Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Front Physiol ; 11: 673, 2020.
Article in English | MEDLINE | ID: mdl-32625118

ABSTRACT

Atherogenesis and arterial remodeling following mechanical injury are driven by inflammation and mononuclear cell infiltration. The binding of immune complexes (ICs) to immunoglobulin (Ig)-Fc gamma receptors (FcγRs) on most innate and adaptive immune cells induces a variety of inflammatory responses that promote atherogenesis. Here, we studied the role of FcγRIII in neointima formation after arterial injury in atherosclerosis-prone mice and compared the outcome and mechanism to that of FcγRIII in diet-induced "chronic" atherosclerosis. FcγrIII-/-/Apoe-/- and control Apoe-/- mice were subjected to wire-induced endothelial denudation of the carotid artery while on high-fat diet (HFD). FcγrIII deficiency mitigated neointimal plaque formation and lesional macrophage accumulation, and enhanced neointimal vascular smooth muscle cell (VSMC) numbers. This went along with a reduced expression of tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein-1 (MCP-1/CCL2), and vascular cell adhesion molecule-1 (VCAM-1) in the neointimal lesions. Interestingly, in a chronic model of diet-induced atherosclerosis, we unraveled a dichotomic role of FcγRIII in an early versus advanced stage of the disease. While FcγrIII deficiency conferred atheroprotection in the early stage, it promoted atherosclerosis in advanced stages. To this end, FcγrIII deficiency attenuated pro-inflammatory responses in early atherosclerosis but promoted these events in advanced stages. Analysis of the mechanism(s) underlying the athero-promoting effect of FcγrIII deficiency in late-stage atherosclerosis revealed increased serum levels of anti-oxidized-LDL immunoglobulins IgG2c and IgG2b. This was paralleled by enhanced lesional accumulation of IgGs without affecting levels of complement-activated products C5a or C5ar1, FcγRII, and FcγRIV. Moreover, FcγrIII-deficient macrophages expressed more FcγrII, Tnf-α, and Il-1ß mRNA when exposed to IgG1 or oxLDL-IgG1 ICs in vitro, and peripheral CD4+ and CD8+ T-cell levels were altered. Collectively, our data suggest that deficiency of activating FcγRIII limits neointima formation after arterial injury in atherosclerosis-prone mice as well as early stage chronic atherosclerosis, but augments late-stage atherosclerosis suggesting a dual role of FcγRIII in atherogenic inflammation.

2.
Am J Physiol Renal Physiol ; 314(1): F35-F46, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28903945

ABSTRACT

Complement factor C5a has two known receptors, C5aR, which mediates proinflammatory effects, and C5L2, a potential C5a decoy receptor. We previously identified C5a/C5aR signaling as a potent profibrotic pathway in the kidney. Here we tested for the first time the role of C5L2 in renal fibrosis. In unilateral ureteral obstruction (UUO)-induced kidney fibrosis, the expression of C5aR and C5L2 increased similarly and gradually as fibrosis progressed and was particularly prominent in injured dilated tubules. Genetic deficiency of either C5aR or C5L2 significantly reduced UUO-induced tubular injury. Expression of key proinflammatory mediators, however, significantly increased in C5L2- compared with C5aR-deficient mice, but this had no effect on the number of renal infiltrating macrophages or T cells. Moreover, in C5L2-/- mice, the cytokine and matrix metalloproteinase-inhibitor tissue inhibitor of matrix metalloproteinase-1 was specifically enhanced. Consequently, in C5L2-/- mice the degree of renal fibrosis was similar to wild type (WT), albeit with reduced mRNA expression of some fibrosis-related genes. In contrast, C5aR-/- mice had significantly reduced renal fibrosis compared with WT and C5L2-/- mice in UUO. In vitro experiments with primary tubular cells demonstrated that deficiency for either C5aR or C5L2 led to a significantly reduced expression of tubular injury and fibrosis markers. Vice versa, stimulation of WT tubular cells with C5a significantly induced the expression of these markers, whereas the absence of either receptor abolished this induction. In conclusion, in experimental renal fibrosis C5L2 and C5aR both contribute to tubular injury, and, while C5aR acts profibrotic, C5L2 does not play a role in extracellular matrix accumulation, arguing against C5L2 functioning simply as a decoy receptor.


Subject(s)
Complement C5a/metabolism , Fibrosis/immunology , Kidney Diseases/immunology , Receptors, Chemokine/metabolism , Animals , Complement C5a/immunology , Fibrosis/genetics , Kidney/immunology , Kidney/pathology , Kidney Diseases/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Male , Mice , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology
3.
Thromb Haemost ; 114(4): 848-58, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26084965

ABSTRACT

Atherogenic processes and vascular remodelling after arterial injury are controlled and driven by a plethora of factors amongst which the activation of the complement system is pivotal. Recently, we reported a clear correlation between high expressions of the second receptor for complement anaphylatoxin C5a, the C5a receptor-like 2 (C5L2, C5aR2), with high pro-inflammatory cytokine expression in advanced human atherosclerotic plaques. This prompted us to speculate that C5aR2 might have a functional role in atherosclerosis. We, therefore, investigated the role of C5aR2 in atherosclerosis and vascular remodelling. Here, we demonstrate that C5ar2 deletion, in atherosclerosis-prone mice, attenuates atherosclerotic as well as neointimal plaque formation, reduces macrophages and CD3+ T cells and induces features of plaque stability, as analysed by histomorphometry and quantitative immunohistochemistry. As a possible underlying mechanism, C5ar2-deficient plaques showed significantly reduced expression of C5a receptor (C5ar1), Tnf-α as well as Vcam-1, as determined by qPCR and quantitative immunohistochemistry. In addition, in vitro mechanistic studies revealed a reduction of these pro-inflammatory and pro-atherosclerotic mediators in C5ar2-deficient macrophages. Finally, blocking C5ar1 with antagonist JPE1375, in C5ar2(-/-)/Apoe(-/-) mice, led to a further reduction in neointimal plaque formation with reduced inflammation. In conclusion, C5ar2 deficiency attenuates atherosclerosis and neointimal plaque formation after arterial injury. This identifies C5aR2 as a promising target to reduce atherosclerosis and restenosis after vascular interventions.


Subject(s)
Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Carotid Artery Injuries/prevention & control , Receptor, Anaphylatoxin C5a/deficiency , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , CD3 Complex/metabolism , Carotid Artery Injuries/genetics , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Carotid Artery, Common/metabolism , Carotid Artery, Common/pathology , Cells, Cultured , Disease Models, Animal , Genetic Predisposition to Disease , Inflammation Mediators/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Neointima , Phenotype , Plaque, Atherosclerotic , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/metabolism , Rupture, Spontaneous , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Remodeling
4.
Am J Pathol ; 184(7): 2123-33, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24819959

ABSTRACT

The complement anaphylatoxin C5a functions through its two receptors, C5aR (CD88) and C5a receptor-like 2 (C5L2). Their role in atherosclerosis is incompletely understood. We, therefore, analyzed C5aR and probed the yet unknown expression and function of C5L2 in human atherogenesis. Human atherosclerotic plaques obtained by endarterectomy were staged and analyzed for C5L2 and C5aR by IHC and quantitative real-time PCR. C5L2-expressing cells in plaques were mostly macrophages, less neutrophils and endothelial cells, as determined by double immunostaining. Although early influx of C5aR(+) cells was detected, C5L2 levels increased with lesion complexity and colocalized with C5aR and oxidized low-density lipoprotein. Gene expression of C5L2 and C5aR showed similar trends, such as the receptor-expressing cells. The expression of C5L2 in advanced lesions correlated with increased levels of IL-1ß and tumor necrosis factor-α in plaques. Furthermore, in vitro experiments in macrophages from wild-type and C5l2- and C5ar-deficient mice corroborated the contributing role of C5l2 in oxidized low-density lipoprotein-pretreated C5a-induced cytokine expression, as measured by enzyme-linked immunosorbent assay. Finally, C5l2- and C5ar-deficient peripheral blood mononuclear cells showed less arrest on tumor necrosis factor-α-stimulated mouse endothelial cells in vitro when compared with wild-type controls. Taken together, prominent C5L2 expression in advanced atherosclerotic stages directly correlates with high levels of proinflammatory cytokines. This might indicate a proinflammatory role of C5L2 in atherosclerosis that needs to be pursued in the future by applying in vivo mouse models.


Subject(s)
Cytokines/metabolism , Plaque, Atherosclerotic/metabolism , Receptors, Chemokine/metabolism , Animals , Carotid Arteries/pathology , Humans , Interleukin-1beta/metabolism , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Receptor, Anaphylatoxin C5a/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
Arterioscler Thromb Vasc Biol ; 34(6): 1209-20, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24723559

ABSTRACT

OBJECTIVE: The Cxcl12/Cxcr4 chemokine ligand/receptor axis mediates the mobilization of smooth muscle cell progenitors, driving injury-induced neointimal hyperplasia. This study aimed to investigate the role of endothelial Cxcr4 in neointima formation. APPROACH AND RESULTS: ß-Galactosidase staining using bone marrow x kinase (Bmx)-CreER(T2) reporter mice and double immunofluorescence revealed an efficient and endothelial-specific deletion of Cxcr4 in Bmx-CreER(T2+) compared with Bmx-CreER(T2-) Cxcr4-floxed apolipoprotein E-deficient (Apoe(-/-)) mice (referred to as Cxcr4(EC-KO)ApoE(-/-) and Cxcr4(EC-WT) ApoE(-/-), respectively). Endothelial Cxcr4 deficiency significantly increased wire injury-induced neointima formation in carotid arteries from Cxcr4(EC-KO)ApoE(-/-) mice. The lesions displayed a higher number of macrophages, whereas the smooth muscle cell and collagen content were reduced. This was associated with a significant reduction in reendothelialization and endothelial cell proliferation in injured Cxcr4(EC-KO)ApoE(-/-) carotids compared with Cxcr4(EC-WT)ApoE(-/-) controls. Furthermore, stimulation of human aortic endothelial cells with chemokine (C-X-C motif) ligand 12 (CXCL12) significantly enhanced their wound-healing capacity in an in vitro scratch assay, an effect that could be reversed with the CXCR4 antagonist AMD3100. Also, flow cytometric analysis showed a reduced mobilization of Sca1(+)Flk1(+)Cd31(+) and of Lin(-)Sca1(+) progenitors in Cxcr4(EC-KO) ApoE(-/-) mice after vascular injury, although Cxcr4 surface expression was unaltered. No differences could be detected in plasma concentrations of Cxcl12, vascular endothelial growth factor, sphingosine 1-phosphate, or Flt3 (fms-related tyrosine kinase 3) ligand, all cytokines with an established role in progenitor cell mobilization. Nonetheless, double immunofluorescence revealed a significant reduction in local endothelial Cxcl12 staining in injured carotids from Cxcr4(EC-KO)ApoE(-/-) mice. CONCLUSIONS: Endothelial Cxcr4 is crucial for efficient reendothelialization after vascular injury through endothelial wound healing and proliferation, and through the mobilization of Sca1(+)Flk1(+)Cd31(+) cells, often referred to as circulating endothelial progenitor cells.


Subject(s)
Atherosclerosis/pathology , Carotid Artery Injuries/pathology , Endothelial Cells/physiology , Neointima/pathology , Receptors, CXCR4/physiology , Animals , Antigens, Ly/physiology , Apolipoproteins E/physiology , Atherosclerosis/physiopathology , Cell Movement , Chemokine CXCL12/physiology , Female , Hyperplasia , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Protein-Tyrosine Kinases/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology
6.
Thromb Haemost ; 110(1): 141-52, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23636414

ABSTRACT

The COP9 signalosome (CSN), a multifunctional protein complex involved in the regulation of cullin-RING-E3 ubiquitin ligases (CRLs), has emerged as a regulator of NF-κB signalling. As NF-κB drives the expression of pro-inflammatory and pro-atherosclerotic genes, we probed the yet unknown role of the CSN, in particular CSN5, on NF-κB-mediated atherogenic responses in endothelial cells. Co-immunoprecipitation in human umbilical vein endothelial cells (HUVECs) revealed the presence of a super-complex between IKK and CSN, which dissociates upon TNF-α stimulation. Furthermore, CSN5 silencing enhanced TNF-α-induced IκB-α degradation and NF-κB activity in luciferase reporter assays. This was paralleled by an increased NF-κB-driven upregulation of atherogenic chemokines and adhesion molecules, as measured by qPCR and flow cytometry, and translated into an enhanced arrest of THP-1 monocytes on TNF-α-stimulated, CSN5-depleted HUVECs. Reverse effects on NF-κB activity and THP-1 arrest were seen upon CSN5 overexpression. Finally, double-immunostaining confirmed the expression of CSN subunits in the endothelium of human atherosclerotic lesions, and revealed an increased expression of CSN5 which correlated with atheroprogression. In conclusion, endothelial CSN5 attenuates NF-κB-dependent pro-inflammatory gene expression and monocyte arrest on stimulated endothelial cells in vitro, suggesting that CSN5 might serve as a negative regulator of atherogenesis.


Subject(s)
Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Monocytes/metabolism , NF-kappa B/metabolism , Peptide Hydrolases/metabolism , Plaque, Atherosclerotic/metabolism , COP9 Signalosome Complex , Cell Adhesion , Cell Line , Gene Expression Regulation/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Monocytes/immunology , NF-kappa B/genetics , Peptide Hydrolases/genetics , Peptide Hydrolases/immunology , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/immunology , Protein Binding/genetics , RNA, Small Interfering/genetics , Transcriptional Activation/genetics , Transendothelial and Transepithelial Migration , Tumor Necrosis Factor-alpha/immunology
7.
Arterioscler Thromb Vasc Biol ; 32(7): 1613-23, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22556330

ABSTRACT

OBJECTIVE: Inflammatory leukocyte accumulation drives atherosclerosis. Although monocytes/macrophages and polymorphonuclear neutrophilic leukocytes (PMN) contribute to lesion formation, sequelae of myeloproliferative disease remain to be elucidated. METHODS AND RESULTS: We used mice deficient in interferon regulatory factor 8 (IRF8(-/-)) in hematopoietic cells that develop a chronic myelogenous leukemia-like phenotype. Apolipoprotein E-deficient mice reconstituted with IRF8(-/-) or IRF8(-/-) apolipoprotein E-deficient bone marrow displayed an exacerbated atherosclerotic lesion formation compared with controls. The chronic myelogenous leukemia-like phenotype in mice with IRF8(-/-) bone marrow, reflected by an expansion of PMN in the circulation, was associated with an increased lesional accumulation and apoptosis of PMN, and enlarged necrotic cores. IRF8(-/-) compared with IRF8(+/+) PMN displayed unaffected reactive oxygen species formation and discharge of PMN granule components. In contrast, accumulating in equal numbers at sites of inflammation, IRF8(-/-) macrophages were defective in efferocytosis, lipid uptake, and interleukin-10 cytokine production. Importantly, depletion of PMN in low-density lipoprotein receptor or apolipoprotein E-deficient mice with IRF8(-/-) or IRF8(-/-) apolipoprotein E-deficient bone marrow abrogated increased lesion formation. CONCLUSIONS: These findings indicate that a chronic myelogenous leukemia-like phenotype contributes to accelerated atherosclerosis in mice. Among proatherosclerotic effects of other cell types, this, in part, is linked to an expansion of functionally intact PMN.


Subject(s)
Atherosclerosis/etiology , Interferon Regulatory Factors/physiology , Animals , Apolipoproteins E/physiology , Apoptosis , Bone Marrow Transplantation , Capillary Permeability , Female , Interleukin-10/biosynthesis , Macrophages/physiology , Mice , Mice, Inbred C57BL , Neutrophils/physiology , Peroxidase/physiology , Reactive Oxygen Species/metabolism , Receptors, LDL/physiology
8.
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
9.
Sci Signal ; 2(100): ra81, 2009 Dec 08.
Article in English | MEDLINE | ID: mdl-19996457

ABSTRACT

Apoptosis is a pivotal process in embryogenesis and postnatal cell homeostasis and involves the shedding of membranous microvesicles termed apoptotic bodies. In response to tissue damage, the CXC chemokine CXCL12 and its receptor CXCR4 counteract apoptosis and recruit progenitor cells. Here, we show that endothelial cell-derived apoptotic bodies are generated during atherosclerosis and convey paracrine alarm signals to recipient vascular cells that trigger the production of CXCL12. CXCL12 production was mediated by microRNA-126 (miR-126), which was enriched in apoptotic bodies and repressed the function of regulator of G protein (heterotrimeric guanosine triphosphate-binding protein) signaling 16, an inhibitor of G protein-coupled receptor (GPCR) signaling. This enabled CXCR4, a GPCR, to trigger an autoregulatory feedback loop that increased the production of CXCL12. Administration of apoptotic bodies or miR-126 limited atherosclerosis, promoted the incorporation of Sca-1+ progenitor cells, and conferred features of plaque stability on different mouse models of atherosclerosis. This study highlights functions of microRNAs in health and disease that may extend to the recruitment of progenitor cells during other forms of tissue repair or homeostasis.


Subject(s)
Apoptosis/physiology , Chemokine CXCL12/physiology , Endothelium, Vascular/cytology , MicroRNAs/administration & dosage , Animals , Atherosclerosis/pathology , Base Sequence , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , DNA Primers , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Mice , Receptors, CXCR4/metabolism , Reverse Transcriptase Polymerase Chain Reaction
10.
Arterioscler Thromb Vasc Biol ; 29(8): 1161-3, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19520977

ABSTRACT

OBJECTIVE: Although junctional adhesion molecule (JAM)-C has been implicated in the control of inflammatory leukocyte recruitment, its role in neointima formation after arterial injury has not been elucidated. METHODS AND RESULTS: In apolipoprotein E-deficient (Apoe(-/-)) mice fed an atherogenic diet, antibody blockade of JAM-C significantly reduced neointimal hyperplasia after wire injury of carotid arteries without altering medial area and decreased neointimal macrophage but not smooth muscle cell (SMC) content. An increased expression of JAM-C was detected in colocalization with luminal SMCs 1 day after injury and neointimal SMCs after 3 weeks. Blocking JAM-C inhibited monocytic cell arrest and leukocyte adhesion to carotid arteries perfused ex vivo and in vivo. Furthermore, monocyte adhesion to activated coronary artery SMCs under flow conditions in vitro was diminished by blocking JAM-C. CONCLUSIONS: Our data provide the first evidence for a crucial role of JAM-C in accelerated lesion formation and leukocyte recruitment in atherosclerosis-prone mice.


Subject(s)
Atherosclerosis/pathology , Carotid Arteries/pathology , Cell Adhesion Molecules/metabolism , Immunoglobulins/metabolism , Monocytes/pathology , Tunica Intima/pathology , Animals , Atherosclerosis/metabolism , Carotid Arteries/metabolism , Cell Adhesion , Disease Models, Animal , Flow Cytometry , Hyperplasia , Mice , Tunica Intima/metabolism
11.
Blood ; 113(4): 963-72, 2009 Jan 22.
Article in English | MEDLINE | ID: mdl-18971423

ABSTRACT

CX(3)CR1 is a chemokine receptor with a single ligand, the membrane-tethered chemokine CX(3)CL1 (fractalkine). All blood monocytes express CX(3)CR1, but its levels differ between the main 2 subsets, with human CD16(+) and murine Gr1(low) monocytes being CX(3)CR1(hi). Here, we report that absence of either CX(3)CR1 or CX(3)CL1 results in a significant reduction of Gr1(low) blood monocyte levels under both steady-state and inflammatory conditions. Introduction of a Bcl2 transgene restored the wild-type phenotype, suggesting that the CX(3)C axis provides an essential survival signal. Supporting this notion, we show that CX(3)CL1 specifically rescues cultured human monocytes from induced cell death. Human CX(3)CR1 gene polymorphisms are risk factors for atherosclerosis and mice deficient for the CX(3)C receptor or ligand are relatively protected from atherosclerosis development. However, the mechanistic role of CX(3)CR1 in atherogenesis remains unclear. Here, we show that enforced survival of monocytes and plaque-resident phagocytes, including foam cells, restored atherogenesis in CX(3)CR1-deficent mice. The fact that CX(3)CL1-CX(3)CR1 interactions confer an essential survival signal, whose absence leads to increased death of monocytes and/or foam cells, might provide a mechanistic explanation for the role of the CX(3)C chemokine family in atherogenesis.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/pathology , Homeostasis , Monocytes/cytology , Monocytes/metabolism , Receptors, Chemokine/metabolism , Animals , Atherosclerosis/genetics , CX3C Chemokine Receptor 1 , Cell Survival , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics
12.
Arterioscler Thromb Vasc Biol ; 28(11): 1897-908, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18566299

ABSTRACT

The fundamental importance of chemokines for atherogenesis, progression, and destabilization of atherosclerotic plaques is now widely appreciated, but the degree of complexity, specificity, and cooperativity harnessed by these signal molecules to govern atherogenic cell recruitment and homeostasis is still being refined. Since the role of chemokines in atherosclerotic vascular disease has been reviewed in this journal, significant progress has been accomplished in defining the regulation of chemokine expression and function in atherosclerosis. In this update, we will highlight these recent developments, in particular the identification of components regulating the transcriptional machinery of the proatherogenic chemokine CCL5, distinct roles of its receptors CCR1 and CCR5 in plaque formation and immunobalance, and differential site- and stage-specific effects of T cell-activating chemokines and their receptors, eg, CXCL10 and CXCR3. The contribution of the transmembrane chemokines CX(3)CL1 and CXCL16 with their respective receptors CX(3)CR1 and CXCR6 in the recruitment of T cell and monocyte subsets and shear-mediated plaque modulation will be discussed. Finally, the role of CXCR2 and CXCR4, their respective ligands CXCL1 and CXCL12, and the noncanonical dual agonist MIF in atheroprogression will be dissected. The considerable leap in insight over recent years leads us to anticipate further advances in comprehending the role of chemokines in atherosclerosis, allowing targeted interventions for its prevention and therapy.


Subject(s)
Atherosclerosis/immunology , Blood Vessels/immunology , Chemokines/metabolism , Inflammation/immunology , Receptors, Chemokine/metabolism , Animals , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Blood Vessels/pathology , Chemotaxis, Leukocyte , Humans , Inflammation/pathology , Inflammation/prevention & control , Ligands , Macrophage Migration-Inhibitory Factors/metabolism , Monocytes/immunology , T-Lymphocytes/immunology
13.
Circulation ; 117(20): 2645-56, 2008 May 20.
Article in English | MEDLINE | ID: mdl-18474814

ABSTRACT

BACKGROUND: Angiotensin (Ang) II-induced target-organ damage involves innate and acquired immunity. Mice deficient for the helix-loop-helix transcription factor inhibitor of differentiation (Id2(-/-)) lack Langerhans and splenic CD8a+ dendritic cells, have reduced natural killer cells, and have altered CD8 T-cell memory. We tested the hypothesis that an alteration in the number and quality of circulating blood cells caused by Id2 deletion would ameliorate Ang II-induced target-organ damage. METHODS AND RESULTS: We used gene-deleted and transgenic mice. We conducted kidney and bone marrow transplants. In contrast to Ang II-infused Id2(+/-), Id2(-/-) mice infused with Ang II remained normotensive and failed to develop albuminuria or renal damage. Bone marrow transplant of Id2(+/-) bone marrow to Id2(-/-) mice did not restore the blunted blood pressure response to Ang II. Transplantation of Id2(-/-) kidneys to Id2(+/-) mice also could not prevent Ang II-induced hypertension and renal damage. We verified the Ang II resistance in Id2(-/-) mice in a model of local tissue Ang II production by crossing hypertensive mice transgenic for rat angiotensinogen with Id2(-/-) or Id2(+/-) mice. Angiotensinogen-transgenic Id2(+/-) mice developed hypertension, albuminuria, and renal injury, whereas angiotensinogen-transgenic Id2(-/-) mice did not. We also found that vascular smooth muscle cells from Id2(-/-) mice showed an antisenescence phenotype. CONCLUSIONS: Our bone marrow and kidney transplant experiments suggest that alterations in circulating immune cells or Id2 in the kidney are not responsible for Ang II resistance. The present studies identify a previously undefined role for Id2 in the pathogenesis of Ang II-induced hypertension.


Subject(s)
Angiotensin II/pharmacology , Hypertension/etiology , Inhibitor of Differentiation Protein 2/physiology , Animals , Blood Cells/immunology , Bone Marrow Transplantation , Hypertension/chemically induced , Immune System/cytology , Inhibitor of Differentiation Protein 2/deficiency , Kidney Transplantation , Mice , Mice, Knockout , Mice, Transgenic
14.
Circulation ; 117(1): 70-8, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18071075

ABSTRACT

BACKGROUND: Although activation of the complement system has been implicated in the progression of human atherosclerosis, its function during arterial remodeling after injury has not been investigated. Here, we examined the contribution of the complement cascade to neointima formation in apolipoprotein E-deficient mice using a C1-esterase inhibitor (C1-inhibitor). METHODS AND RESULTS: Apolipoprotein E-deficient mice fed an atherogenic diet were subjected to wire-induced endothelial denudation of the carotid artery and treated with C1-inhibitor (Berinert; 15 IU i.v.) or vehicle perioperatively and subsequently every 2 days. The effectiveness of C1-inhibitor treatment was confirmed by measurement of plasma C1-inhibitor activity. A significant reduction in serum triglyceride levels was observed in C1-inhibitor-treated mice, whereas cholesterol levels did not differ. After 3 weeks, neointimal area was significantly reduced in C1-inhibitor-treated mice versus controls, whereas medial area was unaltered. This was associated with a significant decrease in neointimal and medial macrophage and CD3+ T-cell content. Expression of C3 mRNA was significantly reduced in plaques of C1-inhibitor-treated mice 10 days after injury, as assessed by reverse-transcription polymerase chain reaction. The peak in serum C3 levels after injury was markedly downregulated by C1-inhibitor, as evidenced by ELISA. Immunohistochemistry revealed strong expression of C3 and C3c, which colocalized to plaque macrophages and was reduced in C1-inhibitor-treated mice. C1-inhibitor impaired monocyte arrest on activated endothelium and platelets under flow conditions in vitro and leukocyte recruitment to carotid arteries 1 day after injury in vivo. CONCLUSIONS: C1-inhibitor limits neointimal plaque formation and inflammation. This may involve blockade of complement activation, inhibition of leukocyte recruitment, and reduced triglyceride levels, thus providing a multimodal approach to treat arterial disease.


Subject(s)
Arteries/injuries , Atherosclerosis/prevention & control , Complement C1 Inhibitor Protein/pharmacology , Regeneration/drug effects , Tunica Intima/pathology , Animals , Apolipoproteins E/deficiency , Arteries/pathology , Atherosclerosis/etiology , Chemotaxis, Leukocyte/drug effects , Complement Activation/drug effects , Disease Models, Animal , Endothelium, Vascular/injuries , Endothelium, Vascular/pathology , Inflammation/prevention & control , Mice , Triglycerides/blood
15.
Circ Res ; 102(2): 209-17, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17991882

ABSTRACT

The CXC ligand (CXCL)12/CXC receptor (CXCR)4 chemokine-receptor axis controls hematopoiesis, organ development, and angiogenesis, but its role in the inflammatory pathogenesis of atherosclerosis is unknown. Here we show that interference with Cxcl12/Cxcr4 by a small-molecule antagonist, genetic Cxcr4 deficiency, or lentiviral transduction with Cxcr4 degrakine in bone marrow chimeras aggravated diet-induced atherosclerosis in apolipoprotein E-deficient (Apoe-/-) or LDL receptor-deficient (Ldlr-/-) mice. Chronic blockade of Cxcr4 caused leukocytosis and an expansion of neutrophils and increased neutrophil content in plaques, associated with apoptosis and a proinflammatory phenotype. Whereas circulating neutrophils were recruited to atherosclerotic lesions, depletion of neutrophils reduced plaque formation and prevented its exacerbation after blocking Cxcr4. Disrupting Cxcl12/Cxcr4 thus promotes lesion formation through deranged neutrophil homeostasis, indicating that Cxcl12/Cxcr4 controls the important contribution of neutrophils to atherogenesis in mice.


Subject(s)
Atherosclerosis/etiology , Chemokine CXCL12/physiology , Neutrophils/pathology , Receptors, CXCR4/deficiency , Receptors, CXCR4/physiology , Animals , Apolipoproteins E/deficiency , Apoptosis , Atherosclerosis/pathology , Atherosclerosis/therapy , Cell Proliferation , Chemotaxis, Leukocyte , Diet , Inflammation/etiology , Leukocytosis/etiology , Mice , Mice, Knockout , Neutropenia , Receptors, CXCR4/antagonists & inhibitors , Receptors, LDL/deficiency
16.
Circulation ; 116(16): 1812-20, 2007 Oct 16.
Article in English | MEDLINE | ID: mdl-17893273

ABSTRACT

BACKGROUND: The CC chemokine CCL5/Regulated on Activation, Normal T Cell Expressed and Secreted (RANTES) is upregulated in mononuclear cells or deposited by activated platelets during inflammation and has been implicated in atherosclerosis and neointimal hyperplasia. We investigated the influence of the transcriptional regulator Y-box binding protein (YB)-1 on CCL5 expression and wire-induced neointimal hyperplasia. METHODS AND RESULTS: Analysis of the CCL5 promoter revealed potential binding sites for YB-1, and interaction of YB-1 with a sequence at position -204/-173 was confirmed by DNA binding assays. Both YB-1 expression and CC chemokine ligand-5 (CCL5) mRNA expression were increased in neointimal versus medial smooth muscle cells, as analyzed by real-time polymerase chain reaction. Overexpression of YB-1 in smooth muscle cells (but not macrophages) enhanced CCL5 transcriptional activity in reporter assays, mRNA and protein expression, and CCL5-mediated monocyte arrest. Carotid arteries of hyperlipidemic apolipoprotein E-deficient mice were subjected to intraluminal transfection with a lentivirus encoding YB-1 short hairpin RNA or empty vector directly after wire injury. Double immunofluorescence revealed YB-1 expression in neointimal smooth muscle cells but not macrophages and colocalization with neointimal CCL5, which was downregulated by YB-1 short hairpin RNA. Neointima formation was decreased significantly after YB-1 knockdown compared with controls and was associated with a diminished content of lesional macrophages. A reduction of lesion formation by YB-1 knockdown was not observed in apolipoprotein E-deficient mice deficient in the CCL5 receptor CCR5 or after treatment with the CCL5 antagonist Met-RANTES, which indicates that YB-1 effects were dependent on CCL5. CONCLUSIONS: The transcriptional regulator YB-1 mediates CCL5 expression in smooth muscle cells and thereby contributes to neointimal hyperplasia, thus representing a novel target with which to limit vascular remodeling.


Subject(s)
Atherosclerosis/pathology , Atherosclerosis/physiopathology , Chemokine CCL5/genetics , Muscle, Smooth, Vascular/pathology , Y-Box-Binding Protein 1/metabolism , Animals , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Cell Line , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/metabolism , Chemokine CCL5/pharmacology , Coronary Vessels/cytology , Macrophages/cytology , Macrophages/physiology , Mice , Mice, Knockout , Monocytes/cytology , Monocytes/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Promoter Regions, Genetic/physiology , Rats , Rats, Sprague-Dawley , Thoracic Arteries/cytology , Transcription, Genetic/physiology , Tunica Intima/pathology , Y-Box-Binding Protein 1/genetics
17.
Article in English | MEDLINE | ID: mdl-17703434

ABSTRACT

We studied the effects of extremely low-dose human renin inhibition (aliskiren) with low angiotensin II receptor blockade (losartan) in a novel double-transgenic rat model harbouring both human renin and angiotensinogen genes. We found that low-dose aliskiren and low-dose losartan effectively reduced mortality and target-organ damage with minimal, non-significant, effects on blood pressure (BP). Our data suggest that renin-angiotensin system (RAS) inhibition ameliorates target-organ damage in an Ang II-driven model of hypertension. Direct renin inhibition is equally efficacious in this regard. Our study does not fully answer the question of BP-lowering versus RAS inhibition. This question is important and was at least partially addressed with our low-dose model.


Subject(s)
Amides/pharmacology , Angiotensinogen/genetics , Antihypertensive Agents/pharmacology , Fumarates/pharmacology , Hypertension/genetics , Receptor, Angiotensin, Type 1/physiology , Renin/genetics , Angiotensinogen/drug effects , Animals , Animals, Genetically Modified , Humans , Hypertension/drug therapy , Rats , Receptor, Angiotensin, Type 1/drug effects , Renin/drug effects , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology
18.
Cardiovasc Res ; 75(1): 195-204, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17434466

ABSTRACT

OBJECTIVE: Despite the success of antiproliferative therapies, restenosis remains a common problem after percutaneous transluminal coronary angioplasty (PTCA). Longer-term clinical results of brachytherapy (intracoronary radiation), the lack of long-term clinical results after implantation of drug eluting stents, and the occurrence of late thrombosis after both procedures leave room for skepticism. Neointimal proliferation is not substantially inhibited at late time points after brachytherapy, and late lumen loss with a "catch-up" proliferation can occur. We hypothesized that the transcription factors nuclear factor-{kappa}B (NF-kappaB) and activator protein-1 (AP-1) are involved in these processes. We addressed the role of these mediators in a porcine model of coronary restenosis. METHODS: Thirty-nine pigs underwent PTCA in two major coronary arteries. One of the two balloon-injured arteries was randomly assigned to receive immediate 20 Gy beta-irradiation (Brachy group) using a noncentered source train ((90)Sr/Y Beta-Cath, Novoste). Animals were sacrificed after 1 day, 14 days, or 28 days. Proliferating cells were labeled prior to euthanasia. RESULTS: At late time points, lumen area was significantly smaller and the inflammatory response was more pronounced in the Brachy group than in the PTCA group. These findings coincided with sustained activation of MMP-9 and transcription factors like NF-kappaB and AP-1. Initially, cell proliferation was reduced in the Brachy group; however, at late time points, differences between the two treatment groups were no longer significant. CONCLUSIONS: Brachytherapy initially inhibits cell proliferation; however, cellular and molecular inflammatory processes (e.g. activation of NF-kappaB) are enhanced within the arterial wall. This proinflammatory side effect may be responsible for the observed delayed proliferation and the resulting lumen loss.


Subject(s)
Coronary Restenosis/metabolism , Coronary Restenosis/prevention & control , Coronary Vessels/metabolism , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism , Angioplasty, Balloon, Coronary , Animals , Beta Particles , Brachytherapy , Cell Proliferation , Combined Modality Therapy , Coronary Disease/metabolism , Coronary Disease/radiotherapy , Coronary Disease/therapy , Coronary Restenosis/pathology , Coronary Vessels/chemistry , Coronary Vessels/pathology , Electrophoretic Mobility Shift Assay , Immunohistochemistry , Macrophages/pathology , Matrix Metalloproteinase 9/analysis , Matrix Metalloproteinase 9/metabolism , Models, Animal , NF-kappa B/analysis , Random Allocation , Selenium Radioisotopes , Sus scrofa , Swine , T-Lymphocytes/pathology , Time Factors , Transcription Factor AP-1/analysis
19.
Am J Physiol Heart Circ Physiol ; 293(2): H1242-53, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17416596

ABSTRACT

Rats harboring the human renin and angiotensinogen genes (dTGR) feature angiotensin (ANG) II/hypertension-induced cardiac damage and die suddenly between wk 7 and 8. We observed by electrocardiogram (ECG) telemetry that ventricular tachycardia (VT) is a common terminal event in these animals. Our aim was to investigate electrical remodeling. We used ECG telemetry, noninvasive cardiac magnetic field mapping (CMFM) at wk 5 and 7, and performed in vivo programmed electrical stimulation at wk 7. We also investigated whether or not losartan (Los; 30 mg x kg(-1) x day(-1)) would prevent electrical remodeling. Cardiac hypertrophy and systolic blood pressure progressively increased in dTGR compared with Sprague-Dawley (SD) controls. Already by wk 5, untreated dTGR showed increased perivascular and interstitial fibrosis, connective tissue growth factor expression, and monocyte infiltration compared with SD rats, differences that progressed through time. Left-ventricular mRNA expression of potassium channel subunit Kv4.3 and gap-junction protein connexin 43 were significantly reduced in dTGR compared with Los-treated dTGR and SD. CMFM showed that depolarization and repolarization were prolonged and inhomogeneous. Los ameliorated all disturbances. VT could be induced in 88% of dTGR but only in 33% of Los-treated dTGR and could not be induced in SD. Untreated dTGR show electrical remodeling and probably die from VT. Los treatment reduces myocardial remodeling and predisposition to arrhythmias. ANG II target organ damage induces VT.


Subject(s)
Angiotensin II/metabolism , Angiotensinogen/metabolism , Death, Sudden, Cardiac/etiology , Heart Conduction System/physiopathology , Hypertension/physiopathology , Renin/metabolism , Tachycardia, Ventricular/etiology , Ventricular Remodeling , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Angiotensinogen/genetics , Animals , Animals, Genetically Modified , Blood Pressure , Cardiac Pacing, Artificial , Cardiomegaly/complications , Cardiomegaly/etiology , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiomegaly/prevention & control , Connexin 43/genetics , Connexin 43/metabolism , Death, Sudden, Cardiac/prevention & control , Disease Models, Animal , Electrocardiography , Heart Conduction System/drug effects , Heart Conduction System/metabolism , Hypertension/complications , Hypertension/drug therapy , Hypertension/metabolism , Hypertension/pathology , Losartan/pharmacology , Losartan/therapeutic use , Male , Myocardium/metabolism , Myocardium/pathology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley/genetics , Renin/genetics , Shal Potassium Channels/genetics , Shal Potassium Channels/metabolism , Tachycardia, Ventricular/complications , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/physiopathology , Tachycardia, Ventricular/prevention & control , Telemetry , Time Factors , Ventricular Remodeling/drug effects
20.
Curr Opin Nephrol Hypertens ; 16(2): 134-42, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17293689

ABSTRACT

PURPOSE OF REVIEW: Aldosterone and its mineralocorticoid receptor represent an ancient signaling system. Indeed, the mineralocorticoid receptor is older than its agonist. Both have probably served various functions through the eons and salt preservation may be relatively recent. A large body of evidence suggests that aldosterone conducts signaling in vascular cells and contributes substantially to vascular remodeling and target organ damage. A blood pressure and salt balance-independent effect was first observed in two large heart failure trials. RECENT FINDINGS: Mineralocorticoid receptor blockade has now been shown to reduce proteinuria even in the face of angiotensin converting enzyme inhibition and AT1 receptor blockade. Mineralocorticoid receptor blockade effectively reduces target organ damage in every hypertensive model tested, irrespective of circulating renin and aldosterone levels. Protection is also observed in nonhypertensive diabetic and hyperlipidemic models. Signaling in vascular cells involves primarily the mitogen activated protein kinase pathway with participation of the epidermal growth factor receptor. Novel signaling molecules have been shown to participate in aldosterone-mediated actions including the murine double-minute type 2 protein that participates in antiapoptotic and proliferative effects. Clinically, mutations in the mineralocorticoid receptor have shed additional light on its importance. SUMMARY: A resurgence of interest in aldosterone reflects its importance and clinical relevance for vascular remodeling and target organ damage.


Subject(s)
Aldosterone/physiology , Blood Vessels/physiology , Inflammation/physiopathology , Receptors, Mineralocorticoid/metabolism , Animals , Humans , Reactive Oxygen Species/metabolism , Renin/metabolism , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...