Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Cell Biochem Funct ; 38(4): 392-400, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31904875

ABSTRACT

The circumventricular organs (CVOs) are the brain regions that lack the blood-brain barrier and allow free entry of blood-derived molecules, offering specialized niche to initiate rapid and early neuroinflammatory responses in the brain. Complement component 1q (C1q) is shown to be the first recognition component of the complement pathway and has a crucial function in the brain under pathological conditions. In the present study, we found that C1q expression in CX3CR1-positive microglia was increased in the CVOs and their neighbouring brain regions of adult mice at 1 day after a single administration of 1 mg/kg lipopolysaccharide (LPS), whereas it returned to control levels at 3 days after LPS stimulation. C1q expression was also seen to localize at synapsin-positive presynaptic axonal terminals in various brain regions. Thus, the present study demonstrates a transient upregulation of microglial C1q expression in the CVOs and their adjacent brain regions, indicating that a transient upregulation of C1q is possibly concerned with physiological responses at early phase of brain inflammation. SIGNIFICANCE OF THE STUDY: The circumventricular organs (CVOs) are specialized brain regions that lack the blood-brain barrier (BBB) and initiate neuroinflammatory responses in the brains. The present study showed that the expression of complement protein C1q was highly increased in microglia of the CVOs and their adjacent brain regions. Moreover, C1q expression was observed to localize specifically at presynaptic axonal terminals in the CVOs and their neighbouring brain regions. Thus, the present study indicates that C1q is possibly correlated with physiological responses at early phase of brain inflammation.


Subject(s)
Brain/metabolism , Circumventricular Organs/metabolism , Complement C1q/biosynthesis , Gene Expression Regulation/drug effects , Lipopolysaccharides/toxicity , Microglia/metabolism , Animals , Axons/metabolism , Axons/pathology , Brain/pathology , Circumventricular Organs/pathology , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Mice , Microglia/pathology
2.
Mediators Inflamm ; 2020: 8873152, 2020.
Article in English | MEDLINE | ID: mdl-33424438

ABSTRACT

Atrial fibrillation (AF) is a highly prevalent cardiac arrhythmia characterized by atrial remodeling. Complement C1q tumor necrosis factor-related protein 3 (CTRP3) is one of the adipokines associated with obesity, diabetes, and coronary heart disease. The association between plasma CTRP3 levels and AF is uncertain. The aim of this study was to investigate whether plasma CTRP3 concentrations were correlated with AF. Our study included 75 AF patients who underwent catheter ablation at our hospital and 47 sinus rhythm patients to determine the difference in plasma CTRP3 concentrations. Blood samples before the ablation were collected, and ELISA was used to measure the concentrations of CTRP3. Plasma CTRP3 concentrations were significantly lower in AF patients compared with control group (366.9 ± 105.2 ng/ml vs. 429.1 ± 100.1 ng/ml, p = 0.002). In subgroup studies, patients with persistent AF had lower plasma CTRP3 concentrations than those with paroxysmal AF (328.3 ± 83.3 ng/ml vs. 380.0 ± 109.2 ng/ml, p = 0.037). The concentrations of plasma CTRP3 in the recurrence group after radiofrequency catheter ablation of AF were lower than those in the nonrecurrence group (337.9 ± 77.3 ng/ml vs. 386.6 ± 108.1 ng/ml, p = 0.045). Multivariate regression analysis revealed the independent correlation between plasma CTRP3 level and AF. Plasma CTRP3 concentrations were correlated with the presence of AF and AF recurrence.


Subject(s)
Atrial Fibrillation/blood , Complement C1q/biosynthesis , Tumor Necrosis Factors/blood , Aged , Case-Control Studies , Catheter Ablation , Echocardiography , Female , Follow-Up Studies , Humans , Male , Middle Aged , Multivariate Analysis , Prognosis , Recurrence
3.
J Immunol ; 199(3): 1069-1085, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28687659

ABSTRACT

Inflammatory processes play a key role in pathophysiology of many neurologic diseases/trauma, but the effect of immune cells and factors on neurotransplantation strategies remains unclear. We hypothesized that cellular and humoral components of innate immunity alter fate and migration of human neural stem cells (hNSC). In these experiments, conditioned media collected from polymorphonuclear leukocytes (PMN) selectively increased hNSC astrogliogenesis and promoted cell migration in vitro. PMN were shown to generate C1q and C3a; exposure of hNSC to PMN-synthesized concentrations of these complement proteins promoted astrogliogenesis and cell migration. Furthermore, in vitro, Abs directed against C1q and C3a reversed the fate and migration effects observed. In a proof-of-concept in vivo experiment, blockade of C1q and C3a transiently altered hNSC migration and reversed astroglial fate after spinal cord injury. Collectively, these data suggest that modulation of the innate/humoral inflammatory microenvironment may impact the potential of cell-based therapies for recovery and repair following CNS pathology.


Subject(s)
Astrocytes/physiology , Cell Differentiation/physiology , Complement C1q/biosynthesis , Complement C3a/biosynthesis , Neural Stem Cells/physiology , Neutrophils/metabolism , Animals , Astrocytes/drug effects , Cell Movement , Cells, Cultured , Complement C1q/antagonists & inhibitors , Complement C1q/genetics , Complement C1q/immunology , Complement C3a/antagonists & inhibitors , Complement C3a/genetics , Complement C3a/immunology , Culture Media, Conditioned , Humans , Immunity, Innate , Mice , Neural Stem Cells/drug effects , Neural Stem Cells/immunology , Neutrophils/immunology , Spinal Cord Injuries/immunology , Spinal Cord Injuries/physiopathology
4.
J Biol Chem ; 290(38): 23039-49, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26231212

ABSTRACT

Calcium-binding proteins (CaBPs) such as parvalbumin are part of the cellular calcium buffering system that determines intracellular calcium diffusion and influences the spatiotemporal dynamics of calcium signals. In neurons, CaBPs are primarily localized to the cytosol and function, for example, in nerve terminals in short-term synaptic plasticity. However, CaBPs are also expressed in the cell nucleus, suggesting that they modulate nuclear calcium signals, which are key regulators of neuronal gene expression. Here we show that the calcium buffering capacity of the cell nucleus in mouse hippocampal neurons regulates neuronal architecture by modulating the expression levels of VEGFD and the complement factor C1q-c, two nuclear calcium-regulated genes that control dendrite geometry and spine density, respectively. Increasing the levels of nuclear calcium buffers by means of expression of a nuclearly targeted form of parvalbumin fused to mCherry (PV.NLS-mC) led to a reduction in VEGFD expression and, as a result, to a decrease in total dendritic length and complexity. In contrast, mRNA levels of the synapse pruning factor C1q-c were increased in neurons expressing PV.NLS-mC, causing a reduction in the density and size of dendritic spines. Our results establish a close link between nuclear calcium buffering capacity and the transcription of genes that determine neuronal structure. They suggest that the development of cognitive deficits observed in neurological conditions associated with CaBP deregulation may reflect the loss of necessary structural features of dendrites and spines.


Subject(s)
Calcium/metabolism , Dendritic Spines/metabolism , Hippocampus/metabolism , Nerve Net/metabolism , Animals , Complement C1q/biosynthesis , Gene Expression Regulation/physiology , Hippocampus/cytology , Mice , Nerve Net/cytology , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor D/biosynthesis
5.
J Immunol ; 192(3): 1138-53, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24353269

ABSTRACT

The complement system is activated in a wide spectrum of CNS diseases and is suggested to play a role in degenerative phenomena such as elimination of synaptic terminals. Still, little is known of mechanisms regulating complement activation in the CNS. Loss of synaptic terminals in the spinal cord after an experimental nerve injury is increased in the inbred DA strain compared with the PVG strain and is associated with expression of the upstream complement components C1q and C3, in the absence of membrane attack complex activation and neutrophil infiltration. To further dissect pathways regulating complement expression, we performed genome-wide expression profiling and linkage analysis in a large F2(DA × PVG) intercross, which identified quantitative trait loci regulating expression of C1qa, C1qb, C3, and C9. Unlike C1qa, C1qb, and C9, which all displayed distinct coregulation with different cis-regulated C-type lectins, C3 was regulated in a coexpression network immediately downstream of butyrylcholinesterase. Butyrylcholinesterase hydrolyses acetylcholine, which exerts immunoregulatory effects partly through TNF-α pathways. Accordingly, increased C3, but not C1q, expression was demonstrated in rat and mouse glia following TNF-α stimulation, which was abrogated in a dose-dependent manner by acetylcholine. These findings demonstrate new pathways regulating CNS complement expression using unbiased mapping in an experimental in vivo system. A direct link between cholinergic activity and complement activation is supported by in vitro experiments. The identification of distinct pathways subjected to regulation by naturally occurring genetic variability is of relevance for the understanding of disease mechanisms in neurologic conditions characterized by neuronal injury and complement activation.


Subject(s)
Central Nervous System/metabolism , Cholinergic Fibers/physiology , Complement Activation , Complement C3/biosynthesis , Gene Expression Regulation/immunology , Gene Regulatory Networks , Acetylcholine/pharmacology , Acetylcholine/physiology , Animals , Animals, Congenic , Astrocytes/drug effects , Astrocytes/metabolism , Brain Injuries/immunology , Brain Injuries/physiopathology , Butyrylcholinesterase/physiology , Cells, Cultured , Central Nervous System/chemistry , Central Nervous System/pathology , Complement C1q/biosynthesis , Complement C1q/genetics , Complement C3/genetics , Denervation , Forkhead Transcription Factors/metabolism , Genetic Linkage , Genome-Wide Association Study , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Quantitative Trait Loci , Rats , Rhizotomy , Specific Pathogen-Free Organisms , Spinal Nerve Roots/surgery , Synaptophysin/analysis , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/physiology
6.
Nanomedicine ; 12(4): 1033-1043, 2016 May.
Article in English | MEDLINE | ID: mdl-26767511

ABSTRACT

When inhaled nanoparticles deposit in the lungs, they transit through respiratory tract lining fluid (RTLF) acquiring a biomolecular corona reflecting the interaction of the RTLF with the nanomaterial surface. Label-free snapshot proteomics was used to generate semi-quantitative profiles of corona proteins formed around silica (SiO2) and poly(vinyl) acetate (PVAc) nanoparticles in RTLF, the latter employed as an archetype drug delivery vehicle. The evolved PVAc corona was significantly enriched compared to that observed on SiO2 nanoparticles (698 vs. 429 proteins identified); however both coronas contained a substantial contribution from innate immunity proteins, including surfactant protein A, napsin A and complement (C1q and C3) proteins. Functional protein classification supports the hypothesis that corona formation in RTLF constitutes opsonisation, preparing particles for phagocytosis and clearance from the lungs. These data highlight how an understanding of the evolved corona is necessary for the design of inhaled nanomedicines with acceptable safety and tailored clearance profiles. FROM THE CLINICAL EDITOR: Inhaled nanoparticles often acquire a layer of protein corona while they go through the respiratory tract. Here, the authors investigated the identity of these proteins. The proper identification would improve the understanding of the use of inhaled nanoparticles in future therapeutics.


Subject(s)
Drug Delivery Systems , Nanoparticles/administration & dosage , Protein Corona , Respiratory System/metabolism , Adult , Aspartic Acid Endopeptidases/biosynthesis , Aspartic Acid Endopeptidases/isolation & purification , Body Fluids/metabolism , Complement C1q/biosynthesis , Complement C1q/isolation & purification , Complement C3/biosynthesis , Complement C3/isolation & purification , Female , Gene Expression Regulation/drug effects , Humans , Male , Nanoparticles/adverse effects , Proteomics , Pulmonary Surfactant-Associated Protein A/biosynthesis , Pulmonary Surfactant-Associated Protein A/isolation & purification , Respiratory System/drug effects , Silicon Dioxide/administration & dosage , Silicon Dioxide/chemistry
7.
J Neurosci ; 33(33): 13460-74, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-23946404

ABSTRACT

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.


Subject(s)
Aging/metabolism , Brain/metabolism , Complement C1q/biosynthesis , Animals , Behavior, Animal , Blotting, Western , Brain/physiology , Electrophysiology , Excitatory Postsynaptic Potentials , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Immunoelectron
8.
Biochem J ; 447(2): 229-37, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22812635

ABSTRACT

C1q deficiency is the strongest known risk factor for SLE (systemic lupus erythematosus) but its endogenous cellular origin remains limitedly understood. In the present study we investigate the production of C1q by both cultured and endogenous bone osteoclasts. Blood monocytes were cultured with RANKL (receptor activator of nuclear factor κB ligand) and M-CSF (macrophage colony-stimulating factor) to generate osteoclasts and these cells expressed C1Q mRNA and also secreted C1q protein. Intracellular C1q was detectable in developing osteoclasts at day 3 by Western blotting and was also detectable by flow cytometry. By immunofluorescence microscopy, C1q was preferentially detected in immature osteoclasts. By multiple detection methods, C1q expression was markedly increased after IFNγ (interferon γ) treatment. By immunohistochemistry, C1q was also detected in endogenous bone osteoclasts. When osteoclasts were cultured on immobilized C1q, these cells exhibited 2-7-fold increases in the expression of signature osteoclast genes [TRAP (tartrate-resistant acid phosphatase), cathepsin K, calcitonin receptor, carbonic anhydrase II and NFATc1 (nuclear factor of activated T-cells, cytoplasmic, calcineurin-dependent 1)], suggesting an osteoclastogenic capability. This is the first report of C1q production by osteoclasts. Its ability to enhance osteoclast development implies reduced osteoclastogenesis in patients with SLE as they often experience decreased C1q levels. This is consistent with the non-erosive nature of lupus arthritis.


Subject(s)
Complement C1q/biosynthesis , Osteoclasts/metabolism , Cathepsin K/biosynthesis , Cell Differentiation , Cells, Cultured , Humans , Interferon-gamma/pharmacology , Lupus Erythematosus, Systemic/physiopathology , Monocytes/metabolism , Monocytes/physiology , Osteoclasts/cytology , RNA, Messenger/metabolism
9.
J Chemother ; 33(7): 476-485, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33825671

ABSTRACT

This study aims to investigate the effects of C1q-like 1 (C1QL1) on the growth and migration of lung adenocarcinoma (LUAD) cells and the underlying mechanism. The expression of C1QL1 in LUAD tissues and its prognostic value were analyzed using the data from The Cancer Genome Atlas (TCGA) database. To investigate the function of C1QL1, loss-of-function and gain-of-function assays were conducted in Calu-3 cells and LTEP-a-2 cells, respectively. Cell growth was evaluated by CCK-8 and colony formation assays. Transwell assays were performed to assess cell invasive and migratory abilities. qRT-PCR and Western blotting were performed to detect RNA and protein expression, respectively. Firstly, we found that C1QL1 was highly expressed and predicted poor outcomes in LUAD patients from TCGA database. Moreover, the mRNA and protein expression levels of C1QL1 were higher in LUAD cells than that in normal lung cells. Results of functional experiments illustrated that depletion of C1QL1 restrained the growth, invasion and migration of Calu-3 cells, meanwhile over-expression of C1QL1 presented the opposite results in LTEP-a-2 cells. Furthermore, we discovered that down-regulation of C1QL1 elevated the protein level of E-cadherin and reduced the protein levels of N-cadherin, Vimentin and Snail in Calu-3 cells, whereas over-expression of C1QL1 led to the opposite outcomes in LTEP-a-2 cells. Our data indicated that C1QL1 functioned as a crucial driver in LUAD cell growth and motility, which might be achieved by modulating epithelial-mesenchymal transition (EMT). These consequences are of important relevance for the design of therapeutic strategies for LUAD.


Subject(s)
Adenocarcinoma of Lung/pathology , Complement C1q/biosynthesis , Lung Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Computational Biology , Humans , Neoplasm Invasiveness/physiopathology , Prognosis , Up-Regulation
10.
Cell Transplant ; 30: 9636897211033275, 2021.
Article in English | MEDLINE | ID: mdl-34338573

ABSTRACT

Myocardial infarction (MI) is identified as the myocardial necrosis due to myocardial ischemia/reperfusion (I/R) injury and remains a leading cause of mortality. C1q/TNF-related protein 13 (CTRP13) is a member of CTRP family that has been found to be involved in coronary artery disease (CAD). However, the role of CTRP13 in MI remains unclear. We aimed to explore the functional role of CTRP13 in H9c2 cells exposed to hypoxia/reoxygenation (H/R). Our results demonstrated that H/R stimulation significantly decreased the expression of CTRP13 in H9c2 cells. H/R-induced an increase in ROS production and reductions in activities of SOD and CAT were prevented by CTRP13 overexpression but were aggravated by CTRP13 silencing. Moreover, CTRP13 overexpression could reverse the inductive effect of H/R on caspase-3 activity and bax expression, as well as the inhibitory effect of H/R on bcl-2 expression in H9c2 cells. However, CTRP13 silencing presented opposite effects with CTRP13 overexpression. Furthermore, CTRP13 overexpression enhanced the H/R-stimulated the expression levels of p-AMPK and nuclear Nrf2, and Nrf2 transcriptional activity. However, inhibition of AMPK reversed the CTRP13-mediated activation of Nrf2/ARE signaling and the cardiac-protective effect in H/R-exposed H9c2 cells. Additionally, silencing of Nrf2 reversed the protective effects of CTRP13 against H/R-stimulated oxidative stress and apoptosis in H9c2 cells. Finally, recombinant CTRP13 protein attenuated myocardial I/R-induced injury in rats. Taken together, these findings indicated that CTRP13 protected H9c2 cells from H/R-stimulated oxidative stress and apoptosis via regulating the AMPK/Nrf2/ARE signaling pathway. Our results provided evidence for the therapeutic potential of CTRP13 in myocardial I/R injury.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipokines/metabolism , Complement C1q/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , AMP-Activated Protein Kinases/genetics , Adipokines/biosynthesis , Adipokines/genetics , Animals , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Cell Hypoxia/physiology , Cell Line , Complement C1q/biosynthesis , Complement C1q/genetics , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Rats , Signal Transduction
11.
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
12.
Mol Neurodegener ; 13(1): 45, 2018 08 20.
Article in English | MEDLINE | ID: mdl-30126455

ABSTRACT

BACKGROUND: The role of the alternative complement pathway and its mediation by retinal microglia and macrophages, is well-established in the pathogenesis of Age-Related Macular Degeneration (AMD). However, the contribution of the classical complement pathway towards the progression of retinal degenerations is not fully understood, including the role of complement component 1q (C1q) as a critical activator molecule of the classical pathway. Here, we investigated the contribution of C1q to progressive photoreceptor loss and neuroinflammation in retinal degenerations. METHODS: Wild-type (WT), C1qa knockout (C1qa-/-) and mice treated with a C1q inhibitor (ANX-M1; Annexon Biosciences), were exposed to photo-oxidative damage (PD) and were observed for progressive lesion development. Retinal function was assessed by electroretinography, followed by histological analyses to assess photoreceptor degeneration. Retinal inflammation was investigated through complement activation, macrophage recruitment and inflammasome expression using western blotting, qPCR and immunofluorescence. C1q was localised in human AMD donor retinas using immunohistochemistry. RESULTS: PD mice had increased levels of C1qa which correlated with increasing photoreceptor cell death and macrophage recruitment. C1qa-/- mice did not show any differences in photoreceptor loss or inflammation at 7 days compared to WT, however at 14 days after the onset of damage, C1qa-/- retinas displayed less photoreceptor cell death, reduced microglia/macrophage recruitment to the photoreceptor lesion, and higher visual function. C1qa-/- mice displayed reduced inflammasome and IL-1ß expression in microglia and macrophages in the degenerating retina. Retinal neutralisation of C1q, using an intravitreally-delivered anti-C1q antibody, reduced the progression of retinal degeneration following PD, while systemic delivery had no effect. Finally, retinal C1q was found to be expressed by subretinal microglia/macrophages located in the outer retina of early AMD donor eyes, and in mouse PD retinas. CONCLUSIONS: Our data implicate subretinal macrophages, C1q and the classical pathway in progressive retinal degeneration. We demonstrate a role of local C1q produced by microglia/macrophages as an instigator of inflammasome activation and inflammation. Crucially, we have shown that retinal C1q neutralisation during disease progression may slow retinal atrophy, providing a novel strategy for the treatment of complement-mediated retinal degenerations including AMD.


Subject(s)
Complement C1q/biosynthesis , Macrophages/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Animals , Disease Progression , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout
13.
J Leukoc Biol ; 80(1): 87-95, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16617159

ABSTRACT

Pentraxins (PTX) and complement belong to the humoral arm of the innate immune system and have essential functions in immune defense to microbes and in scavenging cellular debris. The prototypic long PTX, PTX3, and the first component of the classical complement pathway, C1q, are innate opsonins involved in the disposal of dying cells by phagocytes. Whether the interaction between various innate opsonins impacts on their function is not fully understood. We show here that characterized Toll-like receptor (TLR) ligands elicit the production of C1q and PTX3 by immature dendritic cells (DC). Moreover, these molecules bind to dying cells with similar kinetics, although they recognize different domains on the cell membranes. PTX3 binds in the fluid phase to C1q, decreasing C1q deposition and subsequent complement activation on apoptotic cells. C1q increases the phagocytosis of apoptotic cells by DC and the release of interleukin-12 in the presence of TLR4 ligands and apoptotic cells; PTX3 inhibits both events. Moreover, PTX3 inhibited the cross-presentation of the MELAN-A/melanoma antigen-reactive T cell 1 (MART-1) tumor antigen expressed by dying cells, even in the presence of C1q. These results suggest that interaction of C1q and PTX3 influences the clearance of apoptotic cells by DC. The coordinated induction by primary, proinflammatory signals of C1q and PTX3 and their reciprocal regulation during inflammation influences the clearance of apoptotic cells by antigen-presenting cells and possibly plays a role in immune homeostasis.


Subject(s)
Apoptosis/immunology , C-Reactive Protein/immunology , Complement Activation/immunology , Complement C1q/immunology , Dendritic Cells/immunology , Phagocytes/immunology , Serum Amyloid P-Component/immunology , Antigen Presentation/immunology , Antigens, Neoplasm/biosynthesis , C-Reactive Protein/biosynthesis , Complement C1q/biosynthesis , Humans , MART-1 Antigen , Neoplasm Proteins/biosynthesis , Phagocytosis/immunology , Serum Amyloid P-Component/biosynthesis
14.
Exp Neurol ; 295: 184-193, 2017 09.
Article in English | MEDLINE | ID: mdl-28601603

ABSTRACT

Microglia-mediated neuroinflammation is widely associated with seizures and epilepsy. Although microglial cells are professional phagocytes, less is known about the status of this phenotype in epilepsy. Recent evidence supports that phagocytosis-associated molecules from the classical complement (C1q-C3) play novel roles in microglia-mediated synaptic pruning. Interestingly, in human and experimental epilepsy, altered mRNA levels of complement molecules were reported. Therefore, to identify a potential role for complement and microglia in the synaptodendritic pathology of epilepsy, we determined the protein levels of classical complement proteins (C1q-C3) along with other phagocytosis signaling molecules in human epilepsy. Cortical brain samples surgically resected from patients with refractory epilepsy (RE) and non-epileptic lesions (NE) were examined. Western blotting was used to determine the levels of phagocytosis signaling proteins such as the complements C1q and C3, MerTK, Trem2, and Pros1 along with cleaved-caspase 3. In addition, immunostaining was used to determine the distribution of C1q and co-localization to microglia and dendrites. We found that the RE samples had significantly increased protein levels of C1q (p=0.034) along with those of its downstream activation product iC3b (p=0.027), and decreased levels of Trem2 (p=0.045) and Pros1 (p=0.005) when compared to the NE group. Protein levels of cleaved-caspase 3 were not different between the groups (p=0.695). In parallel, we found C1q localization to microglia and dendrites in both NE and RE samples, and also observed substantial microglia-dendritic interactions in the RE tissue. These data suggest that aberrant phagocytic signaling occurs in human refractory epilepsy. It is likely that alteration of phagocytic pathways may contribute to unwanted elimination of cells/synapses and/or impaired clearance of dead cells. Future studies will investigate whether altered complement signaling contributes to the hyperexcitability that result in epilepsy.


Subject(s)
Complement Activation/genetics , Complement Pathway, Classical , Epilepsy/genetics , Phagocytosis/genetics , Cells, Cultured , Cerebral Cortex/pathology , Complement C1q/biosynthesis , Complement C1q/genetics , Complement C3/biosynthesis , Complement C3/genetics , Complement Pathway, Classical/genetics , Dendrites/genetics , Dendrites/metabolism , Drug Resistant Epilepsy/genetics , Drug Resistant Epilepsy/pathology , Humans , Microglia/metabolism , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 1/genetics
15.
J Leukoc Biol ; 101(2): 481-491, 2017 02.
Article in English | MEDLINE | ID: mdl-27630215

ABSTRACT

Antibodies against C1q (anti-C1q) are frequently found in patients with systemic lupus erythematosus (SLE). The anti-C1q antibodies strongly correlate with the occurrence of lupus nephritis and low-circulating C1q levels. Previous studies have demonstrated that myeloid cells, i.e., dendritic cells and macrophages, are a major source of C1q. However, a direct effect of anti-C1q on C1q secretion by macrophages has not yet been established. In the present study, we investigated the C1q secretion profile of in vitro human monocyte-derived macrophages (HMDMs) obtained from healthy donors and from patients with SLE. The effect of SLE patient-derived anti-C1q bound to immobilized C1q (imC1q) and imC1q alone on HMDMs was investigated by C1q secretion levels, the expression of membrane-bound and intracellular C1q using flow cytometry and ImageStreamX technology, and testing the ability of secreted C1q to activate the classical pathway (CP) of the complement. Bound anti-C1q induced significantly greater C1q secretion levels as compared with imC1q alone or healthy donor IgG. The extent of C1q secretion by HMDMs correlated with IgG anti-C1q levels of patients with SLE but not of healthy controls. Furthermore, bound autoantibodies and imC1q induced continuous and de novo C1q synthesis as evident by the intracellular C1q content, which correlated with C1q secretion levels. Finally, secreted C1q was able to activate the CP, as reflected by C4b deposition. Interestingly, anti-C1q-dependent C1q secretion could also be observed in SLE patient-derived cells. In conclusion, our data indicate that imC1q-bound anti-C1q strongly stimulate the C1q production by HMDMs. Anti-C1q-induced C1q secretion might be an important immune-modulatory factor in SLE.


Subject(s)
Autoantibodies/immunology , Complement C1q/biosynthesis , Complement C1q/immunology , Lupus Erythematosus, Systemic/immunology , Macrophages/metabolism , Adult , Aged , Female , Flow Cytometry , Humans , Immobilized Proteins/metabolism , Immunoglobulin G/metabolism , Intracellular Space/metabolism , Kinetics , Lipopolysaccharide Receptors/metabolism , Macrophages/pathology , Male , Middle Aged , Monocytes/pathology
16.
Mol Immunol ; 78: 164-170, 2016 10.
Article in English | MEDLINE | ID: mdl-27648858

ABSTRACT

C1q is the initiation molecule of the classical pathway of the complement system and is produced by macrophages and immature dendritic cells. As mast cells share the same myeloid progenitor cells, we have studied whether also mast cells can produce and secrete C1q. Mast cells were generated in vitro from CD34+ progenitor cells from buffy coats or cord blood. Fully differentiated mast cells were shown by both RNA sequencing and qPCR to express C1QA, C1QB and C1QC. C1q produced by mast cells has a similar molecular make-up as serum C1q. Reconstituting C1q depleted serum with mast cell supernatant in haemolytic assays, indicated that C1q secreted by mast cells is functionally active. The level of C1q in supernatants produced under basal conditions was considerably enhanced upon stimulation with LPS, dexamethasone in combination with IFN- γ or via FcεRI triggering. Mast cells in human tissues stained positive for C1q in both healthy and in inflamed tissue. Moreover, mast cells in healthy and diseased skin appear to be the predominant C1q positive cells. Together, our data reveal that mast cells are able to produce and secrete functional active C1q and indicate mast cells as a local source of C1q in human tissue.


Subject(s)
Complement C1q/biosynthesis , Mast Cells/immunology , Blotting, Western , Cell Separation , Cells, Cultured , Complement C1q/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Mast Cells/metabolism
17.
Neuron ; 91(5): 1034-1051, 2016 Sep 07.
Article in English | MEDLINE | ID: mdl-27478018

ABSTRACT

C1ql3 is a secreted neuronal protein that binds to BAI3, an adhesion-class GPCR. C1ql3 is homologous to other gC1q-domain proteins that control synapse numbers, but a role for C1ql3 in regulating synapse density has not been demonstrated. We show in cultured neurons that C1ql3 expression is activity dependent and supports excitatory synapse density. Using newly generated conditional and constitutive C1ql3 knockout mice, we found that C1ql3-deficient mice exhibited fewer excitatory synapses and diverse behavioral abnormalities, including marked impairments in fear memories. Using circuit-tracing tools and conditional ablation of C1ql3 targeted to specific brain regions, we demonstrate that C1ql3-expressing neurons in the basolateral amygdala project to the medial prefrontal cortex, that these efferents contribute to fear memory behavior, and that C1ql3 is required for formation and/or maintenance of these synapses. Our results suggest that C1ql3 is a signaling protein essential for subsets of synaptic projections and the behaviors controlled by these projections.


Subject(s)
Amygdala/physiology , Complement C1q/physiology , Memory/physiology , Nerve Tissue Proteins/physiology , Nucleus Accumbens/physiology , Prefrontal Cortex/physiology , Synapses/physiology , Animals , Cells, Cultured , Complement C1q/biosynthesis , Complement C1q/genetics , Male , Mice , Mice, Knockout , Mutation , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neural Pathways/physiology , Neurons/metabolism , Neurons/physiology , Synapses/metabolism
18.
J Mol Med (Berl) ; 73(9): 465-71, 1995 Sep.
Article in English | MEDLINE | ID: mdl-8528750

ABSTRACT

The levels and cellular localization of mRNA for complement C1q and C3 were examined by RNA gel blot and nonradioactive in situ hybridization in the frontal cortex of patients with Alzheimer's disease (AD) and age-matched controls. We found that the hybridization signal for C1q mRNA was markedly increased (approx. 3.5-fold) in the frontal cortex of AD patients compared to that in age-matched controls. In contrast to previous reports we also found that the levels of C3 mRNA, although well expressed, did not differ significantly between AD cases and age-matched controls. Nonradioactive in situ hybridization using digoxigenin-labeled ribo-probes revealed that transcripts coding for both C1q and C3 were closely associated with neurons. These results support the hypothesis that complement could play a role in neuronal degeneration which has been observed in the brain of AD patients.


Subject(s)
Alzheimer Disease/metabolism , Complement C1q/biosynthesis , Complement C3/biosynthesis , Frontal Lobe/metabolism , Gene Expression , RNA, Messenger/biosynthesis , Aged , Aged, 80 and over , Blotting, Northern , Female , Humans , In Situ Hybridization , Male , Middle Aged , RNA, Messenger/analysis , Reference Values
19.
J Leukoc Biol ; 50(5): 453-63, 1991 Nov.
Article in English | MEDLINE | ID: mdl-1748841

ABSTRACT

We investigated the effects of bacterial lipopolysaccharide (LPS), immune complexes (IC), and C3b opsonized zymosan (AZ) alone and in combination with interferon-gamma (IFN-gamma) priming on macrophage synthesis and secretion of C1q. Our results indicated that LPS, IC, and AZ alone stimulated C1q mRNA and secretion in the absence of IFN-gamma. The increase in mRNA accumulation was detectable after 3 h, peaked at 6 h and was maintained at constitutive levels for 24 h. There was a corresponding early burst of increased secretion of functional C1q after 3 to 6 h which declined rapidly after 9 to 24 h culture of LPS-stimulated macrophages. Priming of macrophages with IFN-gamma and simultaneous triggering with LPS, IC, or AZ produced additive rather than synergistic increases in C1q mRNA accumulation. These same agents inhibited constitutive secretion of C1q in the absence of IFN-gamma priming as determined by autoradiographic analysis of metabolically radiolabeled secretory C1q. Triggering of IFN-gamma primed macrophages with LPS, IC, or AZ also markedly suppressed the increased rate of C1q secretion induced by IFN-gamma in a dose-related fashion. A corresponding dose-dependent increased accumulation of endogenous C1q in cell lysates was detected by Western blot analysis of macrophages which had been stimulated by LPS, IC, or AZ alone or in combination with IFN-gamma. Our findings indicate that LPS as well as FcR and C3bR triggering agents stimulate early and sustained C1q synthesis accompanied by an early and short-lived burst of C1q secretion which rapidly diminished and results in an increased intracellular accumulation of C1q due to ongoing synthesis. IFN-gamma appeared to further amplify the same kinetics of increased C1q mRNA accumulation and decreased extracellular accumulation mediated by LPS, IC, and ZM. Our results suggest that LPS, IC, and AZ alone or in combination with IFN-gamma stimulate early C1q production to modulate macrophage effector functions followed by an inhibition of C1q secretion when the activation process has been culminated.


Subject(s)
Antigen-Antibody Complex/pharmacology , Complement C1q/biosynthesis , Interferon-gamma/pharmacology , Lipopolysaccharides/physiology , Macrophages/metabolism , Zymosan/pharmacology , Animals , Autoradiography , Base Sequence , Blotting, Northern , Blotting, Western , Complement C1q/genetics , Complement C3b/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Macrophages/drug effects , Male , Mice , Mice, Inbred C3H , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
Mol Immunol ; 40(10): 709-16, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14644096

ABSTRACT

A reliable way to visualise the state of microglial activation is to monitor the microglial gene expression profile. Microglia are the only CNS resident cells that synthesise C1q, the recognition sub-component of the classical complement pathway, in vivo. C1q biosynthesis in resting ramified microglia is often low, but it increases dramatically in activated microglia. In this study, the expression of C1q was used to monitor microglial activation at all stages of 3-chloropropanediol-induced neurotoxicity, a new model of blood-brain barrier (BBB) breakdown. In rats, 3-chloropropanediol produces very focused lesions in the brain, characterised by early astrocyte swelling and loss, followed by neuronal death and barrier dysfunction. Using in situ hybridisation, immunohistochemistry, and real-time RT-PCR, we found that increased C1q biosynthesis and microglial activation precede BBB dysfunction by at least 18 and peak 48 h after injection of 3-chloropropanediol, which coincides with the onset of active haemorrhage. Microglial activation is biphasic; an early phase of global activation is followed by a later phase in which microglial activation becomes increasingly focused in the lesions. During the early phase, expression of the pro-inflammatory mediators interleukin-1beta (IL1beta), tumour necrosis factor alpha (TNFalpha) and early growth response-1 (Egr-1) increased in parallel with C1q, but was restricted to the lesions. Expression of C1q (but not IL1beta, TNFalpha or Egr-1) remains high after BBB function is restored, and is accompanied by late up-regulation of the C1q-associated serine proteases, C1r and C1s, suggesting that microglial biosynthesis of the activation complex of the classical pathway may support the removal of cell debris by activation of complement.


Subject(s)
Blood-Brain Barrier/immunology , Complement C1q/biosynthesis , Immediate-Early Proteins , Microglia/immunology , Animals , Base Sequence , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain/immunology , Brain/metabolism , Complement C1q/genetics , DNA, Complementary/genetics , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , In Situ Hybridization , Interleukin-1/genetics , Male , Microglia/drug effects , Microglia/physiology , Models, Neurological , Neurotoxins/toxicity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/genetics , alpha-Chlorohydrin/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL