Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
1.
J Heart Lung Transplant ; 40(10): 1112-1121, 2021 10.
Article in English | MEDLINE | ID: mdl-34334299

ABSTRACT

BACKGROUND: Post-transplant ischemia reperfusion injury (IRI) is a recognized risk factor for subsequent organ dysfunction, alloresponsiveness, and rejection. The complement system is known to play a role in IRI and represents a therapeutic target. Complement is activated in transplanted grafts when circulating IgM antibodies bind to exposed ischemia-induced neoepitopes upon reperfusion, and we investigated the targeting of a human complement inhibitor, CR1, to a post-transplant ischemia-induced neoepitope. METHODS: A fragment of human CR1 was linked to a single chain antibody construct (C2 scFv) recognizing an injury-specific neoepitope to yield C2-CR1. This construct, along with a soluble untargeted counterpart, was characterized in a cardiac allograft transplantation model of IRI in terms of efficacy and safety. RESULTS: CR1 was similarly effective against mouse and human complement. C2-CR1 provided effective protection against cardiac IRI at a lower dose than untargeted CR1. The increased efficacy of C2-CR1 relative to CR1 correlated with decreased C3 deposition, and C2-CR1, but not CR1, targeted to cardiac allografts. At a dose necessary to reduce IRI, C2-CR1 had minimal impact on serum complement activity, in contrast to CR1 which resulted in a high level of systemic inhibition. The circulatory half-life of CR1 was markedly longer than that of C2-CR1, and whereas a minimum therapeutic dose of CR1 severely impaired host susceptibility to infection, C2-CR1 had no impact. CONCLUSION: We show the translational potential of a human complement inhibitor targeted to a universal ischemia-induced graft-specific epitope, and demonstrate advantages compared to an untargeted counterpart in terms of efficacy and safety.


Subject(s)
Complement Activation/physiology , Complement C2/immunology , Complement Inactivating Agents/therapeutic use , Heart Transplantation/adverse effects , Myocardial Reperfusion Injury/prevention & control , Receptors, Complement 3b/immunology , Animals , Disease Models, Animal , Epitopes/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Single-Chain Antibodies
2.
Acta Neuropathol Commun ; 9(1): 126, 2021 07 19.
Article in English | MEDLINE | ID: mdl-34281628

ABSTRACT

Activation of the complement system propagates neuroinflammation and brain damage early and chronically after traumatic brain injury (TBI). The complement system is complex and comprises more than 50 components, many of which remain to be characterized in the normal and injured brain. Moreover, complement therapeutic studies have focused on a limited number of histopathological outcomes, which while informative, do not assess the effect of complement inhibition on neuroprotection and inflammation in a comprehensive manner. Using high throughput gene expression technology (NanoString), we simultaneously analyzed complement gene expression profiles with other neuroinflammatory pathway genes at different time points after TBI. We additionally assessed the effects of complement inhibition on neuropathological processes. Analyses of neuroinflammatory genes were performed at days 3, 7, and 28 post injury in male C57BL/6 mice following a controlled cortical impact injury. We also characterized the expression of 59 complement genes at similar time points, and also at 1- and 2-years post injury. Overall, TBI upregulated the expression of markers of astrogliosis, immune cell activation, and cellular stress, and downregulated the expression of neuronal and synaptic markers from day 3 through 28 post injury. Moreover, TBI upregulated gene expression across most complement activation and effector pathways, with an early emphasis on classical pathway genes and with continued upregulation of C2, C3 and C4 expression 2 years post injury. Treatment using the targeted complement inhibitor, CR2-Crry, significantly ameliorated TBI-induced transcriptomic changes at all time points. Nevertheless, some immune and synaptic genes remained dysregulated with CR2-Crry treatment, suggesting adjuvant anti-inflammatory and neurotropic therapy may confer additional neuroprotection. In addition to characterizing complement gene expression in the normal and aging brain, our results demonstrate broad and chronic dysregulation of the complement system after TBI, and strengthen the view that the complement system is an attractive target for TBI therapy.


Subject(s)
Brain Injuries, Traumatic/genetics , Brain Injury, Chronic/genetics , Brain/drug effects , Complement Activation/genetics , Neuroinflammatory Diseases/genetics , Animals , Brain/immunology , Brain/metabolism , Brain/pathology , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/pathology , Brain Injury, Chronic/immunology , Brain Injury, Chronic/pathology , Complement Activation/immunology , Complement C2/genetics , Complement C2/immunology , Complement C3/antagonists & inhibitors , Complement C3/genetics , Complement C3/immunology , Complement C4/genetics , Complement C4/immunology , Complement Inactivating Agents/pharmacology , Gene Expression Profiling , Mice , Neuroinflammatory Diseases/immunology , Recombinant Fusion Proteins/pharmacology , Transcriptome
3.
Mol Vis ; 24: 201-217, 2018.
Article in English | MEDLINE | ID: mdl-29527116

ABSTRACT

Purpose: Systemic increases in reactive oxygen species, and their association with inflammation, have been proposed as an underlying mechanism linking obesity and age-related macular degeneration (AMD). Studies have found increased levels of oxidative stress biomarkers and inflammatory cytokines in obese individuals; however, the correlation between obesity and retinal inflammation has yet to be assessed. We used the leptin-deficient (ob/ob) mouse to further our understanding of the contribution of obesity to retinal oxidative stress and inflammation. Methods: Retinas from ob/ob mice were compared to age-matched wild-type controls for retinal function (electroretinography) and gene expression analysis of retinal stress (Gfap), oxidative stress (Gpx3 and Hmox1), and complement activation (C3, C2, Cfb, and Cfh). Oxidative stress was further quantified using a reactive oxygen species and reactive nitrogen species (ROS and RNS) assay. Retinal microglia and macrophage migration to the outer retina and complement activation were determined using immunohistochemistry for IBA1 and C3, respectively. Retinas and sera were used for metabolomic analysis using QTRAP mass spectrometry. Results: Retinal function was reduced in ob/ob mice, which correlated to changes in markers of retinal stress, oxidative stress, and inflammation. An increase in C3-expressing microglia and macrophages was detected in the outer retinas of the ob/ob mice, while gene expression studies showed increases in the complement activators (C2 and Cfb) and a decrease in a complement regulator (Cfh). The expression of several metabolites were altered in the ob/ob mice compared to the controls, with changes in polyunsaturated fatty acids (PUFAs) and branched-chain amino acids (BCAAs) detected. Conclusions: The results of this study indicate that oxidative stress, inflammation, complement activation, and lipid metabolites in the retinal environment are linked with obesity in ob/ob animals. Understanding the interplay between these components in the retina in obesity will help inform risk factor analysis for acquired retinal degenerations, including AMD.


Subject(s)
Complement Activation , Gene Expression Regulation/immunology , Obesity/immunology , Oxidative Stress/immunology , Retina/immunology , Retinal Degeneration/immunology , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/immunology , Complement C2/genetics , Complement C2/immunology , Complement C3/genetics , Complement C3/immunology , Complement Factor B/genetics , Complement Factor B/immunology , Complement Factor H/genetics , Complement Factor H/immunology , Electroretinography , Fatty Acids/immunology , Fatty Acids/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/immunology , Glutathione Peroxidase/genetics , Glutathione Peroxidase/immunology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/immunology , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Microfilament Proteins/immunology , Obesity/complications , Obesity/genetics , Obesity/pathology , Retina/pathology , Retinal Degeneration/complications , Retinal Degeneration/genetics , Retinal Degeneration/pathology
4.
PLoS Pathog ; 13(7): e1006531, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28742139

ABSTRACT

Whooping cough, or pertussis, is a contagious disease of the respiratory tract that is re-emerging worldwide despite high vaccination coverage. The causative agent of this disease is the Gram-negative Bordetella pertussis. Knowledge on complement evasion strategies of this pathogen is limited. However, this is of great importance for future vaccine development as it has become apparent that a novel pertussis vaccine is needed. Here, we unravel the effect of Virulence associated gene 8 (Vag8) of B. pertussis on the human complement system at the molecular level. We show that both recombinant and endogenously secreted Vag8 inhibit complement deposition on the bacterial surface at the level of C4b. We reveal that Vag8 binding to human C1-inhibitor (C1-inh) interferes with the binding of C1-inh to C1s, C1r and MASP-2, resulting in the release of active proteases that subsequently cleave C2 and C4 away from the bacterial surface. We demonstrate that the depletion of these complement components in the bacterial surrounding and subsequent decreased deposition on B. pertussis leads to less complement-mediated bacterial killing. Vag8 is the first protein described that specifically prevents C1s, C1r and MASP-2 binding to C1-inh and thereby mediates complement consumption away from the bacterial surface. Unravelling the mechanism of this unique complement evasion strategy of B. pertussis is one of the first steps towards understanding the interactions between the first line of defense complement and B. pertussis.


Subject(s)
Bacterial Proteins/immunology , Bordetella pertussis/immunology , Complement C1/immunology , Complement C2/immunology , Complement C4/immunology , Virulence Factors, Bordetella/immunology , Whooping Cough/immunology , Bacterial Proteins/genetics , Bordetella pertussis/genetics , Humans , Virulence , Virulence Factors, Bordetella/genetics , Whooping Cough/microbiology
5.
J Thromb Haemost ; 13(10): 1843-53, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26278069

ABSTRACT

BACKGROUND: In mild hemophilia A (MHA) patients, the risk of inhibitor development is generally low, but some factor VIII (FVIII) gene missense mutations are associated with a higher inhibitor incidence. OBJECTIVE: To investigate the mechanism(s) of inhibitor development in MHA. METHODS AND RESULTS: A patient, HA78, with MHA with a novel P1809L missense mutation in the A3 domain, exhibited significant residual FVIII activity ( FVIII: C ~10 IU dL(-1) ), despite the development of an inhibitor (5.6 BU mL(-1) ). Purified HA78-IgG significantly depressed FVIII: C from normal plasma but not from patient's plasma without inhibitor, indicating that this IgG inhibited allogeneic but not autologous FVIII. The HA78-IgG blocked thrombin and FXa-catalyzed FVIII cleavage but had little effect on FVIII binding to von Willebrand factor and phospholipid. The IgG recognized a C2 epitope close or overlapping the previously described anti-C2 ESH8 epitope. Similarly, a recombinant FVIII-P1809L mutant was little inactivated by HA78-IgG. This mutant demonstrated ~3-fold lower binding affinities to von Willebrand factor and phospholipid compared with wild-type, while reactions with thrombin or FXa were not impaired. Reaction of FVIII-P1809L with the alternative anti-C2 ESH4 showed only an ~20% inhibition compared with wild-type FVIII but was similar to wild-type after incubation with ESH8. A surface plasmon resonance-based assay demonstrated that anti-C2 ESH4 bound to FVIII-P1809L with ~10(2) -fold lower affinity compared with ESH8. CONCLUSION: These results indicated that the P1809L mutation in A3 induced the conformational change in the FVIII molecule that hampered antigenic determinant(s) located in the C2 domain and might result in the inhibitor development.


Subject(s)
Autoantibodies/blood , Blood Coagulation/genetics , Complement C2/immunology , Factor VIII/genetics , Hemophilia A/genetics , Mutation, Missense , Antibody Specificity , Blood Coagulation Tests , Cross Reactions , DNA Mutational Analysis , Epitope Mapping , Epitopes , Factor VIII/chemistry , Factor VIII/immunology , Factor VIII/metabolism , Genetic Predisposition to Disease , Hemophilia A/blood , Hemophilia A/diagnosis , Humans , Phenotype , Protein Structure, Tertiary , Severity of Illness Index , Structure-Activity Relationship
6.
Tissue Antigens ; 86(2): 122-33, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26176736

ABSTRACT

Complement component 2 (C2), an early member of the classical pathway, mainly participates in apoptotic cell clearance. We hypothesize that C2 polymorphism may confer genetic susceptibility to complement dysfunction in systemic lupus erythematosus (SLE). The major aim of our study was to investigate the clinical and serological associations of C2 variants in Chinese patients with SLE. The single-nucleotide polymorphism (rs2844455, G/A SNP) located in the intron region of C2 gene was genotyped by direct sequencing in 95 SLE patients and 95 matched normal control subjects. The gene expression profiles were generated by quantitative real-time polymerase chain reaction (PCR) and reverse transcription PCR. Our results showed that the AA genotype was observed more frequently in SLE patients than in normal control subjects (22.1% vs 9.5%, P < 0.05). The A allele was strongly associated with the occurrence of hair loss, photosensitivity and anti-cardiolipin antibodies; whereas, the G allele was associated with lower frequencies of these clinical presentations. Relative expression levels were significantly lower in patients with the AA genotype [median: 18.86, interquartile range (IQR) 11.36-22.43, P = 0.002] than in those with the GG genotype (35.76, IQR: 19.33-49.71). As expected, we confirmed the A allele as a risk factor for SLE development in a Chinese population, in contrast, the G allele might be a protective factor against the pathogenic autoantibody formation and cutaneous manifestations in SLE patients.


Subject(s)
Alopecia/genetics , Complement C2/genetics , Lupus Erythematosus, Systemic/genetics , Photophobia/genetics , Polymorphism, Single Nucleotide , Adult , Alleles , Alopecia/ethnology , Alopecia/immunology , Alopecia/pathology , Antibodies, Anticardiolipin/blood , Asian People , Case-Control Studies , Complement C2/immunology , Exons , Female , Gene Expression , Gene Expression Profiling , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Introns , Lupus Erythematosus, Systemic/ethnology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Male , Middle Aged , Photophobia/ethnology , Photophobia/immunology , Photophobia/pathology
7.
Immunol Lett ; 166(2): 103-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26093279

ABSTRACT

We aimed to explore the effects of bromodomain-containing protein 4 (BRD4) inhibition on tumor necrosis factor (TNF)-α-stimulated human rheumatoid arthritis fibroblast-like synoviocytes (RA-FLS) behavior and the therapeutic implications using BRD4 inhibitor JQ1 were explored in vivo. The levels of interleukin (IL)-1ß, IL-6, IL-17 and IL-18 in cultural supernatants from TNFα-stimulated RA-FLS were measured by ELISA. RA-FLS migration and invasion in vitro were investigated using wound healing and Matrigel assay. Expression of signaling pathway proteins was measured by Western blot. The in vivo effects of BRD4 inhibitor JQ1 were elucidated using collagen-induced arthritis (CIA) mice. We found BRD4 silencing reduced the secretion of IL-1ß, IL-6, IL-17 and IL-18 from TNFα-stimulated human RA-FLS. Downregulation of BRD4 inhibited FBS-induced migration and invasion of human RA-FLS. BRD4 silencing decreased the phosphorylation of c-Jun and activation of NFκB in TNFα-stimulated RA-FLS. In vivo, BRD4 inhibitor JQ1 reduced the inflammatory response, autoantibody production and joint damage of CIA model. Our data suggest for the first time that BRD4 inhibition has anti-inflammatory property in RA.


Subject(s)
Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Apoptosis/genetics , Arthritis, Experimental , Arthritis, Rheumatoid/pathology , Autoantibodies/blood , Autoantibodies/immunology , Azepines/pharmacology , Cell Cycle Proteins , Cell Movement/genetics , Cell Proliferation , Cell Survival/genetics , Complement C2/immunology , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Gene Targeting , Humans , Inflammation Mediators/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , NF-kappa B/metabolism , Nuclear Proteins/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/genetics , Transcription Factors/antagonists & inhibitors , Transfection , Triazoles/pharmacology
8.
Immunobiology ; 220(6): 817-22, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25601571

ABSTRACT

OBJECTIVES: Recent studies have suggested immunomodulatory and anti-inflammatory effects of cannabinoid receptor 2 (CB2R) activation, which is devoid of psychoactivity. We have demonstrated the expression of CB2R in synovial tissue from patients with rheumatoid arthritis (RA), and its specific activation shows inhibitory effects on fibroblast-like synoviocytes. However, it is still unclear whether selective activation of CB2R inhibits joint inflammation or protects joint damage in RA. METHODS: A murine model of collagen-induced arthritis (CIA) was used to evaluate the therapeutic efficacy of HU-308, a selective CB2R agonist. The disease severity was evaluated by semi-quantitative scoring of joint swelling, histological assessment of joint inflammation and structure, and radiographic assessment of joint destruction by using digital plain radiographs and micro-CT scans. The concentrations of various isotypes of anti-collagen II antibodies in sera and the levels of cytokines in culture supernatants were determined by ELISA. RESULTS: Compared with vehicle treatment, protective treatment with intraperitoneal injection of HU-308 (0.3-1.0 mg/kg) failed to decrease the incidence of the development of CIA, but it effectively suppressed the severity of the disease. In CIA mice, treatment with HU-308 significantly decreased joint swelling, synovial inflammation, and joint destruction, as well as serum levels of anti-collagen II antibodies. In vitro, HU-308 (1-10 µM) significantly suppressed the production of proinflammatory cytokines IL-6 and TNF-α from lipopolysaccharide-stimulated murine peritoneal macrophages with intact CB2R in dose-dependent manners. HU-308 failed to elicit any inhibitory effect of on lipopolysaccharide-stimulated macrophages from CB2R-knockout mice. CONCLUSIONS: Activation of CB2R by HU-308 has therapeutic potential for RA to suppress synovitis and alleviate joint destruction by inhibiting the production of autoantibodies and proinflammatory cytokines.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Joints/immunology , Joints/metabolism , Receptor, Cannabinoid, CB2/metabolism , Synovitis/immunology , Synovitis/metabolism , Animals , Arthritis, Experimental/diagnosis , Arthritis, Experimental/drug therapy , Autoantibodies/blood , Autoantibodies/immunology , Cannabinoids/administration & dosage , Cannabinoids/pharmacology , Collagen/immunology , Complement C2/immunology , Disease Models, Animal , Interleukin-6/biosynthesis , Joints/pathology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Neutrophil Infiltration , Receptor, Cannabinoid, CB2/agonists , Severity of Illness Index , Tumor Necrosis Factor-alpha/biosynthesis , X-Ray Microtomography
9.
Laryngoscope ; 125(4): 852-5, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25124395

ABSTRACT

Acute supraglottitis is a medical emergency as it can rapidly lead to airway compromise. With routine pediatric immunization for Hemophilus influenzae serotype b, supraglottitis is now more prevalent in adults, with a shift in the causative organisms and a change in the natural history of this disease. Here, we present a case of supraglottitis due to group B streptococcus that occurred in an adult with previously undetected immunoglobulin 4 (IgG4) and complement protein C2 deficiency.


Subject(s)
Immunocompromised Host/immunology , Streptococcal Infections/diagnosis , Streptococcus agalactiae/isolation & purification , Supraglottitis/immunology , Supraglottitis/microbiology , Adrenal Cortex Hormones/therapeutic use , Adult , Anti-Bacterial Agents/therapeutic use , Complement C2/deficiency , Complement C2/immunology , Emergency Service, Hospital , Follow-Up Studies , Humans , IgG Deficiency/immunology , Male , Rare Diseases , Risk Assessment , Severity of Illness Index , Streptococcal Infections/drug therapy , Supraglottitis/diagnosis , Supraglottitis/drug therapy , Treatment Outcome
10.
J Immunol ; 193(12): 6161-6171, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25381436

ABSTRACT

The pathogenic bacterium Staphylococcus aureus actively evades many aspects of human innate immunity by expressing a series of small inhibitory proteins. A number of these proteins inhibit the complement system, which labels bacteria for phagocytosis and generates inflammatory chemoattractants. Although the majority of staphylococcal complement inhibitors act on the alternative pathway to block the amplification loop, only a few proteins act on the initial recognition cascades that constitute the classical pathway (CP) and lectin pathway (LP). We screened a collection of recombinant, secreted staphylococcal proteins to determine whether S. aureus produces other molecules that inhibit the CP and/or LP. Using this approach, we identified the extracellular adherence protein (Eap) as a potent, specific inhibitor of both the CP and LP. We found that Eap blocked CP/LP-dependent activation of C3, but not C4, and that Eap likewise inhibited deposition of C3b on the surface of S. aureus cells. In turn, this significantly diminished the extent of S. aureus opsonophagocytosis and killing by neutrophils. This combination of functional properties suggested that Eap acts specifically at the level of the CP/LP C3 convertase (C4b2a). Indeed, we demonstrated a direct, nanomolar-affinity interaction of Eap with C4b. Eap binding to C4b inhibited binding of both full-length C2 and its C2b fragment, which indicated that Eap disrupts formation of the CP/LP C3 proconvertase (C4b2). As a whole, our results demonstrate that S. aureus inhibits two initiation routes of complement by expression of the Eap protein, and thereby define a novel mechanism of immune evasion.


Subject(s)
Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Complement C3 Convertase, Alternative Pathway/antagonists & inhibitors , Complement Pathway, Classical/immunology , Complement Pathway, Mannose-Binding Lectin/immunology , RNA-Binding Proteins/immunology , RNA-Binding Proteins/metabolism , Staphylococcus aureus/immunology , Staphylococcus aureus/metabolism , Bacterial Proteins/chemistry , Binding Sites , Complement C2/immunology , Complement C2/metabolism , Complement C3b/immunology , Complement C3b/metabolism , Complement C4b/immunology , Complement C4b/metabolism , Cytotoxicity, Immunologic , Humans , Models, Immunological , Neutrophils/immunology , Phagocytosis/immunology , Protein Binding , Protein Interaction Domains and Motifs , RNA-Binding Proteins/chemistry , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism
12.
Clin Immunol ; 144(3): 214-27, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22842196

ABSTRACT

Hereditary C2 deficiency (C2D) is an important susceptibility factor for invasive infections caused by encapsulated bacteria such as pneumococci and Haemophilus influenzae type b. The infections are mostly seen in childhood indicating that antibody-mediated acquired immunity is affected. C2D persons and healthy controls were vaccinated with ActHIB® and Pneumo23®. Analysis of specific antibodies to pneumococci serotype 6B, 7F, and 23F, and Hib was performed. Post-vaccination IgG antibodies against pneumococci serotype 6B and 23F at a concentration ≥1.0mg/L was found in similar frequency in C2D persons and controls. Post-vaccination sera from C2D persons showed poor complement-mediated opsonization and phagocytosis of pneumococci by granulocytes when depending on classical and lectin pathway activation only, but increased (p=0.007) and equaled that of the normal controls when also alternative pathway activation was allowed due to antibody-dependent C2 bypass activation. In conclusion, the C2D persons benefited from the vaccination and achieve an increased phagocytic capacity.


Subject(s)
Bacterial Vaccines/immunology , Complement Activation/immunology , Complement C2/deficiency , Complement C2/immunology , Opsonin Proteins/immunology , Adolescent , Adult , Animals , Antibodies, Bacterial/immunology , Antibody Formation , Child , Child, Preschool , Complement Pathway, Alternative/immunology , Complement Pathway, Classical/immunology , Erythrocytes/immunology , Female , Granulocytes/immunology , Haemophilus Infections/immunology , Haemophilus influenzae type b/immunology , Humans , Immunoglobulin G/immunology , Male , Middle Aged , Phagocytosis/immunology , Pneumococcal Infections/immunology , Sheep , Streptococcus pneumoniae/immunology , Vaccination/methods , Young Adult
13.
PLoS One ; 7(5): e36783, 2012.
Article in English | MEDLINE | ID: mdl-22611464

ABSTRACT

Complement C3 plays an essential role in the opsonization of pathogens in the mammalian complement system, whereas the molecular mechanism underlying C3 activation in invertebrates remains unknown. To understand the molecular mechanism of C3b deposition on microbes, we characterized two types of C2/factor B homologs (designated TtC2/Bf-1 and TtC2/Bf-2) identified from the horseshoe crab Tachypleus tridentatus. Although the domain architectures of TtC2/Bf-1 and TtC2/Bf-2 were identical to those of mammalian homologs, they contained five-repeated and seven-repeated complement control protein domains at their N-terminal regions, respectively. TtC2/Bf-1 and TtC2/Bf-2 were synthesized and glycosylated in hemocytes and secreted to hemolymph plasma, which existed in a complex with C3 (TtC3), and their activation by microbes was absolutely Mg(2+)-dependent. Flow cytometric analysis revealed that TtC3b deposition was Mg(2+)-dependent on Gram-positive bacteria or fungi, but not on Gram-negative bacteria. Moreover, this analysis demonstrated that Ca(2+)-dependent lectins (C-reactive protein-1 and tachylectin-5A) were required for TtC3b deposition on Gram-positive bacteria, and that a Ca(2+)-independent lectin (Tachypleus plasma lectin-1) was definitely indispensable for TtC3b deposition on fungi. In contrast, a horseshoe crab lipopolysaccharide-sensitive protease factor C was necessary and sufficient to deposit TtC3b on Gram-negative bacteria. We conclude that plasma lectins and factor C play key roles in microbe-specific TtC3b deposition in a C2/factor B-dependent or -independent manner.


Subject(s)
Complement C2/immunology , Complement C3b/immunology , Complement Factor B/immunology , Horseshoe Crabs/immunology , Animals , Cloning, Molecular , Complement Activation/immunology , Complement C2/genetics , Complement C2/metabolism , Complement C3b/metabolism , Complement Factor B/genetics , Complement Factor B/metabolism , DNA, Complementary , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Fungi/immunology , Gram-Positive Bacteria/immunology , Horseshoe Crabs/microbiology , Magnesium/metabolism , Models, Biological , Protein Binding/immunology , Protein Structure, Tertiary
14.
Chemotherapy ; 58(2): 110-7, 2012.
Article in English | MEDLINE | ID: mdl-22507917

ABSTRACT

BACKGROUND: Carcinoembryonic antigen (CEA) is frequently overexpressed in various types of human cancers and is associated with cell adhesion. There are three possible mechanisms of cancer therapy that employ anti-CEA antibody (Ab): Ab-dependent cell-mediated cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC) or the prevention of CEA interaction with the extracellular matrix and/or intercellular adhesion molecules resulting in anoikis. In this study, the effect of C2-74, a human anti-CEA monoclonal Ab was evaluated. METHODS: ADCC, CDC and anoikis assays in combination with C2-74 and an anticancer drug (5-fluorouracil or cisplatin) were investigated using tumor cell lines (MKN-45, MKN-74 and KATO III). In the anoikis assay, other human anti-CEA Abs and mouse anti-CEA-related cell adhesion molecule 6 Abs were also investigated using HLC-1 cells. RESULTS: Additive cytotoxicity was observed when the anticancer drug and C2-74 on tumor cells were combined in the CDC assays, whereas in the anoikis assay, no such additive effect was observed. Anti-CEA-related cell adhesion molecule 6 Abs, but not anti-CEA Abs, accelerated anoikis in HLC-1 cells. CONCLUSION: A mechanism for the additive antitumor effect when an anticancer drug and C2-74 are combined is indicated mainly by CDC activity but is irrelevant to anoikis in tumor cells.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Carcinoembryonic Antigen/immunology , Anoikis/drug effects , Antibodies, Monoclonal/immunology , Cell Adhesion Molecules/immunology , Cell Line, Tumor , Cisplatin/pharmacology , Complement C2/immunology , Fluorouracil/pharmacology , Humans
15.
J Immunol ; 188(1): 386-93, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22131330

ABSTRACT

The complement system rapidly detects and kills Gram-negative bacteria and supports bacterial killing by phagocytes. However, bacterial pathogens exploit several strategies to evade detection by the complement system. The alkaline protease (AprA) of Pseudomonas aeruginosa has been associated with bacterial virulence and is known to interfere with complement-mediated lysis of erythrocytes, but its exact role in bacterial complement escape is unknown. In this study, we analyzed how AprA interferes with complement activation and whether it could block complement-dependent neutrophil functions. We found that AprA potently blocked phagocytosis and killing of Pseudomonas by human neutrophils. Furthermore, AprA inhibited opsonization of bacteria with C3b and the formation of the chemotactic agent C5a. AprA specifically blocked C3b deposition via the classical and lectin pathways, whereas the alternative pathway was not affected. Serum degradation assays revealed that AprA degrades both human C1s and C2. However, repletion assays demonstrated that the mechanism of action for complement inhibition is cleavage of C2. In summary, we showed that P. aeruginosa AprA interferes with classical and lectin pathway-mediated complement activation via cleavage of C2.


Subject(s)
Bacterial Proteins/immunology , Exopeptidases/immunology , Neutrophils/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/pathogenicity , Virulence Factors/immunology , Bacterial Proteins/metabolism , Complement C2/immunology , Complement C2/metabolism , Complement C3b/immunology , Complement C3b/metabolism , Complement C5a/immunology , Complement C5a/metabolism , Complement Pathway, Mannose-Binding Lectin , Exopeptidases/metabolism , Humans , Immune Evasion , Neutrophils/metabolism , Phagocytosis/immunology , Pseudomonas Infections/enzymology , Pseudomonas aeruginosa/enzymology , Virulence Factors/metabolism
16.
Microbiol Immunol ; 55(11): 817-21, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21831201

ABSTRACT

MBL is a serum lectin that activates the lectin pathway of the complement system. MBL forms complexes with three types of MASPs. Upon binding to Salmonella serogroup C-specific oligosaccharide, MBL activates the alternative pathway via a C2-bypass pathway without involving MASP-2, C2 or C4. We demonstrate that mannan-bound MBL activates the alternative pathway via a C2-bypass pathway that requires MASP-2 and C4. Thus, depending on the ligands to which MBL binds, there may be two distinct MBL-mediated C2-bypass pathways.


Subject(s)
Complement Activation , Complement C2/immunology , Complement C2/metabolism , Complement Pathway, Alternative/immunology , Mannose-Binding Lectin/immunology , Mannose-Binding Lectin/metabolism , Complement C4/immunology , Complement C4/metabolism , Humans , Mannose-Binding Protein-Associated Serine Proteases/immunology , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Salmonella/immunology
17.
Infect Immun ; 79(7): 2578-85, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21502587

ABSTRACT

We recently reported that the complement system plays a pivotal role in innate immune defense against Streptococcus pneumoniae during acute otitis media (OM) in mice. The current study was designed to determine which of the complement pathways are activated during acute pneumococcal OM and whether components of complement are expressed in the middle ear epithelium. Gene expression was determined by quantitative PCR, enzyme-linked immunosorbent assay (ELISA), and immunofluorescence staining. We found that S. pneumoniae induced increased gene expression of factor B of the alternative complement pathway and C3 in mouse middle ear epithelium. Activation of factor B and C3 in the middle ear lavage fluids was significantly greater than in simultaneously obtained serum samples as determined by Western blotting. Using mice deficient in complement C1qa, factor B, and factor B/C2, we found that complement C3 activation and opsonophagocytosis of S. pneumoniae were greatly attenuated in factor B- and factor B/C2-deficient mice. These findings support the concept that local complement activation is an important host innate immune response and that activation of the alternative complement pathway represents one of the innate immune defense mechanisms against pneumococcal infection during the early stage of acute OM.


Subject(s)
Complement Activation , Complement Factor B/immunology , Complement Pathway, Alternative , Otitis Media/immunology , Phagocytosis , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , Animals , Blotting, Western , Complement C2/biosynthesis , Complement C2/deficiency , Complement C2/genetics , Complement C2/immunology , Complement C3/biosynthesis , Complement C3/deficiency , Complement C3/genetics , Complement C3/immunology , Complement Factor B/biosynthesis , Complement Factor B/genetics , Ear, Middle/immunology , Enzyme-Linked Immunosorbent Assay , Epithelium/immunology , Fluorescent Antibody Technique , Gene Expression , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction
18.
Fish Shellfish Immunol ; 30(4-5): 1131-7, 2011.
Article in English | MEDLINE | ID: mdl-21362481

ABSTRACT

Accumulating evidence suggests that environmental endocrine disrupting chemicals (EDCs) may exert adverse effects on aquatic organisms via the modulation of immune competence in addition to the endocrine system. However, to date, most studies have been undertaken only on biochemical and histopathological endpoints, and few studies have addressed the role of immune response gene transcript abundance in response to estrogen. In the present study, the ontogenetic expression of immune-related genes, including three complement components (C3-1, C3-2 and Bf/C2), two cytokines (IL-21 and type I IFN [IFN]), lysozyme (LZM), novel immune-type receptor (NITR-18), Ikaros (IK) and ceruloplasmin (CP) were characterized during different developmental periods (from 0 to 28 d post-hatch [dph]) in Japanese medaka. Furthermore, the responses of these genes to natural estrogen (i.e., 17ß-estradiol [E2]) were evaluated. E2 exposure at sublethal concentrations (0.1-10 µg/L) down-regulated the gene expression of C3-1, C3-2, Bf/C2, LZM and CP, while up-regulating the expression of IL-21, IFN, NITR-18 and IK. The results demonstrate a very different trend in gene expression in fish larvae exposed to E2 when compared with the ontogenetic changes in control, suggesting that exposure to environmental chemicals with estrogenic activities may interfere with immune-related genes and thus potentially influence the susceptibility of fish to opportunistic infections. These findings confirm the ability of exogenous estrogens to elicit changes in immune-related gene expression, and broaden our understanding about the mechanisms underlying the actions of EDCs. In addition, the expression profiles of immune-related genes can be developed for use as biomarkers for future immunotoxicological studies.


Subject(s)
Endocrine Disruptors/immunology , Estradiol/immunology , Gene Expression Regulation, Developmental/immunology , Oryzias/immunology , Animals , Ceruloplasmin/genetics , Ceruloplasmin/immunology , Complement C2/genetics , Complement C2/immunology , Complement C3/genetics , Complement C3/immunology , Gene Expression Regulation, Developmental/drug effects , Humans , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Interferon Type I/genetics , Interferon Type I/immunology , Interleukins/genetics , Interleukins/immunology , Male , Muramidase/genetics , Muramidase/immunology , Oryzias/genetics , RNA/chemistry , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Statistics, Nonparametric
19.
Hum Immunol ; 72(2): 173-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21093518

ABSTRACT

The complement system is an integral part of the host immune system and plays an immunoregulatory role at the interface of innate and acquired immune responses. Limited data are available on the influence of variations in complement genes in infectious diseases such as pulmonary tuberculosis (PTB). The aim of this study was to investigate the role of genetic variations in complement system components C2, C4, BF, and C3 in PTB (n = 125) compared with healthy controls (n = 125) in the Indian population. The study showed, for the first time, an increased occurrence of null alleles at the C4A, i.e., C4AQ0; an increased frequency of BF*FA and C3*F in patients with PTB compared with healthy individuals, and contributed a risk with odds ratios of 18.16 (95% confidence interval [CI] = 3.0-108.6, p = 0.0004), 2.9 (95% CI = 1.9-4.37, p(c) = 3.15E-06), and 2.26 (95% CI = 1.5-3.3, p(c) = 6.7E-05), respectively. A combinatorial analysis of complement gene variants as risk determinants and their phenotypic effects in various populations may provide unique insights into the genetic basis of susceptibility to PTB.


Subject(s)
Complement C2/genetics , Complement C3/genetics , Complement C4/genetics , Complement Factor B/genetics , Major Histocompatibility Complex/genetics , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/immunology , Adult , Alleles , Complement C2/immunology , Complement C3/immunology , Complement C4/immunology , Complement Factor B/immunology , DNA Fingerprinting , Female , Gene Frequency/immunology , Genetic Predisposition to Disease/ethnology , Genetic Variation/immunology , Humans , India , Linkage Disequilibrium/genetics , Linkage Disequilibrium/immunology , Major Histocompatibility Complex/immunology , Male , Phenotype , Polymerase Chain Reaction , Risk Factors , Tuberculosis, Pulmonary/epidemiology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...