Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Immunol ; 198(1): 472-480, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27895181

RESUMEN

In the atherosclerotic lesion, macrophages ingest high levels of damaged modified low-density lipoproteins (LDLs), generating macrophage foam cells. Foam cells undergo apoptosis and, if not efficiently cleared by efferocytosis, can undergo secondary necrosis, leading to plaque instability and rupture. As a component of the innate immune complement cascade, C1q recognizes and opsonizes modified forms of LDL, such as oxidized or acetylated LDL, and promotes ingestion by macrophages in vitro. C1q was shown to be protective in an atherosclerosis model in vivo. Therefore, this study aimed to investigate whether ingestion of modified LDL in the presence of C1q alters macrophage foam cell survival or function. In an unbiased transcriptome analysis, C1q was shown to modulate expression of clusters of genes involved in cell death and apoptosis pathways in human monocyte-derived macrophages ingesting modified LDL; this was validated by quantitative PCR in human and murine macrophages. C1q downregulated levels and activity of active caspase-3 and PARP-1 in human and mouse macrophages during ingestion of modified LDL. This led to a measurable increase in survival and decrease in cell death, as measured by alamarBlue and propidium iodide assays, respectively. C1q opsonization also increased phagocytosis and efferocytosis in macrophage foam cells. These data suggest that C1q promotes macrophage survival during ingestion of excess cholesterol, as well as improves foam cell efferocytic function. This may be important in slowing disease progression and provides insight into the protective role of C1q in early atherosclerosis.


Asunto(s)
Apoptosis/inmunología , Complemento C1q/inmunología , Células Espumosas/inmunología , Animales , Aterosclerosis/inmunología , Aterosclerosis/patología , Supervivencia Celular/inmunología , Humanos , Ratones , Reacción en Cadena de la Polimerasa
2.
J Neurosci ; 33(33): 13460-74, 2013 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-23946404

RESUMEN

The decline of cognitive function has emerged as one of the greatest health threats of old age. Age-related cognitive decline is caused by an impacted neuronal circuitry, yet the molecular mechanisms responsible are unknown. C1q, the initiating protein of the classical complement cascade and powerful effector of the peripheral immune response, mediates synapse elimination in the developing CNS. Here we show that C1q protein levels dramatically increase in the normal aging mouse and human brain, by as much as 300-fold. This increase was predominantly localized in close proximity to synapses and occurred earliest and most dramatically in certain regions of the brain, including some but not all regions known to be selectively vulnerable in neurodegenerative diseases, i.e., the hippocampus, substantia nigra, and piriform cortex. C1q-deficient mice exhibited enhanced synaptic plasticity in the adult and reorganization of the circuitry in the aging hippocampal dentate gyrus. Moreover, aged C1q-deficient mice exhibited significantly less cognitive and memory decline in certain hippocampus-dependent behavior tests compared with their wild-type littermates. Unlike in the developing CNS, the complement cascade effector C3 was only present at very low levels in the adult and aging brain. In addition, the aging-dependent effect of C1q on the hippocampal circuitry was independent of C3 and unaccompanied by detectable synapse loss, providing evidence for a novel, complement- and synapse elimination-independent role for C1q in CNS aging.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Complemento C1q/biosíntesis , Animales , Conducta Animal , Western Blotting , Encéfalo/fisiología , Electrofisiología , Potenciales Postsinápticos Excitadores , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Inmunoelectrónica
3.
Inflamm Res ; 63(10): 885-93, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25091012

RESUMEN

OBJECTIVE: Innate immune protein C1q plays a dual role in the chronic inflammatory disease of atherosclerosis. Complement activation via C1q exacerbates pathology in the atherosclerotic lesion in later stages of the disease. However, in early stages of disease C1q is protective. We hypothesize that complement-independent activities of C1q are involved in reprogramming macrophage inflammatory polarization. METHODS: The influence of C1q on macrophage inflammatory responses during clearance of oxLDL was examined. Changes in cytokines at the gene and protein level were measured by quantitative PCR and ELISA assay. RESULTS: C1q modulated cytokine expression in Raw264.7 macrophages during ingestion of oxLDL. Levels of pro-inflammatory cytokines IL-1ß and IL-6 were downregulated by C1q, whereas levels of the anti-inflammatory cytokine IL-10 were increased. In addition, data from an NFκB-luciferase gene reporter assay suggest that C1q suppresses activation of NFκB during lipoprotein clearance in macrophages, providing one mechanism by which C1q downregulates pro-inflammatory cytokine production. CONCLUSIONS: C1q-polarization of macrophages toward an anti-inflammatory (M2-like) phenotype may be important in dampening inflammation in the early atherosclerotic lesion. Further investigation of molecular pathways targeted by C1q may provide novel therapeutic targets for this disease.


