Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
Zh Evol Biokhim Fiziol ; 45(2): 202-7, 2009.
Article in Russian | MEDLINE | ID: mdl-19435263

ABSTRACT

Obstructive apnea during sleep is accompanied by intermittent hypoxia (IH) leading to hypertension and other cardiovascular disturbances. A comparative evaluation of long-term effects of the neonatal IH on the cardiovascular function was performed in normotensive Sprague-Dawley and spontaneously hypertensive rats (SHR). The newborn rats were placed for 30 days to conditions of IH (8 and 21% O2, alternating every 90 s for 12 h/day). Control groups of rats were constantly kept in normoxia. By 6 months, in the spontaneously hypertensive rats submitted to IH at the period of wakefulness there was a statistically significant increase (as compared with control) of the systolic (correspondingly 185.8 +/- 1.7 and 169.9 +/- 1.4 mm Hg, p < 0.01) and diastolic pressure (correspondingly 96.2 +/- 4.9 and 86.0 +/- 2.6 mm Hg, p < 0.01). During sleep, the systolic and diastolic pressure in these rats was higher than in control animals by 10 mm Hg (p < 0.01) and 12 mm Hg (p < 0.01), its decrease during sleep being absent. SHR submitted to IH had an increase in low- to the high-frequency power ratio of the heart rate variability from 0.9 +/- 0.15 to 1.5 +/- 0.17, which indicates a shift of the sympatho-parasympathetic balance in this group towards predominance of the sympathetic component. In the Sprague-Dawley rats submitted to neonatal hypoxia, the above changes were not pronounced. These peculiarities of the hypertensive rats allow establishing connection of the genetic factor with the sympathetic mechanism providing long-term consequences of the neonatal IH for the cardiovascular control in these rats.


Subject(s)
Hypertension/etiology , Hypertension/physiopathology , Hypoxia/complications , Hypoxia/physiopathology , Animals , Animals, Newborn , Blood Pressure/physiology , Hypertension/genetics , Hypoxia/genetics , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/genetics , Sleep Apnea, Obstructive/physiopathology
2.
Circ Res ; 92(5): 493-500, 2003 Mar 21.
Article in English | MEDLINE | ID: mdl-12600886

ABSTRACT

During angiogenesis, microvascular endothelial cells (ECs) secrete proteinases that permit penetration of the vascular basement membrane as well as the interstitial extracellular matrix. This study tested the hypothesis that cathepsin S (Cat S) contributes to angiogenesis. Treatment of cultured ECs with inflammatory cytokines or angiogenic factors stimulated the expression of Cat S, whereas inhibition of Cat S activity reduced microtubule formation by impairing cell invasion. ECs from Cat S-deficient mice showed reduced collagenolytic activity and impaired invasion of collagens type I and IV. Cat S-deficient mice displayed defective microvessel development during wound repair. This abnormal angiogenesis occurred despite normal vascular endothelial growth factor and basic fibroblast growth factor levels, implying an essential role for extracellular matrix degradation by Cat S during microvessel formation. These results demonstrate a novel function of endothelium-derived Cat S in angiogenesis.


Subject(s)
Cathepsins/physiology , Endothelium, Vascular/enzymology , Endothelium, Vascular/growth & development , Animals , Capillaries/cytology , Cathepsins/genetics , Cell Adhesion , Cell Movement , Cells, Cultured , Collagen/metabolism , Elastin/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Humans , Mice , Mice, Knockout , Wound Healing
4.
Circulation ; 104(16): 1899-904, 2001 Oct 16.
Article in English | MEDLINE | ID: mdl-11602491

ABSTRACT

BACKGROUND: Loss of interstitial collagen, particularly type I collagen, the major load-bearing molecule of atherosclerotic plaques, renders atheroma prone to rupture. Initiation of collagen breakdown requires interstitial collagenases, a matrix metalloproteinase (MMP) subfamily consisting of MMP-1, MMP-8, and MMP-13. Previous work demonstrated the overexpression of MMP-1 and MMP-13 in human atheroma. However, no study has yet evaluated the expression of MMP-8, known as "neutrophil collagenase," the enzyme that preferentially degrades type I collagen, because granulocytes do not localize in plaques. METHODS AND RESULTS: Transcriptional profiling and reverse transcription-polymerase chain reaction analysis revealed inducible expression of MMP-8 transcripts in CD40 ligand-stimulated mononuclear phagocytes. Western blot analysis demonstrated that 3 atheroma-associated cell types, namely, endothelial cells, smooth muscle cells, and mononuclear phagocytes, expressed MMP-8 in vitro upon stimulation with proinflammatory cytokines such as interleukin-1beta, tumor necrosis factor-alpha, or CD40 ligand. MMP-8 protein elaborated from these atheroma-associated cell types migrated as 2 immunoreactive bands, corresponding to the molecular weights of the zymogen and the active molecule. Extracts from atherosclerotic, but not nondiseased arterial tissue, contained similar immunoreactive bands. Moreover, all 3 cell types expressed MMP-8 mRNA and protein in human atheroma in situ. Notably, MMP-8 colocalized with cleaved but not intact type I collagen within the shoulder region of the plaque, a frequent site of rupture. CONCLUSIONS: These data point to MMP-8 as a previously unsuspected participant in collagen breakdown, an important determinant of the vulnerability of human atheroma.


Subject(s)
Arteriosclerosis/enzymology , Collagen/metabolism , Gene Expression Profiling , Matrix Metalloproteinase 8/biosynthesis , Matrix Metalloproteinase 8/genetics , Aorta/enzymology , Aorta/pathology , Arteriosclerosis/pathology , CD40 Ligand , Carotid Arteries/enzymology , Carotid Arteries/pathology , Cells, Cultured , Cytokines/pharmacology , Endothelium, Vascular/enzymology , Endothelium, Vascular/pathology , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/enzymology , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Phagocytes/enzymology , Phagocytes/pathology , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
5.
J Clin Invest ; 107(9): 1117-26, 2001 May.
Article in English | MEDLINE | ID: mdl-11342575

ABSTRACT

Degradation of ECM, particularly interstitial collagen, promotes plaque instability, rendering atheroma prone to rupture. Previous studies implicated matrix metalloproteinases (MMPs) in these processes, suggesting that dysregulated MMP activity, probably due to imbalance with endogenous inhibitors, promotes complications of atherosclerosis. We report here that the serine proteinase inhibitor tissue factor pathway inhibitor-2 (TFPI-2) can function as an MMP inhibitor. TFPI-2 diminished the ability of the interstitial collagenases MMP-1 and MMP-13 to degrade triple-helical collagen, the primary load-bearing molecule of the ECM within human atheroma. In addition, TFPI-2 also reduced the activity of the gelatinases MMP-2 and MMP-9. In contrast to the "classical" tissue inhibitors of MMPs (TIMPs), TFPI-2 expression in situ correlated inversely with MMP levels in human atheroma. TFPI-2 colocalized primarily with smooth muscle cells in the normal media as well as the plaque's fibrous cap. Conversely, the macrophage-enriched shoulder region, the prototypical site of matrix degradation and plaque rupture, stained only weakly for TFPI-2 but intensely for gelatinases and interstitial collagenases. Evidently, human mononuclear phagocytes, an abundant source of MMPs within human atheroma, lost their ability to express this inhibitor during differentiation in vitro. These findings establish a new, anti-inflammatory function of TFPI-2 of potential pathophysiological significance for human diseases, including atherosclerosis.


