina Proteasas , Lipoproteínas/genética
7.
Int J Mol Sci ; 23(16)2022 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-36012546

RESUMEN

Autoantibodies against the complement component C1q (anti-C1q) are among the main biomarkers in lupus nephritis (LN) known to contribute to renal injury. C1q, the recognition subcomponent of the complement classical pathway, forms a heterotetrameric complex with C1r and C1s, and can also associate a central complement regulator and C1 Inhibitor (C1-Inh). However, the frequency and the pathogenic relevance of anti-C1r, anti-C1s and anti-C1-Inh autoantibodies remain poorly studied in LN. In this paper, we screened for anti-C1q, anti-C1r, anti-C1s and anti-C1-Inh autoantibodies and evaluated their association with disease activity and severity in 74 LN patients followed up for 5 years with a total of 266 plasma samples collected. The presence of anti-C1q, anti-C1r, anti-C1s and anti-C1-Inh was assessed by ELISA. IgG was purified by Protein G from antigen-positive plasma and their binding to purified C1q, C1r and C1s was examined by surface plasmon resonance (SPR). The abilities of anti-C1q, anti-C1r and anti-C1s binding IgG on C1 complex formation were analyzed by ELISA. The screening of LN patients' plasma revealed 14.9% anti-C1q positivity; only 4.2%, 6.9% and 0% were found to be positive for anti-C1r, anti-C1s and anti-C1-Inh, respectively. Significant correlations were found between anti-C1q and anti-dsDNA, and anti-nuclear antibodies, C3 and C4, respectively. High levels of anti-C1q antibodies were significantly associated with renal histologic lesions and correlated with histological activity index. Patients with the most severe disease (A class according to BILAG Renal score) had higher levels of anti-C1q antibodies. Anti-C1r and anti-C1s antibodies did not correlate with the clinical characteristics of the LN patients, did not interfere with the C1 complex formation, and were not measurable via SPR. In conclusion, the presence of anti-C1q, but not anti-C1s or anti-C1r, autoantibodies contribute to the autoimmune pathology and the severity of LN.


Asunto(s)
Complemento C1r , Nefritis Lúpica , Autoanticuerpos , Activación de Complemento , Complemento C1q/metabolismo , Complemento C1r/genética , Complemento C1s/metabolismo , Humanos , Inmunoglobulina G
8.
Drugs ; 82(7): 817-823, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35412113

RESUMEN

Sutimlimab (sutimlimab-jome; ENJAYMO™) is a humanized monoclonal antibody developed by Sanofi for the treatment of cold agglutinin disease (CAD). Sutimlimab is an immunoglobulin G, subclass 4 (IgG4) monoclonal antibody that inhibits the classical complement pathway by binding to complement protein component 1, s subcomponent (C1s), a serine protease which cleaves C4 and C2 to form the C3 convertase. Inhibition of the classical complement pathway at the level of C1s prevents deposition of complement opsonins on the surface of red blood cell (RBCs), leading to inhibition of haemolysis in patients with CAD. In February 2022, sutimlimab received its first approval in the USA to decrease the need for RBC transfusion due to haemolysis in adults with CAD. Sutimlimab is under regulatory review in Japan and the EU for CAD. This article summarizes the milestones in the development of sutimlimab leading to this first approval for CAD.


Asunto(s)
Anemia Hemolítica Autoinmune , Complemento C1s , Adulto , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Complemento C1s/metabolismo , Hemólisis , Humanos
9.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34155115

RESUMEN

Complement is an important effector mechanism for antibody-mediated clearance of infections and tumor cells. Upon binding to target cells, the antibody's constant (Fc) domain recruits complement component C1 to initiate a proteolytic cascade that generates lytic pores and stimulates phagocytosis. The C1 complex (C1qr2s2) consists of the large recognition protein C1q and a heterotetramer of proteases C1r and C1s (C1r2s2). While interactions between C1 and IgG-Fc are believed to be mediated by the globular heads of C1q, we here find that C1r2s2 proteases affect the capacity of C1q to form an avid complex with surface-bound IgG molecules (on various 2,4-dinitrophenol [DNP]-coated surfaces and pathogenic Staphylococcus aureus). The extent to which C1r2s2 contributes to C1q-IgG stability strongly differs between human IgG subclasses. Using antibody engineering of monoclonal IgG, we reveal that hexamer-enhancing mutations improve C1q-IgG stability, both in the absence and presence of C1r2s2 In addition, hexamer-enhanced IgGs targeting S. aureus mediate improved complement-dependent phagocytosis by human neutrophils. Altogether, these molecular insights into complement binding to surface-bound IgGs could be important for optimal design of antibody therapies.


Asunto(s)
Membrana Celular/metabolismo , Complemento C1q/metabolismo , Complemento C1r/metabolismo , Complemento C1s/metabolismo , Inmunoglobulina G/metabolismo , Activación de Complemento , Humanos , Microscopía de Fuerza Atómica , Mutación/genética , Fagocitosis , Unión Proteica , Multimerización de Proteína , Estabilidad Proteica , Staphylococcus aureus/inmunología
10.
Sci Rep ; 11(1): 11926, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099740

RESUMEN

Endometriosis (EMS) is a disease that shows immune dysfunction and chronic inflammation characteristics, suggesting a role of complement system in its pathophysiology. To find out the hub genes and pathways involved in the pathogenesis of EMs, three raw microarray datasets were recruited from the Gene Expression Omnibus database (GEO). Then, a series of bioinformatics technologies including gene ontology (GO), Hallmark pathway enrichment, protein-protein interaction (PPI) network and gene co-expression correlation analysis were performed to identify hub genes. The hub genes were further verified by the Real-time quantitative polymerase chain reaction (RT-PCR) and Western Blot (WB). We identified 129 differentially expressed genes (DEGs) in EMs, of which 78 were up-regulated and 51 were down-regulated. Through GO functional enrichment analysis, we found that the DEGs are mainly enriched in cell adhesion, extracellular matrix remodeling, chemokine regulation, angiogenesis regulation, epithelial cell proliferation, et al. In Hallmark pathway enrichment analysis, coagulation pathway showed great significance and the terms in which included the central complement factors. Moreover, the genes were dominating in PPI network. Combined co-expression analysis with experimental verification, we found that the up-regulated expression of complement (C1S, C1QA, C1R, and C3) was positively related to tissue factor (TF) in EMs. In this study, we discovered the over expression complement and the positive correlation between complement and TF in EMs, which suggested that interaction of complement and coagulation system may play a role within the pathophysiology of EMS.


Asunto(s)
Factores de Coagulación Sanguínea/genética , Proteínas del Sistema Complemento/genética , Endometriosis/genética , Perfilación de la Expresión Génica/métodos , Factores de Coagulación Sanguínea/metabolismo , Complemento C1q/genética , Complemento C1q/metabolismo , Complemento C1r/genética , Complemento C1r/metabolismo , Complemento C1s/genética , Complemento C1s/metabolismo , Complemento C3/genética , Complemento C3/metabolismo , Proteínas del Sistema Complemento/metabolismo , Endometriosis/metabolismo , Femenino , Ontología de Genes , Redes Reguladoras de Genes , Humanos , Mapas de Interacción de Proteínas/genética , Transducción de Señal/genética , Tromboplastina/genética , Tromboplastina/metabolismo
11.
Cancer Immunol Res ; 9(8): 891-908, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34039653

RESUMEN

The complement system plays a complex role in cancer. In clear cell renal cell carcinoma (ccRCC), local production of complement proteins drives tumor progression, but the mechanisms by which they do this are poorly understood. We found that complement activation, as reflected by high plasma C4d or as C4d deposits at the tumor site, was associated with poor prognosis in two cohorts of patients with ccRCC. High expression of the C4-activating enzyme C1s by tumor cells was associated with poor prognosis in three cohorts. Multivariate Cox analysis revealed that the prognostic value of C1s was independent from complement deposits, suggesting the possibility of complement cascade-unrelated, protumoral functions for C1s. Silencing of C1s in cancer cell lines resulted in decreased proliferation and viability of the cells and in increased activation of T cells in in vitro cocultures. Tumors expressing high levels of C1s showed high infiltration of macrophages and T cells. Modification of the tumor cell phenotype and T-cell activation were independent of extracellular C1s levels, suggesting that C1s was acting in an intracellular, noncanonical manner. In conclusion, our data point to C1s playing a dual role in promoting ccRCC progression by triggering complement activation and by modulating the tumor cell phenotype and tumor microenvironment in a complement cascade-independent, noncanonical manner. Overexpression of C1s by tumor cells could be a new escape mechanism to promote tumor progression.See related Spotlight by Magrini and Garlanda, p. 855. See article by Daugan et al., p. 909 (40).


Asunto(s)
Biomarcadores de Tumor/metabolismo , Complemento C1s/metabolismo , Complemento C4/metabolismo , Neoplasias Renales/genética , Animales , Estudios de Casos y Controles , Humanos , Ratones , Pronóstico , Estudios Prospectivos , Transfección
13.
Front Immunol ; 11: 583754, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193398

RESUMEN

LRP1 is a large endocytic modular receptor that plays a crucial role in the scavenging of apoptotic material through binding to pattern-recognition molecules. It is a membrane anchored receptor of the LDL receptor family with 4 extracellular clusters of ligand binding modules called cysteine rich complement-type repeats that are involved in the interaction of LRP1 with its numerous ligands. Complement C1q was shown to interact with LRP1 and to be implicated in the phagocytosis of apoptotic cells. The present work aimed at exploring how these two large molecules interact at the molecular level using a dissection strategy. For that purpose, recombinant LRP1 clusters II, III and IV were produced in mammalian HEK293F cells and their binding properties were investigated. Clusters II and IV were found to interact specifically and efficiently with C1q with K Ds in the nanomolar range. The use of truncated C1q fragments and recombinant mutated C1q allowed to localize more precisely the binding site for LRP1 on the collagen-like regions of C1q (CLRs), nearby the site that is implicated in the interaction with the cognate protease tetramer C1r2s2. This site could be a common anchorage for other ligands of C1q CLRs such as sulfated proteoglycans and Complement receptor type 1. The use of a cellular model, consisting in CHO LRP1-null cells transfected with full-length LRP1 or a cluster IV minireceptor (mini IV) confirmed that mini IV interacts with C1q at the cell membrane as well as full-length LRP1. Further cellular interaction studies finally highlighted that mini IV can endorse the full-length LRP1 binding efficiency for apoptotic cells and that C1q has no impact on this interaction.


Asunto(s)
Complemento C1q/metabolismo , Complemento C1r/metabolismo , Complemento C1s/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Péptido Hidrolasas/metabolismo , Animales , Apoptosis/fisiología , Sitios de Unión/fisiología , Células CHO , Línea Celular , Membrana Celular/metabolismo , Cricetulus , Células HEK293 , Humanos , Ligandos , Dominios Proteicos/fisiología
14.
Ann N Y Acad Sci ; 1478(1): 75-91, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32761624

RESUMEN

Neural tube defects (NTDs) are serious congenital malformations. In this study, we aimed to identify more specific and sensitive maternal serum biomarkers for noninvasive NTD screenings. We collected serum from 37 pregnant women carrying fetuses with NTDs and 38 pregnant women carrying normal fetuses. Isobaric tags for relative and absolute quantitation were conducted for differential proteomic analysis, and an enzyme-linked immunosorbent assay was used to validate the results. We then used a support vector machine (SVM) classifier to establish a disease prediction model for NTD diagnosis. We identified 113 differentially expressed proteins; of these, 23 were either up- or downregulated 1.5-fold or more, including five complement proteins (C1QA, C1S, C1R, C9, and C3); C3 and C9 were downregulated significantly in NTD groups. The accuracy rate of the SVM model of the complement factors (including C1QA, C1S, and C3) was 62.5%, with 60% sensitivity and 67% specificity, while the accuracy rate of the SVM model of alpha-fetoprotein (AFP, an established biomarker for NTDs) was 62.5%, with 75% sensitivity and 50% specificity. Combination of the complement factor and AFP data resulted in the SVM model accuracy of 75%, and receiver operating characteristic curve analysis showed 75% sensitivity and 75% specificity. These data suggest that a disease prediction model based on combined complement factor and AFP data could serve as a more accurate method of noninvasive prenatal NTD diagnosis.


Asunto(s)
Biomarcadores , Defectos del Tubo Neural/sangre , Defectos del Tubo Neural/diagnóstico , alfa-Fetoproteínas/metabolismo , Adulto , Biomarcadores/sangre , Complemento C1q/metabolismo , Complemento C1s/metabolismo , Complemento C3/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/patología , Pruebas Prenatales no Invasivas , Embarazo , Transcriptoma/genética
15.
Mol Immunol ; 126: 8-13, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32717572

RESUMEN

The serpin, C1-inhibitor (also known as SERPING1), plays a vital anti-inflammatory role in the body by controlling pro-inflammatory pathways such as complement and coagulation. The inhibitor's action is enhanced in the presence of polyanionic cofactors, such as heparin and polyphosphate, by increasing the rate of association with key enzymes such as C1s of the classical pathway of complement. The cofactor binding site of the serpin has never been mapped. Here we show that residues Lys284, Lys285 and Arg287 of C1-inhibitor play key roles in binding heparin and delivering the rate enhancement seen in the presence of polyanions and thus most likely represent the key cofactor binding residues for the serpin. We also show that simultaneous binding of the anion binding site of C1s by the polyanion is required to deliver the rate enhancement. Finally, we have shown that it is unlikely that the two positively charged zones of C1-inhibitor and C1s interact in the encounter complex between molecules as ablation of the charged zones did not in itself deliver a rate enhancement as might have been expected if the zones interacted. These insights provide crucial information as to the mechanism of action of this key serpin in the presence and absence of cofactor molecules.


Asunto(s)
Proteína Inhibidora del Complemento C1/metabolismo , Complemento C1s/antagonistas & inhibidores , Polímeros/metabolismo , Sitios de Unión/genética , Proteína Inhibidora del Complemento C1/genética , Proteína Inhibidora del Complemento C1/aislamiento & purificación , Complemento C1s/metabolismo , Heparina/metabolismo , Mutagénesis Sitio-Dirigida , Mutación , Polielectrolitos , Polifosfatos/metabolismo , Unión Proteica/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo
16.
Am J Physiol Renal Physiol ; 317(5): F1293-F1304, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31509012

RESUMEN

We have previously reported that complement activation precedes the development of kidney fibrosis; however, little is known about the cellular mechanisms involved in this transition. We hypothesized that increased expression of C1 complex protease C1r, the initiator of complement activation, contributes to tubulointerstitial fibrosis and tested this idea in mice with global deletion of C1r. Although expression of C1r in untreated wild-type (WT) mice was higher in the liver compared with kidney tissue, administration of folic acid (FA) led to upregulation of C1r mRNA and protein levels only in kidney tissue. Immunohistochemistry and in situ hybridization experiments localized increased expression of C1r and C1s proteases to renal tubular epithelial cells. C1r-null mice had reduced acute tubular injury and inflammation measured 2 days after FA administration compared with WT mice. C1r deletion reduced expression of C1s, C3 fragment formation, and organ fibrosis measured 14 days after FA administration. Differential gene expression performed in kidney tissue demonstrated that C1r-null mice had reduced expression of genes associated with the acute phase response, complement, proliferation of connective tissue cells (e.g., platelet-derived growth factor receptor-ß), and reduced expression of genes associated with inflammation compared with FA-treated WT mice. In vitro experiments in renal epithelial cells demonstrated that C1s expression is dependent on increased C1r expression and that interferon-γ induces the expression of these two proteases. We conclude that increased expression of C1 complex proteases is associated with increased tissue inflammation and complement C3 formation and represents an important pathogenic mechanism leading to FA-mediated tubulointerstitial fibrosis.


Asunto(s)
Complemento C1r/metabolismo , Enfermedades Renales/enzimología , Animales , Línea Celular , Complemento C1r/genética , Complemento C1s/genética , Complemento C1s/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Ácido Fólico/farmacología , Regulación Enzimológica de la Expresión Génica , Humanos , Inflamación , Riñón/citología , Enfermedades Renales/genética , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo
17.
Biotechnol Bioeng ; 116(9): 2130-2145, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31087560

RESUMEN

Proteolysis associated with recombinant protein expression in Chinese Hamster Ovary (CHO) cells has hindered the development of biologics including HIV vaccines. When expressed in CHO cells, the recombinant HIV envelope protein, gp120, undergoes proteolytic clipping by a serine protease at a key epitope recognized by neutralizing antibodies. The problem is particularly acute for envelope proteins from clade B viruses that represent the major genetic subtype circulating in much of the developed world, including the US and Europe. In this paper, we have identified complement Component 1's (C1s), a serine protease from the complement cascade, as the protease responsible for the proteolysis of gp120 in CHO cells. CRISPR/Cas9 knockout of the C1s protease in a CHO cell line was shown to eliminate the proteolytic activity against the recombinantly expressed gp120. In addition, the C1s-/- MGAT1- CHO cell line, with the C1s protease and the MGAT1 glycosyltransferase knocked out, enabled the production of unclipped gp120 from a clade B isolate (BaL-rgp120) and enriched for mannose-5 glycans on gp120 that are required for the binding of multiple broadly neutralizing monoclonal antibodies (bN-mAbs). The availability of this technology will allow for the scale-up and testing of multiple vaccine concepts in regions of the world where clade B viruses are in circulation. Furthermore, the proteolysis issues caused by the C1s protease suggests a broader need for a C1s-deficient CHO cell line to express other recombinant proteins that are susceptible to serine protease activity in CHO cells. Similarly, the workflow described here to identify and knockout C1s in a CHO cell line can be applied to remedy the proteolysis of biologics by other CHO proteases.


Asunto(s)
Sistemas CRISPR-Cas , Complemento C1s/genética , Complemento C1s/metabolismo , Técnicas de Inactivación de Genes , Proteína gp120 de Envoltorio del VIH/biosíntesis , VIH-1 , Proteolisis , Animales , Células CHO , Cricetulus , Proteína gp120 de Envoltorio del VIH/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética
18.
Acta Med Acad ; 47(1): 11-17, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29957967

RESUMEN

OBJECTIVE: Angioedema (AE) is a potentially life-threatening event. We investigated the etiology of AE, with the emphasis on bradykinininduced angioedema treatment in emergency medicine. METHODS: The retrospective study included 237 patients with AE, who were examined and treated in two hospitals (group A and B) in Croatia from 2009 to 2016. The location and duration of AE, data about chronic diseases and treatment, potential causative agents (food, drugs, insect bites and chemicals), physical examination data and the subsequent treatment were analyzed. RESULTS: There was no statistical difference regarding age or comorbidities but there was a statistically significant difference in etiology between the groups (Chi-square, P=0.03). Renin-angiotensin-aldosterone system (RAAS) blocker induced AE was the main cause of emergency attendance in group A (37.5%) and among the leading causes in group B (18.8%). Bradykinin-induced AE (hereditary angioedema (HAE) and RAAS-AE) were the leading causes in a total of 75 (31.5%) patients. RAAS-AE was treated with glucocorticoids and antihistamines. HAE attacks in both groups (2/7 patients, 1.5/6%) were treated with specific therapy. Other causes of AE in groups A/B were insect bites (15/23 patients, 13.5/20%), use of antibiotics/analgetics (11/17 patients, 9/15%), gastroesophageal reflux disease (10/11 patients, 8/9%), neoplasms (5/6 patients, 4/5%) and idiopatic (32/31 patients, 26.5/26%). 21% of patients were hospitalized. CONCLUSION: Bradykinin-mediated AE was the main cause of emergency attendance associated with AE. Advances in the treatment of HAE, with case reports of patients with RAAS-AE treated with C1 esterase inhibitor concentrate or bradykinin receptor antagonist, may prove to be a new, reliable and efficacious therapy option.


Asunto(s)
Angioedema/etiología , Bradiquinina/metabolismo , Complemento C1s/metabolismo , Medicina de Emergencia , Adulto , Anciano , Angioedema/tratamiento farmacológico , Angioedemas Hereditarios/epidemiología , Angioedemas Hereditarios/terapia , Bradiquinina/análogos & derivados , Bradiquinina/uso terapéutico , Antagonistas de los Receptores de Bradiquinina/uso terapéutico , Proteína Inhibidora del Complemento C1/uso terapéutico , Croacia , Femenino , Reflujo Gastroesofágico/complicaciones , Hospitalización , Hospitales , Humanos , Incidencia , Mordeduras y Picaduras de Insectos , Masculino , Persona de Mediana Edad , Neoplasias/complicaciones , Prevalencia , Sistema Renina-Angiotensina , Estudios Retrospectivos
19.
Proc Natl Acad Sci U S A ; 115(4): 768-773, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29311313

RESUMEN

The multiprotein complex C1 initiates the classical pathway of complement activation on binding to antibody-antigen complexes, pathogen surfaces, apoptotic cells, and polyanionic structures. It is formed from the recognition subcomponent C1q and a tetramer of proteases C1r2C1s2 as a Ca2+-dependent complex. Here we have determined the structure of a complex between the CUB1-EGF-CUB2 fragments of C1r and C1s to reveal the C1r-C1s interaction that forms the core of C1. Both fragments are L-shaped and interlock to form a compact antiparallel heterodimer with a Ca2+ from each subcomponent at the interface. Contacts, involving all three domains of each protease, are more extensive than those of C1r or C1s homodimers, explaining why heterocomplexes form preferentially. The available structural and biophysical data support a model of C1r2C1s2 in which two C1r-C1s dimers are linked via the catalytic domains of C1r. They are incompatible with a recent model in which the N-terminal domains of C1r and C1s form a fixed tetramer. On binding to C1q, the proteases become more compact, with the C1r-C1s dimers at the center and the six collagenous stems of C1q arranged around the perimeter. Activation is likely driven by separation of the C1r-C1s dimer pairs when C1q binds to a surface. Considerable flexibility in C1s likely facilitates C1 complex formation, activation of C1s by C1r, and binding and activation of downstream substrates C4 and C4b-bound C2 to initiate the reaction cascade.


Asunto(s)
Complemento C1r/metabolismo , Complemento C1s/metabolismo , Animales , Células CHO , Cricetulus , Dimerización , Dominios Proteicos
20.
Dev Comp Immunol ; 81: 279-290, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29247723

RESUMEN

C1r and C1s are serine proteases responsible for activating the classical complement pathway to initiate the complement cascade, which plays a crucial role in eliminating invading pathogenic microbes. In this study, cDNA sequences of C1r and C1s were identified from black rockfish and designated as SsC1r and SsC1s, respectively. In both sequences, two CUB domains, an EGF-like domain, two CCP domains, and a trypsin-like serine protease domain were identified. Multiple sequence alignments with known vertebrate homologs demonstrated that both sequences were highly conserved and, especially, the catalytic and substrate binding residues were completely conserved. In the constructed phylogenetic tree, C1r and C1s formed two separate clusters, which further branched into groups of related organisms. SsC1r and SsC1s joined with their respective teleostean clusters. Transcriptional analysis showed that the highest mRNA expression level was in the liver under normal physiological conditions. Significantly upregulated expression of both mRNAs in spleen and liver after pathologic stress, by intraperitoneal injection with different stimuli, suggested their vital role in immunity. The serine protease domains of SsC1r and SsC1s were cloned and the recombinant proteins were expressed and purified. A protease assay, conducted to confirm their functionality, indicated that both recombinant proteins had proteolytic activity. Taken together, these results indicate that SsC1r and SsC1s have significant properties to aid in the immunity of black rockfish by activating the complement system by proteolytic cleavage.


Asunto(s)
Complemento C1r/genética , Complemento C1s/genética , Proteínas de Peces/genética , Hígado/fisiología , Perciformes/inmunología , Animales , Clonación Molecular , Complemento C1r/metabolismo , Complemento C1s/metabolismo , Secuencia Conservada/genética , Proteínas de Peces/metabolismo , Inmunidad Innata , Perciformes/genética , Filogenia , Dominios Proteicos/genética , Proteolisis , Alineación de Secuencia , Transcriptoma , Vertebrados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...