Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cardiovasc Res ; 118(14): 2932-2945, 2022 11 10.
Article in English | MEDLINE | ID: mdl-34897380

ABSTRACT

AIMS: Atherosclerosis is a chronic inflammatory disease of the vessel wall controlled by local and systemic immune responses. The role of interleukin-23 receptor (IL-23R), expressed in adaptive immune cells (mainly T-helper 17 cells) and γδ T cells, in atherosclerosis is only incompletely understood. Here, we investigated the vascular cell types expressing IL-23R and addressed the function of IL-23R and γδ T cells in atherosclerosis. METHODS AND RESULTS: IL-23R+ cells were frequently found in the aortic root in contrast to the aorta in low-density lipoprotein receptor deficient IL-23R reporter mice (Ldlr-/-Il23rgfp/+), and mostly identified as γδ T cells that express IL-17 and GM-CSF. scRNA-seq confirmed γδ T cells as the main cell type expressing Il23r and Il17a in the aorta. Ldlr-/-Il23rgfp/gfp mice deficient in IL-23R showed a loss of IL-23R+ cells in the vasculature, and had reduced atherosclerotic lesion formation in the aortic root compared to Ldlr-/- controls after 6 weeks of high-fat diet feeding. In contrast, Ldlr-/-Tcrδ-/- mice lacking all γδ T cells displayed unaltered early atherosclerotic lesion formation compared to Ldlr-/- mice. In both HFD-fed Ldlr-/-Il23rgfp/gfp and Ldlr-/-Tcrδ-/- mice a reduction in the plaque necrotic core area was noted as well as an expansion of splenic regulatory T cells. In vitro, exposure of bone marrow-derived macrophages to both IL-17A and GM-CSF induced cell necrosis, and necroptotic RIP3K and MLKL expression, as well as inflammatory mediators. CONCLUSIONS: IL-23R+ γδ T cells are predominantly found in the aortic root rather than the aorta and promote early atherosclerotic lesion formation, plaque necrosis, and inflammation at this site. Targeting IL-23R may thus be explored as a therapeutic approach to mitigate atherosclerotic lesion development.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Receptors, Interleukin , Animals , Mice , Atherosclerosis/metabolism , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Mice, Inbred C57BL , Mice, Knockout , Necrosis/metabolism , Plaque, Atherosclerotic/metabolism , Receptors, LDL , Th17 Cells , Receptors, Interleukin/genetics
2.
Antibodies (Basel) ; 10(1)2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33430104

ABSTRACT

The complement system has demonstrated roles in regulating tumor growth, although these may differ between tumor types. The current study used two murine breast cancer models (EMT6 and 4T1) to investigate whether pharmacological targeting of receptors for complement proteins C3a (C3aR) and C5a (C5aR1) is protective in murine breast cancer models. In contrast to prior studies in other tumor models, treatment with the selective C5aR1 antagonist PMX53 had no effect on tumor growth. However, treatment of mice with a dual C3aR/C5aR1 agonist (YSFKPMPLaR) significantly slowed mammary tumor development and progression. Examination of receptor expression by quantitative polymerase chain reaction (qPCR) analysis showed very low levels of mRNA expression for either C3aR or C5aR1 by EMT6 or 4T1 mammary carcinoma cell lines compared with the J774 macrophage line or bone marrow-derived macrophages. Moreover, flow cytometric analysis found no evidence of C3aR or C5aR1 protein expression by either EMT6 or 4T1 cells, leading us to hypothesize that the tumor inhibitory effects of the dual agonist are indirect, possibly via regulation of the anti-tumor immune response. This hypothesis was supported by flow cytometric analysis of tumor infiltrating leukocyte populations, which demonstrated a significant increase in T lymphocytes in mice treated with the C3aR/C5aR1 agonist. These results support an immunoregulatory role for complement receptors in primary murine mammary carcinoma models. They also suggest that complement activation peptides can influence the anti-tumor response in different ways depending on the cancer type, the host immune response to the tumor and levels of endogenous complement activation within the tumor microenvironment.

4.
FASEB J ; 33(10): 11060-11071, 2019 10.
Article in English | MEDLINE | ID: mdl-31298935

ABSTRACT

The canonical complement component 5a (C5a) receptor (C5aR) 1 has well-described roles in tumorigenesis but the contribution of the second receptor, C5aR2, is unclear. The present study demonstrates that B16.F0 melanoma cells express mRNA for both C5aR1 and C5aR2 and signal through ERK and p38 MAPKs in response to C5a. Despite this, C5a had no impact on melanoma cell proliferation or migration in vitro. In vivo studies demonstrated that the growth of B16.F0 melanoma tumors was increased in C5aR2-/- mice but reduced in C5aR1-/- mice and wild-type mice treated with a C5aR1 antagonist. Analysis of tumor-infiltrating leukocyte populations showed no significant differences between wild-type and C5aR2-/- mice. Conversely, percentages of myeloid-derived suppressor cells, macrophages, and regulatory T lymphocytes were lower in tumors from C5aR1-/- mice, whereas total (CD3+) T lymphocytes and CD4+ subsets were higher. Analysis of cytokine and chemokine levels also showed plasma IFN-γ was higher and tumor C-C motif chemokine ligand 2 was lower in the absence of C5aR1. The results suggest that C5aR1 signaling supports melanoma growth by promoting infiltration of immunosuppressive leukocyte populations into the tumor microenvironment, whereas C5aR2 has a more restricted but beneficial role in limiting tumor growth. Overall, these data support the potential of C5aR1-inhibitory therapies for melanoma.-Nabizadeh, J. A., Manthey, H. D., Panagides, N., Steyn, F. J., Lee, J. D., Li, X. X., Akhir, F. N. M., Chen, W., Boyle, G. M., Taylor, S. M., Woodruff, T. M., Rolfe, B. E. C5a receptors C5aR1 and C5aR2 mediate opposing pathologies in a mouse model of melanoma.


Subject(s)
Cell Movement , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/genetics , Receptor, Anaphylatoxin C5a/genetics , Animals , Cell Proliferation , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Complement C5a/immunology , Female , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , MAP Kinase Signaling System , Male , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Tumor Microenvironment
5.
Thromb Haemost ; 117(1): 176-187, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27786338

ABSTRACT

Atherosclerosis is considered a chronic inflammatory disease of the vessel wall. Coagulation pathways and immune responses contribute to disease development. The role of coagulation factor XII (FXII) in vascular inflammation, however, remains controversial. We here investigated the function of FXII in atherosclerosis using apolipoprotein E and FXII-deficient (F12-/-Apoe-/-) mice. Compared to F12+/+Apoe-/- controls, atherosclerotic lesion formation was reduced in F12-/-Apoe-/- mice. This was associated with a decrease in serum interleukin (IL)-1ß and IL-12 levels and reduced expression of pro-inflammatory cytokines in the aorta in atherosclerotic F12-/-Apoe-/- mice, as well as diminished Th1-cell differentiation in the aorta, blood, and lymphoid organs. No changes in circulating bradykinin, thrombin-antithrombin-complexes or plasminogen were observed. Mechanistically, activated FXII (FXIIa) was revealed to directly induce bone marrow-derived macrophages to secrete pro-inflammatory cytokines, including tumour necrosis factor-α, IL-1ß, IL-12, and IL-6. Exposure of bone marrow-derived antigen presenting cells to FXIIa similarly induced pro-inflammatory cytokines, and an enhanced capacity to trigger antigen-specific interferon γ-production in CD4+ T cells. Notably, bone-marrow derived macrophages were capable of directly activating FXII. Moreover, the induction of cytokine expression by FXIIa in macrophages occurred independently of FXII protease enzymatic activity and was decreased upon phospholipase C treatment, suggesting urokinase-type plasminogen activator receptor (uPAR) to confer FXIIa-induced cell signalling. These data reveal FXII to play an important role in atherosclerotic lesion formation by functioning as a strong inducer of pro-inflammatory cytokines in antigen-presenting cells. Targeting of FXII may thus be a promising approach for treating cardiovascular disease.


Subject(s)
Antigen-Presenting Cells/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Cytokines/metabolism , Factor XII Deficiency/metabolism , Factor XII/metabolism , Inflammation Mediators/metabolism , Macrophages/metabolism , Animals , Antigen-Presenting Cells/immunology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/immunology , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/immunology , Cell Proliferation , Cytokines/immunology , Disease Models, Animal , Factor XII/genetics , Factor XII Deficiency/blood , Factor XII Deficiency/genetics , Factor XII Deficiency/immunology , Factor XIIa/genetics , Factor XIIa/metabolism , Genetic Predisposition to Disease , Inflammation Mediators/immunology , Lymphocyte Activation , Macrophages/immunology , Mice, Inbred C57BL , Mice, Knockout, ApoE , Phenotype , Plaque, Atherosclerotic , Receptors, Urokinase Plasminogen Activator/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Time Factors
6.
J Immunol ; 196(11): 4783-92, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27183625

ABSTRACT

The complement peptide C3a is a key component of the innate immune system and a major fragment produced following complement activation. We used a murine model of melanoma (B16-F0) to identify a hitherto unknown role for C3a-C3aR signaling in promoting tumor growth. The results show that the development and growth of B16-F0 melanomas is retarded in mice lacking C3aR, whereas growth of established melanomas can be arrested by C3aR antagonism. Flow cytometric analysis showed alterations in tumor-infiltrating leukocytes in the absence of C3aR. Specifically, neutrophils and CD4(+) T lymphocyte subpopulations were increased, whereas macrophages were reduced. The central role of neutrophils was confirmed by depletion experiments that reversed the tumor inhibitory effects observed in C3aR-deficient mice and returned tumor-infiltrating CD4(+) T cells to control levels. Analysis of the tumor microenvironment showed upregulation of inflammatory genes that may contribute to the enhanced antitumor response observed in C3aR-deficient mice. C3aR deficiency/inhibition was also protective in murine models of BRAF(V600E) mutant melanoma and colon and breast cancer, suggesting a tumor-promoting role for C3aR signaling in a range of tumor types. We propose that C3aR activation alters the tumor inflammatory milieu, thereby promoting tumor growth. Therapeutic inhibition of C3aR may therefore be an effective means to trigger an antitumor response in melanoma and other cancers.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Carcinogenesis/immunology , Melanoma/immunology , Melanoma/pathology , Neutrophils/immunology , Receptors, Complement/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , Cells, Cultured , Female , Melanoma/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Neutrophils/pathology , Receptors, Complement/deficiency
7.
Arterioscler Thromb Vasc Biol ; 35(11): 2316-25, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26404487

ABSTRACT

OBJECTIVE: Although immune responses drive the pathogenesis of atherosclerosis, mechanisms that control antigen-presenting cell (APC)-mediated immune activation in atherosclerosis remain elusive. We here investigated the function of hypoxia-inducible factor (HIF)-1α in APCs in atherosclerosis. APPROACH AND RESULTS: We found upregulated HIF1α expression in CD11c(+) APCs within atherosclerotic plaques of low-density lipoprotein receptor-deficient (Ldlr(-/-)) mice. Conditional deletion of Hif1a in CD11c(+) APCs in high-fat diet-fed Ldlr(-/-) mice accelerated atherosclerotic plaque formation and increased lesional T-cell infiltrates, revealing a protective role of this transcription factor. HIF1α directly controls Signal Transducers and Activators of Transcription 3 (Stat3), and a reduced STAT3 expression was found in HIF1α-deficient APCs and aortic tissue, together with an upregulated interleukin-12 expression and expansion of type 1 T-helper (Th1) cells. Overexpression of STAT3 in Hif1a-deficient APCs in bone marrow reversed enhanced atherosclerotic lesion formation and reduced Th1 cell expansion in chimeric Ldlr(-/-) mice. Notably, deletion of Hif1a in LysM(+) bone marrow cells in Ldlr(-/-) mice did not affect lesion formation or T-cell activation. In human atherosclerotic lesions, HIF1α, STAT3, and interleukin-12 protein were found to colocalize with APCs. CONCLUSIONS: Our findings identify HIF1α to antagonize APC activation and Th1 T cell polarization during atherogenesis in Ldlr(-/-) mice and to attenuate the progression of atherosclerosis. These data substantiate the critical role of APCs in controlling immune mechanisms that drive atherosclerotic lesion development.


Subject(s)
Antigen-Presenting Cells/metabolism , Aorta/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Antigen-Presenting Cells/immunology , Aorta/immunology , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , CD11c Antigen/genetics , CD11c Antigen/metabolism , Carotid Artery Diseases/metabolism , Cells, Cultured , Coculture Techniques , Diet, High-Fat , Disease Models, Animal , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-12/metabolism , Lymphocyte Activation , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes, Helper-Inducer/immunology
8.
Thromb Haemost ; 110(6): 1267-77, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24114205

ABSTRACT

The chemokine receptor CCR6 is expressed by various cell subsets implicated in atherogenesis, such as monocytes, Th17 and regulatory T cells. In order to further define the role of CCR6 in atherosclerosis, CCR6-deficient (Ccr6-/-) mice were crossed with low-density lipoprotein receptor-deficient (Ldlr-/-) mice to generate atherosclerosis-prone mice deficient in CCR6. Compared to Ldlr-/- controls, atherosclerotic burden in the aortic sinus and aorta were reduced in Ccr6-/-Ldlr-/- mice fed a high fat diet, associated with a profound depression in lesional macrophage accumulation. Local and systemic distributions of T cells, including frequencies of Th1, Th17 and regulatory T cells were unaltered. In contrast, circulating counts of both Gr-1(high) and Gr1(low) monocytes were reduced in Ccr6-/-Ldlr-/- mice. Moreover, CCR6 was revealed to promote monocyte adhesion to inflamed endothelium in vitro and leukocyte adhesion to carotid arteries in vivo. Finally, CCR6 selectively recruited monocytes but not T cells in an acute inflammatory air pouch model. We here show that CCR6 functions on multiple levels and regulates the mobilisation, adhesion and recruitment of monocytes/macrophages to the inflamed vessel, thereby promoting atherosclerosis, but is dispensable for hypercholesterolaemia-associated adaptive immune priming. Targeting CCR6 or its ligand CCL20 may therefore be a promising therapeutic strategy to alleviate atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Macrophages/immunology , Monocytes/immunology , Receptors, CCR6/metabolism , T-Lymphocytes/immunology , Animals , Cell Adhesion/genetics , Cell Movement/genetics , Cells, Cultured , Diet, High-Fat , Disease Models, Animal , Disease Susceptibility , Inflammation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR6/genetics , Receptors, CCR6/immunology , Receptors, Cell Surface/metabolism , Receptors, LDL/genetics
9.
Int J Cardiol ; 167(4): 1282-8, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-22525349

ABSTRACT

BACKGROUND: The molecular mechanisms of exercise-induced cardioprotection are poorly understood. We recently reported that exercise training down-regulated gene expression of the Ras homolog gene family member A (RhoA). RhoA and its first effectors, the Rho-kinases (ROCK), have already been implicated in the pathogenesis of cardiovascular disease. The aim of this study was to compare the effects of a RhoA/ROCK inhibitor (fasudil) and exercise in the Apolipoprotein E knockout (ApoE(-/-)) mouse model of atherosclerosis. METHODS: Four groups of 14 week old ApoE(-/-) mice were randomised as follows (n=12/group): i) sedentary controls (Cont); ii) fasudil (Fas) treatment (100mg/kg bodyweight/day) for 8 weeks; iii) exercise intervention (Ex:free access to running wheel for 8 weeks) and iv) exercise intervention and fasudil treatment (ExFas) for 8 weeks. RESULTS: Phosphorylation of myosin light chain was significantly reduced in the brachiocephalic artery of all treatment groups compared with sedentary controls, implying an inhibitory effect of exercise and fasudil on the RhoA/ROCK pathway. Furthermore, atherosclerotic lesions were significantly smaller in all treatment and intervention groups compared with the control group (Fas: 34.7%, Ex: 48.3%, ExFas: 40.9% less than Control). The intima:media ratio was reduced by both exercise intervention and fasudil treatment alone or in combination (Fas: 23.6%, Ex: 35.5%, ExFas: 43.9% less than Control). Exercise alone and fasudil treatment alone also showed similar effects on plaque composition, increasing both smooth muscle cell and macrophage density. CONCLUSION: These results suggest that the protective effects of exercise on atherogenesis are similar to the inhibitory effects on the RhoA/ROCK signalling pathway.


Subject(s)
Physical Conditioning, Animal/methods , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/enzymology , Protein Kinase Inhibitors/therapeutic use , rho GTP-Binding Proteins/antagonists & inhibitors , rho-Associated Kinases/antagonists & inhibitors , Animals , Apolipoproteins E , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Physical Conditioning, Animal/physiology , Plaque, Atherosclerotic/prevention & control , Protein Kinase Inhibitors/pharmacology , Random Allocation , Treatment Outcome , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein
10.
Arterioscler Thromb Vasc Biol ; 32(10): 2350-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22879583

ABSTRACT

OBJECTIVE: Noninvasive imaging of atherosclerosis remains challenging in clinical applications. Here, we applied noninvasive molecular imaging to detect vascular cell adhesion molecule-1 in early and advanced atherosclerotic lesions of apolipoprotein E-deficient mice. METHODS AND RESULTS: Ultrasmall superparamagnetic iron oxide particles functionalized with (P03011) or without (P3007) vascular cell adhesion molecule-1-binding peptide were visualized by ultra high-field (17.6 T) magnetic resonance. Injection of P03011 resulted in a marked signal loss in the aortic root of apolipoprotein E-deficient mice fed a Western diet for 8 and 26 weeks in vivo and ex vivo, compared with preinjection measurements, P3007-injected mice, and P03011- or P3007-injected age-matched C57BL/6 controls. Histological analyses revealed iron accumulations in the intima, in colocalization with vascular cell adhesion molecule-1-expressing macrophages and endothelial cells. Coherent anti-Stokes Raman scattering microscopy demonstrated iron signals in the intima and media of the aortic root in the P03011-injected but not untreated apolipoprotein E-deficient mice, localized to macrophages, luminal endothelial-like cells, and medial regions containing smooth muscle cells. Electron microscopy confirmed iron particles enclosed in endothelial cells and in the vicinity of smooth muscle cells. CONCLUSIONS: Using a combination of innovative imaging modalities, in this study, we demonstrate the feasibility of applying P03011 as a contrast agent for imaging of atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/pathology , Ferric Compounds/metabolism , Nanoparticles , Vascular Cell Adhesion Molecule-1/metabolism , Vasculitis/metabolism , Vasculitis/pathology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Disease Models, Animal , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Macrophages/metabolism , Macrophages/pathology , Magnetic Resonance Imaging/methods , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Spectrum Analysis, Raman , Tunica Intima/metabolism , Tunica Intima/pathology
11.
Circulation ; 125(13): 1673-83, 2012 Apr 03.
Article in English | MEDLINE | ID: mdl-22388324

ABSTRACT

BACKGROUND: Inflammation has been closely linked to auto-immunogenic processes in atherosclerosis. Plasmacytoid dendritic cells (pDCs) are specialized to produce type-I interferons in response to pathogenic single-stranded nucleic acids, but can also sense self-DNA released from dying cells or in neutrophil extracellular traps complexed to the antimicrobial peptide Cramp/LL37 in autoimmune disease. However, the exact role of pDCs in atherosclerosis remains elusive. METHODS AND RESULTS: Here we demonstrate that pDCs can be detected in murine and human atherosclerotic lesions. Exposure to oxidatively modified low-density lipoprotein enhanced the capacity of pDCs to phagocytose and prime antigen-specific T cell responses. Plasmacytoid DCs can be stimulated to produce interferon-α by Cramp/DNA complexes, and we further identified increased expression of Cramp and formation of neutrophil extracellular traps in atherosclerotic arteries. Whereas Cramp/DNA complexes aggravated atherosclerotic lesion formation in apolipoprotein E-deficient mice, pDC depletion and Cramp-deficiency in bone marrow reduced atherosclerosis and anti-double-stranded DNA antibody titers. Moreover, the specific activation of pDCs and interferon-α treatment promoted plaque growth, associated with enhanced anti-double-stranded-DNA antibody titers. Accordingly, anti-double-stranded DNA antibodies were elevated in patients with symptomatic versus asymptomatic carotid artery stenosis. CONCLUSIONS: Self-DNA (eg, released from dying cells or in neutrophil extracellular traps) and an increased expression of the antimicrobial peptide Cramp/LL37 in atherosclerotic lesions may thus stimulate a pDC-driven pathway of autoimmune activation and the generation of anti-double-stranded-DNA antibodies, critically aggravating atherosclerosis lesion formation. These key factors may thus represent novel therapeutic targets.


Subject(s)
Atherosclerosis/immunology , Autoantigens/immunology , DNA/immunology , Dendritic Cells/immunology , Proteins/genetics , Proteins/immunology , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Autoantigens/genetics , Carotid Stenosis/genetics , Carotid Stenosis/immunology , Carotid Stenosis/metabolism , DNA/genetics , Dendritic Cells/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout
12.
FASEB J ; 25(7): 2447-55, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21490292

ABSTRACT

The complement C5a receptor, CD88, is present on many of the cells found within human atherosclerotic plaques, but little is known about the role of C5a in atherogenesis. Using real-time PCR, we determined that ApoE(-/-) mice fed a normal diet express more aortic CD88 mRNA compared with controls, and this increase coincides with atherosclerotic lesion development (P<0.001 for 3- vs. 25-wk-old animals). Conversely, mRNA expression of the alternative C5a receptor, C5L2, in aortas of ApoE(-/-) mice, was lower than controls at all time points. Using immunohistochemistry, we confirmed the presence of CD88 on macrophages, smooth muscle cells, and activated endothelial cells in plaques from brachiocephalic arteries. Treatment of ApoE(-/-) mice with a CD88 antagonist (PMX53; 3 mg/kg s.c. 3 ×/wk plus 1 mg/kg/d p.o.) for 25 wk reduced lesion size and lipid content in the plaque by ∼ 40% (P<0.05). Our study provides evidence for a proatherogenic role for C5a and identifies the CD88 antagonist PMX53 as a potential antiatherosclerotic drug.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Complement C5a/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Brachiocephalic Trunk/drug effects , Brachiocephalic Trunk/metabolism , Brachiocephalic Trunk/pathology , Cholesterol/blood , Complement C5a/antagonists & inhibitors , Endothelial Cells/metabolism , Female , Immunohistochemistry , Lipids/analysis , Lipids/blood , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Peptides, Cyclic/pharmacology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/genetics , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Reverse Transcriptase Polymerase Chain Reaction
13.
Int J Biochem Cell Biol ; 41(11): 2114-7, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19464229

ABSTRACT

The 74 amino acid glycoprotein, complement component 5a (C5a), is a potent pro-inflammatory mediator cleaved enzymatically from its precursor, C5, upon activation of the complement cascade. C5a is quickly metabolised by carboxypeptidases, forming the less potent C5adesArg. Acting via a classical G protein-coupled receptor, CD88, C5a and C5adesArg exert a number of effects essential to the innate immune response, while their actions at the more recently discovered non-G protein-coupled receptor, C5L2 (or GPR77), remain unclear. The widespread expression of C5a receptors throughout the body allows C5a to elicit a broad range of effects. Thus, C5a has been found to be a significant pathogenic driver in a number of immuno-inflammatory diseases, making C5a inhibition an attractive therapeutic strategy.


Subject(s)
Complement C5a/immunology , Amino Acid Sequence , Animals , Complement C5a/chemistry , Complement C5a/metabolism , Humans , Molecular Sequence Data
14.
J Pharmacol Exp Ther ; 314(2): 811-7, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15879003

ABSTRACT

We have previously shown that complement factor 5a (C5a) plays a role in the pathogenesis of 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis in rats by using the selective, orally active C5a antagonist AcF-[OP(d-Cha)WR]. This study tested the efficacy and potency of a new C5a antagonist, hydrocinnamate (HC)-[OP(d-Cha)WR], which has limited intestinal lumenal metabolism, in this model of colitis. Analogs of AcF-[OP(d-Cha)WR] were examined for their susceptibility to alimentary metabolism in the rat using intestinal mucosal washings. One metabolically stable analog, HC-[OP(d-Cha)WR], was then evaluated pharmacokinetically and investigated at a range of doses (0.03-10 mg/kg/day p.o.) in the 8-day rat TNBS-colitis model, against the comparator drug AcF-[OP(d-Cha)WR]. Using various amino acid substitutions, it was determined that the AcF moiety of AcF-[OP(d-Cha)WR] was responsible for the metabolic instability of the compound in intestinal mucosal washings. The analog HC-[OP(d-Cha)WR], equiactive in vitro to AcF-[OP(d-Cha)WR], was resistant to intestinal metabolism, but it displayed similar oral bioavailability to AcF-[OP(d-Cha)WR]. However, in the rat TNBS-colitis model, HC-[OP(d-Cha)WR] was effective at reducing mortality, colon edema, colon macroscopic scores, and increasing food consumption and body weights, at 10- to 30-fold lower oral doses than AcF-[OP(d-Cha)WR]. These studies suggest that resistance to intestinal metabolism by HC-[OP(d-Cha)WR] may result in increased local concentrations of the drug in the colon, thus affording efficacy with markedly lower oral doses than AcF-[OP(d-Cha)WR] against TNBS-colitis. This large increase in potency and high efficacy of this compound makes it a potential candidate for clinical development against intestinal diseases such as inflammatory bowel disease.


Subject(s)
Complement C5a/metabolism , Inflammatory Bowel Diseases/drug therapy , Peptides, Cyclic/therapeutic use , Receptors, Complement/antagonists & inhibitors , Animals , Biological Availability , Biotransformation , Digestive System/metabolism , Eating/drug effects , Edema/chemically induced , Edema/pathology , Edema/prevention & control , Female , Humans , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Peptides, Cyclic/pharmacokinetics , Peptides, Cyclic/pharmacology , Rats , Rats, Wistar , Trinitrobenzenesulfonic Acid , Ulcer/chemically induced , Ulcer/pathology , Ulcer/prevention & control , Weight Loss
SELECTION OF CITATIONS
SEARCH DETAIL
...