Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Biomolecules ; 13(2)2023 01 22.
Article in English | MEDLINE | ID: mdl-36830586

ABSTRACT

The terminal complement complex (TCC) has been described as a potential driver in the pathogenesis of posttraumatic osteoarthritis (PTOA). However, sublytic TCC deposition might also play a crucial role in bone development and regeneration. Therefore, we elucidated the effects of TCC on joint-related tissues using a rabbit PTOA model. In brief, a C6-deficient rabbit breed was characterized on genetic, protein, and functional levels. Anterior cruciate ligament transection (ACLT) was performed in C6-deficient (C6-/-) and C6-sufficient (C6+/-) rabbits. After eight weeks, the progression of PTOA was determined histologically. Moreover, the structure of the subchondral bone was evaluated by µCT analysis. C6 deficiency could be attributed to a homozygous 3.6 kb deletion within the C6 gene and subsequent loss of the C5b binding site. Serum from C6-/- animals revealed no hemolytic activity. After ACLT surgery, joints of C6-/- rabbits exhibited significantly lower OA scores, including reduced cartilage damage, hypocellularity, cluster formation, and osteophyte number, as well as lower chondrocyte apoptosis rates and synovial prostaglandin E2 levels. Moreover, ACLT surgery significantly decreased the trabecular number in the subchondral bone of C6-/- rabbits. Overall, the absence of TCC protected from injury-induced OA progression but had minor effects on the micro-structure of the subchondral bone.


Subject(s)
Cartilage, Articular , Osteoarthritis , Animals , Rabbits , Complement Membrane Attack Complex/pharmacology , Cartilage, Articular/pathology , Osteoarthritis/pathology , Anterior Cruciate Ligament/pathology , Anterior Cruciate Ligament/surgery , Chondrocytes/pathology
2.
J Immunol ; 209(9): 1760-1767, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36104112

ABSTRACT

Bacterial and mitochondrial DNA, sharing an evolutionary origin, act as danger-associated molecular patterns in infectious and sterile inflammation. They both contain immunomodulatory CpG motifs. Interactions between CpG motifs and the complement system are sparsely described, and mechanisms of complement activation by CpG remain unclear. Lepirudin-anticoagulated human whole blood and plasma were incubated with increasing concentrations of three classes of synthetic CpGs: CpG-A, -B, and -C oligodeoxynucleotides and their GpC sequence controls. Complement activation products were analyzed by immunoassays. Cytokine levels were determined via 27-plex beads-based immunoassay, and CpG interactions with individual complement proteins were evaluated using magnetic beads coated with CpG-B. In whole blood and plasma, CpG-B and CpG-C (p < 0.05 for both), but not CpG-A (p > 0.8 for all), led to time- and dose-dependent increase of soluble C5b-9, the alternative complement convertase C3bBbP, and the C3 cleavage product C3bc. GpC-A, -B, and -C changed soluble fluid-phase C5b-9, C3bBbP, and C3bc to the same extent as CpG-A, -B, and -C, indicating a DNA backbone-dependent effect. Dose-dependent CpG-B binding was found to C1q (r = 0.83; p = 0.006) and factor H (r = 0.93; p < 0.001). The stimulatory complement effect was partly preserved in C2-deficient plasma and completely preserved in MASP-2-deficient serum. CpG-B increased levels of IL-1ß, IL-2, IL-6, IL-8, MCP-1, and TNF in whole blood, which were completely abolished by inhibition of C5 and C5aR1 (p < 0.05 for all). In conclusion, synthetic analogs of bacterial and mitochondrial DNA activate the complement system via the DNA backbone. We suggest that CpG-B interacts directly with classical and alternative pathway components, resulting in complement-C5aR1-dependent cytokine release.


Subject(s)
Cytokines , Oligodeoxyribonucleotides , Humans , Complement Activation , Complement C1q , Complement Factor H , Complement Membrane Attack Complex/pharmacology , Complement System Proteins/metabolism , Cytokines/metabolism , DNA, Mitochondrial , Interleukin-2/pharmacology , Interleukin-6/pharmacology , Interleukin-8 , Mannose-Binding Protein-Associated Serine Proteases , Oligodeoxyribonucleotides/pharmacology , CpG Islands
3.
J Pathol ; 257(1): 29-38, 2022 05.
Article in English | MEDLINE | ID: mdl-35038170

ABSTRACT

Activation of the alternative complement pathway is an initiating event in the pathology of age-related macular degeneration (AMD). Unchecked complement activation leads to the formation of a pro-lytic pore, the membrane attack complex (MAC). MAC deposition is observed on the choriocapillaris of AMD patients and likely causes lysis of choroidal endothelial cells (CECs). Complement factor H (FH, encoded by the gene CFH) is an inhibitor of complement. Both loss of function of FH and reduced choroidal levels of FH have been reported in AMD. It is plausible that reduced local FH availability promotes MAC deposition on CECs. FH is produced primarily in the liver; however, cells including the retinal pigment epithelium can produce FH locally. We hypothesized that CECs produce FH locally to protect against MAC deposition. We aimed to investigate the effect of reduced FH levels in the choroid to determine whether increasing local FH could protect CECs from MAC deposition. We demonstrated that siRNA knockdown of FH (CFH) in human immortalized CECs results in increased MAC deposition. We generated AMD iPSC-derived CECs and found that overexpression of FH protects against MAC deposition. These results suggest that local CEC-produced FH protects against MAC deposition, and that increasing local FH protein may be beneficial in limiting MAC deposition in AMD. © 2022 The Pathological Society of Great Britain and Ireland.


Subject(s)
Complement Factor H , Macular Degeneration , Choroid/metabolism , Complement Factor H/genetics , Complement Membrane Attack Complex/metabolism , Complement Membrane Attack Complex/pharmacology , Endothelial Cells/metabolism , Humans , Macular Degeneration/genetics , Macular Degeneration/pathology , Retinal Pigment Epithelium/metabolism
4.
PLoS Pathog ; 17(11): e1010051, 2021 11.
Article in English | MEDLINE | ID: mdl-34752492

ABSTRACT

Complement proteins can form membrane attack complex (MAC) pores that directly kill Gram-negative bacteria. MAC pores assemble by stepwise binding of C5b, C6, C7, C8 and finally C9, which can polymerize into a transmembrane ring of up to 18 C9 monomers. It is still unclear if the assembly of a polymeric-C9 ring is necessary to sufficiently damage the bacterial cell envelope to kill bacteria. In this paper, polymerization of C9 was prevented without affecting binding of C9 to C5b-8, by locking the first transmembrane helix domain of C9. Using this system, we show that polymerization of C9 strongly enhanced damage to both the bacterial outer and inner membrane, resulting in more rapid killing of several Escherichia coli and Klebsiella strains in serum. By comparing binding of wildtype and 'locked' C9 by flow cytometry, we also show that polymerization of C9 is impaired when the amount of available C9 per C5b-8 is limited. This suggests that an excess of C9 is required to efficiently form polymeric-C9. Finally, we show that polymerization of C9 was impaired on complement-resistant E. coli strains that survive killing by MAC pores. This suggests that these bacteria can specifically block polymerization of C9. All tested complement-resistant E. coli expressed LPS O-antigen (O-Ag), compared to only one out of four complement-sensitive E. coli. By restoring O-Ag expression in an O-Ag negative strain, we show that the O-Ag impairs polymerization of C9 and results in complement-resistance. Altogether, these insights are important to understand how MAC pores kill bacteria and how bacterial pathogens can resist MAC-dependent killing.


Subject(s)
Blood Bactericidal Activity , Cell Wall/pathology , Complement C9/chemistry , Complement Membrane Attack Complex/pharmacology , Escherichia coli/growth & development , Klebsiella/growth & development , Polymerization , Cell Wall/drug effects , Escherichia coli/drug effects , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Humans , Klebsiella/drug effects , Klebsiella Infections/drug therapy , Klebsiella Infections/microbiology
5.
Am J Physiol Cell Physiol ; 320(3): C355-C364, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33296289

ABSTRACT

Podocytes are epithelial cells adhering glomerular capillaries, which regulate the integrity of glomerular filtration barrier. Irreversible podocyte injury induces glomerular inflammation and causes chronic renal diseases. Kcnq1ot1, a long noncoding RNA, participates in the pathogenesis of diabetic retinopathy and cardiomyopathy. However, its function in podocyte injury is elusive. Pyroptosis of murine podocyte MPC5 was triggered by sublytic complement C5b-9 (sC5b-9) for subsequent in vitro functional and mechanistic investigation. Gain/loss-of-function analysis was conducted to examine the functional role of Kcnq1ot1 in podocyte pyroptosis. Meanwhile, the molecular mechanism of Kcnq1ot1's effect on podocyte injury was explored by identifying downstream molecules and their intermediate interactions. Kcnq1ot1 was upregulated in sC5b-9-induced podocytes, and silencing Kcnq1ot1 could inhibit sC5b-9's effect on podocyte pyroptosis. We also identified the interaction between Kcnq1ot1 and miR-486a-3p, through which Kcnq1ot1 mediated miR-486a-3p inhibition by sC5b-9. Furthermore, miR-486a-3p reduced the transcriptional activity of NLRP3, while the overexpression of NLRP3 enhanced sC5b-9's effect on podocyte pyroptosis through activating NLRP3 inflammasome. sC5b-9 induces pyroptosis in podocytes through modulating the Kcnq1ot1/miR-486a-3p/NLRP3 regulatory axis, and these uncovered key molecules might facilitate podocyte-targeted treatment for renal inflammatory diseases.


Subject(s)
Complement Membrane Attack Complex/pharmacology , MicroRNAs/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Podocytes/drug effects , Potassium Channels, Voltage-Gated/genetics , Pyroptosis/genetics , RNA, Long Noncoding/genetics , Up-Regulation/genetics , Animals , Cells, Cultured , Mice , Pyroptosis/drug effects , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Up-Regulation/drug effects
6.
Front Immunol ; 11: 619, 2020.
Article in English | MEDLINE | ID: mdl-32328069

ABSTRACT

Sublytic levels of C5b-9 increase the survival of oligodendrocytes (OLGs) and induce the cell cycle. We have previously observed that SIRT1 co-localizes with surviving OLGs in multiple sclerosis (MS) plaques, but it is not yet known whether SIRT1 is involved in OLGs survival after exposure to sublytic C5b-9. We have now investigated the role of SIRT1 in OLGs differentiation and the effect of sublytic levels of C5b-9 on SIRT1 and phosphorylated-SIRT1 (Ser27) expression. We also examined the downstream effects of SIRT1 by measuring histone H3 lysine 9 trimethylation (H3K9me3) and the expression of cyclin D1 as a marker of cell cycle activation. OLG progenitor cells (OPCs) purified from the brain of rat pups were differentiated in vitro and treated with sublytic C5b-9 or C5b6. To investigate the signaling pathway activated by C5b-9 and required for SIRT1 expression, we pretreated OLGs with a c-jun antisense oligonucleotide, a phosphoinositide 3-kinase (PI3K) inhibitor (LY294002), and a protein kinase C (PKC) inhibitor (H7). Our data show a significant reduction in phospho-SIRT1 and SIRT1 expression during OPCs differentiation, associated with a decrease in H3K9me3 and a peak of cyclin D1 expression in the first 24 h. Stimulation of OLGs with sublytic C5b-9 resulted in an increase in the expression of SIRT1 and phospho-SIRT1, H3K9me3, cyclin D1 and decreased expression of myelin-specific genes. C5b-9-stimulated SIRT1 expression was significantly reduced after pretreatment with c-jun antisense oligonucleotide, H7 or LY294002. Inhibition of SIRT1 with sirtinol also abolished C5b-9-induced DNA synthesis. Taken together, these data show that induction of SIRT1 expression by C5b-9 is required for cell cycle activation and is mediated through multiple signaling pathways.


Subject(s)
Complement Membrane Attack Complex/pharmacology , Oligodendroglia/drug effects , Sirtuin 1/physiology , Animals , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Myelin Sheath/drug effects , Oligodendroglia/physiology , Phosphatidylinositol 3-Kinases/physiology , Protein Kinase C/physiology , Rats , Rats, Sprague-Dawley
7.
Cell Physiol Biochem ; 49(5): 1898-1917, 2018.
Article in English | MEDLINE | ID: mdl-30235450

ABSTRACT

BACKGROUND/AIMS: The activation of complement system and the formation of C5b-9 complex have been confirmed in the glomeruli of patients with mesangioproliferative glomerulonephritis (MsPGN). However, the role and mechanism of C5b-9-induced injury in glomerular mesangial cell (GMC) are poorly understood. Rat Thy-1N is an animal model for studying MsPGN. It has been revealed that the attack of C5b-9 to the GMC in rat Thy-1N is sublytic, and sublytic C5b-9 can cause GMC apoptosis, but the underlying mechanism is not fully elucidated. To explore the role and regulatory mechanism of C5b-9 in MsPGN lesion, we used rat Thy-1N model and first detected the change of microRNA (miRNA) profiles both in Thy-1N rat renal tissues (in vivo) and in the cultured GMCs with sublytic C5b-9 stimulation (in vitro). Then we determined the effect of miR-3546, which increased both in vivo and in vitro, on GMC apoptosis upon sublytic C5b-9 as well as the involved mechanism. METHODS: Rat Thy-1N model was established and GMCs were treated with sublytic C5b-9. The rat renal cortex and the stimulated GMCs were obtained for miRNA microarray detection. Subsequently, the increased miRNAs were verified by real-time PCR. Meanwhile, to ascertain the ability of some miRNAs to upregulate cleaved caspase 3 and induce GMC apoptosis, the corresponding miRNA mimics were transfected into GMCs, followed by western blotting (WB) and flow cytometry mesurement. Thereafter, the miR-3546-targeted gene (SOX4) was predicted using bioinformatics approaches, and SOX4 expression in Thy-1N tissues and in the GMCs upon sublytic C5b-9 stimulation or miR-3546 mimic/inhibitor transfection were detected using real-time PCR and WB. To prove that miR-3546 can affect SOX4 gene transcription and SOX4 can regulate survivin expression, dual luciferase reporter assay, real-time PCR, WB and chromatin immunoprecipitation (ChIP) assays were performed. Furthermore, the role of miR-3546/SOX4/survivin axis in the GMC apoptosis induced by sublytic C5b-9 was examined using WB and flow cytometry. RESULTS: Compared with normal renal tissues and untreated GMCs, there were 43 and 62 upregulated miRNAs (> 2-fold) in Thy-1N tissues and sublytic C5b-9-stimulated GMCs respectively. A total of 17 miRNAs were increased both in vivo and in vitro, 11 of which were validated by real-time PCR. Among them, miR-3546 could markedly promote GMC apoptosis and inhibit SOX4 or survivin expression in response to sublytic C5b-9, and either SOX4 or survivin overexpression markedly rescued the GMC apoptosis mediated by miR-3546 mimic. Additionally, SOX4 overexpression could reverse the survivin suppression by miR-3546 mimic, and SOX4 could bind to survivin promoter (-1,278 to -853 nt) and activate survivin gene transcription. CONCLUSION: MiR-3546/ SOX4/survivin axis has a promoting role in the GMC apoptosis triggered by sublytic C5b-9, and our findings may provide a new insight into the pathogenesis of rat Thy-1N and human MsPGN.


Subject(s)
Apoptosis/drug effects , Complement Membrane Attack Complex/pharmacology , Isoantibodies/pharmacology , MicroRNAs/metabolism , Microtubule-Associated Proteins/metabolism , SOXC Transcription Factors/metabolism , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Caspase 3/metabolism , Cell Line , Gene Expression Regulation/drug effects , Male , Mesangial Cells/cytology , Mesangial Cells/drug effects , Mesangial Cells/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Microtubule-Associated Proteins/genetics , Nephritis/metabolism , Nephritis/pathology , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , SOXC Transcription Factors/antagonists & inhibitors , SOXC Transcription Factors/genetics , Survivin
8.
Redox Biol ; 12: 571-581, 2017 08.
Article in English | MEDLINE | ID: mdl-28390315

ABSTRACT

Antioxidant glutathione (GSH) plays an important role in the regulation of immunity. However, little is known about its effects on humoral immunity, especially its action on effector molecules like antibody and complement. Given that these molecules contain abundant disulfide bonds, we speculated that GSH might influence the action of these proteins via its thiol function. Using a model of a glomerular mesangial cell (MC) lysis induced by antibodies plus complement, we addressed this hypothesis. Exposure of rat MCs to anti-Thy-1 antibody plus complement or anti-MC rabbit serum caused a complement-dependent cell lysis, which was completely blocked by GSH. Moreover, GSH potently prevented the antibody-mediated agglutination of red blood cells and aggregation of antibody-sensitized microspheres. Further analysis revealed that GSH inhibited antibody binding to antigens and promoted the conversion of the antibodies to its reduced forms. GSH also potently inhibited the formation and deposition of C5b-9 in MCs and suppressed both the classic and alternative complement activation pathway. Lastly, GSH attenuated P38 activation, an oxidative sensitive kinase that partially mediated the antibody- and complement-dependent MC lysis. Depletion of GSH via inhibiting gamma-glutamylcysteine synthetase or xCT transporter augmented P38 activation and sensitized MCs to the cell lysis. Collectively, our results indicate that GSH protects cells from immunological cell damage via mechanisms involving inhibition of antibody binding to the antigens, suppression of complement activation and augmentation of cellular defense mechanism. Our study provides novel mechanistic insights into the actions of GSH in the regulation of immune responses and suggests that GSH might be used to treat certain immune disorders.


Subject(s)
Complement System Proteins/pharmacology , Glutathione/pharmacology , Isoantibodies/pharmacology , Mesangial Cells/drug effects , Animals , Cells, Cultured , Complement Activation , Complement Membrane Attack Complex/pharmacology , MAP Kinase Signaling System/drug effects , Mesangial Cells/immunology , Mesangial Cells/metabolism , Protein Binding/drug effects , Rabbits , Rats
9.
J Immunol ; 198(3): 1104-1118, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28039298

ABSTRACT

The apoptosis of glomerular mesangial cells (GMCs) in the early phase of rat Thy-1 nephritis (Thy-1N), a model of human mesangioproliferative glomerulonephritis (MsPGN), is primarily triggered by sublytic C5b-9. However, the mechanism of GMC apoptosis induced by sublytic C5b-9 remains unclear. In this study, we demonstrate that expressions of TNFR1-associated death domain-containing protein (TRADD) and IFN regulatory factor-1 (IRF-1) were simultaneously upregulated in the renal tissue of Thy-1N rats (in vivo) and in GMCs under sublytic C5b-9 stimulation (in vitro). In vitro, TRADD was confirmed to be a downstream gene of IRF-1, because IRF-1 could bind to TRADD gene promoter to promote its transcription, leading to caspase 8 activation and GMC apoptosis. Increased phosphorylation of p38 MAPK was verified to contribute to IRF-1 and TRADD production and caspase 8 activation, as well as to GMC apoptosis induced by sublytic C5b-9. Furthermore, phosphorylation of MEK kinase 2 (MEKK2) mediated p38 MAPK activation. More importantly, three sites (Ser153/164/239) of MEKK2 phosphorylation were identified and demonstrated to be necessary for p38 MAPK activation. In addition, silencing of renal MEKK2, IRF-1, and TRADD genes or inhibition of p38 MAPK activation in vivo had obvious inhibitory effects on GMC apoptosis, secondary proliferation, and urinary protein secretion in rats with Thy-1N. Collectively, these findings indicate that the cascade axis of MEKK2-p38 MAPK-IRF-1-TRADD-caspase 8 may play an important role in GMC apoptosis following exposure to sublytic C5b-9 in rat Thy-1N.


Subject(s)
Apoptosis/drug effects , Caspase 8/physiology , Complement Membrane Attack Complex/pharmacology , Glomerulonephritis, Membranoproliferative/etiology , Interferon Regulatory Factor-1/physiology , MAP Kinase Kinase Kinase 2/physiology , Mesangial Cells/drug effects , TNF Receptor-Associated Death Domain Protein/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Glomerulonephritis, Membranoproliferative/pathology , Male , Mesangial Cells/pathology , Phosphorylation , Rats , Rats, Sprague-Dawley
10.
PLoS One ; 11(1): e0145945, 2016.
Article in English | MEDLINE | ID: mdl-26741681

ABSTRACT

Salmonella enterica serovar Typhi expresses a capsule of Vi polysaccharide, while most Salmonella serovars, including S. Enteritidis and S. Typhimurium, do not. Both S. Typhi and S. Enteritidis express the lipopolysaccharide O:9 antigen, yet there is little evidence of cross-protection from anti-O:9 antibodies. Vaccines based on Vi polysaccharide have efficacy against typhoid fever, indicating that antibodies against Vi confer protection. Here we investigate the role of Vi capsule and antibodies against Vi and O:9 in antibody-dependent complement- and phagocyte-mediated killing of Salmonella. Using isogenic Vi-expressing and non-Vi-expressing derivatives of S. Typhi and S. Typhimurium, we show that S. Typhi is inherently more sensitive to serum and blood than S. Typhimurium. Vi expression confers increased resistance to both complement- and phagocyte-mediated modalities of antibody-dependent killing in human blood. The Vi capsule is associated with reduced C3 and C5b-9 deposition, and decreased overall antibody binding to S. Typhi. However, purified human anti-Vi antibodies in the presence of complement are able to kill Vi-expressing Salmonella, while killing by anti-O:9 antibodies is inversely related to Vi expression. Human serum depleted of antibodies to antigens other than Vi retains the ability to kill Vi-expressing bacteria. Our findings support a protective role for Vi capsule in preventing complement and phagocyte killing of Salmonella that can be overcome by specific anti-Vi antibodies, but only to a limited extent by anti-O:9 antibodies.


Subject(s)
Antibodies, Bacterial/pharmacology , Phagocytes/drug effects , Salmonella typhi/immunology , Salmonella typhimurium/immunology , Typhoid Fever/prevention & control , Typhoid-Paratyphoid Vaccines/administration & dosage , Antibodies, Bacterial/biosynthesis , Antigens, Bacterial/immunology , Bacterial Capsules/chemistry , Bacterial Capsules/immunology , Complement C3/chemistry , Complement C3/pharmacology , Complement Membrane Attack Complex/chemistry , Complement Membrane Attack Complex/pharmacology , Humans , Immune Sera/chemistry , Immunity, Humoral , Immunization , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/blood , Lipopolysaccharides/immunology , Phagocytes/immunology , Phagocytes/microbiology , Phagocytosis/drug effects , Phagocytosis/immunology , Polysaccharides, Bacterial/antagonists & inhibitors , Polysaccharides, Bacterial/blood , Polysaccharides, Bacterial/immunology , Primary Cell Culture , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella Infections/prevention & control , Species Specificity , Typhoid Fever/immunology , Typhoid Fever/microbiology , Typhoid-Paratyphoid Vaccines/antagonists & inhibitors , Typhoid-Paratyphoid Vaccines/blood , Typhoid-Paratyphoid Vaccines/immunology
11.
J Neuroimmunol ; 267(1-2): 16-9, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24434076

ABSTRACT

Soluble C5b-9 has been described as a pro-inflammatory mediator that triggers cell activation rather than inducing cell death. Microglia is the most important immune cell involved in inflammatory response in the CNS. Although microglia activation induced by various stimuli has been well characterized, the role of C5b-9 in microglia has not been well studied. In the current experiment, we utilized assembled functional C5b-9 to treat microglia and analyzed the function. We found that soluble C5b-9 could promote microglia activation by up-regulation of costimulatory molecules and increase cytokine secretion. Our results suggested that soluble C5b-9 possessed immunoregulatory potential on microglia.


Subject(s)
Complement Membrane Attack Complex/pharmacology , Immunologic Factors/pharmacology , Microglia/drug effects , Apoptosis/drug effects , Cell Membrane/drug effects , Cell Membrane/enzymology , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Matrix Metalloproteinase 2 , Microglia/cytology , Microglia/enzymology , Signal Transduction/drug effects , Temporal Lobe/cytology , Up-Regulation/drug effects
12.
Cell Biochem Biophys ; 67(3): 1421-31, 2013.
Article in English | MEDLINE | ID: mdl-23760612

ABSTRACT

Mechanical ventilation with large tidal volumes can increase lung alveolar permeability and initiate inflammatory responses, termed ventilator-induced lung injury (VILI). VILI is characterized by an influx of inflammatory cells, increased pulmonary permeability, and endothelial and epithelial cell death. But the underlying molecular mechanisms that regulate VILI remain unclear. The purpose of this study was to investigate the mechanisms that regulate pulmonary endothelial barrier in an animal model of VILI. These data suggest that SC5b-9, as the production of the complement activation, causes increase in rat pulmonary microvascular permeability by inducing activation of RhoA and subsequent phosphorylation of myosin light chain and contraction of endothelial cells, resulting in gap formation. In general, the complement-mediated increase in pulmonary microvascular permeability may participate in VILI.


Subject(s)
Complement Membrane Attack Complex/pharmacology , Endothelial Cells/drug effects , Ventilator-Induced Lung Injury/etiology , Animals , Cell Membrane Permeability/drug effects , Cells, Cultured , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Male , Myosin Light Chains/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Ventilator-Induced Lung Injury/metabolism , rhoA GTP-Binding Protein/metabolism
13.
J Cell Sci ; 126(Pt 13): 2903-13, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23613465

ABSTRACT

The membrane attack complex of complement (MAC), apart from its classical role of lysing cells, can also trigger a range of non-lethal effects on cells, acting as a drive to inflammation. In the present study, we chose to investigate these non-lethal effects on inflammasome activation. We found that, following sublytic MAC attack, there is increased cytosolic Ca(2+) concentration, at least partly through Ca(2+) release from the endoplasmic reticulum lumen via the inositol 1,4,5-triphosphate receptor (IP3R) and ryanodine receptor (RyR) channels. This increase in intracellular Ca(2+) concentration leads to Ca(2+) accumulation in the mitochondrial matrix via the 'mitochondrial calcium uniporter' (MCU), and loss of mitochondrial transmembrane potential, triggering NLRP3 inflammasome activation and IL-1ß release. NLRP3 co-localises with the mitochondria, probably sensing the increase in calcium and the resultant mitochondrial dysfunction, leading to caspase activation and apoptosis. This is the first study that links non-lethal effects of sublytic MAC attack with inflammasome activation and provides a mechanism by which sublytic MAC can drive inflammation and apoptosis.


Subject(s)
Calcium/metabolism , Carrier Proteins/genetics , Cell Membrane/drug effects , Complement Membrane Attack Complex/pharmacology , Epithelial Cells/drug effects , Inflammasomes/drug effects , Respiratory Mucosa/drug effects , Calcium/agonists , Calcium Channels/genetics , Calcium Channels/metabolism , Carrier Proteins/immunology , Cell Membrane/immunology , Endoplasmic Reticulum/metabolism , Epithelial Cells/cytology , Epithelial Cells/immunology , Humans , Inflammasomes/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/immunology , Mitochondria/drug effects , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Primary Cell Culture , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism
14.
J Biol Chem ; 288(6): 3871-85, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23258543

ABSTRACT

In experimental membranous nephropathy, complement C5b-9-induces glomerular epithelial cell (GEC) injury and proteinuria. The effects of C5b-9 are mediated via signaling pathways, including calcium-independent phospholipase A(2)γ (iPLA(2)γ), and mitogen-activated protein kinases (MAPKs) such as extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38. The iPLA(2)γ pathway is cytoprotective. This study addresses the mechanisms of iPLA(2)γ activation. iPLA(2)γ activity was monitored by quantifying prostaglandin E(2) (PGE(2)) production. In GECs, iPLA(2)γ localized at the endoplasmic reticulum and mitochondria. Complement-mediated production of PGE(2) was amplified in GECs that overexpress iPLA(2)γ, compared with control cells, and was blocked by the iPLA(2)γ inhibitor bromoenol lactone in both iPLA(2)γ-overexpressing and control GECs. In GECs that overexpress iPLA(2)γ, complement-mediated PGE(2) production was reduced by inhibitors of MAP/ERK kinase 1 (MEK1) and p38 but not JNK. In COS-1 cells that overexpress iPLA(2)γ and cyclooxygenase-1, PGE(2) production was induced by co-expression of constitutively active MEK1 or MAPK-interacting kinase 1 (MNK1) as well as by stimulation with epidermal growth factor (EGF) + ionomycin. Complement- and EGF + ionomycin-stimulated iPLA(2)γ activity was attenuated by the S511A/S515A double mutation. Moreover, complement and EGF + ionomycin enhanced phosphorylation of Ser-511. Thus, complement-mediated activation of iPLA(2)γ is mediated via ERK and p38 pathways, and phosphorylation of Ser-511 and/or Ser-515 plays a key role in the catalytic activity and signaling of iPLA(2)γ. Defining the mechanisms by which complement activates iPLA(2)γ provides opportunities for development of novel therapeutic approaches to GEC injury and proteinuria.


Subject(s)
Complement Membrane Attack Complex/metabolism , Glomerulonephritis, Membranous/enzymology , Group VI Phospholipases A2/metabolism , Kidney Glomerulus/enzymology , Amino Acid Substitution , Animals , COS Cells , Calcium Ionophores/pharmacology , Cell Line , Chlorocebus aethiops , Complement Membrane Attack Complex/pharmacology , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Dinoprostone/genetics , Dinoprostone/metabolism , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/genetics , Epidermal Growth Factor/pharmacology , Glomerulonephritis, Membranous/genetics , Glomerulonephritis, Membranous/pathology , Group VI Phospholipases A2/genetics , Humans , Immunologic Factors/metabolism , Immunologic Factors/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Ionomycin/pharmacology , Kidney Glomerulus/injuries , Kidney Glomerulus/pathology , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Mutation, Missense , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteinuria/enzymology , Proteinuria/genetics , Rats
15.
J Pathol ; 226(4): 619-32, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21984198

ABSTRACT

The proliferation of glomerular mesangial cells (GMCs) and secretion of extracellular matrix (ECM) in rat Thy-1 nephritis (Thy-1N), resembling human mesangioproliferative glomerulonephritis (MsPGN), have been studied for many years, but the mechanisms, especially the role of signalling pathway activation and its regulation in GMCs triggered by sublytic C5b-9 complexes in Thy-1N rats remain largely unclear. In the study, the proliferation of GMCs and production of ECM as well as the role of PI3K/Akt and its regulation, both in GMCs induced by sublytic C5b-9 (in vitro) and in the renal tissues of rats with Thy-1N (in vivo), were determined and the results revealed that GMCs proliferation and ECM secretion, both in vitro and in vivo, were notably increased, and that PI3K/Akt1 activation and its regulation, such as TNF receptor-associated factor 6 (TRAF6)-mediated Akt1 ubiquitination and PI3K-dependent Akt1 phosphorylation, were involved in the process of Thy-1N induction. On the other hand, silence of the TRAF6, PI3K or Akt1 genes could obviously diminish the proliferative damages and urinary protein secretion of Thy-1N rats. Together, these data implicated that sublytic C5b-9 complexes in Thy-1N rats could promote GMCs proliferation and ECM production through TRAF6-mediated PI3K-dependent Akt1 activation, in which the ubiquitination and phosphorylation of the Akt1 signal molecule played an important role in the initiation and development of the proliferative changes in the rats with Thy-1N.


Subject(s)
Complement Membrane Attack Complex/pharmacology , Mesangial Cells/pathology , Nephritis/pathology , Phosphatidylinositol 3-Kinases/biosynthesis , Proto-Oncogene Proteins c-akt/biosynthesis , TNF Receptor-Associated Factor 6/metabolism , Animals , Cell Proliferation/drug effects , Gene Silencing , Glomerular Mesangium/drug effects , Glomerular Mesangium/metabolism , Glomerular Mesangium/pathology , Isoantibodies/pharmacology , Male , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Nephritis/chemically induced , Nephritis/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proteinuria/metabolism , Proto-Oncogene Proteins c-akt/genetics , Rats , Rats, Sprague-Dawley , TNF Receptor-Associated Factor 6/genetics , Thy-1 Antigens/immunology , Ubiquitination/drug effects
16.
Immunobiology ; 216(8): 871-81, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21396734

ABSTRACT

The sublytic C5b-9 complexes can result in glomerular mesangial cells (GMCs) apoptosis, which involved in the initiation and development of rat Thy-1 nephritis. Activating transcription factor 3 (ATF3) is an immediate early gene for cells to cope with a variety of stress signals, and our previous study revealed that ATF3 could promote GMCs apoptosis attacked by sublytic C5b-9. But the mechanism of ATF3 promoting GMCs apoptosis triggered by sublytic C5b-9 attack has not been elucidated. In this study, the data showed that the expression of ATF3, growth arrest and DNA damage-45 alpha (Gadd45α), Krüppel-like factor 6 (KLF6) and proliferating cell nuclear antigen (PCNA) in the GMCs in response to sublytic C5b-9 stimulation for the indicated time was significantly increased, and ATF3 expression could lead to GMCs apoptosis through up-regulation of Gadd45α and KLF6, but not up-regulation of PCNA. Furthermore, Gadd45α was identified as a downstream target gene regulated by ATF3 directly, and KLF6 might be regulated by ATF3 in an indirect manner.


Subject(s)
Activating Transcription Factor 3/genetics , Cell Cycle Proteins/genetics , Complement Membrane Attack Complex/immunology , Kruppel-Like Transcription Factors/genetics , Mesangial Cells/immunology , Mesangial Cells/metabolism , Nephritis/immunology , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Activating Transcription Factor 3/immunology , Activating Transcription Factor 3/metabolism , Animals , Apoptosis/immunology , Cell Cycle Proteins/immunology , Cell Cycle Proteins/metabolism , Cells, Cultured , Complement Membrane Attack Complex/adverse effects , Complement Membrane Attack Complex/metabolism , Complement Membrane Attack Complex/pharmacology , DNA Damage , Gene Expression , Gene Silencing/drug effects , Kruppel-Like Factor 6 , Kruppel-Like Transcription Factors/immunology , Kruppel-Like Transcription Factors/metabolism , Luciferases/analysis , Mesangial Cells/cytology , Nephritis/genetics , Nephritis/metabolism , Nephritis/pathology , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Plasmids , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/immunology , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/pharmacology , Rats , Signal Transduction/genetics , Signal Transduction/immunology , Transcriptional Activation/immunology , Transfection , Up-Regulation
17.
Exp Mol Pathol ; 86(2): 87-94, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19162005

ABSTRACT

Proliferation of vascular endothelial cells (EC) and smooth muscle cells (SMC) is a critical event in angiogenesis and atherosclerosis. We previously showed that the C5b-9 assembly during complement activation induces cell cycle in human aortic EC (AEC) and SMC. C5b-9 can induce the expression of Response Gene to Complement (RGC)-32 and over expression of this gene leads to cell cycle activation. Therefore, the present study was carried out to test the requirement of endogenous RGC-32 for the cell cycle activation induced by C5b-9 by knocking-down its expression using siRNA. We identified two RGC-32 siRNAs that can markedly reduce the expression of RGC-32 mRNA in AEC. RGC-32 silencing in these cells abolished DNA synthesis induced by C5b-9 and serum growth factors, indicating the requirement of RGC-32 activity for S-phase entry. RGC-32 siRNA knockdown also significantly reduced the C5b-9 induced CDC2 activation and Akt phosphorylation. CDC2 does not play a role in G1/S transition in HeLa cells stably overexpressing RGC-32. RGC-32 was found to physically associate with Akt and was phosphorylated by Akt in vitro. Mutation of RGC-32 protein at Ser 45 and Ser 47 prevented Akt mediated phosphorylation. In addition, RGC-32 was found to regulate the release of growth factors from AEC. All these data together suggest that cell cycle induction by C5b-9 in AEC is RGC-32 dependent and this is in part through regulation of Akt and growth factor release.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle/drug effects , Complement Membrane Attack Complex/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Muscle Proteins/metabolism , Nerve Tissue Proteins/metabolism , Adult , Angiogenesis Inducing Agents/metabolism , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Endothelial Cells/enzymology , Enzyme Activation/drug effects , HeLa Cells , Humans , Membrane Proteins/metabolism , Muscle Proteins/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/metabolism
18.
Rheumatology (Oxford) ; 48(3): 293-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19168833

ABSTRACT

OBJECTIVE: Complement activation products contribute to a large number of inflammatory diseases, including RA. We have investigated whether osteoprotegerin (OPG) may concur with the soluble terminal complement complex (SC5b-9) to the inflammatory cascade characterizing RA. METHODS: Levels of SC5b-9 and OPG in the plasma and SF of patients with active RA were determined by ELISA. The presence of SC5b-9 and OPG in RA synovial lesions was analysed by immunohistochemistry. Cultured endothelial cells were used for in vitro leucocyte/endothelial cell adhesion assays. In addition, endothelial cells were exposed to SC5b-9 in order to evaluate the effects on the production of OPG protein, as well as the activation of the OPG promoter. RESULTS: Patients affected by active RA are characterized by elevated levels of both SC5b-9 and OPG in plasma and/or SF. Of note, we have observed a co-localization of SC5b-9 and OPG in endothelial cells of post-capillary venules of RA synovial lesions. Data on endothelial cell cultures showed that exposure to SC5b-9 induced the up-regulation of OPG expression/release, stimulating the transcriptional activity of the OPG promoter, and synergized with TNF-alpha in up-regulating OPG production. CONCLUSIONS: Our findings demonstrate that SC5b-9 induces OPG production by endothelial cells and we propose that the SC5b-9-mediated up-regulation of OPG may be an important mechanism whereby complement contributes in promoting and/or enhancing the inflammation in RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Complement Membrane Attack Complex/physiology , Endothelial Cells/metabolism , Osteoprotegerin/biosynthesis , Adult , Aged , Cell Adhesion/physiology , Cells, Cultured , Complement Membrane Attack Complex/pharmacology , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelium, Vascular/metabolism , Female , Humans , Male , Middle Aged , Neutrophil Infiltration/physiology , Neutrophils/physiology , Synovial Membrane/metabolism , Up-Regulation/drug effects
19.
J Am Soc Nephrol ; 17(3): 707-15, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16467447

ABSTRACT

Complement activation in the kidney after ischemia/reperfusion (I/R) seems to occur primarily via the alternative complement pathway. The ability of an inhibitory mAb to mouse factor B, a necessary component of the alternative pathway, to protect mice from ischemic acute renal failure was tested. Treatment with the mAb prevented the deposition of C3b on the tubular epithelium and the generation of systemic C3a after renal I/R. Treated mice had significantly lower increases in serum urea nitrogen and developed significantly less morphologic injury of the kidney after I/R. For gaining insight into potential mechanisms of protection, the activity of caspases within the kidney also was measured, and it was found that caspases-2, -3, and -9 increased in a complement-dependent manner after renal I/R. Apoptotic cells were detected by terminal deoxynucleotidyl transferase catalyzed labeling of DNA fragments, and mice in which the alternative pathway was inhibited demonstrated significantly less apoptosis than control mice. Thus, use of an inhibitory mAb to mouse factor B effectively prevented activation of complement in the kidney after I/R and protected the mice from necrotic and apoptotic injury of the tubules.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Complement Factor B/drug effects , Complement Membrane Attack Complex/pharmacology , Kidney Diseases/prevention & control , Reperfusion Injury/prevention & control , Analysis of Variance , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/physiology , Blotting, Western , Complement Activation , Complement Factor B/metabolism , Disease Models, Animal , Fluorescent Antibody Technique , Kidney Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Probability , Random Allocation , Renal Circulation , Reperfusion Injury/pathology
20.
Am J Physiol Renal Physiol ; 289(3): F593-603, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15855657

ABSTRACT

Extracellular signals may be transmitted to nuclear or cytoplasmic effectors via the mitogen-activated protein kinases. In the passive Heymann nephritis (PHN) model of membranous nephropathy, complement C5b-9 induces glomerular epithelial cell (GEC) injury, proteinuria, and activation of phospholipases and protein kinases. This study addresses the complement-mediated activation of the extracellular signal-regulated kinase (ERK). C5b-9 induced ERK threonine202/tyrosine204 phosphorylation (which correlates with activation) in GEC in culture and PHN in vivo. Expression of a dominant-inhibitory mutant of Ras reduced complement-mediated activation of ERK, but activation was not affected significantly by downregulation of protein kinase C. Complement-induced ERK activation resulted in phosphorylation of cytosolic phospholipase A2 and was, in part, responsible for phosphorylation of mitogen-activated protein kinase-associated protein kinase-2, but did not induce phosphorylation of the transcription factor, Elk-1. Activation of ERK was attenuated by drugs that disassemble the actin cytoskeleton (cytochalasin D, latrunculin B), and these compounds interfered with the activation of ERK by mitogen-activated protein kinase kinase (MEK). Overexpression of a constitutively active RhoA as well as inhibition of Rho-associated kinase blocked complement-mediated ERK activation. Complement cytotoxicity was enhanced after disassembly of the actin cytoskeleton but was unaffected after inhibition of complement-induced ERK activation. However, complement cytotoxicity was enhanced in GEC that stably express constitutively active MEK. Thus complement-induced ERK activation depends on cytoskeletal remodelling and affects the regulation of distinct downstream substrates, while chronic, constitutive ERK activation exacerbates complement-mediated GEC injury.


Subject(s)
Complement Membrane Attack Complex/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Glomerulonephritis/immunology , Glomerulonephritis/metabolism , Kidney Glomerulus/enzymology , Actins/metabolism , Animals , Cells, Cultured , Cytoskeleton/metabolism , Kidney Glomerulus/cytology , Kidney Glomerulus/immunology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Phosphorylation , Rats , ras Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...