Asunto(s)
Complemento C1q/farmacología , Lipoproteínas LDL/metabolismo , Macrófagos/efectos de los fármacos , Animales , Aterosclerosis/metabolismo , Línea Celular , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética
4.
J Immunol ; 188(11): 5682-93, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22523386

RESUMEN

Deficiency in C1q, the recognition component of the classical complement cascade and a pattern recognition receptor involved in apoptotic cell clearance, leads to lupus-like autoimmune diseases characterized by auto-antibodies to self proteins and aberrant innate immune cell activation likely due to impaired clearance of apoptotic cells. In this study, we developed an autologous system using primary human lymphocytes and human monocyte-derived macrophages (HMDMs) to characterize the effect of C1q on macrophage gene expression profiles during the uptake of apoptotic cells. C1q bound to autologous apoptotic lymphocytes modulated expression of genes associated with JAK/STAT signaling, chemotaxis, immunoregulation, and NLRP3 inflammasome activation in LPS-stimulated HMDMs. Specifically, C1q sequentially induced type I IFNs, IL-27, and IL-10 in LPS-stimulated HMDMs and IL-27 in HMDMs when incubated with apoptotic lymphocyte conditioned media. Coincubation with C1q tails prevented the induction of type I IFNs and IL-27 in a dose-dependent manner, and neutralization of type I IFNs partially prevented IL-27 induction by C1q. Finally, C1q decreased procaspase-1 cleavage and caspase-1-dependent cleavage of IL-1ß suggesting a potent inhibitory effect of C1q on inflammasome activation. These results identify specific molecular pathways induced by C1q to suppress macrophage inflammation and provide potential therapeutic targets to control macrophage polarization and thus inflammation and autoimmunity.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Polaridad Celular/inmunología , Complemento C1q/fisiología , Inflamasomas/inmunología , Macrófagos/inmunología , Caspasa 1/metabolismo , Inhibidores de Caspasas , Adhesión Celular/inmunología , Células Cultivadas , Quimiocinas/biosíntesis , Citocinas/biosíntesis , Humanos , Inflamasomas/antagonistas & inhibidores , Interleucina-1beta/metabolismo , Lipopolisacáridos/metabolismo , Linfocitos/citología , Linfocitos/inmunología , Macrófagos/citología , Macrófagos/metabolismo
5.
Biomacromolecules ; 13(4): 974-81, 2012 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-22416762

RESUMEN

Self-assembling protein nanocapsules can be engineered for various bionanotechnology applications. Using the dodecahedral scaffold of the E2 subunit from pyruvate dehydrogenase, we introduced non-native surface cysteines for site-directed functionalization. The modified nanoparticle's structural, assembly, and thermostability properties were comparable to the wild-type scaffold (E2-WT), and after conjugation of poly(ethylene glycol) (PEG) to these cysteines, the nanoparticle remained intact and stable up to 79.7 ± 1.8 °C. PEGylation of particles reduced uptake by human monocyte-derived macrophages and MDA-MB-231 breast cancer cells, with decreased uptake as PEG chain length is increased. In vitro C4-depletion and C5a-production assays yielded 97.6 ± 10.8% serum C4 remaining and 40.1 ± 6.0 ng/mL C5a for E2-WT, demonstrating that complement activation is weak for non-PEGylated E2 nanoparticles. Conjugation of PEG to these particles moderately increased complement response to give 79.7 ± 6.0% C4 remaining and 87.6 ± 10.1 ng/mL C5a. Our results demonstrate that PEGylation of the E2 protein nanocapsules can modulate cellular uptake and induce low levels of complement activation, likely via the classical/lectin pathways.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Activación de Complemento , Macrófagos/metabolismo , Nanocápsulas/química , Línea Celular Tumoral , Cisteína/química , Cisteína/metabolismo , Acetiltransferasa de Residuos Dihidrolipoil-Lisina/química , Acetiltransferasa de Residuos Dihidrolipoil-Lisina/metabolismo , Femenino , Humanos , Macrófagos/citología , Polietilenglicoles/química , Polietilenglicoles/metabolismo , Transporte de Proteínas , Propiedades de Superficie
6.
J Immunol ; 185(7): 3932-9, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20833838

RESUMEN

Atherosclerosis is a chronic inflammatory disorder that is characterized by the accumulation of modified lipoproteins in the arterial intima. C1q and mannan-binding lectin (MBL) are not only recognition components involved in activation of inflammation via the complement cascade, but they are also able to directly modulate phagocyte activation. Studies in C1q(-/-) and MBL(-/-) mice suggest that these molecules play a protective role in the early atherosclerotic lesion in the absence of, or prior to, expression of other complement components. However, in later stages, complement activation becomes an inappropriate inflammatory response, contributing to disease pathology. Therefore, to investigate possible molecular interactions of C1q and MBL in atherosclerotic lesions, we examined the influence of C1q and MBL in the clearance of native and modified lipoproteins by human monocytes and monocyte-derived macrophages. Both C1q and MBL are shown to bind and enhance the monocyte/monocyte-derived macrophage clearance of modified forms of low-density lipoprotein (LDL), including oxidized LDL and acetylated LDL, but not native LDL. Modified forms of LDL activate the classical complement pathway, but no lectin pathway activation was detected. Interestingly, monocytes that ingested modified LDL in the presence of C1q or MBL upregulated surface CD80 and CD31, as well as CCL2 chemokine gene expression. However, C1q and MBL also significantly reduced levels of free cholesterol accumulation in monocytes and human monocyte-derived macrophages that ingested oxidized LDL, while enhancing high-density lipoprotein-specific cholesterol efflux from these cells. These results suggest a novel pathway in which C1q and MBL influence removal and metabolism of atherogenic forms of LDL in the early stages of atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Complemento C1q/metabolismo , Lipoproteínas/metabolismo , Macrófagos/metabolismo , Lectina de Unión a Manosa/metabolismo , Monocitos/metabolismo , Aterosclerosis/inmunología , Separación Celular , Activación de Complemento/inmunología , Complemento C1q/inmunología , Citometría de Flujo , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Inmunidad Innata/inmunología , Lipoproteínas/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Lectina de Unión a Manosa/inmunología , Monocitos/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
J Neurosci ; 30(6): 2077-87, 2010 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-20147536

RESUMEN

Hypoxic-ischemic (HI) brain injury in infants is a leading cause of lifelong disability. We report a novel pathway mediating oxidative brain injury after hypoxia-ischemia in which C1q plays a central role. Neonatal mice incapable of classical or terminal complement activation because of C1q or C6 deficiency or pharmacologically inhibited assembly of membrane attack complex were subjected to hypoxia-ischemia. Only C1q(-/-) mice exhibited neuroprotection coupled with attenuated oxidative brain injury. This was associated with reduced production of reactive oxygen species (ROS) in C1q(-/-) brain mitochondria and preserved activity of the respiratory chain. Compared with C1q(+/+) neurons, cortical C1q(-/-) neurons exhibited resistance to oxygen-glucose deprivation. However, postischemic exposure to exogenous C1q increased both mitochondrial ROS production and mortality of C1q(-/-) neurons. This C1q toxicity was abolished by coexposure to antioxidant Trolox (6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid). Thus, the C1q component of complement, accelerating mitochondrial ROS emission, exacerbates oxidative injury in the developing HI brain. The terminal complement complex is activated in the HI neonatal brain but appeared to be nonpathogenic. These findings have important implications for design of the proper therapeutic interventions against HI neonatal brain injury by highlighting a pathogenic priority of C1q-mediated mitochondrial oxidative stress over the C1q deposition-triggered terminal complement activation.


Asunto(s)
Complemento C1q/fisiología , Hipoxia-Isquemia Encefálica/metabolismo , Mitocondrias/fisiología , Estrés Oxidativo , Animales , Animales Recién Nacidos , Infarto Encefálico/metabolismo , Infarto Encefálico/patología , Antígenos CD59/farmacología , Células Cultivadas , Activación de Complemento , Complemento C1q/genética , Citosol/metabolismo , Femenino , Glucosa/deficiencia , Hipoxia-Isquemia Encefálica/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Oxígeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo
8.
J Biol Chem ; 285(44): 33826-33, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-20736175

RESUMEN

This study was designed to explore the effect of recombinant, membrane-targeted CD59 (rCD59-APT542) on the growth and size of fully developed neovascular complex using the murine model of laser-induced choroidal neovascularization (CNV). CNV was induced by laser photocoagulation in C57BL/6 mice using an argon laser, and the animals received rCD59-APT542 via intravitreal (ivt) route. Western blot analysis, immunohistochemistry, and total complement hemolytic assay demonstrated that exogenously administered rCD59-APT542 was incorporated as well as retained in RPE and choroid and was functionally active in vivo. Single ivt injection during the growth of the CNV (i.e. at day 3 post-laser) resulted in ∼79% inhibition of the further growth of neovascular complex. The size of the CNV complex was significantly (p < 0.05) reduced by the administration of rCD59-APT542 after the CNV complex has fully developed (i.e. at day 7 post-laser). Treatment with rCD59-APT542 blocked the formation of membrane attack complex (MAC), increased apoptosis and decreased cell proliferation in the neovascular complex. On the basis of results presented here we conclude that recombinant membrane targeted CD59 inhibited the growth of the CNV complex and reduced the size of fully developed CNV in the laser-induced mouse model. We propose that a combination of two mechanisms: increased apoptosis and decreased cell proliferation, both resulting from local inhibition of MAC, may be responsible for inhibition of CNV by rCD59-APT542.


Asunto(s)
Antígenos CD59/metabolismo , Neovascularización Coroidal/metabolismo , Neovascularización Patológica/metabolismo , Proteínas Recombinantes/química , Animales , Apoptosis , Membrana Celular/metabolismo , Proliferación Celular , Proteínas del Sistema Complemento/química , Inmunohistoquímica , Inflamación , Degeneración Macular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
9.
J Immunol ; 183(10): 6175-85, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19864605

RESUMEN

C1q, the first component of the classical complement pathway, is also a pattern recognition receptor involved in the recognition and clearance of apoptotic cells. C1q deficiency in humans leads to development of lupus-like autoimmune disease, and it has been speculated that impaired clearance of apoptotic cells may contribute to disease development. Since phagocytes initiate specific and appropriate immune responses as a result of initial ligand-receptor interactions, regulation of gene expression by C1q may also contribute to the sculpting of an immune response to the ingested "self-Ags." In this study, the role of C1q in apoptotic cell clearance and subsequent modulation of cytokine release by phagocytes was assessed including donor matched human monocytes, monocyte-derived macrophages (HMDMs), and dendritic cells (DCs). First, C1q binding is much greater to late compared with early apoptotic cells. Second, C1q binding to apoptotic cells significantly enhanced the levels of ingestion by monocytes but had no effect on HMDM and DC uptake. Third, in the presence of serum, C1q bound to apoptotic cells, activated the complement pathway, leading to C3b deposition, and enhancement of uptake of apoptotic cells by monocytes, HMDMs, and DCs. Finally, although C1q, either immobilized on a plate or bound to apoptotic cells, modulates the LPS-induced cytokine levels released by human monocytes, HMDMs, and DCs toward a more limited immune response, both the degree and direction of modulation differed significantly depending on the differentiation state of the phagocyte, providing further evidence of the integration of these cell- and environment-specific signals in determining appropriate immune responses.


Asunto(s)
Complemento C1q/inmunología , Complemento C3/inmunología , Células Dendríticas/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Fagocitosis/inmunología , Apoptosis/inmunología , Complemento C1q/metabolismo , Complemento C3/metabolismo , Vía Clásica del Complemento/inmunología , Citocinas/efectos de los fármacos , Citocinas/inmunología , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Humanos , Células Jurkat , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Fagocitosis/efectos de los fármacos
10.
J Neurochem ; 112(3): 733-43, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19919576

RESUMEN

The expression of C1q, a recognition molecule of the complement system, is up-regulated following neuronal injury and is detected early in neurodegenerative disorders such as Alzheimer's disease. This multimeric protein triggers an enhancement of phagocytosis of suboptimally opsonized targets by microglia, the phagocytic cells of the CNS, similar to other phagocytes, enhances the uptake of apoptotic cells in peripheral phagocytes, and suppresses inflammatory cytokine production in human monocytes, macrophages and dendritic cells in the absence of activation of the entire complement cascade. The goal of this study was to determine if C1q could influence the inflammatory response to injury in the CNS, using primary rat microglia and neurons. The data show that microglia preferentially ingest apoptotic cells in comparison to live cells, like other professional phagocytes, that microglial ingestion of apoptotic neurons and neuronal blebs is enhanced by the presence of normal serum and that these enhanced levels of uptake are diminished in serum depleted of C1q. In addition, purified C1q bound to apoptotic neurons and neuronal blebs in a dose dependent manner, and alone triggered a significant enhancement of uptake by microglia. Microglia added to C1q coated wells or fed apoptotic neurons or neuronal blebs coated with C1q suppressed the lipopolysaccharide-induced production of proinflammatory cytokines interleukin (IL)-1alpha, IL-1beta, IL-6 and TNF-alpha, while the presence of C1q enhanced levels of the chemokine MCP-1/CCL2. The data are consistent with a protective role for C1q in the CNS during early stages of cell death by enhancing microglial clearance of apoptotic cells and suppressing proinflammatory cytokines.


Asunto(s)
Apoptosis/fisiología , Vesícula/metabolismo , Complemento C1q/farmacología , Citocinas/metabolismo , Neuroglía/fisiología , Neuronas/efectos de los fármacos , Fagocitosis/fisiología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Antígeno CD11b/metabolismo , Células Cultivadas , Complemento C1q/genética , Complemento C1q/metabolismo , Medio de Cultivo Libre de Suero/farmacología , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Lipopolisacáridos/farmacología , Neuronas/fisiología , Fagocitosis/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
11.
Mol Immunol ; 117: 65-72, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31739194

RESUMEN

Complement protein C1q plays a dual role in a number of inflammatory diseases such as atherosclerosis. While in later stages classical complement pathway activation by C1q exacerbates disease progression, C1q also plays a beneficial role in early disease. Independent of its role in complement activation, we and others have identified a number of potentially beneficial interactions of C1q with phagocytes in vitro, including triggering phagocytosis of cellular and molecular debris and polarizing macrophages toward an anti-inflammatory phenotype. These interactions may also be important in preventing autoimmunity. Here, we characterize variants of recombinant human C1q (rC1q) which no longer initiate complement activation, through mutation of the C1r2C1s2 interaction site. For insight into the structural location of the site of C1q that is important for interaction with phagocytes, we investigated the effect of these mutations on phagocytosis and macrophage inflammatory polarization, as compared to wild-type C1q. Phagocytosis of antibody coated sheep erythrocytes and oxidized LDL was measured in human monocytes and monocyte-derived macrophages (HMDM) respectively that had interacted with rC1q wild-type or variants. Secreted levels of cytokines were also measured in C1q stimulated HMDM. All variants of C1q increased phagocytosis in HMDM compared to controls, similar to native or wild-type rC1q. In addition, levels of certain pro-inflammatory cytokines and chemokines secreted by HMDM were modulated in cells that interacted with C1q variants, similar to wild-type rC1q and native C1q. This includes downregulation of IL-1α, IL-1ß, TNFα, MIP-1α, and IL-12p40 by native and rC1q in both resting and M1-polarized HMDM. This suggests that the site responsible for C1q interaction with phagocytes is independent of the C1r2C1s2 interaction site. Further studies with these classical pathway-null variants of C1q should provide greater understanding of the complement-independent role of C1q, and allow for potential therapeutic exploitation.


Asunto(s)
Complemento C1q/química , Complemento C1q/inmunología , Vía Clásica del Complemento/inmunología , Macrófagos/inmunología , Fagocitosis/inmunología , Humanos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
12.
Mol Immunol ; 45(11): 3244-52, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18400300

RESUMEN

Evidence has been accumulating for a role of inflammation in the development of Alzheimer's disease (AD), a progressive neurodegenerative disorder causing a common form of dementia in the elderly. C1q, part of the initiation component of the classical complement pathway (CCP), is associated with beta-sheet, fibrillar amyloid plaques in AD brain. In vitro, beta-amyloid peptide in fibrillar beta-sheet conformation (fAbeta) can activate CCP via interaction of specific negatively charged amino acids of the beta-amyloid fibril with human C1q. Previous results using peptide inhibitors led to the hypothesis that a highly positively charged domain consisting of three arginine residues, such as that present in the N-terminal collagen-like region of the human C1q A chain, may be critical for the activation event. However, mouse C1q A chain lacks two of the three arginines in the corresponding C1q A chain collagen-like region. To test the hypothesis that this divergent activation domain results in a weaker C' activation and thus may contribute to the lower neuronal loss observed in transgenic mouse models of AD, a partially humanized C1q A chain knock-in mouse was generated. The mouse C1q A chain gene was modified by homologous recombination to replace 4 residues in the 13-20 amino acid region to mimic the corresponding sequence from human A chain. No significant differences in the expression of C1q were found in sera from mice homozygous for the humanized C1q A chain compared to littermate wild type mice. Two distinct C1 activation assays demonstrated that activation by fAbeta was not significantly different in the homozygous humanized C1q A chain mice. Activation of C1 by DNA, previously hypothesized to interact with this C1q A chain arginine-rich sequence was also not significantly different in the knock-in mouse. Molecular modeling based on the published crystal structure of human C1q B chain globular head and a beta-sheet model for fibrillar amyloid suggests an alternative arginine ladder in the globular head domain may provide the functional C1 activating interaction domains. The humanized C1q mouse generated here should provide a better animal model for assessing the mechanisms of C1 activation and the contribution of C1q to human health and disease.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Activación de Complemento/efectos de los fármacos , Complemento C1q/genética , Complemento C1q/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos/inmunología , Complemento C1q/química , Complemento C1q/aislamiento & purificación , ADN , Hemólisis/efectos de los fármacos , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular
13.
Sci Rep ; 9(1): 16875, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31728030

RESUMEN

The higher-order architecture observed in biological systems, like viruses, is very effective in nucleic acid transport. The replications of this system has been attempted with both synthetic and naturally occurring polymers with mixed results. Here we describe a peptide/siRNA quaternary complex that functions as an siRNA delivery system. The rational design of a peptide assembly is inspired by the viral capsids, but not derived from them. We selected the collagen peptide (COL) to provide the structural stability and the folding framework, and hybridize it with the cell penetrating peptide (CPP) that allows for effective penetration of biological barriers. The peptide/siRNA quaternary complex forms stoichiometric, 10 nm nanoparticles, that show fast cellular uptake (<30 min), effective siRNA release, and gene silencing. The complex provides capsid-like protection for siRNA against nucleases without being immunostimulatory, or cytotoxic. Our data suggests that delivery vehicles based on synthetic quaternary structures that exhibit higher-order architecture may be effective in improving delivery and release of nucleic acid cargo.


Asunto(s)
Péptidos de Penetración Celular/metabolismo , Colágeno/metabolismo , Silenciador del Gen , Técnicas de Transferencia de Gen , Polímeros/metabolismo , ARN Interferente Pequeño/genética , Animales , Transporte Biológico , Materiales Biomiméticos/química , Materiales Biomiméticos/metabolismo , Cápside/química , Carbocianinas/química , Carbocianinas/metabolismo , Péptidos de Penetración Celular/química , Colágeno/química , Fluoresceína-5-Isotiocianato/química , Fluoresceína-5-Isotiocianato/metabolismo , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Genes Reporteros , Proteínas Fluorescentes Verdes/antagonistas & inhibidores , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Hidroxiprolina/química , Hidroxiprolina/metabolismo , Ratones , Conformación Molecular , Células 3T3 NIH , Nanopartículas/química , Polímeros/química , ARN Interferente Pequeño/metabolismo
14.
Mol Immunol ; 44(1-3): 33-43, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16908067

RESUMEN

C1q and mannose binding lectin, members of the "defense collagen" family, are pattern recognition molecules that can trigger rapid enhanced phagocytosis resulting in efficient containment of pathogens or clearance of cellular debris, apoptotic cells and immune complexes. In addition, interaction of C1q and mannose binding lectin with the phagocyte alters subsequent phagocyte cytokine synthesis, and thus may have important implications in directing acute inflammation as well as long-term protective immunity. The importance of the role of defense collagens in phagocytosis of apoptotic cells is highlighted by studies in vivo of mice deficient in C1q, pulmonary surfactant D and mannose binding lectin in which there is delayed clearance of apoptotic cells. Indeed, deficiency of C1q is a risk factor for the development of autoimmunity in both humans and mice, consistent with the hypothesis that inefficient clearance of apoptotic cells results in release of autoantigens and contributes to the pathology associated with autoimmune diseases such as systemic lupus erythematosus. Further understanding of the importance of C1q and mannose binding lectin in the clearance of apoptotic cells and regulation of cytokine synthesis and identification of the receptors implicated in mediating these processes should provide novel targets for therapeutic intervention in the control and manipulation of the immune response in terms of both host defense against infectious disease and tissue repair and remodeling.


Asunto(s)
Complemento C1q/fisiología , Inmunidad Innata , Lectina de Unión a Manosa/fisiología , Receptores de Reconocimiento de Patrones/fisiología , Transducción de Señal , Animales , Antígenos CD/fisiología , Calreticulina/fisiología , Colágeno/metabolismo , Complemento C1q/genética , Citocinas/biosíntesis , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Lectina de Unión a Manosa/genética , Fagocitosis
15.
J Leukoc Biol ; 80(1): 107-16, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16617157

RESUMEN

It has recently been recognized that the innate immune response, the powerful first response to infection, has significant influence in determining the nature of the subsequent adaptive immune response. C1q, mannose-binding lectin (MBL), and other members of the defense collagen family of proteins are pattern recognition molecules, able to enhance the phagocytosis of pathogens, cellular debris, and apoptotic cells in vitro and in vivo. Humans deficient in C1q inevitably develop a lupus-like autoimmune disorder, and studies in C1q knockout mice demonstrate a deficiency in the clearance of apoptotic cells with a propensity for autoimmune responses. The data presented here show that under conditions in which phagocytosis is enhanced, C1q and MBL modulate cytokine production at the mRNA and protein levels. Specifically, these recognition molecules of the innate immune system contribute signals to human peripheral blood mononuclear cells, leading to the suppression of lipopolysaccharide-induced proinflammatory cytokines, interleukin (IL)-1alpha and IL-1beta, and an increase in the secretion of cytokines IL-10, IL-1 receptor antagonist, monocyte chemoattractant protein-1, and IL-6. These data support the hypothesis that defense collagen-mediated suppression of a proinflammatory response may be an important step in the avoidance of autoimmunity during the clearance of apoptotic cells.


Asunto(s)
Complemento C1q/inmunología , Citocinas/biosíntesis , Inmunidad Innata/inmunología , Lectina de Unión a Manosa/inmunología , Monocitos/inmunología , Animales , Complemento C1q/deficiencia , Citocinas/inmunología , Humanos , Lipopolisacáridos/inmunología , Ratones , Ratones Noqueados , ARN Mensajero/biosíntesis , ARN Mensajero/inmunología , Regulación hacia Arriba/inmunología
16.
Environ Pollut ; 223: 42-50, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28153417

RESUMEN

Chronic organochlorine (OC) exposure has been shown to cause immune impairment in numerous vertebrate species. To determine if elasmobranchs exhibited compromised immunity due to high OC contamination along the coastal mainland of southern California, innate immune function was compared in round stingrays (Urobatis halleri) collected from the mainland and Santa Catalina Island. Proliferation and phagocytosis of peripheral blood, splenic, and epigonal leukocytes were assessed. Percent phagocytosis and mean fluorescence intensity (MFI) were evaluated by quantifying % leukocytes positive for, and relative amounts of ingested fluorescent E. coli BioParticles. Total cell proliferation differed between sites, with mainland rays having a higher cell concentration in whole blood. ∑PCB load explained significantly higher % phagocytosis in blood of mainland rays, while ∑PCB and ∑pesticide loads described increased splenic % phagocytosis and MFI in the mainland population. Data provides evidence of strong OC-correlated immunostimulation; however, other site-specific environmental variables may be contributing to the observed effects.


Asunto(s)
Hidrocarburos Clorados/toxicidad , Rajidae , Contaminantes Químicos del Agua/toxicidad , Animales , California , Elasmobranquios , Monitoreo del Ambiente , Escherichia coli , Masculino
17.
Data Brief ; 9: 362-367, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27699187

RESUMEN

We characterized the transcriptional effects of complement opsonization on foam cell formation in human monocyte-derived macrophages (HMDM). RNA-sequencing was used to identify the pathways modulated by complement protein C1q during HMDM ingestion of the atherogenic lipoproteins oxidized low density lipoprotein (oxLDL) and acetylated low density lipoprotein (acLDL). All raw data were submitted to the MIAME-compliant database Gene Expression Omnibus (accession number GEO: GSE80442; http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE80442). Data presented here include Venn diagram overviews of up- and down-regulated genes for each condition tested, gene ontology analyses of biological processes, molecular functions and cellular components and KEGG pathway analysis. Further investigation of the pathways modulated by C1q in HMDM during ingestion of atherogenic lipoproteins and their functional relevance are described in "Macrophage molecular signaling and inflammatory responses during ingestion of atherogenic lipoproteins are modulated by complement protein C1q" (M.M. Ho, A. Manughian-Peter, W.R. Spivia, A. Taylor, D.A. Fraser, 2016) [1].

18.
Atherosclerosis ; 253: 38-46, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27573737

RESUMEN

BACKGROUND AND AIMS: This study investigated the effect of innate immune protein C1q on macrophage programmed responses during the ingestion of atherogenic lipoproteins. C1q plays a dual role in atherosclerosis where activation of complement by C1q is known to drive inflammation and promote disease progression. However, C1q is atheroprotective in early disease using mouse models. Our previous studies have highlighted a non-complement associated role for C1q in polarizing macrophages towards an M2-like anti-inflammatory phenotype during ingestion of targets such as atherogenic lipoproteins. This study aims to investigate the molecular mechanisms involved. METHODS: We investigated the molecular signaling mechanisms involved in macrophage polarization using an unbiased examination of gene expression profiles in human monocyte derived macrophages ingesting oxidized or acetylated low density lipoproteins in the presence or absence of C1q. RESULTS: Expression of genes involved in Janus kinase and signal transducer and activator of transcription (JAK-STAT) signaling, peroxisome proliferator activating receptor (PPAR) signaling and toll-like receptor (TLR) signaling were modulated by C1q in this screen. C1q was also shown to significantly suppress JAK-STAT pathway activation (a maximum 55% ± 13% reduction, p = 0.044) and increase transcriptional activation of PPARs (a maximum 229% ± 54% increase, p = 0.0002), consistent with an M2-like polarized response. These pathways were regulated in macrophages by C1q bound to different types of modified atherogenic lipoprotein and led to a reduction in the release of pro-inflammatory cytokine IL-6. CONCLUSIONS: This study identifies potential molecular mechanisms for the beneficial role C1q plays in early atherosclerosis.


Asunto(s)
Complemento C1q/metabolismo , Lipoproteínas/metabolismo , Macrófagos/metabolismo , Transducción de Señal , Animales , Aterosclerosis , Núcleo Celular/metabolismo , Progresión de la Enfermedad , Humanos , Inmunidad Innata , Inflamación , Interleucina-6/metabolismo , Ratones , Oxígeno/química , PPAR gamma/metabolismo , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Fenotipo , Células RAW 264.7 , Factor de Transcripción STAT1/metabolismo , Transcripción Genética
19.
Protein Sci ; 11(10): 2512-21, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12237472

RESUMEN

Inappropriate or unregulated activation of complement can contribute to pathology in inflammatory diseases. Previous studies have shown that soluble recombinant regulators of complement are effective in animal models and some human diseases. However, limitations include cost, rapid clearance, and unwanted systemic effects. To avoid some of these problems, bacterial expression of regulators has been optimized and methods for the addition of a membrane-targeting moiety to the complement regulator developed. When administered directly to sites of inflammation, membrane-targeted human regulators are retained and inhibit complement-activation locally. To test the efficacy of membrane-targeted complement regulators in vivo, we have undertaken the expression and membrane targeting of the rat-complement regulator Crry. A soluble recombinant form of Crry, containing only the first four short consensus repeats, was expressed in a mammalian expression system and shown to be functional as a fluid phase regulator. To generate the quantities required for testing in vivo, Crry was expressed in bacteria and refolded successfully. Refolded protein had full-complement regulatory activity in vitro. Attachment of a membrane address tag conferred membrane-binding capacity and greatly increased complement regulatory function in vitro. This novel anticomplement agent can now be applied to rat models of arthritis and other inflammatory diseases.


Asunto(s)
Membrana Celular/metabolismo , Receptores de Complemento/genética , Animales , Antígenos de Superficie , Células CHO , Clonación Molecular , Proteínas del Sistema Complemento/metabolismo , Cricetinae , Escherichia coli , Señales de Clasificación de Proteína/genética , Ratas , Receptores de Superficie Celular , Receptores de Complemento/biosíntesis , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética
20.
Front Immunol ; 5: 402, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25191325

RESUMEN

Complement is a critical system of enzymes, regulatory proteins, and receptors that regulates both innate and adaptive immune responses. Natural mutations in complement molecules highlight their requirement in regulation of a variety of human conditions including infectious disease and autoimmunity. As sentinels of the immune system, macrophages are specialized to respond to infectious microbes, as well as normal and altered self, and dictate appropriate immune responses. Complement components such as anaphylatoxins (C3a and C5a) and opsonins [C3b, C1q, mannan binding lectin (MBL)] influence macrophage responses. While anaphylatoxins C3a and C5a trigger inflammasome activation, opsonins such as C1q and related molecules (MBL and adiponectin) downregulate inflammasome activation and inflammation, and upregulate engulfment of apoptotic cells consistent with a pro-resolving or M2 macrophage phenotype. This review summarizes our current understanding of the influence of the complement system on macrophage polarization with an emphasis on C1q and related molecules.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA