Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Nat Med ; 12(2): 178-80, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16462800

ABSTRACT

Atherosclerosis is an immunoinflammatory disease elicited by accumulation of lipids in the artery wall and leads to myocardial infarction and stroke. Here, we show that naturally arising CD4(+)CD25(+) regulatory T cells, which actively maintain immunological tolerance to self and nonself antigens, are powerful inhibitors of atherosclerosis in several mouse models. These results provide new insights into the immunopathogenesis of atherosclerosis and could lead to new therapeutic approaches that involve immune modulation using regulatory T cells.


Subject(s)
Atherosclerosis/etiology , T-Lymphocytes, Regulatory/immunology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , B7-1 Antigen/genetics , CD28 Antigens/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/pathology
2.
Am J Pathol ; 174(2): 693-700, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19131590

ABSTRACT

Rupture of the collagenous, fibrous cap of an atherosclerotic plaque commonly causes thrombosis. Activated immune cells can secrete mediators that jeopardize the integrity of the fibrous cap. This study aimed to determine the relationship between T-cell-mediated inflammation and collagen turnover in a mouse model of experimental atherosclerosis. Both Apoe(-/-) x CD4dnTbetaRII mice with defective transforming growth factor-beta receptors in T cells (and hence released from tonic suppression of T-cell activation) and lesion size-matched Apoe(-/-) mice were used. Picrosirius red staining showed a lower content of thick mature collagen fibers in lesions of Apoe(-/-) x CD4dnTbetaRII mice, although both groups had similar levels of procollagen type I or III mRNA and total collagen content in lesions. Analysis of both gene expression and protein content showed a significant decrease of lysyl oxidase, the extracellular enzyme needed for collagen cross-linking, in aortas of Apoe(-/-)--CD4dnTbetaRII mice. T-cell-driven inflammation provoked a selective and limited increase in the expression of proteinases that catabolize the extracellular matrix. Atheromata of Apoe(-/-)--CD4dnTbetaRII mice had increased levels of matrix metalloproteinase-13 and cathepsin S mRNAs and of the active form of cathepsin S protein but no increase was detected in collagen fragmentation. Our results suggest that exaggerated T-cell-driven inflammation limits collagen maturation in the atherosclerotic plaque while having little effect on collagen degradation.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/immunology , Collagen/metabolism , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/pathology , Blotting, Western , Collagen/genetics , Collagen/immunology , Fluorescent Antibody Technique , Gene Expression , Immunohistochemistry , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Knockout , Protein-Lysine 6-Oxidase/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
3.
Arterioscler Thromb Vasc Biol ; 27(3): 614-20, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17170367

ABSTRACT

OBJECTIVE: Based on their role in T-cell homing into nonlymphoid tissue, we examined the role of the homeostatic chemokines CCL19 and CCL21 and their common receptor CCR7 in coronary artery disease (CAD). METHODS AND RESULTS: We performed studies in patients with stable (n=40) and unstable (n=40) angina and healthy controls (n=20), in vitro studies in T-cells and macrophages, and studies in apolipoprotein-E-deficient (ApoE-/-) mice and human atherosclerotic carotid plaques. We found increased levels of CCL19 and CCL21 within the atherosclerotic lesions of the ApoE-/- mice, in human atherosclerotic carotid plaques, and in plasma of CAD patients. Whereas strong CCR7 expression was seen in T-cells from murine and human atherosclerotic plaques, circulating T-cells from angina patients showed decreased CCR7 expression. CCL19 and CCL21 promoted an inflammatory phenotype in T-cells and macrophages and increased matrix metalloproteinase (MMP) and tissue factor levels in the latter cell type. Although aggressive statin therapy increased CCR7 and decreased CCL19/CCL21 levels in peripheral blood from CAD patients, conventional therapy did not. CONCLUSIONS: The abnormal regulation of CCL19 and CCL21 and their common receptor in atherosclerosis could contribute to disease progression by recruiting T-cells and macrophages to the atherosclerotic lesions and by promoting inflammatory responses in these cells.


Subject(s)
Chemokines, CC/metabolism , Coronary Disease/blood , Coronary Disease/drug therapy , Heptanoic Acids/therapeutic use , Pyrroles/therapeutic use , Simvastatin/therapeutic use , Angioplasty, Balloon, Coronary/methods , Animals , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Atorvastatin , Biopsy, Needle , Cells, Cultured , Chemokine CCL19 , Chemokine CCL21 , Chemokines, CC/genetics , Coronary Disease/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Gene Expression Regulation , Humans , Immunohistochemistry , In Vitro Techniques , Leukocytes, Mononuclear , Mice , Mice, Transgenic , Prognosis , RNA, Messenger/analysis , Receptors, CCR7 , Receptors, Chemokine/metabolism , Reference Values , Risk Factors , Sensitivity and Specificity , Treatment Outcome
4.
J Clin Invest ; 112(9): 1342-50, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14568988

ABSTRACT

Increasing evidence suggests that atherosclerosis is an inflammatory disease promoted by hypercholesterolemia. The role of adaptive immunity has been controversial, however. We hypothesized that proatherogenic T cells are controlled by immunoregulatory cytokines. Among them, TGF-beta has been implied in atherosclerosis, but its mechanism of action remains unclear. We crossed atherosclerosis-prone ApoE-knockout mice with transgenic mice carrying a dominant negative TGF-beta receptor II in T cells. The ApoE-knockout mice with disrupted TGF-beta signaling in T cells exhibited a sixfold increase in aortic lesion surface area, a threefold increase in aortic root lesion size, and a 125-fold increase in aortic IFN-gamma mRNA when compared with age-matched ApoE-knockout littermates. When comparing size-matched lesions, those of mice with T cell-specific blockade of TGF-beta signaling displayed increased T cells, activated macrophages, and reduced collagen, consistent with a more vulnerable phenotype. Ab's to oxidized LDL, circulating T cell cytokines, and spleen T cell activity were all increased in ApoE-knockout mice with dominant negative TGF-beta receptors in T cells. Taken together, these results show that abrogation of TGF-beta signaling in T cells increases atherosclerosis and suggest that TGF-beta reduces atherosclerosis by dampening T cell activation. Inhibition of T cell activation may therefore represent a strategy for antiatherosclerotic therapy.


Subject(s)
Arteriosclerosis/etiology , Signal Transduction/physiology , T-Lymphocytes/physiology , Transforming Growth Factor beta/physiology , Animals , Apolipoproteins E/genetics , Cytokines/biosynthesis , Female , Interferon-gamma/biosynthesis , Lipoproteins/metabolism , Lipoproteins, LDL/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL
5.
Circ Res ; 96(4): 427-34, 2005 Mar 04.
Article in English | MEDLINE | ID: mdl-15662027

ABSTRACT

Atherosclerosis is a complex disease, bearing many of the characteristics of a chronic inflammatory process. Both cellular and humoral immune responses may be involved in the disease development. Oxidized low-density lipoprotein (oxLDL) is suggested to be an autoantigen in atherosclerosis. A protective effect against atherosclerosis has been demonstrated in animals immunized with oxLDL. Such a protection is associated with elevation of T cell-dependent IgG antibodies against oxLDL. In addition, it has been shown that immunization with Freund adjuvant alone also confers protection against development of atherosclerosis. We therefore hypothesized that CD4+ T cells are critical in the development of atherosclerosis and that they are involved in protective immune reactions after immunization. The development of atherosclerosis was studied in apolipoprotein E knockout (apoE KO) mice and CD4/apoE double knockout (dKO) mice that were immunized with either oxLDL in Freund adjuvant or adjuvant alone, or left untreated. Our results show that (1) the absence of CD4+ cells in apoE KO mice leads to reduced atherosclerosis, indicating that CD4+ cells constitute a major proatherogenic cell population, and (2) the atheroprotective effect of LDL immunization does not depend on CD4+ cells, whereas (3) the atheroprotective effect of adjuvant injection is CD4-dependent. These findings demonstrate complex roles of immune cell-cell interactions in the regulation of the atherosclerotic process and point to several possible targets in the treatment and prevention of atherosclerosis.


Subject(s)
Aortic Diseases/immunology , Arteriosclerosis/immunology , Autoantigens/immunology , CD4 Antigens/physiology , CD4-Positive T-Lymphocytes/immunology , Immunization , Lipoproteins, LDL/immunology , Malondialdehyde/analogs & derivatives , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/biosynthesis , Antigens, Differentiation, Myelomonocytic/genetics , Aortic Diseases/blood , Aortic Diseases/etiology , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arteriosclerosis/blood , Arteriosclerosis/etiology , Arteriosclerosis/pathology , Arteriosclerosis/prevention & control , Autoantibodies/biosynthesis , Autoantibodies/blood , Autoantibodies/immunology , CD4 Antigens/genetics , CD4 Antigens/immunology , Cholesterol/blood , Disease Progression , Female , Freund's Adjuvant/pharmacology , Freund's Adjuvant/therapeutic use , Genes, MHC Class II , Histocompatibility Antigens Class II/biosynthesis , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Immunoglobulin G/immunology , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Interleukin-4/genetics , Lipoproteins, LDL/therapeutic use , Malondialdehyde/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Random Allocation , Receptors, Immunologic/metabolism , Receptors, Scavenger , T-Cell Antigen Receptor Specificity , Triglycerides/blood , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics , Vasculitis/blood
6.
Arterioscler Thromb Vasc Biol ; 26(11): 2421-32, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16973967

ABSTRACT

The idea that atherosclerosis is an inflammatory disease is no longer controversial. Instead, much of the current research is now focused on understanding what drives this inflammation and how it is regulated. Adaptive immunity, in particular T cells, is highly involved in atherogenesis. It is well known that different subsets of T cells can drive or dampen inflammatory processes, but we still have much to learn about the regulation of this balance in the context of atherosclerosis. This review summarizes our knowledge of T cells in atherogenesis, their potential antigens, their contact-dependent activities, and their secretion of inflammatory and antiinflammatory mediators, aiming to illustrate how T cells can aggravate or attenuate this disease through cross-talk with other cells within or outside the atherosclerotic plaque.


Subject(s)
Atherosclerosis/immunology , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/immunology , Antigens/immunology , Antigens, CD/immunology , Cytokines/metabolism , Disease Models, Animal , Humans , Immunity , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
7.
Arterioscler Thromb Vasc Biol ; 26(4): 864-70, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16456097

ABSTRACT

OBJECTIVE: Atherosclerosis is associated with immune responses to oxidized low-density lipoprotein (oxLDL). The presence of activated macrophages and T cells in lesions suggests that cell-mediated immune reactions are taking place during the disease process. However, the role of specific immune responses has remained unclear. We have previously shown that transfer of CD4+ T cells from apolipoprotein E knockout mice (apoE(-/-)) into immunodeficient apoE(-/-) scid/scid mice accelerates disease. METHODS AND RESULTS: To test whether this effect is dependent on specific disease-associated antigens, purified CD4+ T cells from oxLDL-immunized mice were transferred into apoE(-/-) scid/scid mice. CD4+ T cells from mice immunized with a nonrelevant antigen, keyhole limpet hemocyanin (KLH), and naïve CD4+ T cells were used as controls. After 12 weeks, all mice that received T cells had larger lesions than untouched apoE(-/-) scid/scid controls. However, mice receiving CD4+ T cells from oxLDL immunized mice had substantially accelerated lesion progression compared with those receiving naive or KLH-primed T cells. Circulating levels of interferon-gamma were increased in proportion to the acceleration of atherosclerosis. CONCLUSIONS: These data show that adoptive transfer of purified CD4+ T cells from oxLDL-immunized mice accelerates atherosclerosis. They support the notion that Th1 cellular immunity is proatherogenic and identify oxLDL as a culprit autoantigen.


Subject(s)
Atherosclerosis/immunology , CD4-Positive T-Lymphocytes/immunology , Lipoproteins, LDL/immunology , Adjuvants, Immunologic , Adoptive Transfer , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/pathology , Autoantigens/immunology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/transplantation , Hemocyanins/immunology , Immunity, Cellular , Interferon-gamma/blood , Interleukin-5/blood , Interleukins/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Th1 Cells/immunology , Th1 Cells/pathology
8.
Arterioscler Thromb Vasc Biol ; 26(4): 857-63, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16424351

ABSTRACT

OBJECTIVE: Based on its role in inflammation and matrix degradation, we hypothesized a role for osteoprotegerin (OPG), RANK, and RANK ligand (RANKL) in coronary artery disease. METHODS AND RESULTS: We examined the expression of various members of the OPG/RANKL/RANK axis in patients with stable and unstable angina and in the atherosclerotic lesions of apolipoprotein E-deficient (apoE(-/-)) mice. Our findings were: (1) Serum levels of OPG were raised in patients with unstable angina (n=40), but not in those with stable angina (n=40), comparing controls (n=20); (2) mRNA levels of RANKL were increased in T-cells in unstable angina patients accompanied by increased expression of RANK in monocytes; (3) strong immunostaining of OPG/RANKL/RANK was seen within thrombus material obtained at the site of plaque rupture during acute myocardial infarction; (4) OPG/RANKL/RANK was expressed in the atherosclerotic plaques of apoE(-/-) mice, with RANKL located specifically to the plaques; and (5) RANKL enhanced the release of monocyte chemoattractant peptide-1 in mononuclear cells from unstable angina patients, and promoted matrix metalloproteinase (MMP) activity in vascular smooth muscle cells. CONCLUSIONS: We show enhanced expression of the OPG/RANKL/RANK system both in clinical and experimental atherosclerosis, with enhanced T-cell expression of RANKL as an important feature of unstable disease.


Subject(s)
Angina, Unstable/metabolism , Atherosclerosis/metabolism , Carrier Proteins/metabolism , Glycoproteins/blood , Glycoproteins/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cytoplasmic and Nuclear/blood , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Tumor Necrosis Factor/blood , Receptors, Tumor Necrosis Factor/metabolism , T-Lymphocytes/metabolism , Adult , Aged , Angina, Unstable/immunology , Angina, Unstable/pathology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Cell Line , Female , Humans , Male , Matrix Metalloproteinases/metabolism , Mice , Mice, Knockout , Middle Aged , Monocytes/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Osteoprotegerin , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Rupture, Spontaneous/immunology , Rupture, Spontaneous/metabolism , T-Lymphocytes/pathology
9.
Int J Cardiol ; 215: 506-15, 2016 Jul 15.
Article in English | MEDLINE | ID: mdl-27135822

ABSTRACT

BACKGROUND: Immunization with oxidized LDL (oxLDL) reduces atherosclerosis in rodents. We tested the hypothesis that treatment with a human recombinant monoclonal antibody against oxLDL will reduce the burden or composition of atherosclerotic lesions in hypercholesterolemic minipigs. METHODS AND RESULTS: Thirty-eight hypercholesterolemic minipigs with defective LDL receptors were injected with an oxLDL antibody or placebo weekly for 12weeks. An 18F-fluorodeoxyglucose positron emission tomography (FDG PET) scan (n=9) was performed before inclusion and after 3months of treatment. Blood samples were obtained prior to each injection. Following the last injection all animals were sacrificed, and the heart, aorta, and iliac arteries were removed. The left anterior descending coronary artery was sectioned at 5mm intervals for quantitative and qualitative assessments of atherosclerosis, including immunohistochemical phenotyping of macrophages using a pan-macrophage marker (CD68) and markers for putative pro-atherogenic (cathepsin S) and atheroprotective (CD163) macrophages. Aorta, right coronary artery, and left iliac artery were stained en face with Sudan IV and the amount of atherosclerosis quantified. There was no effect of treatment on plasma lipid profile, vascular FDG-PET signal or the amount of atherosclerosis in any of the examined arteries. However, immunostaining of coronary lesions revealed reduced cathepsin S positivity in the treated group compared with placebo (4.8% versus 8.2% of intima area, p=0.03) with no difference in CD68 or CD163 positivity. CONCLUSIONS: In hypercholesterolemic minipigs, treatment with a human recombinant monoclonal antibody against oxLDL reduced cathepsin S in coronary lesions without any effect on the burden of atherosclerosis or aortic FDG-PET signal.


Subject(s)
Antibodies, Monoclonal/pharmacology , Atherosclerosis/blood , Atherosclerosis/drug therapy , Cathepsins/blood , Hypercholesterolemia/drug therapy , Recombinant Proteins/pharmacology , Animals , Atherosclerosis/diagnostic imaging , Disease Models, Animal , Female , Fluorodeoxyglucose F18 , Humans , Hypercholesterolemia/diagnostic imaging , Lipids/blood , Positron-Emission Tomography/methods , Swine , Treatment Outcome
10.
Arterioscler Thromb Vasc Biol ; 24(6): 1062-7, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15059809

ABSTRACT

OBJECTIVE: To investigate the effect of complement deficiency on atherogenesis and lipidemia, we used mice deficient in the third complement component (C3-/-) or factor B (FB-/-). METHODS AND RESULTS: Complement-deficient mice were crossed with mice deficient in both apolipoprotein E and the low-density lipoprotein receptor (Apoe-/- LDLR-/-). The percent lesion area in the aorta at 16 weeks, determined by en face analysis, was 84% higher in C3-/- mice than in controls (11.8%+/-0.4% versus 6.4%+/-0.8%, mean+/-SEM, P<0.00005). The C3-/- mice also had 58% higher serum triglyceride levels (P<0.05) and a more proatherogenic lipoprotein profile, with significantly more low-density lipoprotein cholesterol and very-low-density lipoprotein triglycerides than control mice. The C3-/- mice weighed 13% less (P<0.01) and had a lower body fat content (3.5%+/-1.0% versus 13.1%+/-3.0%, P<0.01). There were no differences between FB-/- mice and controls. CONCLUSIONS: Complement activation by the classical or lectin pathway exerts atheroprotective effects, possibly through the regulation of lipid metabolism.


Subject(s)
Aortic Diseases/genetics , Arteriosclerosis/genetics , Complement C3/deficiency , Complement Factor B/deficiency , Animals , Aortic Diseases/blood , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arteriosclerosis/blood , Arteriosclerosis/pathology , Cholesterol, LDL/blood , Complement C3/genetics , Complement C3/physiology , Complement Factor B/genetics , Complement Factor B/physiology , Complement Pathway, Alternative/genetics , Complement Pathway, Classical/genetics , Crosses, Genetic , Female , Genetic Predisposition to Disease , Hyperlipoproteinemia Type II/blood , Hyperlipoproteinemia Type II/genetics , Hyperlipoproteinemia Type IV/blood , Hyperlipoproteinemia Type IV/genetics , Lipoproteins, VLDL/blood , Male , Mice , Mice, Knockout , Receptors, LDL/deficiency , Receptors, LDL/genetics , Triglycerides/blood
12.
Sci Transl Med ; 5(196): 196ra100, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23903754

ABSTRACT

Adaptive immunity has a major impact on atherosclerosis, with pro- and anti-atherosclerotic effects exerted by different subpopulations of T cells. Transforming growth factor-ß (TGF-ß) may promote development either of anti-atherosclerotic regulatory T cells or of T helper 17 (TH17) cells, depending on factors in the local milieu. We have addressed the effect on atherosclerosis of enhanced TGF-ß signaling in T cells. Bone marrow from mice with a T cell-specific deletion of Smad7, a potent inhibitor of TGF-ß signaling, was transplanted into hypercholesterolemic Ldlr(-/-) mice. Smad7-deficient mice had significantly larger atherosclerotic lesions that contained large collagen-rich caps, consistent with a more stable phenotype. The inflammatory cytokine interleukin-6 (IL-6) was expressed in the atherosclerotic aorta, and increased mRNA for IL-17A and the TH17-specific transcription factor RORγt were detected in draining lymph nodes. Treating Smad7-deficient chimeras with neutralizing IL-17A antibodies reversed stable cap formation. IL-17A stimulated collagen production by human vascular smooth muscle cells, and RORγt mRNA correlated positively with collagen type I and α-smooth muscle actin mRNA in a biobank of human atherosclerotic plaques. These data link IL-17A to induction of a stable plaque phenotype, could lead to new plaque-stabilizing therapies, and should prompt an evaluation of cardiovascular events in patients treated with IL-17 receptor blockade.


Subject(s)
Interleukin-17/metabolism , Plaque, Atherosclerotic/pathology , Signal Transduction , T-Lymphocytes/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Aorta/pathology , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Chimera , Collagen/biosynthesis , Humans , Immunohistochemistry , Integrases/metabolism , Interleukin-17/immunology , Interleukin-6/immunology , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/metabolism , Receptors, LDL/deficiency , Receptors, LDL/metabolism , Signal Transduction/drug effects , Smad7 Protein/deficiency , Smad7 Protein/metabolism , T-Lymphocytes/drug effects , Th17 Cells/drug effects , Th17 Cells/metabolism
14.
Annu Rev Pathol ; 1: 297-329, 2006.
Article in English | MEDLINE | ID: mdl-18039117

ABSTRACT

Atherosclerosis, the cause of myocardial infarction, stroke, and ischemic gangrene, is an inflammatory disease. The atherosclerotic process is initiated when cholesterol-containing low-density lipoproteins accumulate in the intima and activate the endothelium. Leukocyte adhesion molecules and chemokines promote recruitment of monocytes and T cells. Monocytes differentiate into macrophages and upregulate pattern recognition receptors, including scavenger receptors and toll-like receptors. Scavenger receptors mediate lipoprotein internalization, which leads to foam-cell formation. Toll-like receptors transmit activating signals that lead to the release of cytokines, proteases, and vasoactive molecules. T cells in lesions recognize local antigens and mount T helper-1 responses with secretion of pro-inflammatory cytokines that contribute to local inflammation and growth of the plaque. Intensified inflammatory activation may lead to local proteolysis, plaque rupture, and thrombus formation, which causes ischemia and infarction. Inflammatory markers are already used to monitor the disease process and anti-inflammatory therapy may be useful to control disease activity.


Subject(s)
Adaptation, Physiological/immunology , Atherosclerosis/immunology , Atherosclerosis/pathology , Inflammation/immunology , Inflammation/pathology , Animals , Arteries/immunology , Arteries/pathology , Atherosclerosis/drug therapy , Autoimmunity , Biomarkers , Disease Models, Animal , Humans , Immunity, Innate , Immunologic Factors/therapeutic use , Inflammation/drug therapy
15.
Annu Rev Immunol ; 24: 99-146, 2006.
Article in English | MEDLINE | ID: mdl-16551245

ABSTRACT

Transforming growth factor-beta (TGF-beta) is a potent regulatory cytokine with diverse effects on hemopoietic cells. The pivotal function of TGF-beta in the immune system is to maintain tolerance via the regulation of lymphocyte proliferation, differentiation, and survival. In addition, TGF-beta controls the initiation and resolution of inflammatory responses through the regulation of chemotaxis, activation, and survival of lymphocytes, natural killer cells, dendritic cells, macrophages, mast cells, and granulocytes. The regulatory activity of TGF-beta is modulated by the cell differentiation state and by the presence of inflammatory cytokines and costimulatory molecules. Collectively, TGF-beta inhibits the development of immunopathology to self or nonharmful antigens without compromising immune responses to pathogens. This review highlights the findings that have advanced our understanding of TGF-beta in the immune system and in disease.


Subject(s)
Transforming Growth Factor beta/immunology , Animals , Atherosclerosis/immunology , Autoimmune Diseases/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation , Cell Proliferation , Cell Survival , Dendritic Cells/immunology , Granulocytes/immunology , Humans , Immunoglobulin A/biosynthesis , Infections/immunology , Killer Cells, Natural/immunology , Macrophages/immunology , Mast Cells/immunology , Models, Immunological , Neoplasms/immunology , Signal Transduction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Transforming Growth Factor beta/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL