Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
PLoS Negl Trop Dis ; 15(7): e0009508, 2021 07.
Article in English | MEDLINE | ID: mdl-34237073

ABSTRACT

The complement system is activated in tuberculous pleural effusion (TPE), with increased levels of the anaphylatoxins stimulating pleural mesothelial cells (PMCs) to secrete chemokines, which recruit nonclassical monocytes to the pleural cavity. The differentiation and recruitment of naive CD4+ T cells are induced by pleural cytokines and PMC-produced chemokines in TPE. However, it is unclear whether anaphylatoxins orchestrate CD4+ T cell response via interactions between PMCs and monocytes in TPE. In this study, CD16+ and CD16- monocytes isolated from TPE patients were cocultured with PMCs pretreated with anaphylatoxins. After removing the PMCs, the conditioned monocytes were cocultured with CD4+ T cells. The levels of the cytokines were measured in PMCs and monocyte subsets treated separately with anaphylatoxins. The costimulatory molecules were assessed in conditioned monocyte subsets. Furthermore, CD4+ T cell response was evaluated in different coculture systems. The results indicated that anaphylatoxins induced PMCs and CD16+ monocytes to secrete abundant cytokines capable of only inducing Th17 expansion, but Th1 was feeble. In addition, costimulatory molecules were more highly expressed in CD16+ than in CD16- monocytes isolated from TPE. The interactions between monocytes and PMCs enhanced the ability of PMCs and monocytes to produce cytokines and that of monocytes to express HLA-DR, CD40, CD80 and CD86, which synergistically induced Th17 expansion. In the above process, anaphylatoxins enhanced the interactions between monocytes and PMCs by increasing the level of the cytokines IL-1ß, IL-6, IL-23 and upregulating the phenotype of CD40 and CD80 in CD16+ monocytes. Collectively, these data indicate that anaphylatoxins play a central role in orchestrating Th17 response mainly via interactions between CD16+ monocytes and PMCs in TPE.


Subject(s)
Anaphylatoxins/immunology , Epithelium/immunology , Monocytes/immunology , Pleural Effusion/immunology , Th17 Cells/immunology , Tuberculosis/immunology , Adult , CD4-Positive T-Lymphocytes , Cytokines/immunology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelium/microbiology , Female , Humans , Male , Middle Aged , Mycobacterium tuberculosis/physiology , Pleural Effusion/microbiology , Receptors, IgG/immunology , Tuberculosis/microbiology
2.
Front Immunol ; 12: 580594, 2021.
Article in English | MEDLINE | ID: mdl-33767691

ABSTRACT

The zoonotic intracellular bacterium Chlamydia psittaci causes life-threatening pneumonia in humans. During mouse lung infection, complement factor C3 and the anaphylatoxin C3a augment protection against C. psittaci by a so far unknown mechanism. To clarify how complement contributes to the early, innate and the late, specific immune response and resulting protection, this study addresses the amount of C3, the timing when its presence is required as well as the anaphylatoxin receptor(s) mediating its effects and the complement-dependent migration of dendritic cells. Challenge experiments with C. psittaci on various complement KO mice were combined with transient decomplementation by pharmacological treatment, as well as the analysis of in vivo dendritic cells migration. Our findings reveal that a plasma concentration of C3 close to wildtype levels was required to achieve full protection. The diminished levels of C3 of heterozygote C3+/- mice permitted already relative effective protection and improved survival as compared to C3-/- mice, but overall recovery of these animals was delayed. Complement was in particular required during the first days of infection. However, additionally, it seems to support protection at later stages. Migration of CD103+ dendritic cells from the infected lung to the draining lymph node-as prerequisite of antigen presentation-depended on C3 and C3aR and/or C5aR. Our results provide unique mechanistic insight in various aspects of complement-dependent immune responses under almost identical, rather physiological experimental conditions. Our study contributes to an improved understanding of the role of complement, and C3a in particular, in infections by intracellular bacteria.


Subject(s)
Cell Movement/immunology , Chlamydiaceae Infections/immunology , Chlamydophila psittaci/immunology , Complement C3a/immunology , Dendritic Cells/immunology , Lung/immunology , Anaphylatoxins/immunology , Anaphylatoxins/metabolism , Animals , Cell Line , Chlamydiaceae Infections/metabolism , Chlamydiaceae Infections/microbiology , Chlamydophila psittaci/physiology , Complement Activation/immunology , Complement C3a/genetics , Complement C3a/metabolism , Dendritic Cells/cytology , Dendritic Cells/microbiology , Lung/metabolism , Lung/microbiology , Mice, Inbred C57BL , Mice, Knockout , Receptors, Complement/genetics , Receptors, Complement/immunology , Receptors, Complement/metabolism , Signal Transduction/immunology , Survival Analysis
3.
FASEB J ; 34(6): 7540-7560, 2020 06.
Article in English | MEDLINE | ID: mdl-32301538

ABSTRACT

The vascular endothelium has been discovered in the past several years to be important in shaping the cellular immune response. During the immune response the vascular endothelium is constantly perturbed by biologically potent molecules, including the complement activation peptides, C3a and C5a. Despite the importance of C3a and C5a in inflammation and immunity, their role in modulating lymphocyte function via activation of vascular endothelial cells is unknown. Accordingly, we investigated the regulated expression of the C3a and C5a receptors (complement anaphylatoxin C3a receptor [C3aR] and complement anaphylatoxin C5a receptor 1 [C5aR1]) on human umbilical vascular endothelial cells (HUVECs) and examined how C3a or C5a activation of HUVECs affects the activation and polarization of lymphatic cells. Our findings demonstrated that C3a and C5a increase C3aR and C5aR1 expression by HUVECs as well as directing their cellular transmigration and spreading through transwell filters. Moreover, C3a- or C5a-stimulated endothelial cells: (1) caused activation of B-lymphoblasts with significant increase in Fas Ligand (CD95L) (FasL), CD69, and IL-R1 expression, and (2) skewed T-lymphoblast cells toward a Th1 subtype, (CD4+ /CCR5+ ) that correlated with significant increase of IFN-γ. Collectively, these data indicate that C3a and C5a signaling is important in the activation and polarization of lymphocytes as they traffic through the vascular endothelium during the immune response.


Subject(s)
Anaphylatoxins/immunology , B-Lymphocytes/immunology , Complement C3a/immunology , Complement C5a/immunology , Peptides/immunology , T-Lymphocytes/immunology , Cells, Cultured , Complement Activation/immunology , Endothelium, Vascular/immunology , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/immunology , Receptor, Anaphylatoxin C5a/immunology , Receptors, Complement/immunology , Signal Transduction/immunology
4.
Virulence ; 10(1): 677-694, 2019 12.
Article in English | MEDLINE | ID: mdl-31274379

ABSTRACT

The complement system is pivotal in the defense against invasive disease caused by Neisseria meningitidis (Nme, meningococcus), particularly via the membrane attack complex. Complement activation liberates the anaphylatoxins C3a and C5a, which activate three distinct G-protein coupled receptors, C3aR, C5aR1 and C5aR2 (anaphylatoxin receptors, ATRs). We recently discovered that C5aR1 exacerbates the course of the disease, revealing a downside of complement in Nme sepsis. Here, we compared the roles of all three ATRs during mouse nasal colonization, intraperitoneal infection and human whole blood infection with Nme. Deficiency of complement or ATRs did not alter nasal colonization, but significantly affected invasive disease: Compared to WT mice, the disease was aggravated in C3ar-/- mice, whereas C5ar1-/- and C5ar2-/- mice showed increased resistance to meningococcal sepsis. Surprisingly, deletion of either of the ATRs resulted in lower cytokine/chemokine responses, irrespective of the different susceptibilities of the mice. This was similar in ex vivo human whole blood infection using ATR inhibitors. Neutrophil responses to Nme were reduced in C5ar1-/- mouse blood. Upon stimulation with C5a plus Nme, mouse macrophages displayed reduced phosphorylation of ERK1/2, when C5aR1 or C5aR2 were ablated or inhibited, suggesting that both C5a-receptors prime an initial macrophage response to Nme. Finally, in vivo blockade of C5aR1 alone (PMX205) or along with C5aR2 (A8Δ71-73) resulted in ameliorated disease, whereas neither antagonizing C3aR (SB290157) nor its activation with a "super-agonist" peptide (WWGKKYRASKLGLAR) demonstrated a benefit. Thus, C5aR1 and C5aR2 augment disease pathology and are interesting targets for treatment, whereas C3aR is protective in experimental meningococcal sepsis.


Subject(s)
Meningococcal Infections/immunology , Neisseria meningitidis/immunology , Receptor, Anaphylatoxin C5a/immunology , Receptors, Complement/immunology , Anaphylatoxins/immunology , Animals , Chemokines/immunology , Cytokines/immunology , Humans , Macrophages/immunology , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neisseria meningitidis/pathogenicity , Neutrophils/immunology , Neutrophils/microbiology , Receptor, Anaphylatoxin C5a/genetics , Receptors, Complement/genetics , Sepsis
5.
Front Immunol ; 9: 2133, 2018.
Article in English | MEDLINE | ID: mdl-30294325

ABSTRACT

Background and aims: Chronic ethanol exposure results in inflammation in adipose tissue; this response is associated with activation of complement as well as the development of alcohol-related liver disease (ALD). Adipose communicates with other organs, including liver, via the release of soluble mediators, such as adipokines and cytokines, characterized as the "adipose secretome." Here we investigated the role of the anaphaylatoxin receptors C3aR and C5aR1 in the development of adipose tissue inflammation and regulation of the adipose secretome in murine ALD (mALD). Methods: Wild-type C57BL/6 (WT), C3aR-/-, and C5aR1-/- mice were fed Lieber-DeCarli ethanol diet for 25 days (6% v/v, 32% kcal) or isocaloric control diets; indicators of inflammation and injury were assessed in gonadal adipose tissue. The adipose secretome was characterized in isolated adipocytes and stromal vascular cells. Results: Ethanol feeding increased the expression of adipokines, chemokines and leukocyte markers in gonadal adipose tissue from WT mice; C3aR-/- were partially protected while C5aR1-/- mice were completely protected. In contrast, induction of CYP2E1 and accumulation of TUNEL-positive cells in adipose in response to ethanol feeding was independent of genotype. Bone marrow chimeras, generated with WT and C5aR1-/- mice, revealed C5aR1 expression on non-myeloid cells, likely to be adipocytes, contributed to ethanol-induced adipose inflammation. Chronic ethanol feeding regulated both the quantity and distribution of adipokines secreted from adipocytes in a C5aR1-dependent mechanism. In WT mice, chronic ethanol feeding induced a predominant release of pro-inflammatory adipokines from adipocytes, while the adipose secretome from C5aR1-/- mice was characterized by an anti-inflammatory/protective profile. Further, the cargo of adipocyte-derived extracellular vesicles (EVs) was distinct from the soluble secretome; in WT EVs, ethanol increased the abundance of pro-inflammatory mediators while EV cargo from C5aR1-/- adipocytes contained a greater diversity and more robust expression of adipokines. Conclusions: C3aR and C5aR1 are potent regulators of ethanol-induced adipose inflammation in mALD. C5aR1 modulated the impact of chronic ethanol on the content of the adipose secretome, as well as influencing the cargo of an extensive array of adipokines from adipocyte-derived EVs. Taken together, our data demonstrate that C5aR1 contributes to ethanol-mediated changes in the adipose secretome, likely contributing to intra-organ injury in ALD.


Subject(s)
Adipose Tissue/metabolism , Ethanol/adverse effects , Liver Diseases, Alcoholic/immunology , Receptor, Anaphylatoxin C5a/metabolism , Receptors, G-Protein-Coupled/metabolism , Adipocytes , Adipokines/immunology , Adipokines/metabolism , Adipose Tissue/cytology , Adipose Tissue/drug effects , Adipose Tissue/immunology , Anaphylatoxins/immunology , Anaphylatoxins/metabolism , Animals , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Ethanol/administration & dosage , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Female , Liver/immunology , Liver/metabolism , Liver Diseases, Alcoholic/etiology , Liver Diseases, Alcoholic/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Primary Cell Culture , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/immunology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology
6.
J Cell Mol Med ; 22(12): 6002-6014, 2018 12.
Article in English | MEDLINE | ID: mdl-30247799

ABSTRACT

The anaphylatoxin C5a is generated upon activation of the complement system, a crucial arm of innate immunity. C5a mediates proinflammatory actions via the C5a receptor C5aR1 and thereby promotes host defence, but also modulates tissue homeostasis. There is evidence that the C5a/C5aR1 axis is critically involved both in physiological bone turnover and in inflammatory conditions affecting bone, including osteoarthritis, periodontitis, and bone fractures. C5a induces the migration and secretion of proinflammatory cytokines of osteoblasts. However, the underlying mechanisms remain elusive. Therefore, in this study we aimed to determine C5a-mediated downstream signalling in osteoblasts. Using a whole-genome microarray approach, we demonstrate that C5a activates mitogen-activated protein kinases (MAPKs) and regulates the expression of genes involved in pathways related to insulin, transforming growth factor-ß and the activator protein-1 transcription factor. Interestingly, using coimmunoprecipitation, we found an interaction between C5aR1 and Toll-like receptor 2 (TLR2) in osteoblasts. The C5aR1- and TLR2-signalling pathways converge on the activation of p38 MAPK and the generation of C-X-C motif chemokine 10, which functions, among others, as an osteoclastogenic factor. In conclusion, C5a-stimulated osteoblasts might modulate osteoclast activity and contribute to immunomodulation in inflammatory bone disorders.


Subject(s)
Chemokine CXCL10/genetics , Complement C5a/genetics , Inflammation/genetics , Receptor, Anaphylatoxin C5a/genetics , Toll-Like Receptor 2/genetics , Anaphylatoxins/genetics , Anaphylatoxins/immunology , Anaphylatoxins/metabolism , Animals , Bone Diseases/genetics , Bone Diseases/immunology , Bone Diseases/pathology , Bone Remodeling/genetics , Complement C5a/immunology , Gene Expression Regulation, Developmental , Humans , Immunity, Innate/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Osteoblasts/immunology , Osteoblasts/metabolism , Osteoclasts/immunology , Osteoclasts/metabolism , Osteogenesis/genetics , Osteogenesis/immunology , Signal Transduction , Transforming Growth Factor beta/genetics , p38 Mitogen-Activated Protein Kinases/genetics
8.
Clin Exp Med ; 18(3): 297-318, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29450785

ABSTRACT

The complement system (CS) has recently been recognized as a bridge between innate and adaptive immunity that constitutes a very complex mechanism controlling the clearance of pathogens, cellular debris, and immune complexes. Out of three known pathways of complement activation, the alternative pathway (AP) plays a critical role in host defense by amplifying the complement response, independently of initiation pathway and continuously maintaining low-level activity in a process called 'thick-over.' A key molecule of the CS is C3, in which the AP is constantly activated. To prevent host cell destruction, a group of the AP regulators tightly controls this pathway of the CS activation. Acquired and genetic abnormalities of the CS may alter the delicate balance between enhancing and inhibiting the AP cascade. These can lead to the uncontrolled CS activation, inflammatory response, and subsequent tissue damage. Since complement components are locally produced and activated in the kidney, the abnormalities targeting the AP may cause glomerular injury. C3 glomerulopathy is a new entity, in which the AP dysregulation has been well established. However, recent studies indicate that the AP may also contribute to a wide range of kidney pathologies, including immune-complex-mediated glomerulonephritis (GN), pauci-immune GN, and primary membranous nephropathy (PMN). This article provides insight into current knowledge on the role of the AP in the pathogenesis of glomerular diseases, focusing mainly on various types of primary and secondary GN and PMN.


Subject(s)
Complement Pathway, Alternative/genetics , Gene Expression Regulation/immunology , Glomerulonephritis, Membranous/immunology , Glomerulonephritis/immunology , Kidney Glomerulus/immunology , Lupus Nephritis/immunology , Adaptive Immunity , Anaphylatoxins/immunology , Anaphylatoxins/metabolism , Autoantibodies/biosynthesis , Complement Activation , Complement C3/genetics , Glomerulonephritis/classification , Glomerulonephritis/genetics , Glomerulonephritis/pathology , Glomerulonephritis, Membranous/genetics , Glomerulonephritis, Membranous/pathology , Humans , Immunity, Innate , Kidney Glomerulus/pathology , Lupus Nephritis/genetics , Lupus Nephritis/pathology , Mutation , Protein Domains , Proteolysis
9.
Drug Discov Today ; 23(3): 487-492, 2018 03.
Article in English | MEDLINE | ID: mdl-29326077

ABSTRACT

A recent study on nanoparticle-induced hypersensitivity reactions in pigs showed robust pulmonary intravascular macrophage clearance of Polybead® carboxylate microspheres in mediating the adverse cardiopulmonary distress, irrespective of the ability of these particles to activate the complement (C) system in vitro. Focusing on this observation, this article highlights the controversies in projecting in vitro C assay data to in vivo conditions and applying data on polystyrene particles to therapeutic nanopharmaceuticals. Based on overwhelming evidence of a role of anaphylatoxins in hypersensitivity reactions, the need to further explore the role of C activation in the reported and other reactions is highlighted. C-activation-related and C-independent pseudoallergies (CARPA and CIPA) can proceed simultaneously, as outlined by the 'double-hit' hypothesis.


Subject(s)
Complement Activation/drug effects , Drug Hypersensitivity/etiology , Drug Hypersensitivity/immunology , Nanoparticles/adverse effects , Nanoparticles/chemistry , Anaphylatoxins/chemistry , Anaphylatoxins/immunology , Animals , Humans , Microspheres , Polystyrenes/adverse effects , Polystyrenes/chemistry , Polystyrenes/immunology , Swine
10.
Toxicol Lett ; 265: 156-169, 2017 Jan 04.
Article in English | MEDLINE | ID: mdl-27932254

ABSTRACT

Cysteine-rich secretory proteins (CRISPs) are commonly described as part of the protein content of snake venoms, nevertheless, so far, little is known about their biological targets and functions. Our study describes the isolation and characterization of Bj-CRP, the first CRISP isolated from Bothrops jararaca snake venom, also aiming at the identification of possible targets for its actions. Bj-CRP was purified using three chromatographic steps (Sephacryl S-200, Source 15Q and C18) and showed to be an acidic protein of 24.6kDa with high sequence identity to other snake venom CRISPs. This CRISP was devoid of proteolytic, hemorrhagic or coagulant activities, and it did not affect the currents from 13 voltage-gated potassium channel isoforms. Conversely, Bj-CRP induced inflammatory responses characterized by increase of leukocytes, mainly neutrophils, after 1 and 4h of its injection in the peritoneal cavity of mice, also stimulating the production of IL-6. Bj-CRP also acted on the human complement system, modulating some of the activation pathways and acting directly on important components (C3 and C4), thus inducing the generation of anaphylatoxins (C3a, C4a and C5a). Therefore, our results for Bj-CRP open up prospects for better understanding this class of toxins and its biological actions.


Subject(s)
Bothrops , Crotalid Venoms/chemistry , Peptides/isolation & purification , Amino Acid Sequence , Anaphylatoxins/biosynthesis , Anaphylatoxins/immunology , Animals , Blood Coagulation/drug effects , Cells, Cultured , Complement Activation/drug effects , Electrophoresis, Polyacrylamide Gel , Hemorrhage/chemically induced , Humans , In Vitro Techniques , Male , Mice, Inbred C57BL , Molecular Weight , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Peptides/pharmacology , Peptides/toxicity , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Reptilian Proteins/isolation & purification , Reptilian Proteins/pharmacology , Reptilian Proteins/toxicity , Viper Venoms/isolation & purification , Viper Venoms/pharmacology , Viper Venoms/toxicity , Xenopus laevis
11.
Mol Immunol ; 75: 133-43, 2016 07.
Article in English | MEDLINE | ID: mdl-27280846

ABSTRACT

A serine protease activity was detected in aqueous peanuts seeds extracts, partially purified and characterized as a thiol-dependent serine protease. The potential role of this proteolytic activity on allergic reaction to peanuts was prospected through complement activation studies in human plasma and serum, and MDCK cells to investigate a possible occludin degradation in tight junctions. The peanut protease activity induced the production of anaphylatoxins C3a and C5a, and of the terminal membrane attack complex SC5b-9 whatever the complement activation pathway. The protease activity was also involved in the partial digestion of occludin within tight junctions, with for result, an increase of the epithelial permeability to antigen absorption.


Subject(s)
Anaphylatoxins/immunology , Arachis/enzymology , Peanut Hypersensitivity/enzymology , Serine Proteases/immunology , Anaphylatoxins/chemistry , Animals , Arachis/chemistry , Chromatography, Affinity , Dogs , Humans , Madin Darby Canine Kidney Cells , Serine Proteases/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Sulfhydryl Compounds
12.
J Neuroimmunol ; 289: 1-7, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26616865

ABSTRACT

Acquired demyelinating syndromes (ADS) in children evolve either as a monophasic disease diagnosed as acute demyelinating encephalomyelitis (ADEM), transverse myelitis (TM) or optic neuritis (ON), or a multiphasic one with several relapses most often leading to the diagnosis of multiple sclerosis (MS) or neuromyelitis optica (NMO). These neuroinflammatory disorders are increasingly associated with autoantibodies against proteins such as aquaporin-4 in rare instances, and more frequently against myelin oligodendrocyte glycoprotein (MOG). Recently, in adult NMO patients, C5a levels were shown to be elevated in cerebrospinal fluid (CSF) during acute exacerbation. We investigated the CSF levels of anaphylatoxins and pro-inflammatory cytokines, and plasma MOG antibodies in onset samples from children with ADS. Thirty four children presenting with a first episode of ADS, 17 with monophasic ADS (9 with ADEM, 4 with TM and 4 with ON) and 17 with MS, who had paired blood and CSF samples at onset were included and compared to 12 patients with other non-inflammatory neurological disorders (OND). Cytokines and anaphylatoxins in CSF were measured by Cytometric Bead Array immunoassay. MOG antibody titers in plasma were tested by flow cytometry using a stable cell line expressing full-length human MOG. We found a significant increase in C5a levels in the CSF of patients with monophasic ADS (n=17) compared to OND (n=12, p=0.0036) and to MS (n=17, p=0.0371). The C5a levels in MS were higher than in OND without reaching significance (p=0.2). CSF IL-6 levels were significantly increased in monophasic ADS compared to OND (p=0.0027) and to MS (p=0.0046). MOG antibody plasma levels were significantly higher in monophasic ADS (p<0.0001) and, to a lesser extent, in MS compared to OND (p=0.0023). Plasma MOG antibodies and CSF IL-6 levels were significantly correlated (r=0.51, p=0.018). CSF C5a and IL-6 levels are increased in monophasic ADS but not in MS when compared to OND, suggesting that these markers may help to predict monophasic or relapsing fate of ADS at onset. MOG antibody titers, which were higher in monophasic ADS than in MS, correlated with IL-6 levels, but not with C5a, suggesting an association between MOG antibodies and neuroinflammation in pediatric ADS.


Subject(s)
Demyelinating Diseases , Immunoglobulin G/blood , Interleukin-6/immunology , Multiple Sclerosis , Myelin-Oligodendrocyte Glycoprotein/immunology , Adolescent , Anaphylatoxins/cerebrospinal fluid , Anaphylatoxins/immunology , Anaphylatoxins/metabolism , Child , Cytokines/metabolism , Demyelinating Diseases/blood , Demyelinating Diseases/cerebrospinal fluid , Demyelinating Diseases/immunology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Interleukin-6/blood , Interleukin-6/cerebrospinal fluid , Longitudinal Studies , Male , Multiple Sclerosis/blood , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/immunology , Myelin-Oligodendrocyte Glycoprotein/blood , Myelin-Oligodendrocyte Glycoprotein/cerebrospinal fluid , Retrospective Studies
13.
Mol Immunol ; 67(2 Pt B): 575-83, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26256795

ABSTRACT

BACKGROUND: Staphylococcus aureus peritonitis is a serious complication of Chronic Peritoneal Dialysis (CPD) and associated with a higher risk for severe and recurrent infections compared with other bacteria. We have previously shown that complement-mediated effectors essential for optimal opsonophagocytosis of S. aureus are inhibited by high glucose concentrations. Since most commonly used peritoneal dialysis (PD) fluids are glucose-based, we hypothesized that glucose-based PD fluids likely inhibit complement host defenses against S. aureus. METHODS: Commercially available PD fluids were tested: glucose-based (Dianeal), Dianeal supplemented with amino acids, icodextrin-based (Extraneal) and amino acid-based (Nutrineal). Control PD fluid was generated to simulate Dianeal excluding the glucose. Three commercially available glucose concentrations were tested: Dianeal 1.5% (15 gm/1000 ml), Dianeal 2.5% (25 gm/1000 ml) and Dianeal 4.25% (42.5 gm/1000 ml). Complement effectors against S. aureus were analyzed including opsonization with C3-fragments, anaphylatoxin generation, and phagocytosis efficiency. We also evaluated clinical strains, including MRSA strains, and specific complement activation pathways. RESULTS: Glucose-based PD fluids inhibited complement opsonization of S. aureus (≥7-fold reduction) and inhibited S. aureus-induced generation of anaphylatoxins C3a and C5a (>10-fold reduction) compared to non-glucose based PD fluids. Dianeal 1.5%, 2.5% and 4.25%, all similarly inhibited C3-mediated opsonization. Glucose-based PD fluids showed a ≥4-fold reduction in opsonization of clinical strains of S.aureus, including MRSA strains. Decreased opsonization of S.aureus in the glucose-based PD fluid compared with non-glucose based fluids correlated with decreased phagocytosis by neutrophils. CONCLUSION: Complement-mediated opsonophagocytosis of S. aureus and anaphylatoxin generation were severely inhibited in glucose-based PD fluids compared with non-glucose-based PD fluids. By inhibiting complement host defenses, glucose-based PD fluids may increase the risk of and severity of S. aureus peritonitis for CPD patients using these fluids.


Subject(s)
Dialysis Solutions/pharmacology , Glucose/pharmacology , Immunity, Innate/drug effects , Renal Dialysis , Staphylococcus aureus/immunology , Anaphylatoxins/immunology , Complement C3/immunology , Complement Pathway, Alternative/drug effects , Humans , Opsonin Proteins/metabolism , Phagocytosis/drug effects , Staphylococcus aureus/drug effects
14.
J Transl Med ; 13: 272, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26289385

ABSTRACT

The complement mediators are the major effectors of the immune balance, which operates at the interface between the innate and adaptive immunity, and is vital for many immunoregulatory functions. Activation of the complement cascade through the classical, alternative or lectin pathways thus generating opsonins like C3b and C5b, anaphylatoxins C3a and C5a, chemotaxin, and inflammatory mediators, which leads to cellular death. Complement mediators that accelerate the airway remodeling are not well defined; however, an uncontrolled Th2-driven adaptive immune response has been linked to the major pathophysiologic features of asthma, including bronchoconstriction, airway hyperresponsiveness, and airway inflammation. The mechanisms leading to complement mediated airway tissue remodeling, and the effect of therapy on preventing and/or reversing it are not clearly understood. This review highlights complement-mediated inflammation, and the mechanism through it triggers the airway tissue injury and remodeling in the airway epithelium that could serve as potential targets for developing a new drug to rescue the asthma patients.


Subject(s)
Airway Remodeling , Asthma/immunology , Complement Activation , Complement C3a/immunology , Complement C5a/immunology , Anaphylatoxins/immunology , Animals , Asthma/pathology , Chemotactic Factors/immunology , Humans , Immunity, Innate , Inflammation/immunology , Inflammation Mediators/immunology , Interleukin-13/immunology , Opsonin Proteins/immunology , Th2 Cells/cytology , Transforming Growth Factor beta/immunology
15.
Mol Immunol ; 66(2): 299-309, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25909494

ABSTRACT

Notothenioidei are typical Antarctic teleosts evolved to adapt to the very low temperatures of the Antarctic seas. Aim of the present paper is to investigate sequence and structure of C3, the third component of the complement system of the notothenioid Trematomus bernacchii and Chionodraco hamatus. We determined the complete nucleotide sequence of two C3 isoforms of T. bernacchii and a single C3 isoform of C. hamatus. These sequences were aligned against other homologous teleost sequences to check for the presence of diversifying selection. Evidence for positive selection was observed in the evolutionary lineage of Antarctic teleost C3 sequences, especially in that of C. hamatus, the most recently diverged species. Adaptive selection affected numerous amino acid positions including three residues located in the anaphylatoxin domain. In an attempt to evaluate the link between sequence variants and specific structural features, we constructed molecular models of Antarctic teleost C3s, of their proteolytic fragments C3b and C3a, and of the corresponding molecules of the phylogenetically related temperate species Epinephelus coioides, using human crystallographic structures as templates. Subsequently, we compared dynamic features of these models by molecular dynamics simulations and found that the Antarctic C3s models show higher flexibility, which likely allows for more pronounced movements of both the TED domain in C3b and the carboxyl-terminal region of C3a. As such dynamic features are associated to positively selected sites, it appears that Antarctic teleost C3 molecules positively evolved toward an increased flexibility, to cope with low kinetic energy levels of the Antarctic marine environment.


Subject(s)
Anaphylatoxins/immunology , Complement C3/immunology , Evolution, Molecular , Fish Proteins/immunology , Perciformes/immunology , Phylogeny , Adaptation, Physiological/genetics , Adaptation, Physiological/immunology , Anaphylatoxins/chemistry , Anaphylatoxins/genetics , Animals , Antarctic Regions , Base Sequence , Cold Temperature , Complement C3/chemistry , Complement C3/genetics , Fish Proteins/chemistry , Fish Proteins/genetics , Gene Expression , Humans , Molecular Dynamics Simulation , Molecular Sequence Data , Perciformes/classification , Perciformes/genetics , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Structure, Tertiary , Proteolysis , Selection, Genetic , Sequence Alignment
16.
J Immunol ; 193(10): 5099-107, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25297874

ABSTRACT

Listeria monocytogenes is a major cause of mortality resulting from food poisoning in the United States. In mice, C5 has been genetically linked to host resistance to listeriosis. Despite this genetic association, it remains poorly understood how C5 and its activation products, C5a and C5b, confer host protection to this Gram-positive intracellular bacterium. In this article, we show in a systemic infection model that the major receptor for C5a, C5aR1, is required for a normal robust host immune response against L. monocytogenes. In comparison with wild-type mice, C5aR1(-/-) mice had reduced survival and increased bacterial burden in their livers and spleens. Infected C5aR1(-/-) mice exhibited a dramatic reduction in all major subsets of splenocytes, which was associated with elevated caspase-3 activity and increased TUNEL staining. Because type 1 IFN has been reported to impede the host response to L. monocytogenes through the promotion of splenocyte death, we examined the effect of C5aR1 on type 1 IFN expression in vivo. Indeed, serum levels of IFN-α and IFN-ß were significantly elevated in L. monocytogenes-infected C5aR1(-/-) mice. Similarly, the expression of TRAIL, a type 1 IFN target gene and a proapoptotic factor, was elevated in NK cells isolated from infected C5aR1(-/-) mice. Treatment of C5aR1(-/-) mice with a type 1 IFNR blocking Ab resulted in near-complete rescue of L. monocytogenes-induced mortality. Thus, these findings reveal a critical role for C5aR1 in host defense against L. monocytogenes through the suppression of type 1 IFN expression.


Subject(s)
Interferon-alpha/genetics , Interferon-beta/genetics , Listeria monocytogenes/immunology , Listeriosis/immunology , Spleen/immunology , Anaphylatoxins/immunology , Animals , Antibodies/pharmacology , Apoptosis , Bacterial Load , Caspase 3/genetics , Caspase 3/immunology , Complement C5a/genetics , Complement C5a/immunology , Complement C5b/genetics , Complement C5b/immunology , Gene Expression , Interferon-alpha/immunology , Interferon-beta/immunology , Listeriosis/drug therapy , Listeriosis/microbiology , Listeriosis/mortality , Liver/immunology , Liver/microbiology , Liver/pathology , Lymphocytes/immunology , Lymphocytes/microbiology , Lymphocytes/pathology , Male , Mice , Mice, Knockout , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/immunology , Receptors, Interferon/antagonists & inhibitors , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Spleen/microbiology , Spleen/pathology , Survival Analysis , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/immunology
17.
Curr Opin Organ Transplant ; 19(1): 8-13, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24316762

ABSTRACT

PURPOSE OF REVIEW: To provide the reader with an up-to-date comprehensive review of recent findings that highlight advances describing how proteins of the complement cascades contribute to the pathogenesis of solid organ rejection. The review is focussed mainly on renal transplantation. RECENT FINDINGS: Of note are recent advances in elucidating the interactions between anaphylatoxins and their receptors in organ transplantation; there is evidence of direct engagement of C5aR on donor tubules and in addition, mechanisms by which the allostimulatory capacity of dendritic cells is modulated by complement are more fully understood. Activation of the lectin pathway is increasingly implicated in allograft rejection and the role of complement in modulating regulatory T cells is being vigorously investigated. As an alternative to systemic complement inhibition, there is continued focus on the design of targeted anti-complement therapies, directed to the donor organ. SUMMARY: Complement has evolved as the first line of defence against pathogens, employing well defined effector mechanisms to rapidly remove infectious material. However, complement effector mechanisms are also triggered during inflammation associated with solid organ transplantation. Hence, complement has a significant role in mediating donor organ injury during both the initial ischaemia/reperfusion phase and the subsequent adaptive immune responses. Research on mechanisms of complement-mediated injury in transplantation provide a basis for the development of therapies that are aimed at transiently blocking complement activation at the site of injury, whereas leaving systemic anti-bacterial complement effector mechanisms intact.


Subject(s)
Complement System Proteins/immunology , Graft Rejection/immunology , Kidney Transplantation , Transplantation Immunology , Anaphylatoxins/immunology , Animals , Complement Activation , Humans
18.
J Diabetes Res ; 2014: 762051, 2014.
Article in English | MEDLINE | ID: mdl-25610878

ABSTRACT

Hyperglycemia from diabetes is associated with increased risk of infection from S. aureus and increased severity of illness. Previous work in our laboratory demonstrated that elevated glucose (>6 mM) dramatically inhibited S. aureus-initiated complement-mediated immune effectors. Here we report in vivo studies evaluating the extent to which a hyperglycemic environment alters complement-mediated control of S. aureus infection in a rat peritonitis model. Rats were treated with streptozocin to induce diabetes or sham-treated and then inoculated i.p. with S. aureus. Rats were euthanized and had peritoneal lavage at 2 or 24 hours after infection to evaluate early and late complement-mediated effects. Hyperglycemia decreased the influx of IgG and complement components into the peritoneum in response to S. aureus infection and decreased anaphylatoxin generation. Hyperglycemia decreased C4-fragment and C3-fragment opsonization of S. aureus recovered in peritoneal fluids, compared with euglycemic or insulin-rescued rats. Hyperglycemic rats showed decreased phagocytosis efficiency compared with euglycemic rats, which correlated inversely with bacterial survival. These results suggest that hyperglycemia inhibited humoral effector recruitment, anaphylatoxin generation, and complement-mediated opsonization of S. aureus, suggesting that hyperglycemic inhibition of complement effectors may contribute to the increased risk and severity of S. aureus infections in diabetic patients.


Subject(s)
Complement Activation , Complement System Proteins/immunology , Diabetes Mellitus, Experimental/immunology , Peritonitis/immunology , Staphylococcal Infections/immunology , Anaphylatoxins/immunology , Anaphylatoxins/metabolism , Animals , Biomarkers/blood , Blood Glucose/metabolism , Complement System Proteins/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Immunity, Humoral , Male , Neutrophil Infiltration , Neutrophils/immunology , Neutrophils/microbiology , Peritoneal Cavity/microbiology , Peritonitis/microbiology , Phagocytosis , Rats, Wistar , Staphylococcal Infections/microbiology , Staphylococcus aureus/growth & development , Staphylococcus aureus/immunology , Streptozocin , Time Factors
19.
Sci Rep ; 3: 2271, 2013.
Article in English | MEDLINE | ID: mdl-23881356

ABSTRACT

Polyreactive antibodies are a major component of the natural antibody repertoire and are capable of binding a variety of structurally unrelated antigens. Many of the properties attributed to natural antibodies, in fact, are turning out to be due to polyreactive antibodies. In humans, each day, billions of cells undergo apoptosis. In the present experiments, we show by ImageStream technology that although polyreactive antibodies do not bind to live T cells they bind to both the plasma membrane and cytoplasm of late apoptotic cells, fix complement, generate the anaphylatoxin C5a and increase by as much as 5 fold complement-mediated phagocytosis by macrophages. Of particular importance, T cells undergoing apoptosis following infection with HIV also bind polyreactive antibodies and are phagocytosed. We conclude that the polyreactive antibodies in the natural antibody repertoire contribute in a major way to the clearance of cells made apoptotic by a variety of natural and infectious processes.


Subject(s)
Antibodies/immunology , Apoptosis/immunology , Apoptosis/radiation effects , Complement System Proteins/immunology , Phagocytosis/immunology , Anaphylatoxins/immunology , Animals , Antibodies/metabolism , Complement C5a/immunology , Complement System Proteins/metabolism , HIV/physiology , Humans , Immunoglobulin M/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , T-Lymphocytes/immunology , T-Lymphocytes/pathology , T-Lymphocytes/radiation effects , Ultraviolet Rays/adverse effects
20.
Mol Immunol ; 56(3): 181-90, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23796748

ABSTRACT

Complement receptors are expressed on cells of the innate and the adaptive immune system. They play important roles in pathogen and danger sensing as they translate the information gathered by complement fluid phase sensors into cellular responses. Further, they control complement activation on viable and apoptotic host cells, clearance of immune complexes and mediate opsonophagocytosis. More recently, evidence has accumulated that complement receptors form a complex network with other innate receptors systems such as the Toll-like receptors, the Notch signaling system, IgG Fc receptors and C-type lectin receptors contributing to the benefit and burden of innate and adaptive immune responses in autoimmune and allergic diseases as well as in cancer and transplantation. Here, we will discuss recent developments and emerging concepts of complement receptor activation and regulation with a particular focus on the differentiation, maintenance and contraction of effector and regulatory T cells.


Subject(s)
Receptors, Complement/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Anaphylatoxins/immunology , Anaphylatoxins/metabolism , Animals , Cell Differentiation/immunology , Complement Activation/immunology , Homeostasis/immunology , Humans , Lymphocyte Activation/immunology , Membrane Cofactor Protein/metabolism , Receptors, Complement/immunology , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...