Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
Add more filters

Country/Region as subject
Publication year range
1.
BMC Nephrol ; 22(1): 43, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33514328

ABSTRACT

BACKGROUND: Immunoglobulin A nephropathy (IgAN) is identified as mesangial IgA deposition and is usually accompanied by other immunofluorescence deposits. The impact of immunofluorescent features in IgAN patients, however, remains unclear. METHODS: Baseline clinicopathologic parameters and renal outcomes of 337 patients diagnosed with IgAN between January 2009 and December 2015 were analyzed. We then categorized these patients into four groups: without immunofluorescence deposits, mesangial-only, mesangial and glomerular capillary loops (GCLs), and GCLs-only. The study endpoint was end-stage kidney disease (ESKD) or a ≥ 50% decline in the estimated glomerular filtration rate (eGFR). Kaplan-Meier and Cox regression analyses were performed to calculate renal survival. RESULTS: Of the 337 IgAN patients, women comprised 57.0%. Compared to patients with IgA deposition in the mesangial-only group, patients with IgA deposition in the mesangial +GCLs group were much heavier, and exhibited higher systolic blood pressure, lower serum IgG levels, and heavier proteinuria (all P < 0.05). Patients with IgG deposition in the mesangial +GCLs group presented with higher levels of cholesterol, heavier proteinuria than IgG deposition in the mesangial-only group (both P < 0.05). Compared with the mesangial-only group exhibiting C3 deposits, patients in the mesangial +GCLs group with C3 deposition had a higher systolic blood pressure (P = 0.028). A total of 38 patients (11.3%) continued to the study endpoint after a median follow-up time of 63.5 months (range,49.8-81.4). Kaplan-Meier analysis and Cox regression analysis showed that C1q deposition in the mesangial +GCLs group predicted a poor renal prognosis. CONCLUSIONS: IgA and IgG deposits in the mesangial region and GCLs were associated with more unfavorable clinical and histopathologic findings in IgAN patients. C1q deposition in the mesangial region and GCLs predicted a poor renal prognosis. However, the impact of the pattern of immunofluorescence deposits on renal outcomes remains to be proven by further investigation.


Subject(s)
Complement C1q/physiology , Glomerular Mesangium/blood supply , Glomerulonephritis, IGA/pathology , Immunoglobulin A/analysis , Kidney Glomerulus/metabolism , Adult , Capillaries , Female , Fluorescent Antibody Technique , Glomerulonephritis, IGA/immunology , Glomerulonephritis, IGA/metabolism , Humans , Male , Prognosis , Retrospective Studies , Young Adult
2.
Mol Psychiatry ; 24(9): 1383-1397, 2019 09.
Article in English | MEDLINE | ID: mdl-30283031

ABSTRACT

TYROBP/DAP12 forms complexes with ectodomains of immune receptors (TREM2, SIRPß1, CR3) associated with Alzheimer's disease (AD) and is a network hub and driver in the complement subnetwork identified by multi-scale gene network studies of postmortem human AD brain. Using transgenic or viral approaches, we characterized in mice the effects of TYROBP deficiency on the phenotypic and pathological evolution of tauopathy. Biomarkers usually associated with worsening clinical phenotype (i.e., hyperphosphorylation and increased tauopathy spreading) were unexpectedly increased in MAPTP301S;Tyrobp-/- mice despite the improved learning behavior and synaptic function relative to controls with normal levels of TYROBP. Notably, levels of complement cascade initiator C1q were reduced in MAPTP301S;Tyrobp-/- mice, consistent with the prediction that C1q reduction exerts a neuroprotective effect. These observations suggest a model wherein TYROBP-KO-(knock-out)-associated reduction in C1q is associated with normalized learning behavior and electrophysiological properties in tauopathy model mice despite a paradoxical evolution of biomarker signatures usually associated with neurological decline.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Alzheimer Disease/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/metabolism , Animals , Animals, Genetically Modified , Brain/metabolism , Complement C1q/metabolism , Complement C1q/physiology , Disease Models, Animal , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Mice, Knockout , Mice, Transgenic , Microglia/metabolism , Phenotype , Phosphorylation , Plaque, Amyloid/metabolism , Tauopathies/genetics , tau Proteins/metabolism
3.
Sheng Li Xue Bao ; 71(3): 471-477, 2019 Jun 25.
Article in Zh | MEDLINE | ID: mdl-31218338

ABSTRACT

The C1q family is one of the subcomponents of the C1 complex that recognizes immune complexes and initiates the classical pathway of the complement system. In addition, as a pattern recognition receptor (PRR), the C1q family binds to a wide variety of ligands. As a member of the C1q family, the secretory C1q includes several subtypes. The main subtypes are cerebellin (Cbln) and C1q-like protein (C1ql). In the nervous system, secretory C1q is involved in the formation and regulation of various types of synapses, thus secretory C1q is closely related to diseases of the central nervous system. In this article, we review the role of secretory C1q in synapse formation and regulation, and its relationship with some diseases of the central nervous system.


Subject(s)
Complement C1q/physiology , Synapses/physiology , Antigen-Antibody Complex , Central Nervous System , Humans
4.
J Immunol ; 197(6): 2051-60, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27591336

ABSTRACT

Complement is a network of soluble and cell surface-associated proteins that gives rise to a self-amplifying, yet tightly regulated system with fundamental roles in immune surveillance and clearance. Complement becomes activated on the surface of nonself cells by one of three initiating mechanisms known as the classical, lectin, and alternative pathways. Evasion of complement function is a hallmark of invasive pathogens and hematophagous organisms. Although many complement-inhibition strategies hinge on hijacking activities of endogenous complement regulatory proteins, an increasing number of uniquely evolved evasion molecules have been discovered over the past decade. In this review, we focus on several recent investigations that revealed mechanistically distinct inhibitors of the classical pathway. Because the classical pathway is an important and specific mediator of various autoimmune and inflammatory disorders, in-depth knowledge of novel evasion mechanisms could direct future development of therapeutic anti-inflammatory molecules.


Subject(s)
Complement Pathway, Classical , Immune Evasion , Animals , Complement C1/physiology , Complement C1q/physiology , Complement C3-C5 Convertases/antagonists & inhibitors , Humans
5.
Proc Natl Acad Sci U S A ; 111(11): 4209-14, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24591625

ABSTRACT

We have previously shown that C1q is expressed on endothelial cells (ECs) of newly formed decidual tissue. Here we demonstrate that C1q is deposited in wound-healing skin in the absence of C4 and C3 and that C1q mRNA is locally expressed as revealed by real-time PCR and in situ hybridization. C1q was found to induce permeability of the EC monolayer, to stimulate EC proliferation and migration, and to promote tube formation and sprouting of new vessels in a rat aortic ring assay. Using a murine model of wound healing we observed that vessel formation was defective in C1qa(-/-) mice and was restored to normal after local application of C1q. The mean vessel density of wound-healing tissue and the healed wound area were significantly increased in C1q-treated rats. On the basis of these results we suggest that C1q may represent a valuable therapeutic agent that can be used to treat chronic ulcers or other pathological conditions in which angiogenesis is impaired, such as myocardial ischemia.


Subject(s)
Complement C1q/physiology , Endothelial Cells/drug effects , Neovascularization, Physiologic/genetics , Wound Healing/genetics , Animals , Cell Proliferation/drug effects , Complement C1q/genetics , Complement C1q/pharmacology , DNA Primers/genetics , Endothelial Cells/physiology , Enzyme-Linked Immunosorbent Assay , Human Umbilical Vein Endothelial Cells , Humans , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic/physiology , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Wound Healing/physiology
6.
J Immunol ; 188(2): 902-15, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22147767

ABSTRACT

Immune complexes (ICs) play a pivotal role in causing inflammation in systemic lupus erythematosus (SLE). Yet, it remains unclear what the dominant blood cell type(s) and inflammation-related gene programs stimulated by lupus ICs are. To address these questions, we exposed normal human PBMCs or CD14(+) isolated monocytes to SLE ICs in the presence or absence of C1q and performed microarray analysis and other tests for cell activation. By microarray analysis, we identified genes and pathways regulated by SLE ICs that are both type I IFN dependent and independent. We also found that C1q-containing ICs markedly reduced expression of the majority of IFN-response genes and also influenced the expression of multiple other genes induced by SLE ICs. Surprisingly, IC activation of isolated CD14(+) monocytes did not upregulate CD40 and CD86 and only modestly stimulated inflammatory gene expression. However, when monocyte subsets were purified and analyzed separately, the low-abundance CD14(dim) ("patrolling") subpopulation was more responsive to ICs. These observations demonstrate the importance of plasmacytoid dendritic cells, CD14(dim) monocytes, and C1q as key regulators of inflammatory properties of ICs and identify many pathways through which they act.


Subject(s)
Antigen-Antibody Complex/physiology , Complement C1q/physiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Dendritic Cells/pathology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interferon-alpha/biosynthesis , Lipopolysaccharide Receptors/biosynthesis , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/biosynthesis , U937 Cells
7.
J Immunol ; 188(11): 5682-93, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22523386

ABSTRACT

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.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Cell Polarity/immunology , Complement C1q/physiology , Inflammasomes/immunology , Macrophages/immunology , Caspase 1/metabolism , Caspase Inhibitors , Cell Adhesion/immunology , Cells, Cultured , Chemokines/biosynthesis , Cytokines/biosynthesis , Humans , Inflammasomes/antagonists & inhibitors , Interleukin-1beta/metabolism , Lipopolysaccharides/metabolism , Lymphocytes/cytology , Lymphocytes/immunology , Macrophages/cytology , Macrophages/metabolism
8.
J Neurosci ; 31(9): 3459-69, 2011 Mar 02.
Article in English | MEDLINE | ID: mdl-21368058

ABSTRACT

Activation of the complement cascade, a powerful effector mechanism of the innate immune system, is associated with neuroinflammation but also with elimination of inappropriate synapses during development. Synthesis of C1q, a recognition component of the complement system, occurs in brain during ischemia/reperfusion and Alzheimer's disease, suggesting that C1q may be a response to injury. In vitro, C1q, in the absence of other complement proteins, improves neuronal viability and neurite outgrowth and prevents ß-amyloid-induced neuronal death, suggesting that C1q may have a direct neuroprotective role. Here, investigating the molecular basis for this neuroprotection in vitro, addition of C1q to rat primary cortical neurons significantly upregulated expression of genes associated with cholesterol metabolism, such as cholesterol-25-hydroxylase and insulin induced gene 2, and transiently decreased cholesterol levels in neurons, known to facilitate neurite outgrowth. In addition, the expression of syntaxin-3 and its functional association with synaptosomal-associated protein 25 was increased. C1q also increased the nuclear translocation of cAMP response element-binding protein and CCAAT/enhancer-binding protein-δ (C/EBP-δ), two transcription factors involved in nerve growth factor (NGF) expression and downregulated specific microRNAs, including let-7c that is predicted to target (and thus inhibit) NGF and neurotrophin-3 (NT-3) mRNA. Accordingly, C1q increased expression of NGF and NT-3, and small interfering RNA inhibition of C/EBP-δ, NGF, or NT-3 expression prevented the C1q-dependent neurite outgrowth. No such neuroprotective effect is seen in the presence of C3a or C5a. Finally, the induced neuronal gene expression required conformationally intact C1q. These results show that C1q can directly promote neuronal survival, thereby demonstrating new interactions between immune proteins and neuronal cells that may facilitate neuroprotection.


Subject(s)
Complement C1q/physiology , Gene Expression Regulation, Developmental , MicroRNAs/biosynthesis , Neurons/physiology , Neuroprotective Agents , Animals , Cell Survival/immunology , Cells, Cultured , Gene Expression Regulation, Developmental/immunology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Neurons/immunology , Neurons/pathology , Rats , Rats, Sprague-Dawley
9.
J Biol Chem ; 286(18): 15652-65, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21378161

ABSTRACT

Members of the C1q/TNF family play important and diverse roles in the immune, endocrine, skeletal, vascular, and sensory systems. Here, we identify and characterize CTRP13, a new and extremely conserved member of the C1q/TNF family. CTRP13 is preferentially expressed by adipose tissue and the brain in mice and predominantly by adipose tissue in humans. Within mouse adipose tissue, CTRP13 is largely expressed by cells of the stromal vascular compartment. Due to sexually dimorphic expression patterns, female mice have higher transcript and circulating CTRP13 levels than males. CTRP13 transcript and circulating levels are elevated in obese male mice, suggesting a potential role in energy metabolism. The insulin-sensitizing drug rosiglitazone also increases the expression of CTRP13 in adipocytes, which correlates with the insulin-sensitizing action of CTRP13. In a heterologous expression system, CTRP13 is secreted as a disulfide-linked oligomeric protein. When co-expressed, CTRP13 forms heteromeric complexes with a closely related family member, CTRP10. This heteromeric association does not involve conserved N-terminal Cys residues. Functional studies using purified recombinant protein demonstrated that CTRP13 is an adipokine that promotes glucose uptake in adipocytes, myotubes, and hepatocytes via activation of the AMPK signaling pathway. CTRP13 also ameliorates lipid-induced insulin resistance in hepatocytes through suppression of the SAPK/JNK stress signaling that impairs the insulin signaling pathway. Further, CTRP13 reduces glucose output in hepatocytes by inhibiting the mRNA expression of gluconeogenic enzymes, glucose-6-phosphatase and the cytosolic form of phosphoenolpyruvate carboxykinase. These results provide the first functional characterization of CTRP13 and establish its importance in glucose homeostasis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipokines/physiology , Complement C1q/physiology , Energy Metabolism/physiology , Fatty Acids/metabolism , MAP Kinase Kinase 4/metabolism , Signal Transduction/physiology , Tumor Necrosis Factors/biosynthesis , AMP-Activated Protein Kinases/genetics , Adipocytes/metabolism , Animals , Enzyme Activation/drug effects , Enzyme Activation/physiology , Fatty Acids/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Glucose/metabolism , Glucose-6-Phosphatase/biosynthesis , Glucose-6-Phosphatase/genetics , Hepatocytes/metabolism , Humans , MAP Kinase Kinase 4/genetics , Male , Mice , Mice, Obese , Phosphoenolpyruvate Carboxylase/biosynthesis , Phosphoenolpyruvate Carboxylase/genetics , Protein Multimerization/drug effects , Protein Multimerization/physiology , Sex Characteristics , Signal Transduction/drug effects , Tumor Necrosis Factors/genetics
10.
J Neurosci ; 30(6): 2077-87, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-20147536

ABSTRACT

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.


Subject(s)
Complement C1q/physiology , Hypoxia-Ischemia, Brain/metabolism , Mitochondria/physiology , Oxidative Stress , Animals , Animals, Newborn , Brain Infarction/metabolism , Brain Infarction/pathology , CD59 Antigens/pharmacology , Cells, Cultured , Complement Activation , Complement C1q/genetics , Cytosol/metabolism , Female , Glucose/deficiency , Hypoxia-Ischemia, Brain/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Oxygen/metabolism , Reactive Oxygen Species/metabolism
11.
J Immunol ; 182(3): 1305-13, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19155476

ABSTRACT

Mice lacking complement components show delayed development of prion disease following peripheral inoculation. The delay could relate to reduced scrapie prion protein (PrP(Sc)) accumulation on follicular dendritic cells (DCs). However conventional DCs (cDCs) play a crucial role in the early pathogenesis of prion diseases and complement deficiency could result in decreased PrP(Sc) uptake by cDCs in the periphery. To explore this possibility, we cultured murine splenic or gut-associated lymph node cDCs with scrapie-infected whole brain homogenate in the presence or absence of complement. Uptake decreased significantly if the serum in the cultures was heat-inactivated. Because heat inactivation primarily denatures C1q, we used serum from C1q(-/-) mice and showed that PrP(Sc) uptake was markedly decreased. PrP(Sc) internalization was saturable and temperature-dependent, suggesting receptor-mediated uptake. Furthermore, uptake characteristics differed from fluid-phase endocytosis. Immunofluorescence showed colocalization of C1q and PrP(Sc), suggesting interaction between these molecules. We evaluated the expression of several complement receptors on cDCs and confirmed that cDCs that take up PrP(Sc) express one of the C1q receptors, calreticulin. Our results show that C1q participates in PrP(Sc) uptake by cDCs, revealing a critical role for cDCs in initial prion capture, an event that takes place before the PrP(Sc) accumulation within the follicular DC network.


Subject(s)
Complement C1q/physiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , PrPSc Proteins/metabolism , Scrapie/immunology , Scrapie/metabolism , Animals , Brain/cytology , Brain/immunology , Brain/metabolism , Cells, Cultured , Coculture Techniques , Complement C1q/deficiency , Complement C1q/genetics , Dendritic Cells/pathology , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/metabolism , Endocytosis/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Transport/immunology , Receptors, Complement/biosynthesis , Scrapie/pathology
12.
J Immunol ; 183(1): 749-58, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19535640

ABSTRACT

The CD20 mAb ofatumumab (OFA) is more effective than rituximab (RTX) in promoting complement-dependent cytotoxicity (CDC) of B cells via the classical pathway (CP) of complement. CP activation is initiated by C1q binding to cell-bound IgG. Therefore, we examined the role of C1q in the dynamics of complement activation and CDC of B cell lines and primary cells from patients with chronic lymphocytic leukemia, reacted with OFA or RTX. C1q binding, complement activation, and colocalization of C1q with cell-bound mAbs were determined by flow cytometry and high-resolution digital imaging. C1q binds avidly to OFA-opsonized Raji and Daudi cells (K(D) = 12-16 nM) and colocalizes substantially with cell-bound OFA. Cells opsonized with OFA undergo high levels of complement activation and CDC in C1q-depleted serum supplemented with low concentrations of C1q. Under comparable conditions, RTX-opsonized cells bind less C1q; in addition, even when higher concentrations of C1q are used to achieve comparable C1q binding to RTX-opsonized cells, less complement activation and CDC are observed. Greater CDC induced by OFA may occur because C1q is bound in close proximity and with high avidity to OFA, resulting in effective CP activation. Moreover, OFA binds to the small, extracellular CD20 loop, placing the mAb considerably closer to the cell membrane than does RTX. This may facilitate effective capture and concentration of activated complement components closer to the cell membrane, potentially shielding them from inactivation by fluid phase agents and promoting efficient generation of the membrane attack complex.


Subject(s)
Antibodies, Monoclonal/metabolism , Antigens, CD20/immunology , B-Lymphocyte Subsets/immunology , Complement C1q/metabolism , Cytotoxicity Tests, Immunologic , Opsonin Proteins/metabolism , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived , B-Lymphocyte Subsets/metabolism , Cell Line, Tumor , Complement C1q/physiology , Complement C3b/metabolism , Complement Pathway, Classical/immunology , Cytotoxicity Tests, Immunologic/methods , Dose-Response Relationship, Immunologic , Humans , Protein Binding/immunology , Rituximab
13.
J Immunol ; 183(12): 8225-31, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-20007586

ABSTRACT

Autoantibodies against complement C1q (anti-C1q Abs) were shown to strongly correlate with the occurrence of severe nephritis in patients with systemic lupus erythematosus (SLE), suggesting a potential pathogenic role by interfering with the complement cascade. To analyze the humoral immune response against C1q at the molecular level, we screened a bone marrow-derived IgGkappa/IgGlambda Fab phage display library from a SLE patient with high anti-C1q Ab titer against purified human C1q. Six Fabs that exhibited strong binding to C1q in ELISA were isolated. The anti-C1q Fabs recognized neoepitopes that were only exposed on bound C1q and not present on soluble C1q mapping to different regions of the collagen-like region of C1q. Analysis of the genes encoding the variable H and L chains of the IgG-derived anti-C1q Fab revealed that all the variable H and L chain regions were highly mutated, with nucleotide and amino acid homologies to the closest germline in the range of 71-97% (average 85 +/- 4) and 72-92% (average 88 +/- 6), respectively. In addition, the variable region of the Fabs exhibited high replacement to silent ratios. The six anti-C1q Fabs were shown to be of high affinity, with a K(d) ranging from of 8.4 x 10(-8) M to 1.4 x 10(-7) M, comparable to an antiviral immune response. Our data underlines the notion that the development of anti-C1q Abs in SLE is the consequence of an Ag-driven, affinity-matured immune response. Those anti-C1q Fabs are unique tools to address how complement C1q is implicated in the pathogenesis of SLE.


Subject(s)
Autoantibodies/biosynthesis , Autoantigens/physiology , Complement C1q/physiology , Lupus Erythematosus, Systemic/immunology , Amino Acid Sequence , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/metabolism , Antibody Affinity , Antibody Specificity , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/isolation & purification , Apoptosis Regulatory Proteins/metabolism , Autoantibodies/isolation & purification , Autoantibodies/metabolism , Autoantigens/immunology , Autoantigens/metabolism , Complement C1q/immunology , Complement C1q/metabolism , Humans , Immunoglobulin Fab Fragments/biosynthesis , Immunoglobulin Fab Fragments/isolation & purification , Immunoglobulin Fab Fragments/metabolism , Molecular Sequence Data
14.
Science ; 373(6560): eabj2685, 2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34516796

ABSTRACT

Although traumatic brain injury (TBI) acutely disrupts the cortex, most TBI-related disabilities reflect secondary injuries that accrue over time. The thalamus is a likely site of secondary damage because of its reciprocal connections with the cortex. Using a mouse model of mild TBI (mTBI), we found a chronic increase in C1q expression specifically in the corticothalamic system. Increased C1q expression colocalized with neuron loss and chronic inflammation and correlated with disruption in sleep spindles and emergence of epileptic activities. Blocking C1q counteracted these outcomes, suggesting that C1q is a disease modifier in mTBI. Single-nucleus RNA sequencing demonstrated that microglia are a source of thalamic C1q. The corticothalamic circuit could thus be a new target for treating TBI-related disabilities.


Subject(s)
Brain Injuries/complications , Complement C1q/physiology , Sleep Stages , Sleep Wake Disorders/etiology , Sleep Wake Disorders/physiopathology , Thalamus/physiopathology , Animals , Brain Injuries/physiopathology , Complement C1q/genetics , Disease Models, Animal , Epilepsy/physiopathology , Mice , Microglia/metabolism , Thalamus/metabolism
15.
J Exp Med ; 178(2): 579-87, 1993 Aug 01.
Article in English | MEDLINE | ID: mdl-7688027

ABSTRACT

C1q receptors (C1qR) have been identified on a variety of somatic and cultured cells including peripheral blood platelets. Since platelets are likely to encounter both circulating C1q multimers and C1q associated with the extracellular matrix after complement activation by the classical pathway, the present study was designed to assess the effect of fluid phase and immobilized C1q on platelet function. Platelet adhesion to C1q-coated surfaces was accompanied by the induction of fibrinogen receptors. Scatchard analysis of fibrinogen binding to adherent platelets revealed the binding of approximately 10,000 molecules of fibrinogen per platelet with a Kd of 0.1 +/- 0.03 microM (mean +/- SD, n = 4). Furthermore, fluid phase C1q multimers were noted to aggregate platelets at doses > 5 micrograms/ml. This aggregation was preceded by a rise in inositol-1,4,5-trisphosphate (IP3) (6.9 +/- 2.4 pmoles/10(9) platelets at 15 s, n = 4), and activation of GPIIb-IIIa complexes supporting fibrinogen binding. Platelet aggregation in response to C1q multimers was accompanied by the aspirin-inhibitable release of granule contents and P-selectin (CD62) expression. Platelet aggregation was inhibited by the collagenous domain of C1q (c-Clq) and a monoclonal antibody directed against C1q receptors, suggesting the direct involvement of the 67-kD platelet C1qR. Antibodies against the very late antigen 2 or CD36 collagen receptors were without effect. Platelet exposure to C1q multimers was also accompanied by the expression of procoagulant activity, as demonstrated by the dose-dependent shortening of the kaolin recalcification time of normal plasma from 108 +/- 12 s in the presence of unstimulated platelets to 62 +/- 14 s in the presence of platelets that had been preincubated (5 min, 37 degrees C) with 100 micrograms/ml multimeric C1q (n = 3). These data suggest that platelet interactions with C1q multimers or immobilized C1q, resulting in the activation of GPIIb-IIIa fibrinogen binding sites and the expression of P-selectin as well as platelet procoagulant activity, are likely to contribute to thrombotic events associated with complement activation and inflammation.


Subject(s)
Blood Coagulation/physiology , Cell Adhesion Molecules/biosynthesis , Complement C1q/physiology , Integrins/biosynthesis , Platelet Activation , Platelet Membrane Glycoproteins/biosynthesis , Cell Adhesion , Cells, Cultured , Humans , In Vitro Techniques , P-Selectin , Platelet Aggregation
16.
Eur J Neurosci ; 32(2): 191-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20646056

ABSTRACT

Several C1q family members, especially the Cbln and C1q-like subfamilies, are highly and predominantly expressed in the central nervous system. Cbln1, a member of the Cbln subfamily, plays two unique roles at parallel fiber (PF)-Purkinje cell synapses in the cerebellum: the formation and stabilization of synaptic contact, and the control of functional synaptic plasticity by regulating the postsynaptic endocytotic pathway. The delta2 glutamate receptor (GluD2), which is predominantly expressed in Purkinje cells, plays similar critical roles in the cerebellum. In addition, viral expression of GluD2 or the application of recombinant Cbln1 induces PF-Purkinje cell synaptogenesis in vitro and in vivo. Antigen-unmasking methods were necessary to reveal the immunoreactivities for endogenous Cbln1 and GluD2 at the synaptic junction of PF synapses. We propose that Cbln1 and GluD2 are located at the synaptic cleft, where various proteins undergo intricate molecular interactions with each other, and serve as a bidirectional synaptic organizer.


Subject(s)
Complement C1q/physiology , Signal Transduction/physiology , Synapses/physiology , Animals , Cerebellum/physiology , Glutamate Dehydrogenase/physiology , Humans , Nerve Tissue Proteins/physiology , Protein Precursors/physiology
17.
Adv Exp Med Biol ; 703: 95-104, 2010.
Article in English | MEDLINE | ID: mdl-20711709

ABSTRACT

Chronic open angle glaucoma is a degenerative optic neuropathy that can lead to blindness. We have shown that one of the major genes with altered expression in the glaucomatous retina is complement component C1q in both animal models of the disease as well as in humans. These observations together with evidence of upregulation of other complement components within the retina suggest a role for complement in the pathogenesis of this disease. We review the current evidence that supports such a role and discuss possible mechanisms through which complement may act. A thorough understanding of these mechanisms is important in allowing us to rationally design new therapeutic approaches.


Subject(s)
Complement System Proteins/physiology , Glaucoma/immunology , Animals , Cell Death , Complement C1q/genetics , Complement C1q/physiology , Complement System Proteins/genetics , Disease Models, Animal , Gene Expression , Glaucoma/etiology , Glaucoma/genetics , Glaucoma/pathology , Humans , Mice , Neuroimmunomodulation , Retinal Ganglion Cells/immunology , Retinal Ganglion Cells/pathology
18.
Front Neural Circuits ; 14: 583391, 2020.
Article in English | MEDLINE | ID: mdl-33177995

ABSTRACT

During development, neurons generate excess processes which are then eliminated in concert with circuit maturation. C1q is the initiating protein in the complement cascade and has been implicated in this process, but whether C1q-mediated elimination is targeted to particular neural compartments is unclear. Using the murine retina, we identify C1q as a specific regulator of horizontal cell neurite confinement. Subsets of horizontal cell dendritic and axonal neurites extend into the outer retina suggesting that complement achieves both cellular and subcellular selectivity. These alterations emerge as outer retina synapses become mature. C1q expression is restricted to retina microglia, and the loss of C1q results in decreased microglia activation. This pathway appears independent of the C3a receptor (C3aR) and complement receptor 3 (CR3), as horizontal cells are normal when either protein is absent. Together, these data identify a new role for C1q in cell and neurite-specific confinement and implicate microglia-mediated phagocytosis in this process.


Subject(s)
Complement C1q/physiology , Microglia/metabolism , Neurites/physiology , Neuronal Plasticity/physiology , Animals , Complement C3a , Mice , Mice, Knockout , Microglia/physiology , Phagocytosis , Receptors, Complement , Retinal Horizontal Cells
19.
Diabetes ; 69(1): 99-111, 2020 01.
Article in English | MEDLINE | ID: mdl-31676569

ABSTRACT

Endothelial dysfunction plays a crucial role in the progress of diabetic vasculopathy. C1q/tumor necrosis factor-related protein 13 (CTRP13) is a secreted adipokine that can ameliorate atherosclerosis and vascular calcification. However, the role of CTRP13 in regulating endothelial function in diabetes has yet to be explored. In this study, CTRP13 treatment improved endothelium-dependent relaxation in the aortae and mesenteric arteries of both db/db mice and streptozotocin-injected mice. CTRP13 supplement also rescued the impaired endothelium-dependent relaxation ex vivo in the db/db mouse aortae and in high glucose (HG)-treated mouse aortae. Additionally, CTRP13 treatment reduced reactive oxygen species overproduction and improved nitric oxide (NO) production and endothelial NO synthase (eNOS) coupling in the aortae of diabetic mice and in HG-treated human umbilical vein endothelial cells. Mechanistically, CTRP13 could increase GTP cyclohydrolase 1 (GCH1) expression and tetrahydrobiopterin (BH4) levels to ameliorate eNOS coupling. More importantly, CTRP13 rescued HG-induced inhibition of protein kinase A (PKA) activity. Increased PKA activity enhanced phosphorylation of the peroxisome proliferator-activated receptor α and its recruitment to the GCH1 promoter, thus activating GCH1 transcription and, ultimately, endothelial relaxation. Together, these results suggest that CTRP13 preserves endothelial function in diabetic mice by regulating GCH1/BH4 axis-dependent eNOS coupling, suggesting the therapeutic potential of CTRP13 against diabetic vasculopathy.


Subject(s)
Adipokines/physiology , Complement C1q/physiology , Diabetes Mellitus/physiopathology , Endothelium, Vascular/physiology , GTP Cyclohydrolase/genetics , Human Umbilical Vein Endothelial Cells/physiology , Animals , Cells, Cultured , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetic Angiopathies/genetics , Diabetic Angiopathies/physiopathology , Diabetic Angiopathies/prevention & control , GTP Cyclohydrolase/metabolism , Gene Expression Regulation, Enzymologic , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
20.
Mol Neurobiol ; 57(5): 2290-2300, 2020 May.
Article in English | MEDLINE | ID: mdl-32008166

ABSTRACT

The complement C1q plays a critical role in microglial phagocytosis of glutamatergic synapses and in the pathogenesis of neuroinflammation in Alzheimer's disease (AD). We recently reported that upregulation of metabotropic glutamate receptor signaling is associated with increased synaptic C1q production and subsequent microglial phagocytosis of synapses in the rodent models of AD. Here, we explored the role of astrocytic glutamate transporter in the synaptic C1q production and microglial phagocytosis of hippocampal glutamatergic synapses in a rat model of AD. Activation of astrocyte and reduction glutamate transporter 1 (GLT1) were noted after bilateral microinjection of amyloid-beta (Aß1-40) fibrils into the hippocampal CA1 area of rats. Ceftriaxone is a ß-lactam antibiotic that upregulates GLT1 expression. Bilateral microinjection of ceftriaxone recovered GLT1 expression, decreased synaptic C1q production, suppressed microglial phagocytosis of glutamatergic synapses in the hippocampal CA1, and attenuated synaptic and cognitive deficits in rats microinjected with Aß1-40. In contrast, artificial suppression of GLT1 activity by DL-threo-beta-benzyloxyaspartate (DL-TBOA) in naïve rats induced synaptic C1q expression and microglial phagocytosis of glutamatergic synapses in the hippocampal CA1 area, resulting in synaptic and cognitive dysfunction. These findings demonstrated that impairment of astrocytic glutamate transporter plays a role in the pathogenesis of AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/toxicity , Astrocytes/drug effects , CA1 Region, Hippocampal/drug effects , Cognition Disorders/chemically induced , Complement C1q/physiology , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Glutamic Acid/physiology , Microglia/physiology , Neurons/metabolism , Peptide Fragments/toxicity , Animals , Aspartic Acid/pharmacology , Astrocytes/metabolism , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Ceftriaxone/pharmacology , Cognition Disorders/metabolism , Cognition Disorders/pathology , Complement C1q/biosynthesis , Complement C1q/genetics , Disease Models, Animal , Excitatory Amino Acid Transporter 2/biosynthesis , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/physiology , Male , Morris Water Maze Test/drug effects , Morris Water Maze Test/physiology , Patch-Clamp Techniques , Phagocytosis/drug effects , Phagocytosis/physiology , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/physiology , Synapses/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL