Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Circulation ; 122(10): 1026-36, 2010 Sep 07.
Article in English | MEDLINE | ID: mdl-20733098

ABSTRACT

BACKGROUND: Receptor binding of complement C5a leads to proinflammatory activation of many cell types, but the role of receptor-mediated action during arterial remodeling after injury has not been studied. In the present study, we examined the contribution of the C5a receptor (C5aR) to neointima formation in apolipoprotein E-deficient mice employing a C5aR antagonist (C5aRA) and a C5aR-blocking monoclonal antibody. METHODS AND RESULTS: Mice fed an atherogenic diet were subjected to wire-induced endothelial denudation of the carotid artery and treated with C5aRA and anti-C5aR-blocking monoclonal antibody or vehicle control. Compared with controls, neointima formation was significantly reduced in mice receiving C5aRA or anti-C5aR-blocking monoclonal antibody for 1 week but not for 3 weeks, attributable to an increased content of vascular smooth muscle cells, whereas a marked decrease in monocyte and neutrophil content was associated with reduced vascular cell adhesion molecule-1. As assessed by immunohistochemistry, reverse transcription polymerase chain reaction, and flow cytometry, C5aR was expressed in lesional and cultured vascular smooth muscle cells, upregulated by injury or tumor necrosis factor-alpha, and reduced by C5aRA. Plasma levels and neointimal plasminogen activator inhibitor-1 peaked 1 week after injury and were downregulated in C5aRA-treated mice. In vitro, C5a induced plasminogen activator inhibitor-1 expression in endothelial cells and vascular smooth muscle cells in a C5aRA-dependent manner, possibly accounting for higher vascular smooth muscle cell immigration. CONCLUSIONS: One-week treatment with C5aRA or anti-C5aR-blocking monoclonal antibody limited neointimal hyperplasia and inflammatory cell content and was associated with reduced vascular cell adhesion molecule-1 expression. However, treatment for 3 weeks failed to reduce but rather stabilized plaques, likely by reducing vascular plasminogen activator inhibitor-1 and increasing vascular smooth muscle cell migration.


Subject(s)
Antibodies, Monoclonal/pharmacology , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Muscle, Smooth, Vascular/drug effects , Oligopeptides/pharmacology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Animals , Apolipoproteins E/genetics , Atherosclerosis/pathology , Carotid Artery Injuries/drug therapy , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Cell Movement/drug effects , Cell Movement/immunology , Complement C5a/metabolism , Disease Models, Animal , Flow Cytometry , Leukocytes/cytology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/immunology , Serpin E2 , Serpins/metabolism , Tunica Intima/drug effects , Tunica Intima/immunology , Tunica Intima/pathology , Vascular Cell Adhesion Molecule-1/metabolism
2.
J Am Soc Nephrol ; 19(12): 2302-12, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18753257

ABSTRACT

Complement activation plays a key role in mediating apoptosis, inflammation, and transplant rejection. In this study, the role of the complement 5a receptor (C5aR) was examined in human renal allografts and in an allogenic mouse model of renal transplant rejection. In human kidney transplants with acute rejection, C5aR expression was increased in renal tissue and in cells infiltrating the tubulointerstitium. Similar findings were observed in mice. When recipient mice were treated once daily with a C5aR antagonist before transplantation, long-term renal allograft survival was markedly improved compared with vehicle-treatment (75 versus 0%), and apoptosis was reduced. Furthermore, treatment with a C5aR antagonist significantly attenuated monocyte/macrophage infiltration, perhaps a result of reduced levels of monocyte chemoattractant protein 1 and the intercellular adhesion molecule 1. In vitro, C5aR antagonism inhibited intercellular adhesion molecule 1 upregulation in primary mouse aortic endothelial cells and reduced adhesion of peripheral blood mononuclear cells. Furthermore, C5aR blockade markedly reduced alloreactive T cell priming. These results demonstrate that C5aR plays an important role in mediating acute kidney allograft rejection, suggesting that pharmaceutical targeting of C5aR may have potential in transplantation medicine.


Subject(s)
Graft Survival , Kidney Transplantation/methods , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Animals , Apoptosis , Disease Models, Animal , Gene Expression Regulation , Humans , Intercellular Adhesion Molecule-1/metabolism , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/metabolism , T-Lymphocytes/metabolism
3.
J Am Soc Nephrol ; 18(5): 1508-15, 2007 May.
Article in English | MEDLINE | ID: mdl-17389734

ABSTRACT

Renal fibrosis is the final common pathway of most progressive renal diseases. C5 was recently identified as a risk factor for liver fibrosis. This study investigated the role of C5 in the development of renal tubulointerstitial fibrosis by (1) induction of renal fibrosis in wild-type and C5(-/-) mice by unilateral ureteral ligation (UUO) and (2) investigation of the effects of a C5a receptor antagonist (C5aRA) in UUO. In C5(-/-) mice, when compared with wild-type controls, markers of renal fibrosis (Sirius Red, type I collagen, fibronectin, alpha-smooth muscle actin, vimentin, and infiltrating macrophages) were significantly reduced on day 5 of UUO. On day 10, fibronectin mRNA and protein expression were still reduced in the C5(-/-) mice. Cortical mRNA of all PDGF isoforms and of TGF-beta(1) (i.e., central mediators of renal disease) were significantly reduced in C5(-/-) mice when compared with controls. Renal tubular cell expression of the C5aR was sparse in normal cortex but markedly upregulated after UUO. Treatment of wild-type UUO mice with C5aRA also led to a significant reduction of cortical Sirius Red staining, fibronectin protein expression, and PDGF-B mRNA expression on day 5. Neither genetic C5 deficiency nor C5aRA treatment caused any histologic changes in the nonobstructed kidneys. In cultured murine cortical tubular cells, C5a stimulated production of TGF-beta(1), and this was inhibited by C5aRA. Using a combined genetic and pharmacologic approach, C5, in particular C5a, is identified as a novel profibrotic factor in renal disease and as a potential new therapeutic target.


Subject(s)
Complement C5/genetics , Disease Models, Animal , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney/pathology , Animals , Cells, Cultured , Complement C5a/genetics , Complement C5a/metabolism , Fibrosis , Gene Expression Regulation , Humans , Kidney Diseases/drug therapy , Kidney Diseases/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Complement/therapeutic use , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/drug therapy , Ureteral Obstruction/genetics , Ureteral Obstruction/pathology , Wound Healing/drug effects
4.
Bioorg Med Chem Lett ; 16(19): 5088-92, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16876401

ABSTRACT

A new class of peptidomimetic C5a receptor antagonists characterized by C-terminal amino acids with hydrophobic side chains is presented. Systematic optimization of the first hits led to JPE1375 (36), which was intensively characterized in vitro and in vivo. Compound 36 exhibits high microsomal stability and receptor specificity and is highly active in an immune complex mediated peritonitis model (reverse passive Arthus reaction) in mice.


Subject(s)
Oligopeptides/chemistry , Oligopeptides/pharmacology , Peritonitis/drug therapy , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Animals , Antigen-Antibody Complex/adverse effects , Disease Models, Animal , Hydrophobic and Hydrophilic Interactions , Inhibitory Concentration 50 , Mice , Molecular Mimicry , Peritonitis/chemically induced , Structure-Activity Relationship , Substrate Specificity
5.
J Clin Invest ; 116(3): 783-96, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16511606

ABSTRACT

Complement component 5 (C5) has been described as either promoting or protecting against airway hyperresponsiveness (AHR) in experimental allergic asthma, suggesting pleomorphic effects of C5. Here we report that local pharmacological targeting of the C5a receptor (C5aR) prior to initial allergen sensitization in murine models of inhalation tolerance or allergic asthma resulted in either induction or marked enhancement of Th2-polarized immune responses, airway inflammation, and AHR. Importantly, C5aR-deficient mice exhibited a similar, increased allergic phenotype. Pulmonary allergen exposure in C5aR-targeted mice resulted in increased sensitization and accumulation of CD4+ CD69+ T cells associated with a marked increase in pulmonary myeloid, but not plasmacytoid, DC numbers. Pulmonary DCs from C5aR-targeted mice produced large amounts of CC chemokine ligand 17 (CCL17) and CCL22 ex vivo, suggesting a negative impact of C5aR signaling on pulmonary homing of Th2 cells. In contrast, C5aR targeting in sensitized mice led to suppressed airway inflammation and AHR but was still associated with enhanced production of Th2 effector cytokines. These data suggest a dual role for C5a in allergic asthma, i.e., protection from the development of maladaptive type 2 immune responses during allergen sensitization at the DC/T cell interface but enhancement of airway inflammation and AHR in an established inflammatory environment.


Subject(s)
Allergens/administration & dosage , Allergens/immunology , Asthma/immunology , Bronchial Hyperreactivity/immunology , Receptor, Anaphylatoxin C5a/physiology , Th2 Cells/immunology , Animals , Asthma/genetics , Bronchi/immunology , Bronchi/metabolism , Bronchial Hyperreactivity/genetics , Dendritic Cells/immunology , Disease Models, Animal , Immunity, Innate/genetics , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/physiology , Lung/cytology , Lung/immunology , Lung/metabolism , Mice , Mice, Inbred BALB C , Mice, Transgenic , Mucus/metabolism , Ovalbumin/administration & dosage , Ovalbumin/immunology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Signal Transduction/genetics , Signal Transduction/immunology , Th2 Cells/metabolism
6.
Mol Immunol ; 43(1-2): 13-21, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16019071

ABSTRACT

The innate immune system provides sophisticated defense mechanisms to protect complex macroorganisms from the attack of microorganisms. Among those, the complement system and Toll-like receptors are of paramount importance to discriminate between infectious non-self and non-infectious self and to provide critical danger signals instructing adaptive immune responses. Here, we will discuss recent advances in our understanding of the mechanisms underlying complement and TLR-mediated regulation of adaptive immunity. We will focus on the regulation of T cell immunity and discuss recent findings on the cross-talk between complement receptor and TLR signaling pathways. Such cross-talk is likely to affect the outcome of infections with intracellular pathogens, as well as the initiation and maintenance of aberrant immune responses leading to autoimmunity and atopy.


Subject(s)
Autoimmunity/physiology , Complement System Proteins/immunology , Immunity, Cellular/physiology , T-Lymphocytes/immunology , Toll-Like Receptors/immunology , Animals , Autoimmune Diseases/immunology , Humans
7.
Immunity ; 22(4): 415-26, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15845447

ABSTRACT

The complement system and the Toll-like receptors (TLRs) are two central arms of innate immunity that are critical to host defense as well as the development of adaptive immunity. Most pathogens activate both complement and TLRs, suggesting the potential for crosstalk between the two systems. We show here that the complement-derived C5a anaphylatoxin negatively regulates TLR4- and CD40-induced synthesis of IL-12 family cytokines (IL-12, IL-23, and IL-27) from inflammatory macrophages (M phi s) by extracellular signal-regulated kinase- and phosphoinositide 3 kinase-dependent pathways. This decreased cytokine response translates into a decreased T helper type 1 (Th1) response in vitro and in vivo. Accordingly, we found enhanced Th1 immunity in C5a receptor-deficient mice, something that conferred protection from Leishmania major infection. Our findings identify the negative impact of C5a on IL-12 family cytokines as an important mechanism for regulating Th1 polarization in response to innate and adaptive immune network activation.


Subject(s)
Complement C5a/physiology , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Base Sequence , DNA-Binding Proteins/metabolism , Down-Regulation , Interferon Regulatory Factor-1 , Interferon Regulatory Factors , Interleukin-12/biosynthesis , Leishmania/pathogenicity , Membrane Glycoproteins/immunology , Mice , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Receptors, Cell Surface/immunology , Repressor Proteins/metabolism , Toll-Like Receptor 4 , Toll-Like Receptors , Up-Regulation
8.
J Immunol ; 173(5): 3437-45, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15322209

ABSTRACT

Immune complex (IC)-induced inflammation is integral to the pathogenesis of several autoimmune diseases. ICs activate the complement system and interact with IgG FcgammaR. In this study, we demonstrate that activation of the complement system, specifically generation of C5a, initiates the neutrophilic inflammation in IC peritonitis. We show that ablation of C5a receptor signaling abrogates neutrophil recruitment in wild-type mice and prevents the enhancement of neutrophil migration seen in FcgammaRIIB(-/-) mice, suggesting that C5aR signaling is the crucial initial event upstream of FcgammaR signaling. We also provide evidence that C5a initiates the inflammatory cascade both directly, through C5aR-mediated effector functions on infiltrating and resident peritoneal cells, and indirectly, through shifting the balance between activating and inhibitory FcgammaRs on resident cells toward an inflammatory phenotype. We conclude that complement activation and C5a generation are prerequisites for IC-induced inflammation through activating FcgammaR, which amplifies complement-induced inflammation in autoimmunity.


Subject(s)
Complement C5a/metabolism , Immune Complex Diseases/metabolism , Inflammation/metabolism , Peritonitis/metabolism , Animals , Cell Movement/immunology , Chemokines, CXC/immunology , Chemokines, CXC/metabolism , Female , Immune Complex Diseases/immunology , Inflammation/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Neutrophils/immunology , Peritonitis/immunology , Receptor, Anaphylatoxin C5a/immunology , Receptor, Anaphylatoxin C5a/metabolism , Receptors, IgG/immunology , Receptors, IgG/metabolism , Signal Transduction/immunology , Signal Transduction/physiology
9.
Mol Immunol ; 41(2-3): 123-31, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15159057

ABSTRACT

The complement system has long been recognized for its role as a lytic effector system that protects against microbial pathogens, as well as for its role in mediating acute and chronic inflammatory responses. Many of the inflammatory sequelae of complement activation can be related to the complement cleavage fragments C3a and C5a, the so-called anaphylatoxins (ATs). Cloning and subsequent gene targeting of their corresponding receptors, as well as generation of specific C3a and C5a inhibitors, have fueled new interest in studies aimed at defining the roles of the anaphylatoxins in inflammatory diseases. Traditionally, the anaphylatoxins have been considered mediators of end-stage effector mechanisms. However, recent data from animal models of allergic asthma suggest that C3a and C5a provide a critical link between innate and adaptive immunity. This review is aimed at outlining our current knowledge of when and where anaphylatoxins contribute to and control the development of allergic asthma. The accumulated data suggest a model in which C3a and C5a play important but opposing roles during allergen-induced T-cell polarization: C3a promotes Th2 responses, whereas C5a prevents Th2 polarization. During the effector phase, both anaphylatoxins trigger the inflammatory response and contribute to bronchoconstriction.


Subject(s)
Anaphylatoxins/immunology , Asthma/immunology , Allergens/immunology , Animals , Complement System Proteins/immunology , Humans , Immunity, Cellular , Immunity, Innate , T-Lymphocytes, Helper-Inducer/immunology
10.
J Biol Chem ; 279(1): 142-51, 2004 Jan 02.
Article in English | MEDLINE | ID: mdl-14570896

ABSTRACT

The anaphylatoxin C5a exerts a plethora of biologic activities critical in the pathogenesis of systemic inflammatory diseases. Recently, we reported on a C5a mutant, jun/fos-A8, as a potent antagonist for the human and mouse C5a receptor (CD88). Addressing the molecular mechanism accounting for CD88 receptor antagonism by site-directed mutagenesis, we found that a positively charged amino acid at position 69 is crucial. Replacements by either hydrophobic or negatively charged amino acids switched the CD88 antagonist jun/fos-A8 to a CD88 agonist. In addition to CD88, the seven-transmembrane receptor C5L2 has recently been found to provide high affinity binding sites for C5a and its desarginated form, C5adesArg74. A jun/fos-A8 mutant in which the jun/ fos moieties and amino acids at positions 71-73 were deleted, A8Delta71-73, blocked C5a and C5adesArg74 binding to CD88 and C5L2. In contrast, the cyclic C5a C-terminal analog peptide AcF-[OP-d-ChaWR] inhibited binding of the two anaphylatoxins to CD88 but not to C5L2, suggesting that the C5a core segment is important for high affinity binding to C5L2. Both receptors are coexpressed on human monocytes and the human mast cell line HMC-1; however, C5L2 expression on monocytes is weaker as compared with HMC-1 cells and highly variable. In contrast, no C5L2 expression was found on human neutrophils. A8Delta71-73 is the first antagonist that blocks C5a and C5adesArg74 binding to both C5a receptors, CD88 and C5L2, making it a valuable tool for studying C5L2 functions and for blocking the biological activities of C5a and C5adesArg74 in mice and humans.


Subject(s)
Complement C5a/genetics , Complement C5a/pharmacology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptors, Chemokine/antagonists & inhibitors , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Binding Sites , Cell Line , Chromatography, Affinity , Complement C5a/isolation & purification , DNA Primers , Humans , Mast Cells , Mice , Molecular Sequence Data , Monocytes/physiology , Mutagenesis, Site-Directed , Neutrophils/physiology , Polymerase Chain Reaction , Receptors, Chemokine/isolation & purification , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL
...