Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 522
Filtrar
1.
J Immunol ; 212(4): 689-701, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38149922

RESUMEN

The classical pathway (CP) is a potent mechanism for initiating complement activity and is a driver of pathology in many complement-mediated diseases. The CP is initiated via activation of complement component C1, which consists of the pattern recognition molecule C1q bound to a tetrameric assembly of proteases C1r and C1s. Enzymatically active C1s provides the catalytic basis for cleavage of the downstream CP components, C4 and C2, and is therefore an attractive target for therapeutic intervention in CP-driven diseases. Although an anti-C1s mAb has been Food and Drug Administration approved, identifying small-molecule C1s inhibitors remains a priority. In this study, we describe 6-(4-phenylpiperazin-1-yl)pyridine-3-carboximidamide (A1) as a selective, competitive inhibitor of C1s. A1 was identified through a virtual screen for small molecules that interact with the C1s substrate recognition site. Subsequent functional studies revealed that A1 dose-dependently inhibits CP activation by heparin-induced immune complexes, CP-driven lysis of Ab-sensitized sheep erythrocytes, CP activation in a pathway-specific ELISA, and cleavage of C2 by C1s. Biochemical experiments demonstrated that A1 binds directly to C1s with a Kd of ∼9.8 µM and competitively inhibits its activity with an inhibition constant (Ki) of ∼5.8 µM. A 1.8-Å-resolution crystal structure revealed the physical basis for C1s inhibition by A1 and provided information on the structure-activity relationship of the A1 scaffold, which was supported by evaluating a panel of A1 analogs. Taken together, our work identifies A1 as a new class of small-molecule C1s inhibitor and lays the foundation for development of increasingly potent and selective A1 analogs for both research and therapeutic purposes.


Asunto(s)
Complemento C1s , Vía Clásica del Complemento , Animales , Ovinos , Péptido Hidrolasas , Complemento C1/metabolismo , Endopeptidasas , Piridinas/farmacología
2.
Cells ; 11(19)2022 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-36231090

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal disease in which motor neurons gradually degenerate. The mutation of the C9orf72 gene is the main genetic cause of ALS (C9-ALS). One of its specific pathological features is the production of proline-arginine (PR) dipeptide repeat protein (DPR). In this study, we developed a PR-DPR (PR50)-expressing human HMC3 microglial cell model. We found that PR50 mainly aggregates into spots in the nucleus and induces significant NLRP3 inflammasome activity. Moreover, mouse NSC-34 motor neuron cells treated with a conditional medium of PR50-expressing HMC3 cells (PR-CM) caused cell damage and apoptosis activity. However, R50-expressing HMC cells treated with MCC950 (an NLRP3 inhibitor) reversed this result. Furthermore, we identified complement component 1 q subcomponent-binding protein (C1QBP) as one of the interaction partners of PR50. The downregulation of C1QBP in HMC3 cells induces NLRP3 inflammasome activity similar to PR50 expression. Finally, we found that syringin can block the interaction between PR50 and C1QBP, and effectively reduce the PR50-induced NLRP3 inflammasome activity in HMC3 cells. This improves the apoptosis of NSC-34 cells caused by PR-CM. This study is the first to link PR50, C1QBP, and NLRP3 inflammasome activity in microglia and develop potential therapeutic strategies for syringin intervention in C9-ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Arginina , Proteína C9orf72/genética , Proteínas Portadoras , Complemento C1/metabolismo , Dipéptidos/metabolismo , Dipéptidos/farmacología , Glucósidos , Humanos , Inflamasomas/metabolismo , Ratones , Microglía/metabolismo , Proteínas Mitocondriales/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Fenilpropionatos , Prolina , Proteínas/metabolismo
3.
FASEB J ; 36(6): e22347, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35579659

RESUMEN

C1q/TNF-related proteins (CTRP1-15) constitute a conserved group of secreted proteins of the C1q family with diverse functions. In vitro studies have shown that CTRP11/C1QL4 can inhibit adipogenesis, antagonize myoblast fusion, and promote testosterone synthesis and secretion. Whether CTRP11 is required for these processes in vivo remains unknown. Here, we show that knockout (KO) mice lacking CTRP11 have normal skeletal muscle mass and function, and testosterone level, suggesting that CTRP11 is dispensable for skeletal muscle development and testosterone production. We focused our analysis on whether this nutrient-responsive secreted protein plays a role in controlling sugar and fat metabolism. At baseline when mice are fed a standard chow, CTRP11 deficiency affects metabolic parameters in a sexually dimorphic manner. Only Ctrp11-KO female mice have significantly higher fasting serum ketones and reduced physical activity. In the refeeding phase following food withdrawal, Ctrp11-KO female mice have reduced food intake and increased metabolic rate and energy expenditure, highlighting CTRP11's role in fasting-refeeding response. When challenged with a high-fat diet to induce obesity and metabolic dysfunction, CTRP11 deficiency modestly exacerbates obesity-induced glucose intolerance, with more pronounced effects seen in Ctrp11-KO male mice. Switching to a low-fat diet after obesity induction results in greater fat loss in wild type relative to KO male mice, suggesting impaired response to obesity reversal and reduced metabolic flexibility in the absence of CTRP11. Collectively, our data provide genetic evidence for novel sex-dependent metabolic regulation by CTRP11, but note the overall modest contribution of CTRP11 to systemic energy homeostasis.


