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













Base de datos
Intervalo de año de publicación
1.
Front Biosci (Landmark Ed) ; 29(4): 138, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38682192

RESUMEN

BACKGROUND: Mounting evidence indicates that complement components play a crucial role in cancer progression. Recent findings indicate that certain complement components display a significant rise in expression within esophageal squamous cell carcinoma (ESCC). However, the specific tumorigenic functions of these components remain unclear. This study focuses on investigating the expression pattern of C1r, elucidating a role for C1r in ESCC, as well as exploring underlying mechanisms controlled by C1r. METHODS: The expression of C1r in ESCC tissues, malignant epithelial cells, and its relationship with survival were analyzed using the Gene Expression Omnibus (GEO) database and tissue microarrays. Single-cell RNA sequencing (scRNA-seq) was used to study the expression of C1r in malignant epithelial cells. C1r knockdown or C1r overexpression in cultured ESCC cells were used to assess the effects of C1r on proliferation, migration, invasion, cell-matrix adhesion, apoptosis, and growth of xenografted tumors in immunocompromised (nude) mice. Western blotting was used to detect the expression of MMP-1 and MMP-10 in C1r knockdown or C1r overexpressing ESCC cells. RESULTS: C1r was highly expressed in ESCC tissues, malignant epithelial cells, and cultured ESCC cell lines. High C1r expression indicated a poor prognosis. Knockdown of C1r significantly suppressed the proliferation, migration, invasion, cell-matrix adhesion, and promoted apoptosis in cultured ESCC cells. Additionally, knockdown of C1r markedly inhibited tumor growth in nude mice. Overexpression of C1r had the opposite effects. C1r induced the expression of MMP-1 and MMP-10. CONCLUSIONS: C1r is highly expressed in ESCC and promotes the progression of this tumor type. Our findings suggest that C1r may serve as a novel prognostic biomarker and therapeutic target in ESCC.


Asunto(s)
Biomarcadores de Tumor , Proliferación Celular , Complemento C1r , Progresión de la Enfermedad , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Ratones Desnudos , Humanos , Animales , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/metabolismo , Pronóstico , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/patología , Carcinoma de Células Escamosas de Esófago/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Complemento C1r/genética , Complemento C1r/metabolismo , Proliferación Celular/genética , Movimiento Celular/genética , Apoptosis/genética , Ratones , Masculino , Femenino , Regulación Neoplásica de la Expresión Génica , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 1 de la Matriz/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología
2.
J Proteome Res ; 23(4): 1232-1248, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38407963

RESUMEN

The aim of this study was to identify serum diagnostic biomarkers associated with the severity of obstructive sleep apnea (OSA) during pregnancy. Differentially expressed proteins (DEPs) were identified in the control (C), mild (O), and moderate (MO) OSA groups (n = 3 in each group). Bioinformatics analysis was conducted to identify the underlying functions, pathways, and networks of the proteins. Receiver operating characteristic curves were used to assess the diagnostic value of the identified DEPs. The enzyme-linked immunoassay was performed to detect serum levels of the complement C1r subcomponent (C1R) and alpha-2-macroglobulin (A2M) in 79 pregnant women with OSA (mild OSA [n = 32]; moderate OSA [n = 29], and severe OSA [n = 18]) and 65 healthy pregnant women without OSA. Pearson's correlation analysis was conducted to analyze the correlation between C1R and A2M levels and OSA clinicopathological factors. In total, 141 DEPs, 29 DEPs, and 103 DEPs were identified in the three groups (i.e., the mild OSA vs control group, the moderate OSA vs mild apnea group, and the moderate OSA vs control group, respectively). C1R and A2M were identified as continuously up-regulated proteins, and the levels of C1R and A2M were associated with OSA severity. C1R and A2M were found to be correlated with body mass index, systolic blood pressure, apnea-hypopnea index, oxygen desaturation index, time with saturation below 90%, and lowest SaO2. Adverse maternal and neonatal outcomes were observed in pregnant women with OSA. C1R and A2M have been identified as diagnostic biomarkers and are associated with the severity of OSA during pregnancy.


Asunto(s)
Mujeres Embarazadas , Apnea Obstructiva del Sueño , Femenino , Humanos , Recién Nacido , Embarazo , alfa-Macroglobulinas , Biomarcadores , Complemento C1r/metabolismo , Polisomnografía , Proteoma , Apnea Obstructiva del Sueño/diagnóstico , Apnea Obstructiva del Sueño/complicaciones , Factores de Transcripción
3.
J Invest Dermatol ; 142(5): 1478-1488.e9, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34756877

RESUMEN

Cutaneous squamous cell carcinoma (cSCC) is the most common metastatic skin cancer, with increasing incidence worldwide. Previous studies have shown the role of the complement system in cSCC progression. In this study, we have investigated the mechanistic role of serine proteinase C1r, a component of the classical pathway of the complement system, in cSCC. Knockout of C1r in cSCC cells using CRISPR/Cas9 resulted in a significant decrease in their proliferation, migration, and invasion through collagen type I compared with that of wild-type cSCC cells. Knockout of C1r suppressed the growth and vascularization of cSCC xenograft tumors and promoted apoptosis of tumor cells in vivo. mRNA-sequencing analysis after C1r knockdown revealed significantly regulated Gene Ontology terms cell-matrix adhesion, extracellular matrix component, basement membrane, and metalloendopeptidase activity and Kyoto Encyclopedia of Genes and Genomes pathway extracellular matrix‒receptor interaction. Among the significantly regulated genes were invasion-associated matrix metalloproteinases (MMPs) MMP1, MMP13, MMP10, and MMP12. Knockout of C1r resulted in decreased production of MMP-1, MMP-13, MMP-10, and MMP-12 by cSCC cells in culture. Knockout of C1r inhibited the expression of MMP-13 by tumor cells, suppressed invasion, and reduced the amount of degraded collagen in vivo in xenografts. These results provide evidence for the role of C1r in promoting the invasion of cSCC cells by increasing MMP production.


Asunto(s)
Carcinoma de Células Escamosas , Complemento C1r , Metaloproteinasa 13 de la Matriz , Neoplasias Cutáneas , Animales , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Complemento C1r/genética , Complemento C1r/metabolismo , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Neoplasias Cutáneas/patología
4.
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
5.
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
6.
Int J Mol Sci ; 22(1)2020 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-33375205

RESUMEN

The complement system is involved in promoting secondary injury after traumatic brain injury (TBI), but the roles of the classical and lectin pathways leading to complement activation need to be clarified. To this end, we aimed to determine the ability of the brain to activate the synthesis of classical and lectin pathway initiators in response to TBI and to examine their expression in primary microglial cell cultures. We have modeled TBI in mice by controlled cortical impact (CCI), a clinically relevant experimental model. Using Real-time quantitative polymerase chain reaction (RT-qPCR) we analyzed the expression of initiators of classical the complement component 1q, 1r and 1s (C1q, C1r, and C1s) and lectin (mannose binding lectin A, mannose binding lectin C, collectin 11, ficolin A, and ficolin B) complement pathways and other cellular markers in four brain areas (cortex, striatum, thalamus and hippocampus) of mice exposed to CCI from 24 h and up to 5 weeks. In all murine ipsilateral brain structures assessed, we detected long-lasting, time- and area-dependent significant increases in the mRNA levels of all classical (C1q, C1s, C1r) and some lectin (collectin 11, ficolin A, ficolin B) initiator molecules after TBI. In parallel, we observed significantly enhanced expression of cellular markers for neutrophils (Cd177), T cells (Cd8), astrocytes (glial fibrillary acidic protein-GFAP), microglia/macrophages (allograft inflammatory factor 1-IBA-1), and microglia (transmembrane protein 119-TMEM119); moreover, we detected astrocytes (GFAP) and microglia/macrophages (IBA-1) protein level strong upregulation in all analyzed brain areas. Further, the results obtained in primary microglial cell cultures suggested that these cells may be largely responsible for the biosynthesis of classical pathway initiators. However, microglia are unlikely to be responsible for the production of the lectin pathway initiators. Immunofluorescence analysis confirmed that at the site of brain injury, the C1q is localized in microglia/macrophages and neurons but not in astroglial cells. In sum, the brain strongly reacts to TBI by activating the local synthesis of classical and lectin complement pathway activators. Thus, the brain responds to TBI with a strong, widespread and persistent upregulation of complement components, the targeting of which may provide protection in TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/genética , Activación de Complemento/genética , Lectina de Unión a Manosa de la Vía del Complemento/genética , Lectinas/genética , Animales , Lesiones Traumáticas del Encéfalo/metabolismo , Células Cultivadas , Corteza Cerebral/metabolismo , Complemento C1/genética , Complemento C1/metabolismo , Complemento C1q/genética , Complemento C1q/metabolismo , Complemento C1r/genética , Complemento C1r/metabolismo , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Hipocampo/metabolismo , Humanos , Lectinas/metabolismo , Masculino , Ratones Endogámicos C57BL , Microglía/metabolismo , Neostriado/metabolismo , Tálamo/metabolismo , Factores de Tiempo
7.
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
8.
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
9.
PLoS Pathog ; 15(3): e1007659, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30897158

RESUMEN

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


Asunto(s)
Proteínas Bacterianas/genética , Borrelia burgdorferi/genética , Vía Clásica del Complemento/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/ultraestructura , Borrelia burgdorferi/inmunología , Grupo Borrelia Burgdorferi , Complemento C1r/metabolismo , Vía Clásica del Complemento/fisiología , Proteínas del Sistema Complemento/metabolismo , Humanos , Enfermedad de Lyme/fisiopatología , Dominios Proteicos/fisiología , Proteínas Recombinantes , Análisis de Secuencia de Proteína
10.
J Vasc Surg ; 68(6S): 209S-221S.e2, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29395422

RESUMEN

OBJECTIVE: Ischemia-reperfusion (I/R) injury is a major clinical problem linked to vascular surgery. Currently, no drugs to prevent or to treat I/R injury are approved for clinical use. C1 inhibitor (C1 INH) is known to reduce activation of the plasma cascade systems that are involved in the pathophysiologic process of I/R injury. The aim of this study was therefore to investigate the effect of C1 INH on complement deposition and endothelial cell activation in a rat model of hind limb I/R injury. METHODS: Male Wistar rats (wild type, bred at the central animal facility, University of Bern), weighing 250 to 320 g, were used. The rats underwent 2-hour ischemia and 24-hour reperfusion by unilateral clamping of the femoral artery and additional use of a tourniquet. Five groups were divided according to intravenous treatment 5 minutes before ischemia: 50 IU/kg C1 INH (n = 5); 100 IU/kg C1 INH (n = 7); vehicle control (n = 5); nontreated control (n = 7); and normal, healthy control without intervention (n = 4). At the end, muscle edema, tissue viability, and histologic features were assessed. Deposition of immunoglobulin M, C1r, C4d, and fibrin and expression of plasminogen activator inhibitor 1, heparan sulfate (HS), E-selectin, and vascular cell adhesion molecule 1 were evaluated by fluorescence staining. In addition, high-mobility group box 1 protein was measured in plasma. RESULTS: Edema formation was reduced by C1 INH at two dosages, mirrored by improved histologic injury scores and preserved muscle viability. Deposition of immunoglobulin M, C4d, and fibrin was significantly decreased by 100 IU/kg C1 INH compared with nontreated controls. Pretreatment with 100 IU/kg C1 INH also significantly reduced HS shedding and expression of plasminogen activator inhibitor 1 as well as plasma levels of high-mobility group box 1 protein. CONCLUSIONS: Pretreatment with both 50 and 100 IU/kg C1 INH attenuated reperfusion injury of rat hind limbs. Pretreatment with 100 IU/kg also preserved the endothelial HS layer as well as the natural, profibrinolytic phenotype of the endothelium. Prevention of endothelial cell activation by C1 INH may therefore be a promising strategy to prevent I/R injury in the clinical setting of peripheral vascular diseases and elective surgery on extremities.


Asunto(s)
Activación de Complemento/efectos de los fármacos , Proteína Inhibidora del Complemento C1/farmacología , Inactivadores del Complemento/farmacología , Células Endoteliales/efectos de los fármacos , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/efectos de los fármacos , Daño por Reperfusión/prevención & control , Animales , Complemento C1r/metabolismo , Complemento C4b/metabolismo , Modelos Animales de Enfermedad , Selectina E/metabolismo , Edema/inmunología , Edema/metabolismo , Edema/patología , Edema/prevención & control , Células Endoteliales/metabolismo , Células Endoteliales/patología , Fibrina/metabolismo , Proteína HMGB1/metabolismo , Heparitina Sulfato/metabolismo , Miembro Posterior , Inmunoglobulina M/metabolismo , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Fragmentos de Péptidos/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Ratas Wistar , Daño por Reperfusión/inmunología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Supervivencia Tisular/efectos de los fármacos , Molécula 1 de Adhesión Celular Vascular/metabolismo
11.
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
12.
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
13.
J Immunol ; 199(12): 3981-3990, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29070672

RESUMEN

Anti-nuclear autoantibodies, which frequently target the nucleoli, are pathogenic hallmarks of systemic lupus erythematosus (SLE). Although the causes of these Abs remain broad and ill-defined, a genetic deficiency in C1 complex (C1qC1r2C1s2) or C4 is able to induce these Abs. Considering a recent finding that, in dead cells, nucleoli were targeted by C1q and two nucleolar autoantigens were degraded by C1r/C1s proteases, we considered that C1 could help protect against antinuclear autoimmunity by broadly degrading nucleolar proteins or autoantigens. Nucleoli were isolated to homogeneity and structurally defined. After C1 treatment, cleaved nucleolar proteins were identified by proteomic two-dimensional fluorescence difference gel electrophoresis and mass spectrometry, and further verified by Western blotting using specific Abs. The extent of nucleolar autoantigen degradation upon C1 treatment was estimated using SLE patient autoantibodies. The isolated nucleoli were broadly reactive with SLE patient autoantibodies. These nucleoli lacked significant autoproteolysis, but many nucleolar proteins and autoantigens were degraded by C1 proteases; >20 nucleolar proteins were identified as C1 cleavable. These were further validated by Western blotting using specific Abs. The broad autoantigenicity of the nucleoli may attribute to their poor autoproteolysis, causing autologous immune stimulation upon necrotic exposure. However, C1q targets at these nucleoli to cause C1 protease activation and the cleavage of many nucleolar proteins or autoantigens. This may represent one important surveillance mechanism against antinuclear autoimmunity because C1 genetic deficiency causes anti-nuclear autoantibodies and SLE disease.


Asunto(s)
Autoanticuerpos/inmunología , Autoantígenos/metabolismo , Nucléolo Celular/inmunología , Complemento C1r/metabolismo , Complemento C1s/metabolismo , Inmunoglobulina G/inmunología , Proteínas Nucleares/metabolismo , Autoanticuerpos/sangre , Nucléolo Celular/ultraestructura , Células HeLa , Humanos , Inmunoglobulina G/sangre , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/inmunología , Proteolisis , Proteómica , Especificidad por Sustrato
14.
Fish Shellfish Immunol ; 70: 240-251, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28882800

RESUMEN

The complement components C1r and C1s play a vital role in immunity with the activation of C1 complex in the classical complement pathway against pathogen infection. In this study, Nile tilapia (Oreochromis niloticus) C1r and C1s orthologs (OnC1r and OnC1s) were identified and characterized. The cDNA of OnC1r and OnC1s ORFs consisted of 1902 bp and 2100 bp of nucleotide sequence encoding polypeptides of 633 and 699 amino acids, respectively. The deduced OnC1r and OnC1s proteins both possessed CUB, EGF, CCP and SP domains, which were significantly homology to teleost. Spatial mRNA expression analysis revealed that the OnC1r and OnC1s were highly expressed in liver. After the in vivo challenges of Streptococcus agalactiae (S. agalactiae) and lipopolysaccharide (LPS), the mRNA expressions of OnC1r and OnC1s were significantly up-regulated in liver and spleen, which were consistent with immunohistochemical detection at the protein level. The up-regulation of OnC1r and OnC1s expressions were also demonstrated in head kidney monocytes/macrophages in vitro stimulated with LPS, S. agalactiae, and recombinant OnIFN-γ. Taken together, the results of this study indicated that OnC1r and OnC1s were likely to get involved in the immune response of Nile tilapia against bacterial infection.


Asunto(s)
Cíclidos/genética , Cíclidos/inmunología , Complemento C1r/genética , Complemento C1s/genética , Enfermedades de los Peces/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Secuencia de Aminoácidos , Animales , Complemento C1r/química , Complemento C1r/metabolismo , Complemento C1s/química , Complemento C1s/metabolismo , Biología Computacional , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Perfilación de la Expresión Génica/veterinaria , Lipopolisacáridos/farmacología , Especificidad de Órganos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia/veterinaria , Infecciones Estreptocócicas/veterinaria , Streptococcus agalactiae/fisiología , Regulación hacia Arriba
15.
Proc Natl Acad Sci U S A ; 114(5): 986-991, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28104818

RESUMEN

The complement system is an important antimicrobial and inflammation-generating component of the innate immune system. The classical pathway of complement is activated upon binding of the 774-kDa C1 complex, consisting of the recognition molecule C1q and the tetrameric protease complex C1r2s2, to a variety of activators presenting specific molecular patterns such as IgG- and IgM-containing immune complexes. A canonical model entails a C1r2s2 with its serine protease domains tightly packed together in the center of C1 and an intricate intramolecular reaction mechanism for activation of C1r and C1s, induced upon C1 binding to the activator. Here, we show that the serine protease domains of C1r and C1s are located at the periphery of the C1r2s2 tetramer both when alone or within the nonactivated C1 complex. Our structural studies indicate that the C1 complex adopts a conformation incompatible with intramolecular activation of C1, suggesting instead that intermolecular proteolytic activation between neighboring C1 complexes bound to a complement activating surface occurs. Our results rationalize how a multitude of structurally unrelated molecular patterns can activate C1 and suggests a conserved mechanism for complement activation through the classical and the related lectin pathway.


Asunto(s)
Complemento C1r/química , Complemento C1s/química , Vía Clásica del Complemento/fisiología , Complemento C1r/genética , Complemento C1r/metabolismo , Complemento C1s/genética , Complemento C1s/metabolismo , Activación Enzimática , Genes Sintéticos , Células HEK293 , Humanos , Inmunidad Innata , Microscopía Electrónica , Modelos Moleculares , Conformación Proteica , Proteínas Recombinantes/química , Dispersión del Ángulo Pequeño , Relación Estructura-Actividad , Difracción de Rayos X
16.
Mol Cell Neurosci ; 79: 23-33, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28064059

RESUMEN

Protease-activated receptor-1 (PAR1) is the prototypic member of a family of four G-protein-coupled receptors that signal in response to extracellular proteases. In the peripheral nervous system, the expression and/or the role of PARs are still poorly investigated. High PAR1 mRNA expression was found in the rat dorsal root ganglia and the signal intensity of PAR1 mRNA increased in response to sciatic nerve transection. In the sciatic nerve, functional PAR1 receptor was reported at the level of non-compacted Schwann cell myelin microvilli of the nodes of Ranvier. Schwann cells are the principal population of glial cells of the peripheral nervous system which myelinate axons playing an important role during axonal regeneration and remyelination. The present study was undertaken in order to determine if the activation of PAR1 affects the neurotrophic properties of Schwann cells. Our results suggest that the stimulation of PAR1 could potentiate the Schwann cell ability to favour nerve regeneration. In fact, the conditioned medium obtained from Schwann cell cultures challenged with a specific PAR1 activating peptide (PAR1 AP) displays increased neuroprotective and neurotrophic properties with respect to the culture medium from untreated Schwann cells. The proteomic analysis of secreted proteins in untreated and PAR1 AP-treated Schwann cells allowed the identification of factors differentially expressed in the two samples. Some of them (such as macrophage migration inhibitory factor, matrix metalloproteinase-2, decorin, syndecan 4, complement C1r subcomponent, angiogenic factor with G patch and FHA domains 1) appear to be transcriptionally regulated after PAR1 AP treatment as shown by RT-PCR.


Asunto(s)
Factores de Crecimiento Nervioso/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Células de Schwann/metabolismo , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/metabolismo , Animales , Células Cultivadas , Complemento C1q/genética , Complemento C1q/metabolismo , Complemento C1r/genética , Complemento C1r/metabolismo , Medios de Cultivo Condicionados/farmacología , Decorina/genética , Decorina/metabolismo , Femenino , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Factores de Crecimiento Nervioso/farmacología , Regeneración Nerviosa/efectos de los fármacos , Células PC12 , Ratas , Ratas Wistar , Nervio Ciático/citología , Nervio Ciático/metabolismo , Nervio Ciático/fisiología , Sindecano-4/genética , Sindecano-4/metabolismo
17.
Dev Comp Immunol ; 65: 314-320, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27514577

RESUMEN

Marsupials are born immunologically premature, relying on cells and molecules in maternal milk for immune protection. Both immunoglobulin and complement proteins have been identified in marsupial milk, but the expression of specific complement proteins remains largely unexplored. We report partial cDNA sequences for two complement-activating proteins, C3, C1r, CFP and MASP2, in liver tissues from red-tailed phascogale (Phascogale calura). Conservation of functionally relevant motifs were identified in the translated cDNA sequences from phascogale C3, CFP and MASP2 and their eutherian homologues. Gene expression of representative molecules from each of the major complement pathways was also investigated in whole body tissues from 1 to 18 day old animals and liver tissues from 31-day to 14-month old animals. Average complement expression in whole bodies and liver tissues of C1r, CFP, MASP2 and C3 increased significantly in juveniles compared to pouch young, presumably due to the maturation of the young's own complement system. Comparing expression in liver tissues only, we found that the average CFP expression were higher in pouch young compared to juveniles, while results were still statistically similar to the average expression of all tissues for C1r, MASP2 and C3. The average complement expression then significantly decreased as the animals aged into adulthood.


Asunto(s)
Complemento C1r/metabolismo , Complemento C3/metabolismo , Hígado/fisiología , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/metabolismo , Marsupiales/inmunología , Animales , Complemento C1r/genética , Complemento C3/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Inmunidad Innata , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/genética , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Transactivadores/genética
18.
Fish Shellfish Immunol ; 46(2): 656-68, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26241508

RESUMEN

The complement components C1r and C1s play a crucial role in innate immunity via activation of the classical complement cascade system. As initiators of the pathogen-induced signaling cascade, C1r and C1s modulate innate immunity. In order to understand the immune responses of teleost C1r and C1s, Oplegnathus fasciatus C1r and C1s genes (OfC1r and OfC1s) were identified and characterized. The genomic sequence of OfC1r was enclosed with thirteen exons that represented a putative peptide with 704 amino acids (aa), whereas eleven exons of OfC1s represented a 691 aa polypeptide. In addition, genomic analysis revealed that both OfC1r and OfC1s were located on a single chromosome. These putative polypeptides were composed of two CUB domains, an EGF domain, two CCP domains, and a catalytically active serine protease domain. Phylogenetic analysis of C1r and C1s showed that OfC1r and OfC1s were evolutionary close to the orthologs of Pundamilia nyererei (identity = 73.4%) and Oryzias latipes (identity = 58.0%), respectively. Based on the results of quantitative real-time qPCR analysis, OfC1r and OfC1s transcripts were detected in all the eleven different tissues, with higher levels of OfC1r in blood and OfC1s in liver. The putative roles of OfC1r and OfC1s in response to pathogenic bacteria (Edwardsiella tarda and Streptococcus iniae) and virus (rock bream iridovirus, RBIV) were investigated in liver and head kidney tissues. The transcription of OfC1r and OfC1s was found to be significantly upregulated in response to pathogenic bacterial and viral infections. Overall findings of the present study demonstrate the potential immune responses of OfC1r and OfC1s against invading microbial pathogens and the activation of classical signaling cascade in rock bream.


Asunto(s)
Complemento C1r/genética , Complemento C1s/genética , Enfermedades de los Peces/inmunología , Proteínas de Peces/genética , Inmunidad Innata , Perciformes , Secuencia de Aminoácidos , Animales , Complemento C1r/química , Complemento C1r/metabolismo , Complemento C1s/química , Complemento C1s/metabolismo , Infecciones por Virus ADN/inmunología , Infecciones por Virus ADN/virología , Edwardsiella tarda/fisiología , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/microbiología , Enfermedades de los Peces/microbiología , Enfermedades de los Peces/virología , Proteínas de Peces/química , Proteínas de Peces/metabolismo , Riñón Cefálico/virología , Iridoviridae/fisiología , Hígado/virología , Datos de Secuencia Molecular , Especificidad de Órganos , Filogenia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Alineación de Secuencia/veterinaria , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus/fisiología
19.
J Biol Chem ; 290(37): 22570-80, 2015 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-26231209

RESUMEN

In infection, complement C1q recognizes pathogen-congregated antibodies and elicits complement activation. Among endogenous ligands, C1q binds to DNA and apoptotic cells, but whether C1q binds to nuclear DNA in apoptotic cells remains to be investigated. With UV irradiation-induced apoptosis, C1q initially bound to peripheral cellular regions in early apoptotic cells. By 6 h, binding concentrated in the nuclei to the nucleolus but not the chromatins. When nucleoli were isolated from non-apoptotic cells, C1q also bound to these structures. In vivo, C1q exists as the C1 complex (C1qC1r2C1s2), and C1q binding to ligands activates the C1r/C1s proteases. Incubation of nucleoli with C1 caused degradation of the nucleolar proteins nucleolin and nucleophosmin 1. This was inhibited by the C1 inhibitor. The nucleoli are abundant with autoantigens. C1q binding and C1r/C1s degradation of nucleolar antigens during cell apoptosis potentially reduces autoimmunity. These findings help us to understand why genetic C1q and C1r/C1s deficiencies cause systemic lupus erythematosus.


Asunto(s)
Apoptosis/efectos de la radiación , Nucléolo Celular/metabolismo , Complemento C1q/metabolismo , Complemento C1r/metabolismo , Complemento C1s/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteolisis/efectos de la radiación , Proteínas de Unión al ARN/metabolismo , Rayos Ultravioleta , Células HeLa , Humanos , Nucleofosmina , Nucleolina
20.
Proc Natl Acad Sci U S A ; 110(21): 8650-5, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23650384

RESUMEN

Complement C1q is a hexameric molecule assembled from 18 polypeptide chains of three different types encoded by three genes. This versatile recognition protein senses a wide variety of immune and nonimmune ligands, including pathogens and altered self components, and triggers the classical complement pathway through activation of its associated proteases C1r and C1s. We report a method for expression of recombinant full-length human C1q involving stable transfection of HEK 293-F mammalian cells and fusion of an affinity tag to the C-terminal end of the C chain. The resulting recombinant (r) C1q molecule is similar to serum C1q as judged from biochemical and structural analyses and exhibits the characteristic shape of a bunch of flowers. Analysis of its interaction properties by surface plasmon resonance shows that rC1q retains the ability of serum C1q to associate with the C1s-C1r-C1r-C1s tetramer, to recognize physiological C1q ligands such as IgG and pentraxin 3, and to trigger C1r and C1s activation. Functional analysis of rC1q variants carrying mutations of LysA59, LysB61, and/or LysC58, in the collagen-like stems, demonstrates that LysB61 and LysC58 each play a key role in the interaction with C1s-C1r-C1r-C1s, with LysA59 being involved to a lesser degree. We propose that LysB61 and LysC58 both form salt bridges with outer acidic Ca(2+) ligands of the C1r and C1s CUB (complement C1r/C1s, Uegf, bone morphogenetic protein) domains. The expression method reported here opens the way for deciphering the molecular basis of the unusual binding versatility of C1q by mapping the residues involved in the sensing of its targets and the binding of its receptors.


Asunto(s)
Activación de Complemento/fisiología , Complemento C1q/metabolismo , Complemento C1r/metabolismo , Complemento C1s/metabolismo , Sustitución de Aminoácidos , Sitios de Unión , Proteína C-Reactiva/química , Proteína C-Reactiva/metabolismo , Calcio/metabolismo , Complemento C1q/química , Complemento C1q/genética , Complemento C1r/química , Complemento C1r/genética , Complemento C1s/química , Complemento C1s/genética , Expresión Génica , Células HEK293 , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/metabolismo , Mutación Missense , Unión Proteica/fisiología , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Componente Amiloide P Sérico/química , Componente Amiloide P Sérico/metabolismo , Resonancia por Plasmón de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA