Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Immunity ; 42(6): 1100-15, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26084025

ABSTRACT

Tertiary lymphoid organs (TLOs) emerge during nonresolving peripheral inflammation, but their impact on disease progression remains unknown. We have found in aged Apoe(-/-) mice that artery TLOs (ATLOs) controlled highly territorialized aorta T cell responses. ATLOs promoted T cell recruitment, primed CD4(+) T cells, generated CD4(+), CD8(+), T regulatory (Treg) effector and central memory cells, converted naive CD4(+) T cells into induced Treg cells, and presented antigen by an unusual set of dendritic cells and B cells. Meanwhile, vascular smooth muscle cell lymphotoxin ß receptors (VSMC-LTßRs) protected against atherosclerosis by maintaining structure, cellularity, and size of ATLOs though VSMC-LTßRs did not affect secondary lymphoid organs: Atherosclerosis was markedly exacerbated in Apoe(-/-)Ltbr(-/-) and to a similar extent in aged Apoe(-/-)Ltbr(fl/fl)Tagln-cre mice. These data support the conclusion that the immune system employs ATLOs to organize aorta T cell homeostasis during aging and that VSMC-LTßRs participate in atherosclerosis protection via ATLOs.


Subject(s)
Aging/immunology , Atherosclerosis/immunology , Lymphotoxin beta Receptor/metabolism , Myocytes, Smooth Muscle/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adventitia/immunology , Aging/genetics , Animals , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Cell Differentiation/genetics , Cell Movement/genetics , Cells, Cultured , Choristoma/immunology , Immunologic Memory , Lymphocyte Activation/genetics , Lymphoid Tissue/immunology , Lymphotoxin beta Receptor/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microfilament Proteins/genetics , Muscle Proteins/genetics
2.
Arterioscler Thromb Vasc Biol ; 36(6): 1174-85, 2016 06.
Article in English | MEDLINE | ID: mdl-27102965

ABSTRACT

OBJECTIVE: Explore aorta B-cell immunity in aged apolipoprotein E-deficient (ApoE(-/-)) mice. APPROACH AND RESULTS: Transcript maps, fluorescence-activated cell sorting, immunofluorescence analyses, cell transfers, and Ig-ELISPOT (enzyme-linked immunospot) assays showed multilayered atherosclerosis B-cell responses in artery tertiary lymphoid organs (ATLOs). Aging-associated aorta B-cell-related transcriptomes were identified, and transcript atlases revealed highly territorialized B-cell responses in ATLOs versus atherosclerotic lesions: ATLOs showed upregulation of bona fide B-cell genes, including Cd19, Ms4a1 (Cd20), Cd79a/b, and Ighm although intima plaques preferentially expressed molecules involved in non-B effector responses toward B-cell-derived mediators, that is, Fcgr3 (Cd16), Fcer1g (Cd23), and the C1q family. ATLOs promoted B-cell recruitment. ATLO B-2 B cells included naive, transitional, follicular, germinal center, switched IgG1(+), IgA(+), and IgE(+) memory cells, plasmablasts, and long-lived plasma cells. ATLOs recruited large numbers of B-1 cells whose subtypes were skewed toward interleukin-10(+) B-1b cells versus interleukin-10(-) B-1a cells. ATLO B-1 cells and plasma cells constitutively produced IgM and IgG and a fraction of plasma cells expressed interleukin-10. Moreover, ApoE(-/-) mice showed increased germinal center B cells in renal lymph nodes, IgM-producing plasma cells in the bone marrow, and higher IgM and anti-MDA-LDL (malondialdehyde-modified low-density lipoprotein) IgG serum titers. CONCLUSIONS: ATLOs orchestrate dichotomic, territorialized, and multilayered B-cell responses in the diseased aorta; germinal center reactions indicate generation of autoimmune B cells within the diseased arterial wall during aging.


Subject(s)
Aging/immunology , Aorta/immunology , Aortic Diseases/immunology , Apolipoproteins E/deficiency , Atherosclerosis/immunology , B-Lymphocytes/immunology , Tertiary Lymphoid Structures/immunology , Aging/genetics , Aging/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Autoantibodies/blood , Autoimmunity , B-Lymphocytes/metabolism , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation , Genetic Predisposition to Disease , Germinal Center/immunology , Germinal Center/metabolism , Immunoglobulins/blood , Immunologic Memory , Lipoproteins, LDL/immunology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Male , Malondialdehyde/analogs & derivatives , Malondialdehyde/immunology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Plasma Cells/immunology , Plasma Cells/metabolism , Signal Transduction , Tertiary Lymphoid Structures/metabolism , Tertiary Lymphoid Structures/pathology , Transcriptome
3.
J Biol Chem ; 290(25): 15687-15696, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25922067

ABSTRACT

The NF-κB transcriptional response is tightly regulated by a number of processes including the phosphorylation, ubiquitination, and subsequent proteasomal degradation of NF-κB subunits. The IκB family protein BCL-3 stabilizes a NF-κB p50 homodimer·DNA complex through inhibition of p50 ubiquitination. This complex inhibits the binding of the transcriptionally active NF-κB subunits p65 and c-Rel on the promoters of NF-κB target genes and functions to suppress inflammatory gene expression. We have previously shown that the direct interaction between p50 and BCL-3 is required for BCL-3-mediated inhibition of pro-inflammatory gene expression. In this study we have used immobilized peptide array technology to define regions of BCl-3 that mediate interaction with p50 homodimers. Our data show that BCL-3 makes extensive contacts with p50 homodimers and in particular with ankyrin repeats (ANK) 1, 6, and 7, and the N-terminal region of Bcl-3. Using these data we have designed a BCL-3 mimetic peptide based on a region of the ANK1 of BCL-3 that interacts with p50 and shares low sequence similarity with other IκB proteins. When fused to a cargo carrying peptide sequence this BCL-3-derived peptide, but not a mutated peptide, inhibited Toll-like receptor-induced cytokine expression in vitro. The BCL-3 mimetic peptide was also effective in preventing inflammation in vivo in the carrageenan-induced paw edema mouse model. This study demonstrates that therapeutic strategies aimed at mimicking the functional activity of BCL-3 may be effective in the treatment of inflammatory disease.


Subject(s)
Anti-Inflammatory Agents , Biomimetic Materials , NF-kappa B p50 Subunit , Peptides , Proto-Oncogene Proteins , Transcription Factors , Animals , Ankyrin Repeat , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , B-Cell Lymphoma 3 Protein , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Disease Models, Animal , Edema/drug therapy , Edema/genetics , Edema/metabolism , Edema/pathology , Gene Expression Regulation , HeLa Cells , Humans , Mice , NF-kappa B p50 Subunit/chemistry , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , Peptide Mapping , Peptides/chemistry , Peptides/pharmacology , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Circulation ; 130(16): 1363-73, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25223984

ABSTRACT

BACKGROUND: Plasmacytoid dendritic cells (pDCs) bridge innate and adaptive immune responses and are important regulators of immuno-inflammatory diseases. However, their role in atherosclerosis remains elusive. METHODS AND RESULTS: Here, we used genetic approaches to investigate the role of pDCs in atherosclerosis. Selective pDC deficiency in vivo was achieved using CD11c-Cre × Tcf4(-/flox) bone marrow transplanted into Ldlr(-/-) mice. Compared with control Ldlr(-/-) chimeric mice, CD11c-Cre × Tcf4(-/flox) mice had reduced atherosclerosis levels. To begin to understand the mechanisms by which pDCs regulate atherosclerosis, we studied chimeric Ldlr(-/-) mice with selective MHCII deficiency on pDCs. Significantly, these mice also developed reduced atherosclerosis compared with controls without reductions in pDC numbers or changes in conventional DCs. MHCII-deficient pDCs showed defective stimulation of apolipoprotein B100-specific CD4(+) T cells in response to native low-density lipoprotein, whereas production of interferon-α was not affected. Finally, the atheroprotective effect of selective MHCII deficiency in pDCs was associated with significant reductions of proatherogenic T cell-derived interferon-γ and lesional T cell infiltration, and was abrogated in CD4(+) T cell-depleted animals. CONCLUSIONS: This study supports a proatherogenic role for pDCs in murine atherosclerosis and identifies a critical role for MHCII-restricted antigen presentation by pDCs in driving proatherogenic T cell immunity.


Subject(s)
Antigen-Presenting Cells/immunology , Atherosclerosis/immunology , Atherosclerosis/pathology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Adaptive Immunity/immunology , Animals , Aorta/cytology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/immunology , CD4-Positive T-Lymphocytes/cytology , Cell Communication/immunology , Cells, Cultured , Dendritic Cells/cytology , Flow Cytometry , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/genetics , Receptors, LDL/immunology , Transcription Factor 4
5.
Arterioscler Thromb Vasc Biol ; 34(1): 90-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24158518

ABSTRACT

OBJECTIVE: We investigated the role of matrix metalloproteinase-8 (MMP8) in neointima formation and in vascular smooth muscle cell (VSMC) migration and proliferation. APPROACH AND RESULTS: After carotid artery wire injuring, MMP8(-/-)/apoE(-/-) mice had fewer proliferating cells in neointimal lesions and smaller lesion sizes. Ex vivo assays comparing VSMCs isolated from MMP8 knockout and wild-type mice showed that MMP8 knockout decreased proliferation and migration. Proteomics analysis revealed that a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) had lower concentrations in MMP8 knockout VSMC culture media than in MMP8 wild-type VSMC culture media. Western blot, flow cytometric, and immunocytochemical analyses showed that MMP8 knockout VSMCs contained more pro-ADAM10 but less mature ADAM10, more N-cadherin, and ß-catenin in the plasma membrane but less ß-catenin in the nucleus and less cyclin D1. Treatment of MMP8 wild-type VSMCs with an ADAM10 inhibitor, GI254023X, or siRNA knockdown of ADAM10 in MMP8 wild-type VSMCs inhibited proliferation and migration, increased N-cadherin and ß-catenin in the plasma membrane, reduced ß-catenin in the nucleus, and decreased cyclin D1 expression. Incubation of MMP8 knockout VSMCs with a recombinant ADAM10 rescued the proliferative and migratory ability of MMP8 knockout VSMCs and increased cyclin D1 expression. Furthermore, immunohistochemical analyses showed colocalization of ADAM10 with VSMCs and N-cadherin, and nuclear accumulation of ß-catenin in the neointima in apoE(-/-)/MMP8(+/+) mice. CONCLUSIONS: MMP8 enhances VSMC proliferation via an ADAM10, N-cadherin, and ß-catenin-mediated pathway and plays an important role in neointima formation.


Subject(s)
Carotid Artery Injuries/enzymology , Carotid Artery Injuries/pathology , Cell Proliferation , Matrix Metalloproteinase 8/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Neointima , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM10 Protein , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Cadherins/metabolism , Carotid Artery Injuries/genetics , Cell Movement , Cell Proliferation/drug effects , Cells, Cultured , Culture Media, Conditioned/metabolism , Cyclin D1/metabolism , Disease Models, Animal , Matrix Metalloproteinase 8/deficiency , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase Inhibitors/pharmacology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Proteomics/methods , RNA Interference , Time Factors , Transfection , Wnt Signaling Pathway , Wnt1 Protein/metabolism , beta Catenin/metabolism
6.
Blood ; 119(11): 2545-51, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22271449

ABSTRACT

In vivo imaging has revolutionized understanding of the spatiotemporal complexity that subserves the generation of successful effector and regulatory immune responses. Until now, invasive surgery has been required for microscopic access to lymph nodes (LNs), making repeated imaging of the same animal impractical and potentially affecting lymphocyte behavior. To allow longitudinal in vivo imaging, we conceived the novel approach of transplanting LNs into the mouse ear pinna. Transplanted LNs maintain the structural and cellular organization of conventional secondary lymphoid organs. They participate in lymphocyte recirculation and exhibit the capacity to receive and respond to local antigenic challenge. The same LN could be repeatedly imaged through time without the requirement for surgical exposure, and the dynamic behavior of the cells within the transplanted LN could be characterized. Crucially, the use of blood vessels as fiducial markers also allowed precise re-registration of the same regions for longitudinal imaging. Thus, we provide the first demonstration of a method for repeated, noninvasive, in vivo imaging of lymphocyte behavior.


Subject(s)
Diagnostic Imaging , Immune System/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphatic Diseases/pathology , Animals , Antigen Presentation/immunology , Flow Cytometry , Fluorescent Antibody Technique , Green Fluorescent Proteins/metabolism , Longitudinal Studies , Lymphatic Diseases/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Photons , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/immunology
7.
Arterioscler Thromb Vasc Biol ; 32(11): 2569-79, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22936340

ABSTRACT

OBJECTIVE: Clinical studies have identified that reduced numbers of circulating plasmacytoid dendritic cells (pDCs) act as a predictor of cardiovascular events in coronary artery disease and that pDCs are detectable in the shoulder region of human atherosclerotic plaques, where rupture is most likely to occur. Results from animal models are controversial, with pDCs seen to inhibit or promote lesion development depending on the experimental settings. Here, we investigated the role of pDCs in atherosclerosis in apolipoprotein E-deficient mice. METHODS AND RESULTS: We demonstrated that the aorta and spleen of both apolipoprotein E-deficient and C57BL/6 mice displayed similar numbers of pDCs, with similar activation status. In contrast, assessment of antigen uptake/presentation using the Eα/Y-Ae system revealed that aortic pDCs in apolipoprotein E-deficient(-) mice were capable of presenting in vivo systemically administered antigen. Continuous treatment of apolipoprotein E-deficient mice with anti-mouse plasmacytoid dendritic cell antigen 1 (mPDCA-1) antibody caused specific depletion of pDCs in the aorta and spleen and significantly reduced atherosclerosis formation in the aortic sinus (by 46%; P<0.001). Depletion of pDCs also reduced macrophages (by 34%; P<0.05) and increased collagen content (by 41%; P<0.05) in aortic plaques, implying a more stable plaque phenotype. Additionally, pDC depletion reduced splenic T-cell activation and inhibited interleukin-12, chemokine (C-X-C motif) ligand 1, monokine induced by interferon-γ, interferon γ-induced protein 10, and vascular endothelium growth factor serum levels. CONCLUSIONS: These results identify a critical role for pDCs in atherosclerosis and suggest a potential role for pDC targeting in the control of the pathology.


Subject(s)
Aorta/metabolism , Aortic Diseases/etiology , Apolipoproteins E/deficiency , Atherosclerosis/etiology , Dendritic Cells/metabolism , Animals , Antibodies/administration & dosage , Antigen Presentation , Antigens, Surface/immunology , Aorta/immunology , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Cell Proliferation , Cells, Cultured , Dendritic Cells/immunology , Diet, High-Fat , Disease Models, Animal , Female , Genes, T-Cell Receptor alpha , Genes, T-Cell Receptor beta , Inflammation Mediators/metabolism , Injections, Intraperitoneal , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phenotype , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors
8.
Mediators Inflamm ; 2013: 584715, 2013.
Article in English | MEDLINE | ID: mdl-23401644

ABSTRACT

Macrophages play crucial roles in atherosclerotic immune responses. Recent investigation into macrophage autophagy (AP) in atherosclerosis has demonstrated a novel pathway through which these cells contribute to vascular inflammation. AP is a cellular catabolic process involving the delivery of cytoplasmic contents to the lysosomal machinery for ultimate degradation and recycling. Basal levels of macrophage AP play an essential role in atheroprotection during early atherosclerosis. However, AP becomes dysfunctional in the more advanced stages of the pathology and its deficiency promotes vascular inflammation, oxidative stress, and plaque necrosis. In this paper, we will discuss the role of macrophages and AP in atherosclerosis and the emerging evidence demonstrating the contribution of macrophage AP to vascular pathology. Finally, we will discuss how AP could be targeted for therapeutic utility.


Subject(s)
Atherosclerosis/physiopathology , Autophagy/physiology , Macrophages/physiology , Animals , Humans
9.
J Biol Chem ; 286(15): 12933-43, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21317287

ABSTRACT

Mitogen-activated protein kinase phosphatase-2 (MKP-2) is a type 1 nuclear dual specific phosphatase (DUSP) implicated in a number of cancers. We examined the role of MKP-2 in the regulation of MAP kinase phosphorylation, cell proliferation, and survival responses in mouse embryonic fibroblasts (MEFs) derived from a novel MKP-2 (DUSP-4) deletion mouse. We show that serum and PDGF induced ERK-dependent MKP-2 expression in wild type MEFs but not in MKP-2(-/-) MEFs. PDGF stimulation of sustained ERK phosphorylation was enhanced in MKP-2(-/-) MEFs, whereas anisomycin-induced JNK was only marginally increased. However, marked effects upon cell growth parameters were observed. Cellular proliferation rates were significantly reduced in MKP-2(-/-) MEFs and associated with a significant increase in cell doubling time. Infection with adenoviral MKP-2 reversed the decrease in proliferation. Cell cycle analysis revealed a block in G(2)/M phase transition associated with cyclin B accumulation and enhanced cdc2 phosphorylation. MEFs from MKP-2(-/-) mice also showed enhanced apoptosis when stimulated with anisomycin correlated with increased caspase-3 cleavage and γH2AX phosphorylation. Increased apoptosis was reversed by adenoviral MKP-2 infection and correlated with selective inhibition of JNK signaling. Collectively, these data demonstrate for the first time a critical non-redundant role for MKP-2 in regulating cell cycle progression and apoptosis.


Subject(s)
Cell Division/physiology , Embryo, Mammalian/enzymology , Fibroblasts/enzymology , G2 Phase/physiology , Gene Expression Regulation, Enzymologic/physiology , Protein Tyrosine Phosphatases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Caspase 3/genetics , Caspase 3/metabolism , Cell Division/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cyclin B/genetics , Cyclin B/metabolism , Embryo, Mammalian/cytology , Fibroblasts/cytology , G2 Phase/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Histones/genetics , Histones/metabolism , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Mice, Knockout , Phosphorylation/drug effects , Phosphorylation/genetics , Platelet-Derived Growth Factor/pharmacology , Protein Tyrosine Phosphatases/genetics
10.
Arterioscler Thromb Vasc Biol ; 31(11): 2448-54, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21852559

ABSTRACT

OBJECTIVE: We have previously demonstrated that bindarit, a selective inhibitor of monocyte chemotactic proteins (MCPs), is effective in reducing neointimal formation in rodent models of vascular injury by reducing smooth muscle cell proliferation and migration and neointimal macrophage content, effects associated with the inhibition of MCP-1/CCL2 production. The aim of the current study was to evaluate the efficacy of bindarit on in-stent stenosis in the preclinical porcine coronary stent model. METHODS AND RESULTS: One or 2 bare metal stents (Multi-Link Vision, 3.5 mm) were deployed (1:1.2 oversize ratio) in the coronary arteries of 42 pigs (20 bindarit versus 22 controls). Bindarit (50 mg/kg per day) was administered orally from 2 days before stenting until the time of euthanasia at 7 and 28 days. Bindarit caused a significant reduction in neointimal area (39.4%, P<0.001, n=9 group), neointimal thickness (51%, P<0.001), stenosis area (37%, P<0.001), and inflammatory score (40%, P<0.001) compared with control animals, whereas there was no significant difference in the injury score between the 2 groups. Moreover, treatment with bindarit significantly reduced the number of proliferating cells (by 45%, P<0.05; n=6 group) and monocyte/macrophage content (by 55%, P<0.01; n=5-6 group) in stented arteries at day 7 and 28, respectively. These effects were associated with a significant (P<0.05) reduction of MCP-1 plasma levels at day 28. In vitro data showed that bindarit (10-300 µmol/L) reduced tumor necrosis factor-α (50 ng/mL)-induced pig coronary artery smooth muscle cell proliferation and inhibited MCP-1 production. CONCLUSION: Our results show the efficacy of bindarit in the prevention of porcine in-stent stenosis and support further investigation for clinical application of this compound.


Subject(s)
Coronary Stenosis/prevention & control , Coronary Vessels/pathology , Indazoles/administration & dosage , Indazoles/therapeutic use , Propionates/administration & dosage , Propionates/therapeutic use , Stents , Administration, Oral , Animals , Cell Proliferation/drug effects , Coronary Stenosis/pathology , Coronary Vessels/drug effects , Indazoles/pharmacology , Male , Models, Animal , Monocyte Chemoattractant Proteins/antagonists & inhibitors , Monocyte Chemoattractant Proteins/blood , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Neointima/pathology , Neointima/prevention & control , Propionates/pharmacology , Swine , Treatment Outcome
11.
Arterioscler Thromb Vasc Biol ; 30(12): 2458-66, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20930169

ABSTRACT

OBJECTIVE: The activation of nuclear factor-κB (NF-κB) is a crucial step in the arterial wall's response to injury. The identification and characterization of the NF-κB essential modulator-binding domain (NBD) peptide, which can block the activation of the IκB kinase complex, have provided an opportunity to selectively abrogate the inflammation-induced activation of NF-κB. The aim of the present study was to evaluate the effect of the NBD peptide on neointimal formation. METHODS AND RESULTS: In the rat carotid artery balloon angioplasty model, local treatment with the NBD peptide (300 µg/site) significantly reduced the number of proliferating cells at day 7 (by 40%; P<0.01) and reduced injury-induced neointimal formation (by 50%; P<0.01) at day 14. These effects were associated with a significant reduction of NF-κB activation and monocyte chemotactic protein-1 expression in the carotid arteries of rats treated with the peptide. In addition, the NBD peptide (0.01 to 1 µmol/L) reduced rat smooth muscle cell proliferation, migration, and invasion in vitro. Similar results were observed in apolipoprotein E(-/-) mice in which the NBD peptide (150 µg/site) reduced wire-induced neointimal formation at day 28 (by 47%; P<0.01). CONCLUSIONS: The NBD peptide reduces neointimal formation and smooth muscle cell proliferation/migration, both effects associated with the inhibition of NF-κB activation.


Subject(s)
Carotid Artery Injuries/prevention & control , I-kappa B Kinase/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Myocytes, Smooth Muscle/drug effects , Peptides/pharmacology , Protein Kinase Inhibitors/pharmacology , Tunica Intima/drug effects , Angioplasty, Balloon/adverse effects , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apoptosis/drug effects , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/etiology , Carotid Artery Injuries/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chemokine CCL2/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activation , Female , Hyperplasia , I-kappa B Kinase/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , NF-kappa B/metabolism , Phosphorylation , Rats , Rats, Wistar , Tunica Intima/enzymology , Tunica Intima/pathology
12.
J Sex Med ; 7(5): 1778-86, 2010 May.
Article in English | MEDLINE | ID: mdl-19788716

ABSTRACT

INTRODUCTION: Urotensin II (U-II) is a cyclic peptide originally isolated from the teleost neurosecretory system and subsequently identified in other species, including man. U-II was identified as the natural ligand of an orphan G-protein coupled receptor (UT receptor). U-II and UT receptor are expressed in a variety of peripheral organs and especially in cardiovascular tissue. U-II caused both constrictor and vasodilator effect, depending by vascular bed. The in vivo functional consequences of U-II on the cardiovascular hemodynamics are not clearly understood. AIM: To investigate the presence of UT receptor and the effect of U-II in human corpus cavernosum (HCC) strips. To evaluate the effect of U-II in vivo in anesthetized rats. METHODS: UT receptor expression as protein and as mRNA were assessed by Western blot and reverse transcriptase polymerase chain reaction. Next, the UT receptor localization was evaluated by immunohistochemical analysis. By using HCC strips, with or without endothelium, the effect of U-II (0.1 nM-10 microM) was evaluated. In order to asses the nitric oxide (NO) involvement, the strips were incubated with N (G)-nitro-L-arginine methyl ester (NO synthase inhibitor, 100 microM). U-II (0.1, 0.3, 1.0 nmol/rat) effect in vivo was studied in anesthetized rats by monitoring the intracavernous and systemic blood pressure. MAIN OUTCOME MEASURES: HCC expresses the UT receptor and its activation, by UII, causes an endothelium- and NO-dependent relaxation. RESULTS: UT receptor is expressed in human and rat corpus cavernosum. In HCC UT receptor is localized on endothelial cells. U-II significantly relaxed HCC strips in endothelium- and -NO-dependent fashion. The peptide caused a significant increase in intracavernous pressure in anesthetized rats. CONCLUSION: This study demonstrates that UT receptor is expressed on the endothelium of HCC. U-II/UT receptor system is involved in HCC function and it involves endothelium and NO pathway. Thus, U-II/UT receptor pathway could be involved in erectile function.


Subject(s)
Penis/metabolism , Receptors, G-Protein-Coupled/genetics , Urotensins/physiology , Animals , Blotting, Western , Endothelium, Vascular/metabolism , Humans , Male , Microscopy, Fluorescence , Nitric Oxide/physiology , Penile Erection/physiology , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
13.
Cardiovasc Res ; 116(2): 306-316, 2020 02 01.
Article in English | MEDLINE | ID: mdl-30980670

ABSTRACT

AIMS: Aortic adaptive immunity plays a role in atherosclerosis; however, the precise mechanisms leading to T-cell activation in the arterial wall remain poorly understood. METHODS AND RESULTS: Here, we have identified naïve T cells in the aorta of wild-type and T-cell receptor transgenic mice and we demonstrate that naïve T cells can be primed directly in the vessel wall with both kinetics and frequency of T-cell activation found to be similar to splenic and lymphoid T cells. Aortic homing of naïve T cells is regulated at least in part by the P-selectin glycosylated ligand-1 receptor. In experimental atherosclerosis the aorta supports CD4+ T-cell activation selectively driving Th1 polarization. By contrast, secondary lymphoid organs display Treg expansion. CONCLUSION: Our results demonstrate that the aorta can support T-cell priming and that naïve T cells traffic between the circulation and vessel wall. These data underpin the paradigm that local priming of T cells specific for plaque antigens contributes to atherosclerosis progression.


Subject(s)
Adaptive Immunity , Aorta/immunology , Aortic Diseases/immunology , Atherosclerosis/immunology , Cell Proliferation , Lymphocyte Activation , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Genes, T-Cell Receptor , Kinetics , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Mice, Knockout, ApoE , Phenotype , Plaque, Atherosclerotic , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/metabolism
14.
Front Immunol ; 10: 849, 2019.
Article in English | MEDLINE | ID: mdl-31068936

ABSTRACT

Atherosclerosis is a complex inflammatory pathology underpinning cardiovascular diseases (CVD), which are the leading cause of death worldwide. The interplay between vascular stromal cells and immune cells is fundamental to the progression and outcome of atherosclerotic disease, however, the majority of in vitro studies do not consider the implications of these interactions and predominantly use mono-culture approaches. Here we present a simple and robust methodology involving the co-culture of vascular endothelial (ECs) and smooth muscle cells (SMCs) alongside an inflammatory compartment, in our study containing THP-1 macrophages, for studying these complex interactions. Using this approach, we demonstrate that the interaction between vascular stromal and immune cells produces unique cellular phenotypes and soluble mediator profiles not observed in double-cell 2D cultures. Our results highlight the importance of cellular communication and support the growing idea that in vitro research must evolve from mono-culture systems to provide data more representative of the multi-cellular environment found in vivo. The methodology presented, in comparison with established approaches, has the advantage of being technically simple whilst enabling the isolation of pure populations of ECs, SMCs and immune cells directly from the co-culture without cell sorting. The approach described within would be applicable to those studying mechanisms of vascular inflammation, particularly in relation to understanding the impact cellular interaction has on the cumulative immune-vascular response to atherogenic or inflammatory stimuli.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/pathology , Cell Communication , Cell Culture Techniques , Coculture Techniques , Models, Biological , Biomarkers , Endothelial Cells/immunology , Endothelial Cells/metabolism , Fluorescent Antibody Technique , Humans , Macrophages/immunology , Macrophages/metabolism , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism
15.
Heart ; 104(6): 460-467, 2018 03.
Article in English | MEDLINE | ID: mdl-29061690

ABSTRACT

To accurately predict atherosclerotic plaque progression, a detailed phenotype of the lesion at the molecular level is required. Here, we assess the respective merits and limitations of molecular imaging tools. Clinical imaging includes contrast-enhanced ultrasound, an inexpensive and non-toxic technique but with poor sensitivity. CT benefits from high spatial resolution but poor sensitivity coupled with an increasing radiation burden that limits multiplexing. Despite high sensitivity, positron emission tomography and single-photon emission tomography have disadvantages when applied to multiplex molecular imaging due to poor spatial resolution, signal cross talk and increasing radiation dose. In contrast, MRI is non-toxic, displays good spatial resolution but poor sensitivity. Preclinical techniques include near-infrared fluorescence (NIRF), which provides good spatial resolution and sensitivity; however, multiplexing with NIRF is limited, due to photobleaching and spectral overlap. Fourier transform infrared spectroscopy and Raman spectroscopy are label-free techniques that detect molecules based on the vibrations of chemical bonds. Both techniques offer fast acquisition times with Raman showing superior spatial resolution. Raman signals are inherently weak; however, leading to the development of surface-enhanced Raman spectroscopy (SERS) that offers greatly increased sensitivity due to using metallic nanoparticles that can be functionalised with biomolecules targeted against plaque ligands while offering high multiplexing potential. This asset combined with high spatial resolution makes SERS an exciting prospect as a diagnostic tool. The ongoing refinements of SERS technologies such as deep tissue imaging and portable systems making SERS a realistic prospect for translation to the clinic.


Subject(s)
Cardiovascular Diseases/diagnosis , Plaque, Atherosclerotic/diagnostic imaging , Spectrum Analysis, Raman/methods , Disease Progression , Humans , Molecular Imaging/methods
16.
Theranostics ; 8(22): 6195-6209, 2018.
Article in English | MEDLINE | ID: mdl-30613292

ABSTRACT

Vascular immune-inflammatory responses play a crucial role in the progression and outcome of atherosclerosis. The ability to assess localized inflammation through detection of specific vascular inflammatory biomarkers would significantly improve cardiovascular risk assessment and management; however, no multi-parameter molecular imaging technologies have been established to date. Here, we report the targeted in vivo imaging of multiple vascular biomarkers using antibody-functionalized nanoparticles and surface-enhanced Raman scattering (SERS). Methods: A series of antibody-functionalized gold nanoprobes (BFNP) were designed containing unique Raman signals in order to detect intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1) and P-selectin using SERS. Results: SERS and BFNP were utilized to detect, discriminate and quantify ICAM-1, VCAM-1 and P-selectin in vitro on human endothelial cells and ex vivo in human coronary arteries. Ultimately, non-invasive multiplex imaging of adhesion molecules in a humanized mouse model was demonstrated in vivo following intravenous injection of the nanoprobes. Conclusion: This study demonstrates that multiplexed SERS-based molecular imaging can indicate the status of vascular inflammation in vivo and gives promise for SERS as a clinical imaging technique for cardiovascular disease in the future.


Subject(s)
Coronary Vessels/diagnostic imaging , Coronary Vessels/immunology , Human Umbilical Vein Endothelial Cells/chemistry , Molecular Imaging/methods , Spectrum Analysis, Raman/methods , Animals , Female , Gold/chemistry , Human Umbilical Vein Endothelial Cells/immunology , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Imaging/instrumentation , Nanoparticles/chemistry , P-Selectin/genetics , P-Selectin/immunology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
17.
Circulation ; 114(5): 430-7, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16864728

ABSTRACT

BACKGROUND: Studies in humans and animal models suggest that interleukin-18 (IL-18) plays a crucial role in vascular pathologies. IL-18 is a predictor of cardiovascular death in angina and is involved in atherotic plaque destabilization. Higher IL-18 plasma levels also are associated with restenosis after coronary artery angioplasty performed in patients with acute myocardial infarction. We investigated the effective role of IL-18 in neointimal formation in a balloon-induced rat model of vascular injury. METHODS AND RESULTS: Endothelial denudation of the left carotid artery was performed by use of a balloon embolectomy catheter. Increased expression of IL-18 and IL-18Ralpha/beta mRNA was detectable in carotid arteries from days 2 to 14 after angioplasty. The active form of IL-18 was highly expressed in injured arteries. Strong immunoreactivity for IL-18 was detected in the medial smooth muscle cells at days 2 and 7 after balloon injury and in proliferating/migrating smooth muscle cells in neointima at day 14. Moreover, serum concentrations of IL-18 were significantly higher among rats subjected to vascular injury. Treatment with neutralizing rabbit anti-rat IL-18 immunoglobulin G significantly reduced neointimal formation (by 27%; P < 0.01), reduced the number of proliferating cells, and inhibited interferon-gamma, IL-6, and IL-8 mRNA expression and nuclear factor-kappaB activation in injured arteries. In addition, in vitro data show that IL-18 affects smooth muscle cell proliferation. CONCLUSIONS: These results identify a critical role for IL-18 in neointimal formation in a rat model of vascular injury and suggest a potential role for IL-18 neutralization in the reduction of neointimal development.


Subject(s)
Cardiovascular Diseases/physiopathology , Carotid Artery Injuries/etiology , Carotid Artery Injuries/physiopathology , Interleukin-18/physiology , Tunica Intima/physiopathology , Actins/analysis , Animals , Balloon Occlusion/adverse effects , Carotid Arteries/chemistry , Carotid Arteries/pathology , Carotid Arteries/physiopathology , Carotid Artery Injuries/pathology , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Disease Models, Animal , Endothelium, Vascular/injuries , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Immunoglobulin G/pharmacology , Interferon-gamma/analysis , Interferon-gamma/genetics , Interferon-gamma/physiology , Interleukin-18/analysis , Interleukin-18/blood , Interleukin-18/genetics , Interleukin-6/analysis , Interleukin-6/genetics , Interleukin-6/physiology , Interleukin-8/analysis , Interleukin-8/genetics , Interleukin-8/physiology , Male , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , NF-kappa B/analysis , NF-kappa B/physiology , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Rats, Wistar , Time Factors , Tunica Intima/pathology
18.
Eur J Pharmacol ; 566(1-3): 192-9, 2007 Jul 02.
Article in English | MEDLINE | ID: mdl-17477920

ABSTRACT

Oxidative stress plays an important role in inflammatory process of celiac disease. We have studied the effect of the lycopene, quercetin and tyrosol natural antioxidants on the inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) gene expression in RAW 264.7 macrophages stimulated by gliadin in association with IFN-gamma. The IFN-gamma plus gliadin combination treatment was capable of enhancing iNOS and COX-2 gene expression and nuclear factor-kappaB (NF-kappaB), interferon regulatory factor-1 (IRF-1) and signal transducer and activator of transcription-1alpha (STAT-1alpha) activation induced by reactive oxygen species generation at 24 h. Lycopene, quercetin and tyrosol inhibited all these effects. The results here reported suggest that these compounds may represent non toxic agents for the control of pro-inflammatory genes involved in celiac disease.


Subject(s)
Antioxidants/pharmacology , Carotenoids/pharmacology , Macrophage Activation/drug effects , Phenylethyl Alcohol/analogs & derivatives , Quercetin/pharmacology , Animals , Celiac Disease , Cell Line , Cyclooxygenase 2/genetics , Dinoprostone/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gliadin , Interferon Regulatory Factor-1/metabolism , Interferon-Stimulated Gene Factor 3/metabolism , Interferon-gamma , Lycopene , Mice , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , Nitrites/metabolism , Phenylethyl Alcohol/pharmacology , Reactive Oxygen Species/metabolism
19.
Br J Pharmacol ; 174(22): 3898-3913, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28409825

ABSTRACT

Data from basic science experiments is overwhelmingly supportive of the causal role of immune-inflammatory response(s) at the core of atherosclerosis, and therefore, the theoretical potential to manipulate the inflammatory response to prevent cardiovascular events. However, extrapolation to humans requires care and we still lack definitive evidence to show that interfering in immune-inflammatory processes may safely lessen clinical atherosclerosis. In this review, we discuss key therapeutic targets in the treatment of vascular inflammation, placing basic research in a wider clinical perspective, as well as identifying outstanding questions. LINKED ARTICLES: This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.


Subject(s)
Cardiovascular Diseases/prevention & control , Inflammation/drug therapy , Animals , Anti-Inflammatory Agents/therapeutic use , Anticholesteremic Agents/therapeutic use , Antioxidants/therapeutic use , Biological Products/therapeutic use , Cardiovascular Diseases/immunology , Cardiovascular Diseases/metabolism , Cytokines/immunology , Humans , Phospholipases A2/metabolism , Risk , Vitamins/therapeutic use
20.
Pain ; 157(1): 80-91, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25974242

ABSTRACT

The effects of ultramicronized palmitoylethanolamide were evaluated on pain behaviours and markers of mast cell (MC) activity in a rat model of endometriosis plus ureteral calculosis (ENDO+STONE)-induced viscerovisceral hyperalgesia (VVH). Female Sprague-Dawley rats that underwent surgical induction of endometriosis were randomly assigned to receive active (ultramicronized palmitoylethanolamide 10 mg·kg(-1)·d(-1), orally) or placebo treatment for 25 days. At day 21, they underwent ureteral stone formation and were video-recorded till day 25 to evaluate ureteral and uterine pain behaviours. At autopsy (day 25), ureteral condition and number and diameter of endometrial cysts were evaluated. The following were then measured: number and percentage of degranulating MCs, number of vessels, chymase, nerve growth factor (NGF), vascular endothelial growth factor (VEGF), and Flk-1 (VEGF receptor) in cysts, and NGF in dorsal root ganglia (DRG). Ultramicronized palmitoylethanolamide-treated vs placebo-treated rats showed significantly lower number, duration and complexity of ureteral crises, shorter duration of uterine pain, and smaller cyst diameter (0.0001 < P < 0.004); a significantly higher percentage of expelled stones (P < 0.0001); significantly lower MC number (P < 0.01), vessel number (P < 0.01), chymase (P < 0.05), NGF (P < 0.05), VEGF (P < 0.01), and Flk-1 (P < 0.01) expression in cysts and NGF expression in DRG (P < 0.01). In all animals, the global duration of ureteral crises correlated linearly and directly with cyst diameter, MC number and chymase in cysts, and NGF in cysts and DRG (0.02 < P < 0.0002). Ultramicronized palmitoylethanolamide significantly reduces VVH from ENDO+STONE, probably by modulating MC expression/activity in cysts, thus reducing central sensitization due to noxious signals from endometriotic lesions. The results suggest potential utility of the compound for VVH in clinics.


Subject(s)
Endometriosis/complications , Ethanolamines/therapeutic use , Hyperalgesia/drug therapy , Mast Cells/drug effects , Palmitic Acids/therapeutic use , Ureteral Calculi/complications , Amides , Animals , Chymases/metabolism , Disease Models, Animal , Endometriosis/metabolism , Ethanolamines/pharmacology , Female , Hyperalgesia/complications , Hyperalgesia/metabolism , Mast Cells/metabolism , Nerve Growth Factor/metabolism , Palmitic Acids/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Vascular Endothelial Growth Factor/metabolism , Ureteral Calculi/metabolism , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL