Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 69
1.
Mol Immunol ; 170: 19-25, 2024 Jun.
Article En | MEDLINE | ID: mdl-38598870

The assembly of tissue-damaging membrane attack complexes (MACs; C5b-9) is a major mechanism by which excessive complement activation causes diseases. We previously developed a mouse anti-human C6 monoclonal antibody (mAb) 1C9 that selectively inhibits the assembly of MACs in human and non-human primates. In this project, we found that 1C9 also cross-reacted with rat and guinea pig C6, and determined its binding domains on C6 using different truncated C6 proteins. We then humanized the anti-C6 mAb by molecular modeling and complementarity-determining region grafting. After screening a library of 276 humanized variants with different combinations of humanized light and heavy chains in biophysical assays, we identified clone 3713 with the best developability profile, and an increased affinity against C6 when compared with the parental 1C9 mAb. This humanized 3713 mAb inhibited human, monkey, and rat complement-mediated hemolysis in vitro, and more importantly, it significantly reduced complement-mediated hemolysis in vivo in rats. These results demonstrated the successful humanization of the anti-C6 mAb and suggested that the humanized 3713 mAb could be further developed as a new therapeutic that selectively targets MAC for certain complement-mediated pathological conditions.


Antibodies, Monoclonal , Complement C6 , Hemolysis , Animals , Humans , Rats , Guinea Pigs , Mice , Hemolysis/drug effects , Hemolysis/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Complement C6/immunology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Complement Activation/immunology , Complement Activation/drug effects , Complement Membrane Attack Complex/immunology , Cross Reactions/immunology
2.
Dev Comp Immunol ; 123: 104156, 2021 10.
Article En | MEDLINE | ID: mdl-34077766

The complement component 6 (C6) gene is a component of the membrane attack complex (MAC), which causes rapid lytic destruction of bacteria. MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene stability, including that of immune genes. However, current research on the function of C6 and its regulation by miRNAs is lacking. In the present study, we identified and characterized C6 and a novel miRNA, miR-727 (designated CsC6 and Cse-miR-727, respectively), of the half-smooth tongue sole (Cynoglossus semilaevis) that responded to infection with Vibrio anguillarum, a Gram-negative pathogen of marine fish. The full-length cDNA of CsC6 contained a 256 bp 5' untranslated region (5'-UTR), a 2820 bp open reading frame (ORF) encoding 939 amino acids, and a 205 bp 3'-UTR. SMART analysis showed that CsC6 contains typical C6 domains, including three TSP1 domains, one LDLa domain, one MACPF domain, two CCP domains and two FIMAC domains. CsC6 and Cse-miR-727 are widely expressed in the 13 tissues of half-smooth tongue sole, and their expression in immune tissues is significantly changed after V. anguillarum infection, generally showing an inverse trend. We confirmed that CsC6 was the target gene of Cse-miR-727 using the dual luciferase reporter assay and that Cse-miR-727 regulated CsC6 at the protein level using quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting. The hepatic expression levels of not only the MAC components C7, C8α, C8ß, C8γ and C9 but also the MAPKs, NF-κß, AP-1, IL1ß, IL6 and TNFα, which are involved in many signaling pathways, changed significantly in half-smooth tongue sole following stimulation with the Cse-miR-727 agomir and inhibitor. This evidence suggested that CsC6 could be mediated by Cse-miR-727 to affect MAC assembly and immune signaling pathways in half-smooth tongue soles. To our best knowledge, this study is the first to investigate the regulatory mechanism and immune response of complement genes mediated by miRNAs in fish.


Complement C6/immunology , Fish Diseases/immunology , Fish Proteins/immunology , Flatfishes/immunology , Liver/physiology , MicroRNAs/immunology , Vibrio Infections/immunology , Vibrio/physiology , Animals , Bacteriolysis/genetics , Cloning, Molecular , Complement C6/genetics , Fish Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation , Immunity, Innate , MicroRNAs/genetics
3.
Medicine (Baltimore) ; 99(21): e20362, 2020 May 22.
Article En | MEDLINE | ID: mdl-32481330

RATIONALE: Late complement deficiency increases susceptibility to meningococcal disease and recurrent infections. In Korea, 5 case reports have described meningococcal disease with complement deficiency. However, C6 deficiency has not been described previously. PATIENT CONCERNS: A 21-year-old police trainee presented with recurrent meningococcal meningitis. He was housed in communal living quarters until 20 days before the initial symptom onset. DIAGNOSIS: He was diagnosed with meningococcal meningitis with C6 deficiency. INTERVENTIONS: He was treated with intravenous ceftriaxone. An additional dose of quadrivalent meningococcal conjugate vaccine was administered after discharge. OUTCOMES: He was discharged without complications. LESSONS: Screening for complement deficiency is necessary in patients with a history of recurrent meningococcal infections to provide appropriate care and prevent recurrent infections.


Complement C6/deficiency , Meningitis, Meningococcal/diagnosis , Complement C6/immunology , Exanthema/etiology , Fever/etiology , Headache/etiology , Humans , Male , Meningitis, Meningococcal/drug therapy , Meningitis, Meningococcal/immunology , Meningococcal Vaccines/standards , Meningococcal Vaccines/therapeutic use , Recurrence , Republic of Korea , Young Adult
4.
Mol Immunol ; 118: 91-98, 2020 02.
Article En | MEDLINE | ID: mdl-31862673

The purpose of this study was to identify a membrane-bound complement inhibitor that could be overexpressed on retinal pigment epithelial cells (RPE) providing a potential therapy for age-related macular degeneration (AMD). This type of therapy may allow replacement of damaged RPE with cells that are able to limit complement activation in the retina. Complement Receptor 1 (CR1) is a membrane-bound complement inhibitor commonly found on erythrocytes and immune cells. In this study, QPCR and flow cytometry data demonstrated that CR1 is not well-expressed by RPE, indicating that its overexpression may provide extra protection from complement activation. To screen CR1 for this ability, a stable CR1-expressing ARPE19 line was created using a combination of antibiotic selection and FACS. Cell-based assays were used to demonstrate that addition of CR1 inhibited deposition of complement proteins C3b and C6 on the transfected line. In the end, this study identifies CR1 as a complement inhibitor that may be overexpressed on stem cell-derived RPE to create a potential "enhanced" cell therapy for AMD. A combination cell/complement therapy may create transplantable RPE better suited to avoid complement-mediated lysis and limit chronic inflammation in the retina.


Epithelial Cells/immunology , Macular Degeneration/immunology , Receptors, Complement 3b/immunology , Retina/immunology , Retinal Pigment Epithelium/immunology , Retinal Pigments/immunology , Cell Line , Complement Activation/immunology , Complement C3b/immunology , Complement C6/immunology , Erythrocytes/immunology , Humans
5.
Kidney Int ; 96(1): 67-79, 2019 07.
Article En | MEDLINE | ID: mdl-30910380

Atypical hemolytic uremic syndrome (aHUS) is a form of thrombotic microangiopathy (TMA) caused by dysregulated complement activation. Clinically, aHUS is effectively treated by an anti-C5 monoclonal antibody (mAb) but whether the disease is mediated by the C5a receptor (C5aR) or C5b-9 pathway, or both, is unknown. Here we address this in a factor H mutant mouse (FHR/R) which developed complement-mediated TMA as well as macrovascular thrombosis caused by an aHUS-related factor H point mutation (mouse W1206R, corresponding to human W1183R). C5 deficiency and anti-C5 mAb treatment blocked all disease manifestations in FHR/R mice. C5aR1 gene deficiency prevented macrovascular thrombosis in various organs but did not improve survival or reduce renal TMA. Conversely, C6 or C9 deficiency significantly improved survival and markedly diminished renal TMA but did not prevent macrovascular thrombosis. Interestingly, as they aged both FHR/R C6-/- and FHR/R C9-/- mice developed glomerular disease reminiscent of C3 glomerulonephritis. Thus, C5aR and C5b-9 pathways drove different aspects of disease in FHR/R mice with the C5aR pathway being responsible for macrovascular thrombosis and chronic inflammatory injury while the C5b-9 pathway caused renal TMA. Our data provide new understanding of the pathogenesis of complement-mediated TMA and macrovascular thrombosis in FHR/R mice and suggest that C5 blockade is more effective for the treatment of aHUS than selectively targeting the C5aR or C5b-9 pathway alone.


Atypical Hemolytic Uremic Syndrome/immunology , Complement Factor H/genetics , Complement Membrane Attack Complex/immunology , Kidney Glomerulus/pathology , Receptor, Anaphylatoxin C5a/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Atypical Hemolytic Uremic Syndrome/drug therapy , Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/pathology , Complement Activation/drug effects , Complement Activation/genetics , Complement Activation/immunology , Complement C6/genetics , Complement C6/immunology , Complement C6/metabolism , Complement Factor H/immunology , Complement Membrane Attack Complex/genetics , Complement Membrane Attack Complex/metabolism , Disease Models, Animal , Female , Humans , Kidney Glomerulus/blood supply , Kidney Glomerulus/ultrastructure , Male , Mice , Mice, Transgenic , Microscopy, Electron , Point Mutation , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/metabolism
6.
Fish Shellfish Immunol ; 81: 1-9, 2018 Oct.
Article En | MEDLINE | ID: mdl-29981471

The redlip mullet (Liza haematocheila) is one of the most economically important fish in Korea and other East Asian countries; it is susceptible to infections by pathogens such as Lactococcus garvieae, Argulus spp., Trichodina spp., and Vibrio spp. Learning about the mechanisms of the complement system of the innate immunity of redlip mullet is important for efforts towards eradicating pathogens. Here, we report a comprehensive study of the terminal complement complex (TCC) components that form the membrane attack complex (MAC) through in-silico characterization and comparative spatial and temporal expression profiling. Five conserved domains (TSP1, LDLa, MACPF, CCP, and FIMAC) were detected in the TCC components, but the CCP and FIMAC domains were absent in MuC8ß and MuC9. Expression analysis of four TCC genes from healthy redlip mullets showed the highest expression levels in the liver, whereas limited expression was observed in other tissues; immune-induced expression in the head kidney and spleen revealed significant responses against Lactococcus garvieae and poly I:C injection, suggesting their involvement in MAC formation in response to harmful pathogenic infections. Furthermore, the response to poly I:C may suggest the role of TCC components in the breakdown of the membrane of enveloped viruses. These findings may help to elucidate the mechanisms behind the complement system of the teleosts innate immunity.


Complement Membrane Attack Complex/genetics , Immunity, Innate , Smegmamorpha/immunology , Animals , Complement C6/genetics , Complement C6/immunology , Complement C7/genetics , Complement C7/immunology , Complement C8/genetics , Complement C8/immunology , Complement C9/genetics , Complement C9/immunology , Complement Membrane Attack Complex/immunology , Gene Expression Profiling , Lactococcus , Lipopolysaccharides , Liver/immunology , Poly I-C/pharmacology , Smegmamorpha/genetics , Spleen/immunology
7.
Front Immunol ; 9: 535, 2018.
Article En | MEDLINE | ID: mdl-29616034

Epidermolysis bullosa acquisita (EBA) is an antibody-mediated blistering skin disease associated with tissue-bound and circulating autoantibodies to type VII collagen (COL7). Transfer of antibodies against COL7 into mice results in a subepidermal blistering phenotype, strictly depending on the complement component C5. Further, activation predominantly by the alternative pathway is required to induce experimental EBA, as blistering was delayed and significantly ameliorated only in factor B-/- mice. However, C5 deficiency not only blocked the activation of terminal complement components and assembly of the membrane attack complex (MAC) but also eliminated the formation of C5a. Therefore, in the present study, we first aimed to elucidate which molecules downstream of C5 are relevant for blister formation in this EBA model and could be subsequently pharmaceutically targeted. For this purpose, we injected mice deficient in C5a receptor 1 (C5aR1) or C6 with antibodies to murine COL7. Importantly, C5ar1-/- mice were significantly protected from experimental EBA, demonstrating that C5a-C5aR1 interactions are critical intermediates linking pathogenic antibodies to tissue damage in this experimental model of EBA. By contrast, C6-/- mice developed widespread blistering disease, suggesting that MAC is dispensable for blister formation in this model. In further experiments, we tested the therapeutic potential of inhibitors of complement components which were identified to play a key role in this experimental model. Complement components C5, factor B (fB), and C5aR1 were specifically targeted using complement inhibitors both prophylactically and in mice that had already developed disease. All complement inhibitors led to a significant improvement of the blistering phenotype when injected shortly before anti-COL7 antibodies. To simulate a therapeutic intervention, anti-fB treatment was first administered in full-blown EBA (day 5) and induced significant amelioration only in the final phase of disease evolution, suggesting that early intervention in disease development may be necessary to achieve higher efficacy. Anti-C5 treatment in incipient EBA (day 2) significantly ameliorated disease during the whole experiment. This finding is therapeutically relevant, since the humanized anti-C5 antibody eculizumab is already successfully used in patients. In conclusion, in this study, we have identified promising candidate molecules for complement-directed therapeutic intervention in EBA and similar autoantibody-mediated diseases.


Autoimmune Diseases/immunology , Complement Activation , Epidermolysis Bullosa Acquisita/immunology , Animals , Collagen Type VII/immunology , Complement C6/deficiency , Complement C6/immunology , Female , Hereditary Complement Deficiency Diseases , Immunologic Deficiency Syndromes/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/immunology
8.
Epidemiol Mikrobiol Imunol ; 66(2): 80-85, 2017.
Article Cs | MEDLINE | ID: mdl-28691831

AIM OF THE STUDY: The study was focused on testing the diagnostic value of detection of the chemokine CXCL13 (B lymphocyte chemoattractant) and anti-C6 peptide (synthetic peptide derived from B. burdorferi VlsE protein) antibodies in patients with neuroborreliosis (NB). MATERIAL AND METHODS: One hundred and twenty-nine patients with clinical suspicion of neuroinfection were included in the study. Eighty patients with NB (positive for antibodies in serum and CSF) were subdivided into four groups (A1-A4) based on positivity/negativity of the antibody index (AI) and pleocytosis. The control group was composed of 49 patients with a negative AI and absence of CSF pleocytosis. Chemokine CXCL13 and anti-C6 antibodies were examined by commercial kits (Human CXCL13/BLC/BCA-1 Immunoassay, R&D Systems, INC, USA and C6 B. burgdorferi (Lyme) ELISA, Immunetics Inc. USA). The CXCL13 cut-off values were set to 130 pg/ml for the CSF and 62 pg/ml for the serum. RESULTS: The highest CSF levels of CXCL13 chemokine were found in group A1 (pleocytosis, AI positive), and they were significantly higher (p < 0.001) comparing with other groups except A3 (pleocytosis, AI negative; p = 0.04). Group A3 also showed significantly higher levels of CXCL13 than groups A2 (without pleocytosis, AI positive; p = 0.005), A4 (without pleocytosis, AI negative), and B (p < 0.001). The differences in the serum CXCL13 levels between groups were non-significant. The serum anti-C6 antibodies were detected in all NB groups and the positivity rates did not differ between groups (92%) except for A3 where 55% of the patients were positive. In the CSF, the highest anti-C6 sensitivity was found in the patients with a positive AI (A1 88.6%; A2 76.9%) while in the groups with a negative AI, it was low (A3 25%; A4 0%). In group B, anti-C6 antibodies were not detected. CONCLUSION: The highest CSF CXCL13 levels were found in early stage NB. Elevated CXCL13 concentrations correlate better with pleocytosis than with AI positivity; however, there exist some patients with a positive AI who have low CXCL13 levels. These patients are most probably those in the late - subacute stage of neuroinfection. The CXCL13 testing seems to be the most diagnostically helpful in the acute stage of NB where AI is still negative. The clinical sensitivity of the C6 ELISA test appears to be insufficient for CSF examination under our conditions. On the contrary, the specificity of this test was proven high, because none of the controls tested positive.


Antibodies/blood , Chemokine CXCL13 , Complement C6 , Lyme Neuroborreliosis , Chemokine CXCL13/immunology , Complement C6/immunology , Humans , Lyme Neuroborreliosis/blood , Lyme Neuroborreliosis/diagnosis , Lyme Neuroborreliosis/immunology
9.
J Immunol ; 197(4): 1276-86, 2016 08 15.
Article En | MEDLINE | ID: mdl-27421478

Complement activation occurs during enterohemorrhagic Escherichia coli (EHEC) infection and may exacerbate renal manifestations. In this study, we show glomerular C5b-9 deposits in the renal biopsy of a child with EHEC-associated hemolytic uremic syndrome. The role of the terminal complement complex, and its blockade as a therapeutic modality, was investigated in a mouse model of E. coli O157:H7 infection. BALB/c mice were treated with monoclonal anti-C5 i.p. on day 3 or 6 after intragastric inoculation and monitored for clinical signs of disease and weight loss for 14 d. All infected untreated mice (15 of 15) or those treated with an irrelevant Ab (8 of 8) developed severe illness. In contrast, only few infected mice treated with anti-C5 on day 3 developed symptoms (three of eight, p < 0.01 compared with mice treated with the irrelevant Ab on day 3) whereas most mice treated with anti-C5 on day 6 developed symptoms (six of eight). C6-deficient C57BL/6 mice were also inoculated with E. coli O157:H7 and only 1 of 14 developed disease, whereas 10 of 16 wild-type mice developed weight loss and severe disease (p < 0.01). Complement activation via the terminal pathway is thus involved in the development of disease in murine EHEC infection. Early blockade of the terminal complement pathway, before the development of symptoms, was largely protective, whereas late blockade was not. Likewise, lack of C6, and thereby deficient terminal complement complex, was protective in murine E. coli O157:H7 infection.


Complement C6/antagonists & inhibitors , Complement Membrane Attack Complex/antagonists & inhibitors , Escherichia coli Infections/immunology , Hemolytic-Uremic Syndrome/immunology , Animals , Child, Preschool , Complement C6/immunology , Complement Membrane Attack Complex/immunology , Disease Models, Animal , Enterohemorrhagic Escherichia coli , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
10.
Immunobiology ; 221(11): 1293-303, 2016 11.
Article En | MEDLINE | ID: mdl-27316715

As a potent effector of innate immunity, the complement system has been shown to be involved in the pathogenesis of inflammatory bowel disease (IBD). However, the role of the membrane attack complex (MAC) in the development of IBD is still largely unknown. Here, we used C6-deficient mice in which MAC formation was blocked due to the absence of C6 to develop an acute colitis model by the administration of dextran sulfate sodium (DSS). The results showed that DSS-induced colitis was aggravated in C6-deficient mice compared with wild-type (WT) mice, as represented by the markedly greater weight loss, higher disease activity index (DAI), shortened colon length, more severe histological injury with increased epithelial ulcerations, and massively increased infiltration of leukocytes accompanied by much higher myeloperoxidase (MPO) levels in local inflammatory colonic sites. In addition, the DSS-induced colitis in C6-deficient mice could be significantly ameliorated by the exogenous C6 from WT sera. Furthermore, the significantly enhanced production of pro-inflammatory mediators, including IL-1ß, IL-6, CXCL-1, CCL-3, TGF-ß1 and IL-17F, was also observed in C6-deficient mice. Unexpectedly, the aggravated colitis in C6-deficient mice may be not due to the increase of lipopolysaccharide (LPS) levels in serum. Overall, we demonstrated that MAC exerts a protective role in acute colitis, strongly highlighting the host defense function of the complement system.


Colitis/etiology , Complement C6/deficiency , Dextran Sulfate/adverse effects , Disease Susceptibility , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/immunology , Animals , Colitis/drug therapy , Colitis/metabolism , Colitis/pathology , Complement C6/administration & dosage , Complement C6/immunology , Complement Membrane Attack Complex/immunology , Complement Membrane Attack Complex/metabolism , Complement System Proteins/immunology , Complement System Proteins/metabolism , Cytokines/metabolism , Disease Models, Animal , Hereditary Complement Deficiency Diseases , Humans , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , Mice , Mice, Knockout
11.
Mol Immunol ; 76: 13-21, 2016 08.
Article En | MEDLINE | ID: mdl-27337595

Experimental mouse models have been extensively used to elucidate the role of the complement system in different diseases and injuries. Contribution of gender has revealed an intriguing gender specific difference; female mice often show protection against most complement driven injuries such as ischemia/reperfusion injury, graft rejection and sepsis. Interestingly, early studies to the mouse complement system revealed that female mice have very low total complement activity (CH50), which is related to androgen regulation of hepatic complement synthesis. Here, our aim was to understand at which level the female specific differences in mouse complement resides. We have used recently developed complement assays to study the functional activities of female and male mice at the level of C3 and C9 activation, and furthermore assayed key complement factor levels in serum of age-matched female and male C57BL/6 mice. Our results show that the female mice have normal complement cascade functionality at the level of C3 activation, which was supported by determinations of early complement factors. However, all pathways are strongly reduced at the level of C9 activation, suggesting a terminal pathway specific difference. This was in line with C6 and C9 measurements, showing strongly decreased levels in females. Furthermore, similar gender differences were also found in BALB/cJ mice, but not in CD-1 mice. Our results clearly demonstrate that the complement system in females of frequently used mouse strains is restricted by the terminal pathway components and that the perceived female specific protection against experimental disease and injury might be in part explained by the inability promote inflammation through C5b-9.


Complement Activation/immunology , Complement C3/immunology , Complement C6/immunology , Complement C9/immunology , Sex Characteristics , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
12.
J Autoimmun ; 60: 40-50, 2015 Jun.
Article En | MEDLINE | ID: mdl-25911200

Neuropsychiatric disease is one of the most common manifestations of human systemic lupus erythematosus, but the mechanisms remain poorly understood. In human brain microvascular endothelial cells in vitro, TNF-like weak inducer of apoptosis (TWEAK) decreases tight junction ZO-1 expression and increases the permeability of monolayer cell cultures. Furthermore, knockout (KO) of the TWEAK receptor, Fn14, in the MRL/lpr lupus mouse strain markedly attenuates neuropsychiatric disease, as demonstrated by significant reductions in depressive-like behavior and improved cognitive function. The purpose of the present study was to determine the mechanisms by which TWEAK signaling is instrumental in the pathogenesis of neuropsychiatric lupus (NPSLE). Evaluating brain sections of MRL/lpr Fn14WT and Fn14KO mice, we found that Fn14KO mice displayed significantly decreased cellular infiltrates in the choroid plexus. To evaluate the integrity of the blood brain barrier (BBB) in MRL/lpr mice, Western blot for fibronectin, qPCR for iNOS, and immunohistochemical staining for VCAM-1/ICAM-1 were performed. We found preserved BBB permeability in MRL/lpr Fn14KO mice, attributable to reduced brain expression of VCAM-1/ICAM-1 and iNOS. Additionally, administration of Fc-TWEAK intravenously directly increased the leakage of a tracer (dextran-FITC) into brain tissue. Furthermore, MRL/lpr Fn14KO mice displayed reduced antibody (IgG) and complement (C3, C6, and C4a) deposition in the brain. Finally, we found that MRL/lpr Fn14KO mice manifested reduced neuron degeneration and hippocampal gliosis. Our studies indicate that TWEAK/Fn14 interactions play an important role in the pathogenesis of NPSLE by increasing the accumulation of inflammatory cells in the choroid plexus, disrupting BBB integrity, and increasing neuronal damage, suggesting a novel target for therapy in this disease.


Apoptosis/genetics , Blood-Brain Barrier/physiopathology , Neurons/pathology , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factors/immunology , Animals , Apoptosis/immunology , Choroid Plexus/physiopathology , Cognition , Complement C3/immunology , Complement C4a/immunology , Complement C6/immunology , Cytokine TWEAK , Depression/genetics , Disease Models, Animal , Gliosis/genetics , Immunoglobulin G/immunology , Intercellular Adhesion Molecule-1/metabolism , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Knockout , Nerve Degeneration/genetics , Permeability , Signal Transduction/genetics , Signal Transduction/immunology , TWEAK Receptor , Vascular Cell Adhesion Molecule-1/metabolism , Zonula Occludens-1 Protein/biosynthesis
13.
J Am Soc Nephrol ; 25(2): 225-31, 2014 Feb.
Article En | MEDLINE | ID: mdl-24179165

Necrotizing and crescentic GN (NCGN) with a paucity of glomerular immunoglobulin deposits is associated with ANCA. The most common ANCA target antigens are myeloperoxidase (MPO) and proteinase 3. In a manner that requires activation of the alternative complement pathway, passive transfer of antibodies to mouse MPO (anti-MPO) induces a mouse model of ANCA NCGN that closely mimics human disease. Here, we confirm the importance of C5aR/CD88 in the mediation of anti-MPO-induced NCGN and report that C6 is not required. We further demonstrate that deficiency of C5a-like receptor (C5L2) has the reverse effect of C5aR/CD88 deficiency and results in more severe disease, indicating that C5aR/CD88 engagement enhances inflammation and C5L2 engagement suppresses inflammation. Oral administration of CCX168, a small molecule antagonist of human C5aR/CD88, ameliorated anti-MPO-induced NCGN in mice expressing human C5aR/CD88. These observations suggest that blockade of C5aR/CD88 might have therapeutic benefit in patients with ANCA-associated vasculitis and GN.


Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/prevention & control , Autoantigens/immunology , Glomerulonephritis/prevention & control , Peroxidase/immunology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Administration, Oral , Animals , Complement C6/immunology , Complement Pathway, Alternative , Dose-Response Relationship, Drug , Gene Knock-In Techniques , Glomerulonephritis/complications , Glomerulonephritis/immunology , Hematuria/etiology , Hematuria/prevention & control , Humans , Immunization, Passive , Leukocytes , Metabolism, Inborn Errors/complications , Metabolism, Inborn Errors/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Peroxidase/deficiency , Proteinuria/etiology , Proteinuria/prevention & control , Receptor, Anaphylatoxin C5a/deficiency , Receptor, Anaphylatoxin C5a/genetics , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Receptors, Chemokine/physiology , Recombinant Fusion Proteins , Urine/cytology
14.
Dev Comp Immunol ; 34(5): 485-90, 2010 May.
Article En | MEDLINE | ID: mdl-20067805

Humoral cytotoxicity results from the assembly of terminal components of complement, called membrane attack complex (MAC), which lead to the formation of pores on pathogen membranes. The complement components involved in MAC formation are C5b, C6, C7, C8alpha, C8beta, C8gamma and C9. Among them, C6 protein interacts with C5b through a metastable binding site to form a soluble C5b-6 dimer in the vicinity of the activating cell. Formation of the MAC is controlled by complement regulatory molecules, such as CD59, vitronectin and clusterin. Here, we report the molecular characterization of the C6 complement component, as well as the spatial and temporal expression profile of MAC structural (C6, C7, C8alpha, C8beta, C8gamma) and regulatory (CD59, vitronectin and clusterin) genes in chicken (Gallus gallus). The deduced polypeptide sequence of chicken C6 consists of 935 amino acid residues and exhibits 81%, 58%, 56% and 44% identity with zebra finch, human, frog and trout orthologs, respectively. The 'domain' architecture of chicken C6 resembles that of mammalian counterparts and the cysteine backbone is also conserved. MAC structural and regulatory genes are expressed in a wide range of adult chicken tissues, with the liver being the major source of their produced transcripts. The developmental expression profile of chicken MAC structural genes shows that their transcripts initially appear in the 12th embryonic day in the liver, exhibiting a pick in the 17th, while no expression was detected in the early whole embryo (day 4 and 6), as well as in the 2-day old neonate chicken liver. On the other hand, MAC regulatory genes are expressed in all the developmental stages investigated.


CD59 Antigens/metabolism , Clusterin/metabolism , Complement C6/genetics , Complement Membrane Attack Complex/metabolism , Vitronectin/metabolism , Amino Acid Sequence , Animals , Anura , CD59 Antigens/genetics , Chick Embryo , Chickens/genetics , Chickens/immunology , Cloning, Molecular , Clusterin/genetics , Complement C6/immunology , Complement C6/metabolism , Complement Membrane Attack Complex/genetics , Complement Membrane Attack Complex/immunology , Finches , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Liver/immunology , Liver/metabolism , Molecular Sequence Data , Protein Conformation , Sequence Alignment , Trout , Vitronectin/genetics
15.
Mol Immunol ; 47(5): 1098-105, 2010 Feb.
Article En | MEDLINE | ID: mdl-19959238

AIMS: To examine the roles of the membrane attack complex of complement and its sole membrane regulator, CD59, in atherosclerosis. METHODS: C6 (C6(-/-)) deficient and CD59a (Cd59a(-/-)) knockout mice were separately crossed onto the apolipoprotein E knockout (apoE(-/-)) background. The double knockout mice were fed high-fat diet in order to study the effects of absence of C6 or CD59a on the progression of atherosclerosis. RESULTS: C6 deficiency significantly reduced plaque area and disease severity. CD59a had the opposite effect in that deficiency was associated with a significant increase in plaque area, correlating with increased membrane attack complex (MAC) deposition in the plaque and increased smooth muscle cell proliferation in early plaques. CONCLUSIONS: Our results demonstrate that the MAC contributes to the development of atherosclerosis, C6 deficiency being protective and CD59a deficiency exacerbating disease.


Apolipoproteins E , Atherosclerosis/immunology , CD59 Antigens/immunology , Complement C6/immunology , Complement Membrane Attack Complex/immunology , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , CD59 Antigens/genetics , Complement C6/genetics , Complement Membrane Attack Complex/genetics , Crosses, Genetic , Humans , Mice , Mice, Knockout
16.
Mol Immunol ; 47(2-3): 302-9, 2009 Dec.
Article En | MEDLINE | ID: mdl-19833392

Complement (C) activation is a crucial event in peripheral nerve degeneration but its effect on the subsequent regeneration is unknown. Here we show that genetic deficiency of the sixth C component, C6, accelerates axonal regeneration and recovery in a rat model of sciatic nerve injury. Foot-flick test and Sciatic Function Index monitored up to 5 weeks post-injury showed a significant improvement of sensory and motor function in the C6 deficient animals compared to wildtypes. Retrograde tracing experiments showed a significantly higher number of regenerated neurons at 1 week post-injury in C6 deficient rats than wildtypes. Pathology showed improved nerve regeneration in tibials of C6 deficient animals compared to wildtypes. Reconstitution with purified human C6 protein re-established the wildtype phenotype whereas pharmacological inhibition of C activation with soluble C receptor 1 (sCR1) facilitated recovery and improved pathology similarly to C6 deficient animals. We suggest that a destructive C-mediated event during nerve degeneration hampers the subsequent regenerative process. These findings provide a rationale for the testing of anti-complement agents in human nerve injury.


Complement C6/antagonists & inhibitors , Nerve Regeneration/immunology , Peripheral Nerve Injuries , Peripheral Nerves/immunology , Animals , Complement Activation/immunology , Complement C6/immunology , Disease Models, Animal , Gene Expression Profiling , Hemolysis/immunology , Humans , Nerve Regeneration/genetics , Peripheral Nerves/physiopathology , Peripheral Nerves/ultrastructure , Rats , Receptors, Complement/blood , Receptors, Complement/immunology , Recovery of Function
17.
Fish Shellfish Immunol ; 27(6): 768-72, 2009 Dec.
Article En | MEDLINE | ID: mdl-19716423

The terminal complement components (TCCs) of mammals, C6, C7, C8alpha, C8beta, and C9, are a group of serum proteins involved in the cytolytic killing of microbial pathogens. The mammalian TCCs share a unique core domain structure and were probably generated by the duplication of the ancestral TCC gene and subsequent addition and/or deletion of the N- and C-terminal domains. Proteins and genes for all the TCCs have been identified from bony fish. In contrast, no TCC gene has been identified from cyclostome lamprey using whole-genome shotgun-sequence analysis and liver EST analysis. To clarify the evolutionary origin of TCCs, we performed degenerate RT-PCR and RACE analyses of the cartilaginous fish liver and identified the C6 gene from a shark, Mustelus manazo, and the C8B gene from a chimaera, Chimaera phantasma. The presence of the C6 gene in shark suggests that one of the most crucial steps in the establishment of the cytolytic complement pathway, the addition of the FIM and CCP domains to the primitive TCC, occurred in a common ancestor of the jawed vertebrates. These results also indicate that the gene duplications among TCCs occurred at an early stage of the jawed vertebrate evolution.


Complement C6/genetics , Complement C8/genetics , Lampreys/immunology , Sharks/immunology , Amino Acid Sequence , Animals , Base Sequence , Complement C6/immunology , Complement C8/immunology , Evolution, Molecular , Expressed Sequence Tags , Gene Duplication , Lampreys/genetics , Liver/immunology , Molecular Sequence Data , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Sequence Alignment , Sharks/genetics
18.
Mol Immunol ; 46(5): 803-11, 2009 Feb.
Article En | MEDLINE | ID: mdl-18947875

The rodent-specific complement regulator complement receptor 1-related gene/protein-y (Crry) is critical for complement homeostasis. Gene deletion is 100% embryonically lethal; Crry-deficient (Crry(-/-)) mice were rescued by back-crossing onto C3 deficiency, confirming that embryo loss was complement mediated. In order to rescue viable Crry(-/-) mice without deleting C3, we have tested inhibition of C5 during gestation. Crry(+/-) females were given neutralizing anti-C5 mAb immediately prior to mating with Crry(+/-) males and C5 inhibition maintained through pregnancy. A single, healthy Crry(-/-) female was obtained and mating with Crry(+/-) males yielded healthy litters containing equal numbers of Crry(+/-) and Crry(-/-) pups. Inter-crossing Crry(-/-) mice yielded healthy litters of expected size. Although the mice were not anemic, exposure of Crry(-/-) erythrocytes to normal mouse serum caused C3 deposition and lysis, while transfusion into normal or C6(-/-) mice resulted in rapid clearance. Complement activity and C3 levels in Crry(-/-) mice were markedly reduced. Comparison with factor H deficient (CfH(-/-)) mice revealed similar levels of residual C3; however, unlike the CfH(-/-) mice, Crry(-/-) mice showed no evidence of renal injury, demonstrating distinct roles for these regulators in protecting the kidney.


Complement C3/immunology , Homeostasis/immunology , Kidney/immunology , Receptors, Complement/immunology , Animals , Antibodies, Monoclonal/pharmacology , Complement C3/genetics , Complement C5/antagonists & inhibitors , Complement C5/genetics , Complement C5/immunology , Complement C6/genetics , Complement C6/immunology , Complement Factor H/genetics , Complement Factor H/immunology , Crosses, Genetic , Embryo Loss/genetics , Embryo Loss/immunology , Female , Homeostasis/genetics , Kidney/injuries , Male , Mice , Mice, Knockout , Pregnancy , Receptors, Complement/genetics , Receptors, Complement 3b
20.
Clin Exp Immunol ; 146(2): 278-86, 2006 Nov.
Article En | MEDLINE | ID: mdl-17034580

Myasthenia gravis (MG) is a debilitating and potentially fatal neuromuscular disease characterized by the generation of autoantibodies reactive with nicotinic acetylcholine receptors (AChR) that cause loss of AChR from the neuromuscular endplate with resultant failure of neuromuscular transmission. A role for complement (C) in the pathology of human MG has been suggested based upon identification of C activation products in plasma and deposited at the endplate in MG. In the rat model, experimental autoimmune MG (EAMG), C depletion or inhibition restricts clinical disease, further implicating C in pathology. The mechanisms by which C activation drives pathology in MG and EAMG are unclear. Here we provide further evidence implicating C and specifically the membrane attack complex (MAC) in the Lewis rat passive EAMG model of MG. Rats deficient in C6, an essential component of the MAC, were resistant to disease induction and endplate destruction was reduced markedly compared to C6-sufficient controls. After reconstitution with C6, disease severity and endplate destruction in the C6-deficient rats was equivalent to that in controls. The data confirm the essential role of the MAC in the destruction of the endplate in EAMG and raise the prospect of specific MAC inhibition as an alternative therapy in MG patients resistant to conventional treatments.


Complement C6/immunology , Complement Membrane Attack Complex/immunology , Motor Endplate/immunology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Animals , Complement C3/metabolism , Complement C6/deficiency , Disease Susceptibility , Female , Hemolysis/immunology , Muscle, Skeletal/immunology , Myasthenia Gravis, Autoimmune, Experimental/prevention & control , Rats , Rats, Inbred Lew , Receptors, Cholinergic/metabolism , Severity of Illness Index
...