Asunto(s)
Complemento C1/metabolismo , Complemento C1q , Dieta Alta en Grasa , Animales , Complemento C1q/metabolismo , Metabolismo Energético/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Testosterona
4.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34035177

RESUMEN

Cytoadhesion of Plasmodium falciparum-infected erythrocytes (IEs) to the endothelial lining of blood vessels protects parasites from splenic destruction, but also leads to detrimental inflammation and vessel occlusion. Surface display of the P. falciparum erythrocyte membrane protein 1 (PfEMP1) adhesion ligands exposes them to host antibodies and serum proteins. PfEMP1 are important targets of acquired immunity to malaria, and through evolution, the protein family has expanded and diversified to bind a select set of host receptors through antigenically diversified receptor-binding domains. Here, we show that complement component 1s (C1s) in serum cleaves PfEMP1 at semiconserved arginine motifs located at interdomain regions between the receptor-binding domains, rendering the IE incapable of binding the two main PfEMP1 receptors, CD36 and endothelial protein C receptor (EPCR). Bioinformatic analyses of PfEMP1 protein sequences from 15 P. falciparum genomes found the C1s motif was present in most PfEMP1 variants. Prediction of C1s cleavage and loss of binding to endothelial receptors was further corroborated by testing of several different parasite lines. These observations suggest that the parasites have maintained susceptibility for cleavage by the serine protease, C1s, and provides evidence for a complex relationship between the complement system and the P. falciparum cytoadhesion virulence determinant.


Asunto(s)
Adhesión Bacteriana , Complemento C1/metabolismo , Plasmodium falciparum/fisiología , Proteínas Protozoarias/metabolismo , Secuencia de Aminoácidos , Línea Celular , Secuencia Conservada , Humanos
5.
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
6.
Int Immunopharmacol ; 83: 106526, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32361670

RESUMEN

Hereditary Angioedema (HAE) is a rare, autosomal dominant disease caused by mutations in SERPING1 gene leading to dysfunction/deficiency of C1-esterase inhibitor (C1-INH) protein and subsequent dysregulation of the contact system and bradykinin overproduction. As functional C1-INH (fC1-INH) levels are reduced in HAE types I and II (HAE-I/II), a specific, sensitive and accessible rapid diagnostic method to quantitate fC1-INH is crucial in diagnosing HAE-I/II. Previously, we developed/validated methods to detect fC1-INH levels in human plasma based on functional binding to C1s or FXIIa for C1-INH-based therapies. Quantitative fC1-INH immunoassay methods were converted to the Lateral flow assay (LFA) platform after identifying the best reagent/s pair. The assay was developed and optimized as a first of its kind LFA method for quantifying fC1-INH in human plasma to aid HAE point-of-care diagnosis. Receiver operating characteristic analysis was performed using normal control and HAE subject plasma samples to calculate area-under-curve and a cut-off point to distinguish normal versus HAE subject samples. LFA data was correlated with the conventional diagnostic assay for fC1-INH in HAE plasma samples and profiles matched for individual subjects. Here, we demonstrate a proof-of-concept for the quantitative fC1-INH LFA using normal and HAE plasma samples. We propose that the method could be used as a point-of-care test to diagnose HAE in a variety of settings, such as, a hospital or physician's office, at home or in an ambulance.


Asunto(s)
Angioedemas Hereditarios/diagnóstico , Proteína Inhibidora del Complemento C1/análisis , Angioedemas Hereditarios/genética , Compuestos Cromogénicos , Complemento C1/metabolismo , Humanos , Mutación/genética , Sistemas de Atención de Punto , Unión Proteica
7.
Int J Mol Sci ; 21(5)2020 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-32121610

RESUMEN

Several studies have suggested that there is a link between membrane attack complex (MAC) deposition in the retina and the progression of diabetic retinopathy (DR). Our recent investigation demonstrated that circulating IgG-laden extracellular vesicles contribute to an increase in retinal vascular permeability in DR through activation of the complement system. However, the mechanism through which extracellular vesicle-induced complement activation contributes to retinal vascular cytolytic damage in DR is not well understood. In this study, we demonstrate that IgG-laden extracellular vesicles in rat plasma activate the classical complement pathway, and in vitro Streptozotocin (STZ)-induced rat diabetic plasma results in MAC deposition and cytolytic damage in human retinal endothelial cells (HRECs). Moreover, removal of the plasma extracellular vesicles reduced the MAC deposition and abrogated cytolytic damage seen in HRECs. Together, the results of this study demonstrate that complement activation by IgG-laden extracellular vesicles in plasma could lead to MAC deposition and contribute to endothelium damage and progression of DR.


Asunto(s)
Activación de Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Células Endoteliales/patología , Vesículas Extracelulares/metabolismo , Retina/patología , Animales , Muerte Celular , Supervivencia Celular , Complemento C1/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Vesículas Extracelulares/ultraestructura , Humanos , Inmunoglobulinas/metabolismo , Masculino , Ratas Sprague-Dawley
8.
Cell Host Microbe ; 25(4): 617-629.e7, 2019 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-30926239

RESUMEN

The complement system is vital for anti-microbial defense. In the classical pathway, pathogen-bound antibody recruits the C1 complex (C1qC1r2C1s2) that initiates a cleavage cascade involving C2, C3, C4, and C5 and triggering microbial clearance. We demonstrate a C4-dependent antiviral mechanism that is independent of downstream complement components. C4 inhibits human adenovirus infection by directly inactivating the virus capsid. Rapid C4 activation and capsid deposition of cleaved C4b are catalyzed by antibodies via the classical pathway. Capsid-deposited C4b neutralizes infection independent of C2 and C3 but requires C1q antibody engagement. C4b inhibits capsid disassembly, preventing endosomal escape and cytosolic access. C4-deficient mice exhibit heightened viral burdens. Additionally, complement synergizes with the Fc receptor TRIM21 to block transduction by an adenovirus gene therapy vector but is partially restored by Fab virus shielding. These results suggest that the complement system could be altered to prevent virus infection and enhance virus gene therapy efficacy.


Asunto(s)
Infecciones por Adenovirus Humanos/inmunología , Adenovirus Humanos/inmunología , Cápside/metabolismo , Complemento C4/metabolismo , Inmunidad Humoral , Factores Inmunológicos/metabolismo , Inactivación de Virus , Animales , Anticuerpos Antivirales/metabolismo , Línea Celular , Complemento C1/metabolismo , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Unión Proteica
9.
FASEB J ; 33(4): 4893-4906, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30608882

RESUMEN

C1q-like 4 (C1QL4), a novel member of the C1q- and TNF-related protein family, was found to be highly expressed in rodent and human testis. However, the localization, developmental, and hormonally regulated expression and biologic function of C1ql4 in the testis have not been investigated. Here, we demonstrated that C1ql4 mRNA and protein levels in murine testes gradually increased from the postnatal period to the adult stage and were up-regulated by LH in vivo. In situ hybridization demonstrated that the distribution and expression levels of C1ql4 mRNA varied at different developmental stages, although C1ql4 mRNA was detected in the seminiferous tubule and interstitial Leydig cells. Recombinant C1QL4 did not affect cell proliferation but did increase testosterone production in TM3 Leydig cells, as well as in cultured seminiferous tubules. C1QL4-induced testosterone secretion in Leydig cells was accompanied by increased expression of steroidogenic acute regulatory (StAR) protein and steroidogenic enzymes. During this process, the c-Raf/extracellular signal-regulated protein kinase kinases 1 and 2/ERK1/2/mitogen- and stress-activated protein kinase-1 and cAMP/PKA/cAMP-responsive element binding protein signaling cascades were activated by C1QL4. The cell-adhesion GPCR brain-specific angiogenesis inhibitor 3 (BAI3), a putative receptor of C1QL4, was detected in the seminiferous tubule and interstitial Leydig cells during testicular development. Knockdown of Bai3 expression in Leydig cells led to a reduction in Star expression, accompanied by increases in phosphorylation of ERK1/2 and intercellular cAMP levels. However, C1QL4-induced StAR expression was not completely suppressed in the Bai3-deficient Leydig cells, and phosphorylation of ERK1/2 and intercellular cAMP levels were not significantly changed before and after C1QL4 stimulation. Our results suggested that although BAI3 played a role in C1QL4-induced steroidogenesis, there was an unidentified receptor that mediated C1QL4-activated testosterone secretion in Leydig cells through phosphorylation of ERK1/2 and up-regulation of intracellular cAMP levels. Taken together, our results showed, for the first time to our knowledge, that C1QL4 served as a novel acute regulator of testosterone secretion, and BAI3 functioned as a new receptor that is involved in steroidogenesis in Leydig cells. BAI3-independent ERK1/2 activation and cAMP activation mediated C1QL4-induced testosterone secretion. This study expanded the reproductive roles and mechanisms of C1QL4 and BAI3 signaling pathways.-Tan, A., Ke, S., Chen, Y., Chen, L., Lu, X., Ding, F., Yang, L., Tang, Y., Yu, Y. Expression patterns of C1ql4 and its cell-adhesion GPCR Bai3 in the murine testis and functional roles in steroidogenesis.


Asunto(s)
Complemento C1/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Testículo/citología , Testículo/metabolismo , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Complemento C1/farmacología , AMP Cíclico/metabolismo , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Hibridación in Situ , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Túbulos Seminíferos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Testosterona/metabolismo
10.
Cell Transplant ; 27(4): 666-676, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29871524

RESUMEN

Alzheimer's Disease (AD) is the leading cause of dementia in the elderly. In healthy individuals, amyloid precursor protein (APP) is cleaved by α-secretase, generating soluble α-amyloid precursor protein (sAPPα), which contributes neuroprotective functions in the neuronal environment. In contrast, in the neurodegenerative environment of AD patients, amyloid-ß-peptide (Aß) of either 40 or 42 residues are generated by increased activity of ß- and γ-secretase. These proteins amalgamate in specific regions of the brain, which disrupts neuronal functions and leads to cognitive impairment. Human umbilical cord blood cells (HUCBC) have proven useful as potential immunomodulatory therapies in various models of neurodegenerative diseases, including AD. Our most recent work studied the impact of umbilical cord blood serum (CBS) on modulation of sAPPα production. Heat-sensitive CBS significantly promoted sAPPα production, indicating that heat-sensitive factor(s) play(s) a role in this process. Liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis was used to determine the molecular source of α-secretase in purified CBS and aged blood serum (AgBS) fraction. Of the proteins identified, the subunits of C1 complex (C1q, C1r, and C1s) and alpha-2-macroglobulin showed significantly greater levels in purified α-CBS fraction (α-CBSF) compared with the AgBS fraction (AgBSF). Specifically, C1 markedly increased sAPPα and alpha-carboxyl-terminal fragment (α-CTF) production in a dose-dependent fashion, whereas C1q alone only minimally increased and C3 did not increase sAPPα production in the absence of sera. Furthermore, C1q markedly increased sAPPα and α-CTF, while decreasing Aß, in CHO/APPwt cells cultured in the presence of whole sera. These results confirm our initial assumption that APP α-secretase activity in human blood serum is mediated by complement C1, opening a potential therapeutic modality for the future of AD.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Complemento C1/metabolismo , Sangre Fetal/enzimología , Anciano , Envejecimiento/sangre , Péptidos beta-Amiloides/metabolismo , Animales , Células CHO , Complemento C1/aislamiento & purificación , Complemento C3b/metabolismo , Cricetinae , Cricetulus , Calor , Humanos , Ratones , Proteómica
11.
Biomolecules ; 8(2)2018 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-29735903

RESUMEN

When excessively activated, C1 is insufficiently regulated, which results in tissue damage. Such tissue damage causes the complement system to become further activated to remove the resulting tissue damage, and a vicious cycle of activation/tissue damage occurs. Current Food and Drug Administration approved treatments include supplemental recombinant C1 inhibitor, but these are extremely costly and a more economical solution is desired. In our work, we have utilized an existing data set of 136 compounds that have been previously tested for activity against C1. Using these compounds and the activity data, we have created models using principal component analysis, genetic algorithm, and support vector machine approaches to characterize activity. The models were then utilized to virtually screen the 72 million compound PubChem repository. This first round of virtual high-throughput screening identified many economical and promising inhibitor candidates, a subset of which was tested to validate their biological activity. These results were used to retrain the models and rescreen PubChem in a second round vHTS. Hit rates for the first round vHTS were 57%, while hit rates for the second round vHTS were 50%. Additional structure⁻property analysis was performed on the active and inactive compounds to identify interesting scaffolds for further investigation.


Asunto(s)
Complemento C1/metabolismo , Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Aprendizaje Automático , Bibliotecas de Moléculas Pequeñas/química , Complemento C1/antagonistas & inhibidores , Bases de Datos de Compuestos Químicos , Descubrimiento de Drogas/economía , Ensayos Analíticos de Alto Rendimiento/economía , Humanos , Relación Estructura-Actividad Cuantitativa , Bibliotecas de Moléculas Pequeñas/farmacología
12.
Front Immunol ; 9: 558, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29632531

RESUMEN

Two major aspects of systemic lupus erythematosus (SLE) pathogenesis that have yet to be targeted therapeutically are immune complex-initiated complement activation and neutrophil extracellular trap (NET) formation by neutrophils. Here, we report in vitro testing of peptide inhibitor of complement C1 (PIC1) in assays of immune complex-mediated complement activation in human sera and assays for NET formation by human neutrophils. The lead PIC1 derivative, PA-dPEG24, was able to dose-dependently inhibit complement activation initiated by multiple types of immune complexes (IC), including C1-anti-C1q IC, limiting the generation of pro-inflammatory complement effectors, including C5a and membrane attack complex (sC5b-9). In several instances, PA-dPEG24 achieved complete inhibition with complement effector levels equivalent to background. PA-dPEG24 was also able to dose-dependently inhibit NET formation by human neutrophils stimulated by PMA, MPO, or immune complex activated human sera. In several instances PA-dPEG24 achieved complete inhibition with NETosis with quantitation equivalent to background levels. These results suggest that PA-dPEG24 inhibition of NETs occurs by blocking the MPO pathway of NET formation. Together these results demonstrate that PA-dPEG24 can inhibit immune complex activation of the complement system and NET formation. This provides proof of concept that peptides can potentially be developed to inhibit these two important contributors to rheumatologic pathology that are currently untargeted by available therapies.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Activación de Complemento/inmunología , Proteína Inhibidora del Complemento C1/inmunología , Trampas Extracelulares/inmunología , Neutrófilos/inmunología , Péptidos/inmunología , Complejo Antígeno-Anticuerpo/sangre , Complejo Antígeno-Anticuerpo/metabolismo , Complemento C1/inmunología , Complemento C1/metabolismo , Proteína Inhibidora del Complemento C1/metabolismo , Complemento C1q/inmunología , Trampas Extracelulares/metabolismo , Humanos , Microscopía Fluorescente , Neutrófilos/metabolismo , Péptidos/metabolismo
13.
PLoS One ; 13(3): e0193931, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29499069

RESUMEN

Reactive oxygen species (ROS) are natural byproducts of oxidative respiration that are toxic to organs and tissues. To mitigate ROS damage, organisms have evolved a variety of antioxidant systems to counteract these harmful molecules, however in certain pathological conditions these protective mechanisms can be overwhelmed. We have recently demonstrated that Peptide Inhibitor of Complement C1 (PIC1) mitigates peroxidase activity of the heme bearing proteins myeloperoxidase, hemoglobin, and myoglobin through a reversible process. To determine if this property of PIC1 was antioxidant in nature, we tested PIC1 in a number of well-established antioxidant assays. PIC1 showed dose-dependent antioxidant activity in a total antioxidant (TAC) assay, hydroxyl radical antioxidant capacity (HORAC) assay, oxygen radical antioxidant capacity (ORAC) assay as well as the thiobarbituric acid reactive substances (TBARS) assay to screen for PIC1 antioxidant activity in human plasma. The antioxidant activity of PIC1 in the TAC assay, as well as the HORAC/ORAC assay demonstrated that this peptide acts via the single electron transport (SET) and hydrogen atom transfer (HAT) mechanisms, respectively. Consistent with this mechanism of action, PIC1 did not show activity in a metal chelating activity (MCA) assay. PIC1 contains two vicinal cysteine residues and displayed similar antioxidant activity to the well characterized cysteine-containing tripeptide antioxidant molecule glutathione (GSH). Consistent with the role of the cysteine residues in the antioxidant activity of PIC1, oxidation of these residues significantly abrogated antioxidant activity. These results demonstrate that in addition to its described complement inhibiting activity, PIC1 displays in vitro antioxidant activity.


Asunto(s)
Antioxidantes/metabolismo , Proteína Inhibidora del Complemento C1/metabolismo , Transporte de Electrón/fisiología , Hidrógeno/metabolismo , Péptidos/metabolismo , Complemento C1/metabolismo , Glutatión/metabolismo , Hemoglobinas/metabolismo , Humanos , Radical Hidroxilo/metabolismo , Oxidación-Reducción , Peroxidasa/metabolismo , Especies Reactivas de Oxígeno/metabolismo
14.
Front Immunol ; 9: 63, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29434593

RESUMEN

Leishmania donovani, the causative agent of Indian visceral leishmaniasis has to face several barriers of the immune system inside the mammalian host for its survival. The complement system is one of the first barriers and consists of a well-balanced network of proteases including S1A family serine proteases (SPs). Inhibitor of serine peptidases (ISPs) is considered as inhibitor of S1A family serine peptidases and is reported to be present in trypanosomes, including Leishmania. In our previous study, we have deciphered the role of ISPs [LdISP1 and L. donovani inhibitor of serine peptidases 2 (LdISP2)] in the survival of L. donovani inside the sandfly midgut. However, the role of theses ISPs in the survival of L. donovani inside mammalian host still remains elusive. In the present study, we have deciphered the inhibitory effect of LdISPs on the host complement S1A serine peptidases, such as C1r/C1s and MASP1/MASP2. Our study suggested that although both rLdISP1 and rLdISP2 inferred strong interaction with C1complex and MBL-associated serine proteases (MASPs) but rLdISP2 showed the stronger inhibitory effect on MASP2 than rLdISP1. Moreover, we found that rLdISP2 significantly reduces the formation of C3, C5 convertase, and membrane attacking complex (MAC) by lectin pathway (LP) resulting in significant reduction in serum mediated lysis of the parasites. The role of LdISP2 on neutrophil elastase-mediated C5aR signaling was also evaluated. Notably, our results showed that infection of macrophages with ISP2-overexpressed Leishmania parasites significantly induces the expression of C5aR both at the transcript and translational level. Simultaneously, infection with ISP2KD parasites results in downregulation of host PI3K/AKT phosphorylation and increased in IL-12 production. Taken together, our findings clearly suggest that LdISP2 promotes parasite survival inside host by inhibiting MAC formation and complement-mediated lysis via LP and by upregulation of C5aR signaling.


Asunto(s)
Lectinas/metabolismo , Leishmania donovani/fisiología , Leishmaniasis Visceral/metabolismo , Leishmaniasis Visceral/parasitología , Receptor de Anafilatoxina C5a/metabolismo , Serina Endopeptidasas/metabolismo , Transducción de Señal , Biomarcadores , Línea Celular , Complemento C1/inmunología , Complemento C1/metabolismo , Complemento C4a/inmunología , Complemento C4a/metabolismo , Vía Clásica del Complemento/inmunología , Activación Enzimática , Humanos , Lectinas/química , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/parasitología , Modelos Moleculares , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Conformación Proteica , Proteolisis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina Endopeptidasas/química
15.
Immunobiology ; 223(2): 183-190, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29107384

RESUMEN

Leptospirosis is an important zoonosis of global importance caused by bacteria Leptospira spp. Pathogenic Leptospira is resistant to Complement System killing while non-pathogenic Leptospira is rapidly killed by exposure to normal human serum (NHS). Pathogenic Leptospira interact with Complement Regulators such as Factor H, C4b binding protein and Vitronectin avoiding Complement activation and killing by Alternative and Classical Pathways. One important regulator is C1-inhibitor (C1INH) that interacts with C1s or MASPs controlling the cleavage of C4 and C2 molecules, thereby inhibiting the activation of the Classical and Lectin Pathways. In this study, we demonstrate that attenuated, saprophytic and pathogenic Leptospira interact with C1INH that maintain its regulatory capacity of interaction with C1s preventing the activation of Complement system. Although the interaction with C1INH is not crucial for pathogenic Leptospira survival, it seems to be important for the survival of attenuated and saprophytic Leptospira in normal human serum.


Asunto(s)
Proteína Inhibidora del Complemento C1/metabolismo , Complemento C1/metabolismo , Leptospiraceae/inmunología , Leptospirosis/inmunología , Animales , Activación de Complemento , Proteína Inhibidora del Complemento C1/genética , Proteína de Unión al Complemento C4b/metabolismo , Factor H de Complemento/metabolismo , Cadena Alimentaria , Humanos , Evasión Inmune , Leptospiraceae/patogenicidad , Vacunas Atenuadas , Virulencia , Vitronectina/metabolismo , Zoonosis
16.
J Neuroinflammation ; 14(1): 141, 2017 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-28732515

RESUMEN

BACKGROUND: The necessity of including both males and females in molecular neuroscience research is now well understood. However, there is relatively limited basic biological data on brain sex differences across the lifespan despite the differences in age-related neurological dysfunction and disease between males and females. METHODS: Whole genome gene expression of young (3 months), adult (12 months), and old (24 months) male and female C57BL6 mice hippocampus was analyzed. Subsequent bioinformatic analyses and confirmations of age-related changes and sex differences in hippocampal gene and protein expression were performed. RESULTS: Males and females demonstrate both common expression changes with aging and marked sex differences in the nature and magnitude of the aging responses. Age-related hippocampal induction of neuroinflammatory gene expression was sexually divergent and enriched for microglia-specific genes such as complement pathway components. Sexually divergent C1q protein expression was confirmed by immunoblotting and immunohistochemistry. Similar patterns of cortical sexually divergent gene expression were also evident. Additionally, inter-animal gene expression variability increased with aging in males, but not females. CONCLUSIONS: These findings demonstrate sexually divergent neuroinflammation with aging that may contribute to sex differences in age-related neurological diseases such as stroke and Alzheimer's, specifically in the complement system. The increased expression variability in males suggests a loss of fidelity in gene expression regulation with aging. These findings reveal a central role of sex in the transcriptomic response of the hippocampus to aging that warrants further, in depth, investigations.


Asunto(s)
Envejecimiento , Citocinas/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Hipocampo/metabolismo , Microglía/metabolismo , Caracteres Sexuales , Factores de Edad , Animales , Complemento C1/genética , Complemento C1/metabolismo , Biología Computacional , Citocinas/genética , Femenino , Perfilación de la Expresión Génica , Hipocampo/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Componente Principal , ARN Mensajero/metabolismo , Transducción de Señal/fisiología , Transcriptoma
17.
PLoS One ; 12(1): e0169512, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28107369

RESUMEN

Natural Killer cells (NK) are lymphocytes with the potential to recognize and lyse cells which escaped T-cell mediated lysis due to their aberrant HLA expression profiles. Killer cell immunoglobulin-like receptors (KIR) influence NK-cell activity by mediation of activating or inhibitory signals upon interaction with HLA-C (C1, C2) ligands. Therefore, absence of ligands for donor inhibitory KIRs following hematopoietic stem cell transplantation (HSCT) may have an influence on its outcome. Previous studies showed that C1 negative patients have a decreased HSCT outcome. Our study, based on a cohort of 200 C1-negative patients, confirmed these findings for the endpoints: overall survival (OS: HR = 1.41, CI = 1.14-1.74, p = 0.0012), disease free survival (DFS: HR = 1.27, CI = 1.05-1.53, p = 0.015), treatment related mortality (TRM: HR = 1.41, CI = 1.01-1.96, p = 0.04), and relapse incidence (RI: HR = 1.33, CI = 1.01-1.75, p = 0.04) all being inferior when compared to C1-positive patients (n = 1246). Subsequent analysis showed that these findings applied for patients with myeloid malignancies but not for patients with lymphoproliferative diseases (OS: myeloid: HR = 1.51, CI = 1.15-1.99, p = 0.003; lymphoblastic: HR = 1.26, CI = 0.91-1.75, p = 0.16; DFS: myeloid: HR = 1.31, CI = 1.01-1.70, p = 0.04; lymphoblastic: HR = 1.21, CI = 0.90-1.61, p = 0.21; RI: myeloid: HR = 1.31, CI = 1.01-1.70, p = 0.04; lymphoblastic: HR = 1.21, CI = 0.90-1.61, p = 0.21). Interestingly, within the C1-negative patient group, transplantation with KIR2DS2 resulted in better OS (9/10 matched: HR = 0.24, CI = 0.08-0.67, p = 0.007) as well as DFS (9/10 matched: HR = 0,26, CI = 0.11-0.60, p = 0.002), and transplantation with KIR2DS1 positive donors was associated with a decreased RI (HR = 0.30, CI = 0.13-0.69, p = 0.005). TRM was increased when the donor was positive for KIR2DS1 (HR = 2.61, CI = 1.26-5.41, p = 0.001). Our findings suggest that inclusion of KIR2DS1/2/5 and KIR3DS1-genotyping in the unrelated donor search algorithm of C1-ligand negative patients with myeloid malignancies may prove to be of clinical relevance.


Asunto(s)
Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas , Receptores KIR/genética , Donante no Emparentado , Adolescente , Adulto , Anciano , Estudios de Cohortes , Complemento C1/metabolismo , Femenino , Humanos , Células Asesinas Naturales/inmunología , Ligandos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Resultado del Tratamiento , Adulto Joven
18.
J Immunol ; 197(12): 4829-4837, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27807190

RESUMEN

Human IgG1 type I CD20 Abs, such as rituximab and ofatumumab (OFA), efficiently induce complement-dependent cytotoxicity (CDC) of CD20+ B cells by binding of C1 to hexamerized Fc domains. Unexpectedly, we found that type I CD20 Ab F(ab')2 fragments, as well as C1q-binding-deficient IgG mutants, retained an ability to induce CDC, albeit with lower efficiency than for whole or unmodified IgG. Experiments using human serum depleted of specific complement components demonstrated that the observed lytic activity, which we termed "accessory CDC," remained to be dependent on C1 and the classical pathway. We hypothesized that CD20 Ab-induced clustering of the IgM or IgG BCR was involved in accessory CDC. Indeed, accessory CDC was consistently observed in B cell lines expressing an IgM BCR and in some cell lines expressing an IgG BCR, but it was absent in BCR- B cell lines. A direct relationship between BCR expression and accessory CDC was established by transfecting the BCR into CD20+ cells: OFA-F(ab')2 fragments were able to induce CDC in the CD20+BCR+ cell population, but not in the CD20+BCR- population. Importantly, OFA-F(ab')2 fragments were able to induce CDC ex vivo in malignant B cells isolated from patients with mantle cell lymphoma and Waldenström macroglobulinemia. In summary, accessory CDC represents a novel effector mechanism that is dependent on type I CD20 Ab-induced BCR clustering. Accessory CDC may contribute to the excellent capacity of type I CD20 Abs to induce CDC, and thereby to the antitumor activity of such Abs in the clinic.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD20/metabolismo , Linfocitos B/efectos de los fármacos , Vía Clásica del Complemento , Inmunoterapia Adoptiva/métodos , Linfoma de Células B/terapia , Rituximab/metabolismo , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Antígenos CD20/inmunología , Linfocitos B/inmunología , Línea Celular Tumoral , Complemento C1/metabolismo , Humanos , Fragmentos Fab de Inmunoglobulinas/metabolismo , Inmunoglobulina G/genética , Inmunoglobulina G/metabolismo , Inmunoglobulina M/genética , Inmunoglobulina M/metabolismo , Linfoma de Células B/inmunología , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/metabolismo , Rituximab/genética , Rituximab/uso terapéutico
19.
HLA ; 87(2): 89-99, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26840927

RESUMEN

Presensitized kidney transplant recipients are at high-risk for early antibody-mediated rejection. We studied the impact of pre- and post-transplant donor-specific human leukocyte antigen (HLA) antibodies (DSA) and T-cell-activation on the occurrence of antibody-mediated rejection episodes (AMR) and graft loss (AMR-GL) in a unique cohort of 80 desensitized high-risk kidney transplant recipients. Patients with pre-transplant DSA demonstrated more AMR episodes than patients without DSA, but did not show a significantly increased rate of AMR-GL. The rates of AMR and AMR-GL were not significantly increased in patients with complement split product (C1q)-binding pre-transplant DSA. Pre-transplant C1q-DSA became undetectable post-transplant in 11 of 13 (85%) patients; 2 (18%) of these 11 patients showed AMR but no AMR-GL. In contrast, the post-transplant presence of C1q-DSA was associated with significantly higher rates of AMR (86 vs 33 vs 0%; P < 0.001) and AMR-GL (86 vs 0 vs 0%; log-rank P < 0.001) compared with post-transplant DSA without C1q-binding or the absence of DSA. Patients with both pre-transplant DSA and evidence of pre-transplant T-cell-activation as indicated by soluble CD30-positivity showed a significantly increased risk for AMR-GL [HR = 11.1, 95% confidence interval (CI) = 1.68-73.4; log-rank P = 0.013]. In these high-risk patients, AMR-GL was associated with total DSA in combination with T-cell-activation pre-transplant, and de novo or persistent C1q-binding DSA post-transplant.


Asunto(s)
Rechazo de Injerto/sangre , Isoanticuerpos/sangre , Antígeno Ki-1/sangre , Trasplante de Riñón , Activación de Linfocitos , Periodo Preoperatorio , Linfocitos T/metabolismo , Adulto , Anciano , Complemento C1/inmunología , Complemento C1/metabolismo , Femenino , Rechazo de Injerto/inmunología , Humanos , Isoanticuerpos/inmunología , Antígeno Ki-1/inmunología , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Linfocitos T/inmunología
20.
Sci Rep ; 6: 19300, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26758086

RESUMEN

Blood-feeding insects inject potent salivary components including complement inhibitors into their host's skin to acquire a blood meal. Sand fly saliva was shown to inhibit the classical pathway of complement; however, the molecular identity of the inhibitor remains unknown. Here, we identified SALO as the classical pathway complement inhibitor. SALO, an 11 kDa protein, has no homology to proteins of any other organism apart from New World sand flies. rSALO anti-complement activity has the same chromatographic properties as the Lu. longipalpis salivary gland homogenate (SGH)counterparts and anti-rSALO antibodies blocked the classical pathway complement activity of rSALO and SGH. Both rSALO and SGH inhibited C4b deposition and cleavage of C4. rSALO, however, did not inhibit the protease activity of C1s nor the enzymatic activity of factor Xa, uPA, thrombin, kallikrein, trypsin and plasmin. Importantly, rSALO did not inhibit the alternative or the lectin pathway of complement. In conclusion our data shows that SALO is a specific classical pathway complement inhibitor present in the saliva of Lu. longipalpis. Importantly, due to its small size and specificity, SALO may offer a therapeutic alternative for complement classical pathway-mediated pathogenic effects in human diseases.


Asunto(s)
Inactivadores del Complemento/farmacología , Vía Clásica del Complemento/efectos de los fármacos , Proteínas de Insectos/farmacología , Psychodidae/inmunología , Psychodidae/metabolismo , Saliva/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Activación de Complemento/efectos de los fármacos , Complemento C1/antagonistas & inhibidores , Complemento C1/inmunología , Complemento C1/metabolismo , Complemento C4/antagonistas & inhibidores , Complemento C4/inmunología , Complemento C4/metabolismo , Humanos , Proteínas Recombinantes/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA