Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
2.
BMC Med Genet ; 21(1): 241, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33334325

ABSTRACT

BACKGROUND: Hepatitis B is known to cause several forms of liver diseases including chronic hepatitis B (CHB), and hepatocellular carcinoma. Previous genome-wide association study of CHB risk has demonstrated that rs12614 of complement factor B (CFB) was significantly associated with CHB risk. In this study, fine-mapping study of previously reported GWAS single nucleotide polymorphism (SNP; CFB rs12614) was performed to validate genetic effect of rs12614 on CHB susceptibility and identify possible additional causal variants around rs12614 in a Korean population. This association study was conducted in order to identify genetic effects of CFB single nucleotide polymorphisms (SNPs) and to identify additional independent CHB susceptible causal markers within a Korean population. METHODS: A total of 10 CFB genetic polymorphisms were selected and genotyped in 1716 study subjects comprised of 955 CHB patients and 761 population controls. RESULTS: A non-synonymous variant, rs12614 (Arg32Trp) in exon2 of CFB, had significant associations with risk of CHB (odds ratio = 0.43, P = 5.91 × 10- 10). Additional linkage disequilibrium and conditional analysis confirmed that rs12614 had independent genetic effect on CHB susceptibility with previously identified CHB markers. The genetic risk scores (GRSs) were calculated and the CHB patients had higher GRSs than the population controls. Moreover, OR was found to increase significantly with cumulative GRS. CONCLUSIONS: rs12614 showed significant genetic effect on CHB risk within the Korean population. As such rs12614 may be used as a possible causal genetic variant for CHB susceptibility.


Subject(s)
Complement Factor B/genetics , Exons , Genetic Predisposition to Disease , Hepatitis B, Chronic/genetics , Polymorphism, Single Nucleotide , Adult , Case-Control Studies , Complement Factor B/deficiency , Complement Factor B/immunology , Gene Expression , Hepatitis B, Chronic/diagnosis , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/pathology , Humans , Linkage Disequilibrium , Male , Middle Aged , Odds Ratio , Republic of Korea , Risk
3.
Article in English | MEDLINE | ID: mdl-28894700

ABSTRACT

Nosocomial pathogens that develop multidrug resistance present an increasing problem for healthcare facilities. Due to its rapid rise in antibiotic resistance, Acinetobacter baumannii is one of the most concerning gram-negative species. A. baumannii typically infects immune compromised individuals resulting in a variety of outcomes, including pneumonia and bacteremia. Using a murine model for bacteremia, we have previously shown that the type II secretion system (T2SS) contributes to in vivo fitness of A. baumannii. Here, we provide support for a role of the T2SS in protecting A. baumannii from human complement as deletion of the T2SS gene gspD resulted in a 100-fold reduction in surviving cells when incubated with human serum. This effect was abrogated in the absence of Factor B, a component of the alternative pathway of complement activation, indicating that the T2SS protects A. baumannii against the alternative complement pathway. Because inactivation of the T2SS results in loss of secretion of multiple enzymes, reduced in vivo fitness, and increased sensitivity to human complement, the T2SS may be a suitable target for therapeutic intervention. Accordingly, we developed and optimized a whole-cell high-throughput screening (HTS) assay based on secreted lipase activity to identify small molecule inhibitors of the T2SS. We tested the reproducibility of our assay using a 6,400-compound library. With small variation within controls and a dynamic range between positive and negative controls, the assay had a z-factor of 0.65, establishing its suitability for HTS. Our screen identified the lipase inhibitors Orlistat and Ebelactone B demonstrating the specificity of the assay. To eliminate inhibitors of lipase activity and lipase expression, two counter assays were developed and optimized. By implementing these assays, all seven tricyclic antidepressants present in the library were found to be inhibitors of the lipase, highlighting the potential of identifying alternative targets for approved pharmaceuticals. Although no T2SS inhibitor was identified among the compounds that reduced lipase activity by ≥30%, our small proof-of-concept pilot study indicates that the HTS regimen is simple, reproducible, and specific and that it can be used to screen larger libraries for the identification of T2SS inhibitors that may be developed into novel A. baumannii therapeutics.


Subject(s)
Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Small Molecule Libraries/pharmacology , Type II Secretion Systems/antagonists & inhibitors , Acinetobacter Infections/microbiology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/isolation & purification , Bacteremia/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Complement Factor B/deficiency , Drug Resistance, Multiple/drug effects , Genetic Fitness , Humans , Lactones/pharmacology , Orlistat , Pilot Projects , Reproducibility of Results , Type II Secretion Systems/genetics , Type II Secretion Systems/metabolism
4.
Am J Transplant ; 17(9): 2312-2325, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28276660

ABSTRACT

Despite the introduction of novel and more targeted immunosuppressive drugs, the long-term survival of kidney transplants has not improved satisfactorily. Early antigen-independent intragraft inflammation plays a critical role in the initiation of the alloimmune response and impacts long-term graft function. Complement activation is a key player both in ischemia/reperfusion injury (IRI) as well as in adaptive antigraft immune response after kidney transplantation. Since the alternative pathway (AP) amplifies complement activation regardless of the initiation pathways and renal IR injured cells undergo uncontrolled complement activation, we speculated whether selective blockade of AP could be a strategy for prolonging kidney graft survival. Here we showed that Balb/c kidneys transplanted in factor b deficient C57 mice underwent reduced IRI and diminished T cell-mediated rejection. In in vitro studies, we found that fb deficiency in T cells and dendritic cells conferred intrinsic impaired alloreactive/allostimulatory functions, respectively, both in direct and indirect pathways of alloantigen presentation. By administering anti-fB antibody to C57 wt recipients in the early post Balb/c kidney transplant phases, we documented that inhibition of AP during both ischemia/reperfusion and early adaptive immune response is necessary for prolonging graft survival. These findings may have implication for the use of AP inhibitors in clinical kidney transplantation.


Subject(s)
Complement Activation/immunology , Complement Factor B/deficiency , Graft Rejection/prevention & control , Graft Survival/immunology , Kidney Transplantation/adverse effects , Reperfusion Injury/prevention & control , T-Lymphocytes/immunology , Allografts , Animals , Complement Factor B/genetics , Graft Rejection/etiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Reperfusion Injury/etiology
5.
Sci Rep ; 6: 28445, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27345360

ABSTRACT

Podocyte loss is the initial event in the development of glomerulosclerosis, the structural hallmark of progressive proteinuric nephropathies. Understanding mechanisms underlying glomerular injury is the key challenge for identifying novel therapeutic targets. In mice with protein-overload induced by bovine serum albumin (BSA), we evaluated whether the alternative pathway (AP) of complement mediated podocyte depletion and podocyte-dependent parietal epithelial cell (PEC) activation causing glomerulosclerosis. Factor H (Cfh(-/-)) or factor B-deficient mice were studied in comparison with wild-type (WT) littermates. WT+BSA mice showed podocyte depletion accompanied by glomerular complement C3 and C3a deposits, PEC migration to capillary tuft, proliferation, and glomerulosclerosis. These changes were more prominent in Cfh(-/-) +BSA mice. The pathogenic role of AP was documented by data that factor B deficiency preserved glomerular integrity. In protein-overload mice, PEC dysregulation was associated with upregulation of CXCR4 and GDNF/c-Ret axis. In vitro studies provided additional evidence of a direct action of C3a on proliferation and CXCR4-related migration of PECs. These effects were enhanced by podocyte-derived GDNF. In patients with proteinuric nephropathy, glomerular C3/C3a paralleled PEC activation, CXCR4 and GDNF upregulation. These results indicate that mechanistically uncontrolled AP complement activation is not dispensable for podocyte-dependent PEC activation resulting in glomerulosclerosis.


Subject(s)
Complement C3a/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Adult , Animals , Cattle , Cell Proliferation , Cells, Cultured , Complement Factor B/deficiency , Complement Factor B/genetics , Complement Factor H/deficiency , Complement Factor H/genetics , Disease Models, Animal , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Glomerulosclerosis, Focal Segmental/metabolism , Humans , Kidney/metabolism , Kidney/pathology , Kidney/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Podocytes/cytology , Podocytes/metabolism , Proteinuria/etiology , Serum Albumin, Bovine/administration & dosage , Up-Regulation , Young Adult
6.
PLoS One ; 9(4): e95160, 2014.
Article in English | MEDLINE | ID: mdl-24740152

ABSTRACT

There is considerable evidence that influenza A virus (IAV) promotes adherence, colonization, and superinfection by S. pneumoniae (Spn) and contributes to the pathogenesis of otitis media (OM). The complement system is a critical innate immune defense against both pathogens. To assess the role of the complement system in the host defense and the pathogenesis of acute pneumococcal OM following IAV infection, we employed a well-established transtympanically-induced mouse model of acute pneumococcal OM. We found that antecedent IAV infection enhanced the severity of acute pneumococcal OM. Mice deficient in complement C1qa (C1qa-/-) or factor B (Bf -/-) exhibited delayed viral and bacterial clearance from the middle ear and developed significant mucosal damage in the eustachian tube and middle ear. This indicates that both the classical and alternative complement pathways are critical for the oto-immune defense against acute pneumococcal OM following influenza infection. We also found that Spn increased complement activation following IAV infection. This was characterized by sustained increased levels of anaphylatoxins C3a and C5a in serum and middle ear lavage samples. In contrast, mice deficient in the complement C5a receptor (C5aR) demonstrated enhanced bacterial clearance and reduced severity of OM. Our data support the concept that C5a-C5aR interactions play a significant role in the pathogenesis of acute pneumococcal OM following IAV infection. It is possible that targeting the C5a-C5aR axis might prove useful in attenuating acute pneumococcal OM in patients with influenza infection.


Subject(s)
Eustachian Tube/immunology , Orthomyxoviridae Infections/immunology , Otitis Media/immunology , Pneumococcal Infections/immunology , Receptor, Anaphylatoxin C5a/genetics , Acute Disease , Animals , Coinfection , Complement Activation , Complement C1q/deficiency , Complement C1q/genetics , Complement C3a/genetics , Complement C3a/immunology , Complement C5a/genetics , Complement C5a/immunology , Complement Factor B/deficiency , Complement Factor B/genetics , Eustachian Tube/microbiology , Eustachian Tube/pathology , Eustachian Tube/virology , Female , Gene Deletion , Gene Expression , Immunity, Innate , Influenza A Virus, H1N1 Subtype/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Otitis Media/genetics , Otitis Media/microbiology , Otitis Media/pathology , Pneumococcal Infections/genetics , Pneumococcal Infections/microbiology , Pneumococcal Infections/pathology , Receptor, Anaphylatoxin C5a/deficiency , Severity of Illness Index , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/pathogenicity
8.
J Immunol ; 189(9): 4640-7, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23028050

ABSTRACT

There is mounting evidence indicating an important role for complement in the pathogenesis of cerebral ischemia-reperfusion injury, or ischemic stroke. The role of the alternative complement pathway in ischemic stroke has not been investigated, and there is conflicting data on the role of the terminal pathway. In this study, we show that compared with wild-type mice, mice deficient in the alternative pathway protein factor B or mice treated with the alternative pathway inhibitor CR2-fH have improved outcomes after 60-min middle cerebral artery occlusion and 24-h reperfusion. Factor B-deficient or CR2-fH-treated mice were protected in terms of improved neurologic function and reduced cerebral infarct, demyelination, P-selectin expression, neutrophil infiltration, and microthrombi formation. Mice deficient in both the classical and lectin pathways (C1q/MBL deficient) were also protected from cerebral ischemia-reperfusion injury, and there was no detectable C3d deposition in the ipsilateral brain of these mice. These data demonstrate that the alternative pathway is not alone sufficient to initiate complement activation and indicate that the alternative pathway propagates cerebral injury via amplification of the cascade. Deficiency of C6, a component of the terminal cytolytic membrane attack complex, had no effect on outcome after ischemic stroke, indicating that the membrane attack complex is not involved in mediating injury in this model. We additionally show that the protective effect of factor B deficiency and CR2-fH treatment is sustained in the subacute stage of infarct development, adding to the clinical relevance of these findings.


Subject(s)
Brain Ischemia/immunology , Brain Ischemia/pathology , Complement Pathway, Alternative/immunology , Inflammation Mediators/physiology , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Stroke/immunology , Stroke/pathology , Animals , Brain Ischemia/genetics , CD59 Antigens/genetics , Complement C1q/deficiency , Complement C1q/genetics , Complement C6/deficiency , Complement C6/genetics , Complement Factor B/deficiency , Complement Factor B/genetics , Complement Membrane Attack Complex/deficiency , Complement Membrane Attack Complex/genetics , Complement Pathway, Alternative/genetics , Complement Pathway, Mannose-Binding Lectin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reperfusion Injury/genetics , Stroke/genetics
9.
Mol Immunol ; 52(3-4): 249-57, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22750071

ABSTRACT

The innate immune system causes tissue inflammation and injury after renal ischemia/reperfusion (I/R). The complement system is activated on ischemic tubular epithelial cells (TECs) and induces the cells to produce pro-inflammatory chemokines. TECs also express toll-like receptors (TLRs)-2 and -4. Signaling through the TLRs induces TECs to produce a variety of chemokines, some of which can also be induced by complement activation fragments. We sought to determine whether the effects of complement activation and TLR signaling in TECs are redundant, or whether additive protection can be achieved by blocking both of these innate immune systems. To confirm that the complement system, TLR-2 signaling, and TLR-4 signaling induce production of a similar repertoire of inflammatory chemokines, we stimulated TECs with complement sufficient serum or with TLR-2 and TLR-4 ligands in vitro. We found that all three of these stimuli induce TECs to produce KC, MIP-2, IL-6, and TNF-α, and that there was a trend toward greater production of KC in cells exposed to two stimuli. Based upon these results, we hypothesized that mice deficient in both complement activation and TLR-2 signaling would demonstrate greater protection from I/R than mice deficient only in the complement system. To test this hypothesis we induced ischemic acute kidney injury (AKI) in wild-type mice, mice with targeted deletion of complement factor B (fB(-/-) mice), or mice with targeted deletion of factor B and TLR-2 (fB(-/-)TLR2(-/-) mice). Surprisingly, we found that fB(-/-)TLR2(-/-) mice developed more severe injury than those with single deficiency of factor B. Our results indicate that blockade of the complement system may be more protective than simultaneous blockade of both the complement system and TLR-2 in ischemic AKI.


Subject(s)
Acute Kidney Injury/immunology , Complement Activation , Complement Factor B/immunology , Cytokines/biosynthesis , Kidney Tubules/immunology , Reperfusion Injury/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Acute Kidney Injury/pathology , Animals , Cells, Cultured , Complement Factor B/deficiency , Complement Factor B/genetics , Epithelial Cells/metabolism , Kidney Tubules/metabolism , Kidney Tubules/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Reperfusion Injury/pathology , Signal Transduction , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/biosynthesis
10.
J Immunol ; 187(1): 172-80, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21642543

ABSTRACT

Shiga toxin (Stx)-producing E.coli O157:H7 has become a global threat to public health; it is a primary cause of diarrhea-associated hemolytic uremic syndrome (HUS), a disorder of thrombocytopenia, microangiopathic hemolytic anemia, and acute renal failure with thrombi occluding renal microcirculation. In this study, we explored whether Stx triggers complement-dependent microvascular thrombosis in in vitro and in vivo experimental settings of HUS. Stx induced on human microvascular endothelial cell surface the expression of P-selectin, which bound and activated C3 via the alternative pathway, leading to thrombus formation under flow. In the search for mechanisms linking complement activation and thrombosis, we found that exuberant complement activation in response to Stx generated an increased amount of C3a that caused further endothelial P-selectin expression, thrombomodulin (TM) loss, and thrombus formation. In a murine model of HUS obtained by coinjection of Stx2 and LPS and characterized by thrombocytopenia and renal dysfunction, upregulation of glomerular endothelial P-selectin was associated with C3 and fibrin(ogen) deposits, platelet clumps, and reduced TM expression. Treatment with anti-P-selectin Ab limited glomerular C3 accumulation. Factor B-deficient mice after Stx2/LPS exhibited less thrombocytopenia and were protected against glomerular abnormalities and renal function impairment, indicating the involvement of complement activation via the alternative pathway in the glomerular thrombotic process in HUS mice. The functional role of C3a was documented by data showing that glomerular fibrin(ogen), platelet clumps, and TM loss were markedly decreased in HUS mice receiving C3aR antagonist. These results identify Stx-induced complement activation, via P-selectin, as a key mechanism of C3a-dependent microvascular thrombosis in diarrhea-associated HUS.


Subject(s)
Complement C3a/toxicity , Complement Pathway, Alternative/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/pathology , Shiga Toxin 1/toxicity , Shiga Toxin 2/toxicity , Animals , Cell Line , Complement C3a/biosynthesis , Complement C3a/metabolism , Complement Factor B/deficiency , Complement Factor B/genetics , Disease Models, Animal , Endothelium, Vascular/metabolism , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/pathology , Escherichia coli O157/immunology , Escherichia coli O157/pathogenicity , Hemolytic-Uremic Syndrome/metabolism , Humans , Kidney Glomerulus/blood supply , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microcirculation/immunology , P-Selectin/physiology , Protein Binding/immunology
11.
Circulation ; 122(19): 1948-56, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-20974996

ABSTRACT

BACKGROUND: The early components of the classical and lectin complement pathways have been shown to protect low-density lipoprotein receptor-deficient mice (Ldlr(-/-)) from early atherogenesis. However, the role of the alternative pathway remained unknown, and that was investigated in this study. METHODS AND RESULTS: Mice lacking factor B (Bf(-/-)), the initiator of the alternative pathway, were crossed with Ldlr(-/-) mice and studied under different proatherogenic conditions. There was no statistically significant difference in lipid profiles or atherosclerotic lesion development between Bf(-/-)/Ldlr(-/-) and Ldlr(-/-) mice fed a low-fat diet. However, in these groups, administration of bacterial lipopolysaccharide led to a significant increase in atherosclerosis only in Ldlr(-/-) and not in Bf(-/-)/Ldlr(-/-) mice, indicating that the alternative pathway is necessary for endotoxin-mediated atherogenesis. Bf(-/-)/Ldlr(-/-) mice also had significantly decreased cross-sectional aortic root lesion fraction area and reduced lesion complexity compared with Ldlr(-/-) animals after a 12-week period of high-fat diet, although this was also accompanied by reduced levels of serum cholesterol. Under both experimental conditions, the atherosclerotic changes in the Bf(-/-)/Ldlr(-/-) mice were accompanied by a marked reduction in complement activation in the circulation and in atherosclerotic plaques, with no statistically significant differences in immunoglobulin G deposition or in the serum antibody response to oxidized low-density lipoprotein. CONCLUSIONS: These data demonstrate that amplification of complement activation by the alternative pathway in response to lipopolysaccharide or high-fat diet plays a proatherogenic role.


Subject(s)
Atherosclerosis/immunology , Atherosclerosis/pathology , Complement Pathway, Alternative/immunology , Receptors, LDL/deficiency , Animals , Antibodies/blood , Atherosclerosis/blood , Atherosclerosis/chemically induced , Complement Activation/genetics , Complement Activation/physiology , Complement C3/metabolism , Complement C3a/metabolism , Complement Factor B/deficiency , Complement Pathway, Alternative/physiology , Complement System Proteins/metabolism , Diet, Atherogenic , Disease Models, Animal , Female , Humans , Immunoglobulin G/blood , Inflammation/immunology , Inflammation/physiopathology , Lipopolysaccharides/toxicity , Lipoproteins, LDL/immunology , Mice , Mice, Knockout
12.
Infect Immun ; 78(3): 976-83, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20065024

ABSTRACT

To define the roles of specific complement activation pathways in host defense against Streptococcus pneumoniae in acute otitis media (AOM), we investigated the susceptibility to AOM in mice deficient in complement factor B and C2 (Bf/C2(-/)(-)), C1qa (C1qa(-/)(-)), and factor B (Bf(-)(/)(-)). Bacterial titers of both S. pneumoniae serotype 6A and 14 in the middle ear lavage fluid samples from Bf/C2(-/)(-), Bf(-)(/)(-), and C1qa(-/)(-) mice were significantly higher than in samples from wild-type mice 24 h after transtympanical infection (P < 0.05) and remained persistently higher in samples from Bf/C2(-/)(-) mice than in samples from wild-type mice. Bacteremia occurred in Bf/C2(-/)(-), Bf(-)(/)(-), and C1qa(-/)(-) mice infected with both strains, but not in wild-type mice. Recruitment of inflammatory cells was paralleled by enhanced production of inflammatory mediators in the middle ear lavage samples from Bf/C2(-/)(-) mice. C3b deposition on both strains was greatest for sera obtained from wild-type mice, followed by C1qa(-)(/)(-) and Bf(-)(/)(-) mice, and least for Bf/C2(-)(/)(-) mice. Opsonophagocytosis and whole-blood killing capacity of both strains were significantly decreased in the presence of sera or whole blood from complement-deficient mice compared to wild-type mice. These findings indicate that both the classical and alternative complement pathways are critical for middle ear immune defense against S. pneumoniae. The reduced capacity of complement-mediated opsonization and phagocytosis in the complement-deficient mice appears to be responsible for the impaired clearance of S. pneumoniae from the middle ear and dissemination to the bloodstream during AOM.


Subject(s)
Complement C1q/immunology , Complement C2/immunology , Complement Factor B/immunology , Disease Susceptibility , Otitis Media/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , Animals , Bacteremia/immunology , Bacteremia/microbiology , Blood Bactericidal Activity , Colony Count, Microbial , Complement C1q/deficiency , Complement C2/deficiency , Complement Factor B/deficiency , Ear, Middle/microbiology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbial Viability , Opsonin Proteins/immunology , Otitis Media/genetics , Phagocytosis/immunology , Pneumococcal Infections/complications , Pneumococcal Infections/genetics
13.
J Clin Invest ; 119(4): 986-96, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19273909

ABSTRACT

Myocardial Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibition improves cardiac function following myocardial infarction (MI), but the CaMKII-dependent pathways that participate in myocardial stress responses are incompletely understood. To address this issue, we sought to determine the transcriptional consequences of myocardial CaMKII inhibition after MI. We performed gene expression profiling in mouse hearts with cardiomyocyte-delimited transgenic expression of either a CaMKII inhibitory peptide (AC3-I) or a scrambled control peptide (AC3-C) following MI. Of the 8,600 mRNAs examined, 156 were substantially modulated by MI, and nearly half of these showed markedly altered responses to MI with CaMKII inhibition. CaMKII inhibition substantially reduced the MI-triggered upregulation of a constellation of proinflammatory genes. We studied 1 of these proinflammatory genes, complement factor B (Cfb), in detail, because complement proteins secreted by cells other than cardiomyocytes can induce sarcolemmal injury during MI. CFB protein expression in cardiomyocytes was triggered by CaMKII activation of the NF-kappaB pathway during both MI and exposure to bacterial endotoxin. CaMKII inhibition suppressed NF-kappaB activity in vitro and in vivo and reduced Cfb expression and sarcolemmal injury. The Cfb-/- mice were partially protected from the adverse consequences of MI. Our findings demonstrate what we believe is a novel target for CaMKII in myocardial injury and suggest that CaMKII is broadly important for the genetic effects of MI in cardiomyocytes.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Complement Factor B/genetics , Myocardium/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cell Membrane/metabolism , Complement Factor B/deficiency , Gene Expression , Gene Expression Profiling , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Mice, Transgenic , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , Peptides/genetics , Peptides/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
Infect Immun ; 77(3): 1061-70, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19114546

ABSTRACT

Previous studies have shown that the alternative pathway of complement activation plays an important role in protection against infection with Cryptococcus neoformans. Cryptococcus gattii does not activate the alternative pathway as well as C. neoformans in vitro. The role of complement in C. gattii infection in vivo has not been reported. In this study, we used mice deficient in complement components to investigate the role of complement in protection against a C. gattii isolate from an ongoing outbreak in northwestern North America. While factor B-deficient mice showed an enhanced rate of death, complement component C3-deficient mice died even more rapidly, indicating that the alternative pathway was not the only complement pathway contributing to protection against disease. Both C3- and factor B-deficient mice had increased fungal burdens in comparison to wild-type mice. Histopathology revealed an overwhelming fungal burden in the lungs of these complement-deficient mice, which undoubtedly prevented efficient gas exchange, causing death. Following the fate of radiolabeled organisms showed that both factor B- and C3-deficient mice were less effective than wild-type mice in clearing organisms. However, opsonization of C. gattii with complement components was not sufficient to prolong life in mice deficient in complement. Killing of C. gattii by macrophages in vitro was decreased in the presence of serum from factor B- and C3-deficient versus wild-type mice. In conclusion, we have demonstrated that complement activation is crucial for survival in C. gattii infection. Additionally, we have shown that the alternative pathway of complement activation is not the only complement pathway contributing to protection.


Subject(s)
Complement C3/immunology , Complement Factor B/immunology , Cryptococcosis/immunology , Animals , Complement Activation/immunology , Complement C3/deficiency , Complement Factor B/deficiency , Cryptococcosis/pathology , Cryptococcus/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
15.
J Immunol ; 181(4): 2732-40, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18684964

ABSTRACT

Complement activation is tightly regulated to avoid excessive inflammatory and immune responses. Crry(-/-) is an embryonic lethal phenotype secondary to the maternal complement alternative pathway (AP) attacking a placenta deficient in this inhibitor. In this study, we demonstrate that Crry(-/-) mice could be rescued on a partial as well as on a complete factor B (fB)- or C3-deficient maternal background. The C3 and fB protein concentrations in Crry(-/-)C3(+/-) and Crry(-/-)fB(+/-) mice were substantially reduced for gene dosage secondary to enhanced AP turnover. Based on these observations, a breeding strategy featuring reduced maternal AP-activating capacity rescued the lethal phenotype. It led to a novel, stable line of Crry SKO mice carrying normal alleles for C3 and fB. Crry SKO mice also had accelerated C3 and fB turnover and therefore reduced AP- activating potential. These instructive results represent an example of a membrane regulatory protein being responsible for homeostasis of the complement system. They imply that there is constant turnover on cells of the AP pathway which functions as an immune surveillance system for pathogens and altered self.


Subject(s)
Complement Pathway, Alternative/immunology , Homeostasis/immunology , Membrane Proteins/physiology , Receptors, Complement/physiology , Animals , Cell Line , Complement C3/biosynthesis , Complement C3/deficiency , Complement C3/metabolism , Complement Factor B/biosynthesis , Complement Factor B/deficiency , Complement Factor B/genetics , Complement Pathway, Alternative/genetics , Embryo Loss/genetics , Embryo Loss/immunology , Female , Genotype , Homeostasis/genetics , Humans , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Complement/deficiency , Receptors, Complement/genetics , Receptors, Complement 3b
16.
Mol Immunol ; 45(11): 3125-32, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18452991

ABSTRACT

UNLABELLED: Complement signaling has been implicated as important for normal hepatic regeneration. However, the specific mechanism by which complement is activated during liver regeneration remains undefined. To address this question, we investigated the hepatic regenerative response to partial hepatectomy in wildtype mice, C3-, C4-, and factor B-null mice, and C4-null mice treated with a factor B neutralizing antibody (mAb 1379). The results showed that following partial hepatectomy, C3-null mice exhibit reduced hepatic regeneration compared to wildtype mice as assessed by quantification of hepatic cyclin D1 expression and hepatocellular DNA synthesis and mitosis. In contrast, C4-null mice and factor B-null mice demonstrated normal liver regeneration. Moreover, animals in which all of the traditional upstream C3 activation pathways were disrupted, i.e. C4-null mice treated with mAb 1379, exhibited normal C3 activation and hepatocellular proliferation following partial hepatectomy. In order to define candidate non-traditional mechanisms of C3 activation during liver regeneration, plasmin and thrombin were investigated for their abilities to activate C3 in mouse plasma in vitro. The results showed that both proteases are capable of initiating C3 activation, and that plasmin can do so independent of the classical and alternative pathways. CONCLUSIONS: These results show that C3 is required for a normal hepatic regenerative response, but that disruption of the classical- or lectin-dependent pathways (C4-dependent), the alternative pathway (factor B-dependent), or all of these pathways does not impair the hepatic regenerative response, and indicate that non-traditional mechanisms by which C3 is activated during hepatic regeneration must exist. In vitro analysis raises the possibility that plasmin may contribute to non-traditional complement activation during liver regeneration in vivo.


Subject(s)
Complement Activation , Complement C3/immunology , Liver Regeneration/immunology , Animals , Antibodies, Monoclonal/immunology , Complement C3/deficiency , Complement C4/deficiency , Complement C4/immunology , Complement Factor B/deficiency , Complement Factor B/immunology , Immunoblotting , Liver/cytology , Liver/immunology , Mice , Mice, Inbred C57BL , Neutralization Tests , Protein Processing, Post-Translational
17.
Am J Physiol Endocrinol Metab ; 294(3): E521-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18160458

ABSTRACT

Acylation-stimulating protein (C3adesArg/ASP) is an adipokine that acts on its receptor C5L2 to stimulate triglyceride (TG) synthesis in adipose tissue. The present study investigated ASP levels in mouse models of obesity and leanness and the effect of ASP deficiency in C3 knockout (C3KO) mice on adipose tissue morphology. Plasma ASP levels in wild-type (WT) mice correlated positively with plasma nonesterified fatty acids (NEFA) (R = 0.664, P < 0.001) and total cholesterol (R = 0.515, P < 0.001). Plasma ASP was increased by 85% in obese ob/ob leptin-deficient mice and decreased in lean diacylglycerol acyltransferase 1 (DGAT1) KO mice (-54%) and C/EBPalpha(beta/beta) transgenic mice (-70%) compared with WT. Mice lacking alternative complement factor B or adipsin (FBKO or ADKO), required for ASP production, were also ASP deficient. Both FBKO and C3KO mice had delayed postprandial TG and NEFA clearance on low-fat (LF) and high-fat (HF) diets, suggesting that lack of ASP, not C3, drives the metabolic phenotype. Adipocyte size distribution in C3KO mice was polarized (increased number of both small and large cells), with decreased adipsin expression (-33% gonadal HF), DGAT1 expression (-31% to -50%) and DGAT activity (-41%). Overall, a reduction/deficiency in ASP is associated with an antiadipogenic state and ASP may provide a target for controlling fat storage.


Subject(s)
Adipose Tissue/metabolism , Complement C3a/deficiency , Complement Pathway, Alternative/physiology , Adipocytes/pathology , Adipose Tissue/chemistry , Adipose Tissue/pathology , Animals , Complement C3/deficiency , Complement C3a/analysis , Complement Factor B/deficiency , Complement Factor D/deficiency , Complement Pathway, Alternative/genetics , Diacylglycerol O-Acyltransferase/deficiency , Diacylglycerol O-Acyltransferase/genetics , Fatty Acids, Nonesterified/blood , Female , Leptin/deficiency , Lipoprotein Lipase/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Obesity/blood , RNA, Messenger/analysis , Triglycerides/analysis , Triglycerides/biosynthesis
18.
Allergol Immunopathol (Madr) ; 35(3): 90-4, 2007.
Article in English | MEDLINE | ID: mdl-17594871

ABSTRACT

BACKGROUND: Accumulating data suggest an immunopathogenic role for the complement system as a causative element in pregnancy loss (PL). Formation of pathogenic antibodies with activation of the classical pathway may have a role, but this mechanism fails to characterize the majority of cases with recurrent PL. We established the prevalence of hypocomplementemia without circulating autoantibodies in women with recurrent PL. METHODS: In a retrospective case control study, 201 women with recurrent PL (two or more PL) and 30 healthy women who had normal pregnancies but no PL were studied. Serum levels of C3, C4, and factor B were determined by nephelometry. Total hemolytic activity of the complement system (CH100) was investigated by radial immunodiffusion test. RESULTS: The prevalence of hypocomplementemia [low levels of C3, C4, FB or CH100 (with normal concentrations of C3, C4 and FB)] was significantly higher in women with recurrent PL (22.4%) in comparison with controls (6.6%; p = 0.019). C3, C4, FB hypocomplementemia or low CH100 were observed in 13 (6.5%), 19 (9.4%), 13 (6.5%) and 7 (3.5%) women with recurrent PL, respectively. Among patients with C3, C4, FB or CH100 hypocomplementemia, 10, 18, 12 and 5 patients had no circulating autoantibodies [antinuclear antibodies, anticardiolipin antibodies or antithyroid antibodies], respectively. In all, hypocomplementemia, in the absence of autoantibodies, was observed in 38 (18.9%) women with recurrent PL in a significantly higher frequency than controls (n = 2, p = 0.049). CONCLUSIONS: Hypocomplementemia, in the absence of autoantibodies was observed in a group of women with recurrent PL which might suggest a role of the complement system in the pathogenesis of PL in these patients.


Subject(s)
Abortion, Habitual/immunology , Complement System Proteins/deficiency , Abortion, Habitual/blood , Adult , Autoantibodies/analysis , Cohort Studies , Complement C3/deficiency , Complement C4/deficiency , Complement Factor B/deficiency , Complement System Proteins/analysis , Cross-Sectional Studies , Female , Humans , Immunodiffusion , Pregnancy , Prevalence , Retrospective Studies , Spain/epidemiology
19.
J Clin Invest ; 116(11): 2892-900, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17024247

ABSTRACT

Bullous pemphigoid (BP) is an autoimmune subepidermal blistering disease associated with autoantibodies directed against the hemidesmosomal proteins BP180 and BP230 and inflammation. Passive transfer of antibodies to the murine BP180 (mBP180) induces a skin disease that closely resembles human BP. In the present study, we defined the roles of the different complement activation pathways in this model system. Mice deficient in the alternative pathway component factor B (Fb) and injected with pathogenic anti-mBP180 IgG developed delayed and less intense subepidermal blisters. Mice deficient in the classical pathway component complement component 4 (C4) and WT mice pretreated with neutralizing antibody against the first component of the classical pathway, C1q, were resistant to experimental BP. These mice exhibited a significantly reduced level of mast cell degranulation and polymorphonuclear neutrophil (PMN) infiltration in the skin. Intradermal administration of compound 48/80, a mast cell degranulating agent, restored BP disease in C4(-/-) mice. Furthermore, C4(-/-) mice became susceptible to experimental BP after local injection of PMN chemoattractant IL-8 or local reconstitution with PMNs. These findings provide the first direct evidence to our knowledge that complement activation via the classical and alternative pathways is crucial in subepidermal blister formation in experimental BP.


Subject(s)
Complement C1q/metabolism , Complement C4/metabolism , Complement Factor B/metabolism , Pemphigoid, Bullous/metabolism , Pemphigoid, Bullous/pathology , Signal Transduction , Animals , Autoantigens/immunology , Blister/genetics , Blister/metabolism , Blister/pathology , Cell Nucleus/genetics , Cell Nucleus/pathology , Chemotaxis/drug effects , Complement C1q/immunology , Complement C4/deficiency , Complement C4/genetics , Complement Factor B/deficiency , Complement Factor B/genetics , Disease Models, Animal , Genetic Predisposition to Disease , Injections, Intradermal , Interleukin-8/administration & dosage , Interleukin-8/pharmacology , Mast Cells/immunology , Mast Cells/metabolism , Mast Cells/pathology , Mice , Mice, Knockout , Non-Fibrillar Collagens , Pemphigoid, Bullous/genetics , Pemphigoid, Bullous/immunology , Collagen Type XVII
20.
J Immunol ; 177(3): 1904-12, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16849503

ABSTRACT

Activation of each complement initiation pathway (classical, alternative, and lectin) can lead to the generation of bioactive fragments with resulting inflammation in target organs. The objective of the current study was to determine the role of specific complement activation pathways in the pathogenesis of experimental anti-type II collagen mAb-passive transfer arthritis. C57BL/6 mice were used that were genetically deficient in either the alternative pathway protein factor B (Bf(-/-)) or in the classical pathway component C4 (C4(-/-)). Clinical disease activity was markedly decreased in Bf(-/-) compared with wild-type (WT) mice (0.5 +/- 0.22 (n = 6) in Bf(-/-) vs 8.83 +/- 0.41 (n = 6) in WT mice (p < 0.0001)). Disease activity scores were not different between C4(-/-) and WT mice. Analyses of joints showed that C3 deposition, inflammation, pannus, cartilage, and bone damage scores were all significantly less in Bf(-/-) as compared with WT mice. There were significant decreases in mRNA levels of C3, C4, CR2, CR3, C3aR, and C5aR in the knees of Bf(-/-) as compared with C4(-/-) and WT mice with arthritis; mRNA levels for complement regulatory proteins did not differ between the three strains. These results indicate that the alternative pathway is absolutely required for the induction of arthritis following injection of anti-collagen Abs. The mechanisms by which these target organ-specific mAbs bypass the requirements for engagement of the classical pathway remain to be defined but do not appear to involve a lack of alternative pathway regulatory proteins.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cartilage, Articular/immunology , Cartilage, Articular/pathology , Collagen/immunology , Complement Pathway, Alternative/immunology , Immunization, Passive , Animals , Antibodies, Monoclonal/administration & dosage , Arthritis, Experimental/genetics , Complement C3/chemistry , Complement C4/deficiency , Complement C4/genetics , Complement Factor B/deficiency , Complement Factor B/genetics , Complement Factor H/chemistry , Complement Inactivator Proteins/biosynthesis , Complement Inactivator Proteins/genetics , Cytokines/biosynthesis , Cytokines/genetics , Disease Models, Animal , Immunization, Passive/methods , Immunohistochemistry , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/biosynthesis , Synovial Membrane/immunology , Synovial Membrane/metabolism , Synovial Membrane/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...