Subject(s)
Arteriosclerosis/etiology , Glycoproteins/pharmacology , Matrix Metalloproteinase Inhibitors , Serpins/pharmacology , Aorta/pathology , Carotid Arteries/pathology , Dose-Response Relationship, Drug , Glycoproteins/genetics , Humans , Macrophages/metabolism , Muscle, Smooth, Vascular/metabolism , Procollagen/metabolism , Protein Binding , Protein Processing, Post-Translational/drug effects , Recombinant Proteins/pharmacology
6.
Am J Pathol ; 158(3): 879-91, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238037

ABSTRACT

Inflammation and oxidative stress contribute to the pathogenesis of many human diseases including atherosclerosis. Advanced human atheroma contains high levels of the enzyme myeloperoxidase that produces the pro-oxidant species, hypochlorous acid (HOCl). This study documents increased numbers of myeloperoxidase-expressing macrophages in eroded or ruptured plaques causing acute coronary syndromes. In contrast, macrophages in human fatty streaks contain little or no myeloperoxidase. Granulocyte macrophage colony-stimulating factor, but not macrophage colony-stimulating factor, selectively regulates the ability of macrophages to express myeloperoxidase and produce HOCl in vitro. Moreover, myeloperoxidase-positive macrophages in plaques co-localized with granulocyte macrophage colony-stimulating factor. Pro-inflammatory stimuli known to be present in human atherosclerotic plaque, including CD40 ligand, lysophosphatidylcholine, or cholesterol crystals, could induce release of myeloperoxidase from HOCl production by macrophages in vitro. HOCl-modified proteins accumulated at ruptured or eroded sites of human coronary atheroma. These results identify granulocyte macrophage colony-stimulating factor as an endogenous regulator of macrophage myeloperoxidase expression in human atherosclerosis and support a particular role for the myeloperoxidase-expressing macrophages in atheroma complication and the acute coronary syndromes.


Subject(s)
Arteriosclerosis/etiology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/enzymology , Myocardial Infarction/etiology , Peroxidase/biosynthesis , Arteriosclerosis/enzymology , Arteriosclerosis/pathology , CD40 Antigens/pharmacology , Cell Differentiation , Cells, Cultured , Cholesterol/pharmacology , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Hypochlorous Acid/metabolism , Lysophosphatidylcholines/pharmacology , Macrophages/drug effects , Monocytes/cytology , Monocytes/drug effects , Monocytes/enzymology , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Phenotype , Reactive Oxygen Species/metabolism , Syndrome , Tunica Intima/enzymology
7.
Proc Natl Acad Sci U S A ; 97(13): 7458-63, 2000 Jun 20.
Article in English | MEDLINE | ID: mdl-10861012

ABSTRACT

Interruption of inflammatory pathways may provide a novel approach to the therapy of atherosclerosis. Recently, we and others have implicated the immune mediator dyad CD40/CD40L (CD40 ligand), which is expressed on endothelial and smooth muscle cells, macrophages, and T lymphocytes within human atherosclerotic lesions, in aspects of atherogenesis and the acute coronary syndromes, including regulation of matrix metalloproteinases, procoagulant activity, cytokines, etc. In vivo, interruption of CD40 signaling reduced the initiation and early phases of atheroma formation in hypercholesterolemic mice. However, whether interruption of CD40 signaling can retard the progression or even regress established lesions remains unknown. We report here that anti-CD40L antibody treatment of randomly assigned low-density lipoprotein receptor-deficient mice during the second half of a 26-week regimen of high-cholesterol diet did not regress, but did significantly reduce further evolution of established atherosclerotic lesions within the aortic arch and particularly the thoracic and abdominal aorta, as compared with control treatment (application of rat-IgG or saline; 13 weeks, continued high-cholesterol diet). In addition to limiting lesion progression, anti-CD40L treatment changed the composition of atheroma in manners thought to favor plaque stability, e.g., reduced relative content of macrophages and lipid, as well as increased relative content of smooth muscle cells and collagen. These data implicate CD40/CD40L as crucial mediators not only in the initial events of atherogenesis but also during the evolution of established atheroma. This study lends further support to the importance of this specific inflammatory signaling pathway in atherosclerosis and its complications.


Subject(s)
Arteriosclerosis/immunology , CD40 Antigens/immunology , Membrane Glycoproteins/immunology , Animals , Antibodies/administration & dosage , Antibodies/immunology , Arteriosclerosis/physiopathology , CD40 Ligand , Diet, Atherogenic , Humans , Inflammation/immunology , Inflammation/physiopathology , Mice , Receptors, LDL/deficiency , Receptors, LDL/immunology
8.
J Exp Med ; 191(9): 1535-44, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10790428

ABSTRACT

Interleukin-1beta-converting enzyme (ICE, caspase-1) regulates key steps in inflammation and immunity, by activating the proinflammatory cytokines interleukin (IL-)1beta and IL-18, or mediating apoptotic processes. We recently provided evidence for the regulation of caspase-1 activity via an endogenous inhibitor expressed by human vascular smooth muscle cells (SMCs) (Schönbeck, U., M. Herzberg, A. Petersen, C. Wohlenberg, J. Gerdes, H.-D. Flad, and H. Loppnow. 1997. J. Exp. Med. 185:1287-1294). However, the molecular identity of this endogenous inhibitor remained undefined. We report here that the serine proteinase inhibitor (serpin) PI-9 accounts for the endogenous caspase-1 inhibitory activity in human SMCs and prevents processing of the enzyme's natural substrates, IL-1beta and IL-18 precursor. Treatment of SMC lysates with anti-PI-9 antibody abrogated the caspase-1 inhibitory activity and coprecipitated the enzyme, demonstrating protein-protein interaction. Furthermore, PI-9 antisense oligonucleotides coordinately reduced PI-9 expression and promoted IL-1beta release. Since SMCs comprise the majority of cells in the vascular wall, and because IL-1 is implicated in atherogenesis, we tested the biological validity of our in vitro findings within human atheroma in situ. The unaffected arterial wall contains abundant and homogeneously distributed PI-9. In human atherosclerotic lesions, however, PI-9 expression correlated inversely with immunoreactive IL-1beta, supporting a potential role of the endogenous caspase-1 inhibitor in this chronic inflammatory disease. Thus, our results provide new insights into the regulation of this enzyme involved in immune and inflammatory processes of chronic inflammatory diseases, and point to an endogenous antiinflammatory action of PI-9, dysregulated in a prevalent human disease.


Subject(s)
Caspase Inhibitors , Interleukins/biosynthesis , Muscle, Smooth, Vascular/enzymology , Protein Processing, Post-Translational , Serpins/isolation & purification , Arteries/chemistry , Arteries/pathology , Arteriosclerosis/pathology , Gene Expression Regulation , Humans , Interleukin-1/biosynthesis , Interleukin-18/biosynthesis , Protein Precursors/metabolism
9.
Circ Res ; 86(2): 131-8, 2000 Feb 04.
Article in English | MEDLINE | ID: mdl-10666407

ABSTRACT

Chemokines are chemotactic cytokines that activate and direct the migration of leukocytes. However, their role in modulating platelet function has not been shown. We studied the direct effect of chemokines on human platelets and found that of the 16 tested only stromal cell-derived factor (SDF)-1 induced platelet aggregation, accompanied by a rise in intracellular calcium. Platelets expressed the SDF-1 receptor, CXCR4, and an antibody to CXCR4 and pertussis toxin inhibited SDF-1-induced platelet aggregation, confirming that this effect is mediated through CXCR4, a Galphai-coupled receptor. SDF-1-induced platelet aggregation was also inhibited by wortmannin, LY294002, and genistein, suggesting that phosphatidylinositol 3-kinase and tyrosine kinase are likely involved in SDF-1-induced platelet aggregation. Because chemokines are produced from multiple vascular cells and atherosclerotic vessels are prone to develop platelet-rich thrombi, we examined the expression of SDF-1 in human atheroma. SDF-1 protein was highly expressed in smooth muscle cells, endothelial cells, and macrophages in human atherosclerotic plaques but not in normal vessels. Our studies demonstrate a direct effect of a chemokine in inducing platelet activation and suggest a role for SDF-1 in the pathogenesis of atherosclerosis and thrombo-occlusive diseases.


Subject(s)
Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Chemokines, CXC/pharmacology , Platelet Aggregation/drug effects , Antibodies, Monoclonal , Aortic Diseases/metabolism , Aortic Diseases/pathology , Biological Transport/drug effects , Blotting, Western , Calcium/metabolism , Chemokine CXCL12 , Chemokines, CXC/analysis , Chemokines, CXC/immunology , Chemokines, CXC/metabolism , Female , Flow Cytometry , Humans , Immunoenzyme Techniques , Male , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Signal Transduction/drug effects
10.
Am J Pathol ; 156(1): 7-14, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10623647

ABSTRACT

Tissue factor (TF) instigates the extrinsic pathway of blood coagulation. Plaque disruption and exposure of circulating factor VII/VIIa to subendothelial procoagulants such as TF leads to intravascular thrombosis, a frequent cause of acute atherosclerotic events. Although the expression of TF in the intima of human atherosclerotic lesions is well established, little is known about the mechanisms of TF regulation in vascular smooth muscle cells (SMC). We demonstrate here that TF colocalizes with the receptor CD40 on lesional SMC within atherosclerotic lesions in situ. In cultured vascular SMC, ligation of CD40 with native CD40 ligand (CD40L) derived from activated T lymphocytes or recombinant human CD40L (rCD40L) induced the transient expression of TF on the cell surface (as determined by FACS analysis) in a concentration- and time-dependent manner and enhanced total cell-associated TF (as determined by ELISA). CD40L-induced TF on vascular SMC is functional and activates coagulation. In accordance with the increased TF activity, stimulation of vascular SMC with rCD40L did not affect either protein expression or activity of tissue factor pathway inhibitors. In summary, these findings demonstrate the potential of the CD40/CD40L signaling pathway to augment the procoagulant activity in human vascular SMC. Because TF and CD40 colocalize on lesional SMC in human atheroma, CD40/CD40L signaling may contribute to the TF expression and hence to increased thrombogenicity of plaques during the inflammatory responses of atherogenesis and arterial injury.


Subject(s)
CD40 Antigens/metabolism , Membrane Glycoproteins/metabolism , Muscle, Smooth, Vascular/metabolism , Thromboplastin/metabolism , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , CD40 Ligand , Cell Membrane , Cells, Cultured , Humans , Lipoproteins/metabolism , Muscle, Smooth, Vascular/cytology
11.
J Clin Invest ; 104(9): 1191-7, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10545518

ABSTRACT

The pathogenesis of atherosclerosis and abdominal aortic aneurysm involves breakdown of the elastic laminae. Elastolytic cysteine proteases, including cathepsins S and K, are overexpressed at sites of arterial elastin damage, but whether endogenous local inhibitors counterbalance these proteases is unknown. We show here that, whereas cystatin C is normally expressed in vascular wall smooth muscle cells (SMCs), this cysteine protease inhibitor is severely reduced in both atherosclerotic and aneurysmal aortic lesions. Furthermore, increased abdominal aortic diameter among 122 patients screened by ultrasonography correlated inversely with serum cystatin C levels. In vitro, cytokine-stimulated vascular SMCs secrete cathepsins, whose elastolytic activity could be blocked when cystatin C secretion was induced by treatment with TGF-beta(1). The findings highlight a potentially important role for imbalance between cysteine proteases and cystatin C in arterial wall remodeling and establish that cystatin C deficiency occurs in vascular disease.


Subject(s)
Aortic Aneurysm, Abdominal/metabolism , Arteriosclerosis/metabolism , Cystatins/deficiency , Cysteine Proteinase Inhibitors/deficiency , Aorta/pathology , Aortic Aneurysm, Abdominal/pathology , Arteries/metabolism , Arteries/pathology , Arteriosclerosis/pathology , Cells, Cultured , Cystatin C , Cystatins/blood , Cysteine Proteinase Inhibitors/blood , Dose-Response Relationship, Drug , Humans , Immunoblotting , Immunohistochemistry , Interferon-gamma/metabolism , Muscle, Smooth/metabolism , Transforming Growth Factor beta/metabolism
12.
J Clin Invest ; 104(8): 1041-50, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10525042

ABSTRACT

Activated T lymphocytes accumulate early in atheroma formation and persist at sites of lesion growth and rupture, suggesting that they may play an important role in the pathogenesis of atherosclerosis. Moreover, atherosclerotic lesions contain the Th1-type cytokine IFN-gamma, a potentiator of atherosclerosis. The present study demonstrates the differential expression of the 3 IFN-gamma-inducible CXC chemokines--IFN-inducible protein 10 (IP-10), monokine induced by IFN-gamma (Mig), and IFN-inducible T-cell alpha chemoattractant (I-TAC)--by atheroma-associated cells, as well as the expression of their receptor, CXCR3, by all T lymphocytes within human atherosclerotic lesions in situ. Atheroma-associated endothelial cells (ECs), smooth muscle cells (SMCs), and macrophages (MO) all expressed IP-10, whereas Mig and I-TAC were mainly expressed in ECs and MO, as detected by double immunofluorescence staining. ECs of microvessels within lesions also expressed abundant I-TAC. In vitro experiments supported these results and showed that IL-1beta, TNF-alpha, and CD40 ligand potentiated IP-10 expression from IFN-gamma-stimulated ECs. In addition, nitric oxide (NO) treatment decreased IFN-gamma induction of IP-10. Our findings suggest that the differential expression of IP-10, Mig, and I-TAC by atheroma-associated cells plays a role in the recruitment and retention of activated T lymphocytes observed within vascular wall lesions during atherogenesis.


Subject(s)
Arteriosclerosis/immunology , Chemokines, CXC/analysis , Intercellular Signaling Peptides and Proteins , Animals , Cells, Cultured , Chemokine CXCL10 , Chemokine CXCL11 , Chemokine CXCL9 , Chemokines, CXC/genetics , Humans , Immunohistochemistry , Nitric Oxide/physiology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , RNA, Messenger/analysis , Rabbits , Receptors, CXCR3 , Receptors, Chemokine/analysis
13.
Am J Pathol ; 155(4): 1281-91, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10514410

ABSTRACT

Cyclooxygenase-1 (Cox-1) and Cox-2 convert arachidonic acid to prostaglandin H(2), the precursor of other prostaglandins and thromboxanes, eicosanoids important in vascular pathophysiology. However, knowledge of the expression of cyclooxygenases within atherosclerotic lesions is scant. This study tested the hypothesis that human atheroma and nonatherosclerotic arteries express the two Cox isoforms differentially. Cox-1 mRNA and protein localized on endothelial and medial smooth muscle cells of normal arteries (n = 5), whereas Cox-2 expression was not detectable. In contrast, atheromatous (n = 7) lesions contained both Cox-1 and Cox-2, colocalizing mainly with macrophages of the shoulder region and lipid core periphery, whereas smooth muscle cells showed lower levels, as demonstrated by immunohistochemical and in situ hybridization analysis. Furthermore, microvascular endothelium in plaques showed notable staining for both isoforms. In accord with immunohistochemical studies, Western blot analysis of protein extracts from normal arteries revealed constitutive Cox-1, but not Cox-2, expression. Extracts of atheromatous lesions, however, contained both Cox-1 and Cox-2 protein, detected as two immunoreactive proteins of approximately 70 and 50 kd. Macrophages expressed the short form of Cox-1/-2 constitutively after several days of in vitro culture, rather than the 70-kd protein. These results shed new light on the inflammatory pathways that operate in human atheroma. In particular, the expression of Cox-2 in atheromatous, but not in unaffected, arteries has therapeutic implications, given the advent of selective Cox-2 inhibitors.


Subject(s)
Arteriosclerosis/enzymology , Isoenzymes/biosynthesis , Prostaglandin-Endoperoxide Synthases/biosynthesis , Blotting, Western , Cells, Cultured , Cyclooxygenase 1 , Cyclooxygenase 2 , Endothelium, Vascular/enzymology , Humans , Immunohistochemistry , In Situ Hybridization , Isoenzymes/genetics , Macrophages/enzymology , Membrane Proteins , Microcirculation/enzymology , Muscle, Smooth, Vascular/enzymology , Prostaglandin-Endoperoxide Synthases/genetics , RNA, Messenger/biosynthesis
14.
Am J Pathol ; 155(3): 775-86, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10487835

ABSTRACT

Atherosclerotic plaques are chronic inflammatory lesions composed of dysfunctional endothelium, smooth muscle cells, lipid-laden macrophages, and T lymphocytes. This study analyzed atherosclerotic tissue specimens for expression of CD1 molecules, a family of cell surface proteins that present lipid antigens to T cells, and examined the possibility that CD1+ lipid-laden macrophages might present antigen to T cells. Immunohistochemical studies using a panel of specific monoclonal antibodies demonstrated expression of each of the four previously characterized human CD1 proteins (CD1a, -b, -c, and -d) in atherosclerotic plaques. Expression of CD1 was not observed in normal arterial specimens and appeared to be restricted to the CD68+ lipid-laden foam cells of atherosclerotic lesions. CD1 molecules colocalized in areas of the arterial wall that also contained abundant T lymphocytes, suggesting potential interactions between CD1+ cells and plaque-infiltrating lymphocytes in situ. Using CD1-expressing foam cells derived from macrophages in vitro, we demonstrated the ability of such cells to present lipid antigens to CD1 restricted T cells. Given the abundant T cells, CD1+ macrophages, and lipid accumulation in atherosclerotic plaques, we propose a potential role for lipid antigen presentation by CD1 proteins in the generation of the inflammatory component of these lesions.


Subject(s)
Antigens, CD1/biosynthesis , Arteriosclerosis/metabolism , Foam Cells/metabolism , Lymphocyte Activation/immunology , T-Lymphocytes/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Arteriosclerosis/immunology , Chick Embryo , Flow Cytometry , Foam Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Inflammation/immunology , Interleukin-4/pharmacology , Lipids/immunology , Microscopy, Fluorescence , Monocytes/drug effects , Monocytes/immunology , T-Lymphocytes/immunology
15.
Circulation ; 99(24): 3125-31, 1999 Jun 22.
Article in English | MEDLINE | ID: mdl-10377075

ABSTRACT

BACKGROUND: Adhesion molecule expression on the endothelial cell (EC) surface is critical for leukocyte recruitment to atherosclerotic lesions. Better understanding of transcriptional regulation of adhesion molecules in ECs may provide important insight into plaque formation. Peroxisome proliferator-activated receptor-alpha (PPARalpha), a member of the nuclear receptor family, regulates gene expression in response to certain fatty acids and fibric acid derivatives. The present study investigated PPARalpha expression in human ECs and their regulation of vascular cell adhesion molecule-1 (VCAM-1). METHODS AND RESULTS: Immunohistochemistry revealed that human carotid artery ECs express PPARalpha. Pretreatment of cultured human ECs with the PPARalpha activators fenofibrate or WY14643 inhibited TNF-alpha-induced VCAM-1 in a time- and concentration-dependent manner, an effect not seen with PPARgamma activators. Both PPARalpha activators decreased cytokine-induced VCAM-1 mRNA expression without altering its mRNA half-life. Transient transfection of deletional VCAM-1 promoter constructs and electrophoretic mobility shift assays suggest that fenofibrate inhibits VCAM-1 transcription in part by inhibiting NF-kappaB. Finally, PPARalpha activators significantly reduced adhesion of U937 cells to cultured human ECs. CONCLUSIONS: Human ECs express PPARalpha, a potentially important regulator of atherogenesis through its transcriptional control of VCAM-1 gene expression. Such findings also have implications regarding the clinical use of lipid-lowering agents, like fibric acids, which can activate PPARalpha.


Subject(s)
Endothelium, Vascular/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/genetics , Anticholesteremic Agents/pharmacology , Arteriosclerosis/genetics , Arteriosclerosis/metabolism , Carotid Arteries/chemistry , Carotid Arteries/cytology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Fenofibrate/pharmacology , Gene Expression/drug effects , Gene Expression/physiology , Humans , Hypolipidemic Agents/pharmacology , Monocytes/cytology , Monocytes/physiology , NF-kappa B/metabolism , Promoter Regions, Genetic/physiology , Pyrimidines/pharmacology , RNA, Messenger/analysis , Receptors, Cytoplasmic and Nuclear/metabolism , Saphenous Vein/cytology , Transcription Factors/metabolism , Transfection , U937 Cells
16.
Circulation ; 99(19): 2503-9, 1999 May 18.
Article in English | MEDLINE | ID: mdl-10330380

ABSTRACT

BACKGROUND: Several recent studies attempted to classify plaques as those prone to cause clinical manifestations (vulnerable, atheromatous plaques) or those less frequently associated with acute thrombotic complication (stable, fibrous plaques). Defining the cellular and molecular mechanisms that underlie these morphological features remains a challenge. Because interstitial forms of collagen determine the biomechanical strength of the atherosclerotic lesion, this study investigated expression of the collagen-degrading matrix metalloproteinase (MMP) interstitial collagenase-3 (MMP-13) and the previously studied MMP-1 in human atheroma and used a novel technique to test the hypothesis that collagenolysis in atheromatous lesions exceeds that in fibrous human atherosclerotic lesions. METHODS AND RESULTS: Human carotid atherosclerotic plaques, similar in size, were separated by conventional morphological characteristics into fibrous (n=10) and atheromatous (n=10) lesions. Immunohistochemical and Western blot analysis demonstrated increased levels of MMP-1 and MMP-13 in atheromatous versus fibrous plaques. In addition, collagenase-cleaved type I collagen, demonstrated by a novel cleavage-specific antibody, colocalized with MMP-1- and MMP-13-positive macrophages. Macrophages, rather than endothelial or smooth muscle cells, expressed MMP-13 and MMP-1 on stimulation in vitro. Furthermore, Western blot analysis demonstrated loss of interstitial collagen type I and increased collagenolysis in atheromatous versus fibrous lesions. Finally, atheromatous plaques contained higher levels of proinflammatory cytokines, activators of MMPs. CONCLUSIONS: This report demonstrates that atheromatous rather than fibrous plaques might be prone to rupture due to increased collagenolysis associated with macrophages, probably mediated by the interstitial collagenases MMP-1 and MMP-13.


Subject(s)
Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Collagen/metabolism , Collagenases/metabolism , Blotting, Western , Carotid Arteries/metabolism , Carotid Arteries/pathology , Humans , Immunohistochemistry , Matrix Metalloproteinase 1 , Matrix Metalloproteinase 13 , Rupture
17.
J Exp Med ; 189(5): 843-53, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10049948

ABSTRACT

Stromelysin-3 is an unusual matrix metalloproteinase, being released in the active rather than zymogen form and having a distinct substrate specificity, targeting serine proteinase inhibitors (serpins), which regulate cellular functions involved in atherosclerosis. We report here that human atherosclerotic plaques (n = 7) express stromelysin-3 in situ, whereas fatty streaks (n = 5) and normal arterial specimens (n = 5) contain little or no stromelysin-3. Stromelysin-3 mRNA and protein colocalized with endothelial cells, smooth muscle cells, and macrophages within the lesion. In vitro, usual inducers of matrix metalloproteinases such as interleukin-1, interferon-gamma, or tumor necrosis factor alpha did not augment stromelysin-3 in vascular wall cells. However, T cell-derived as well as recombinant CD40 ligand (CD40L, CD154), an inflammatory mediator recently localized in atheroma, induced de novo synthesis of stromelysin-3. In addition, stromelysin-3 mRNA and protein colocalized with CD40L and CD40 within atheroma. In accordance with the in situ and in vitro data obtained with human material, interruption of the CD40-CD40L signaling pathway in low density lipoprotein receptor-deficient hyperlipidemic mice substantially decreased expression of the enzyme within atherosclerotic plaques. These observations establish the expression of the unusual matrix metalloproteinase stromelysin-3 in human atherosclerotic lesions and implicate CD40-CD40L signaling in its regulation, thus providing a possible new pathway that triggers complications within atherosclerotic lesions.


Subject(s)
Arteriosclerosis/metabolism , CD40 Antigens/metabolism , Membrane Glycoproteins/metabolism , Metalloendopeptidases/biosynthesis , Animals , Aorta/pathology , Arteriosclerosis/pathology , CD40 Ligand , Carotid Arteries/pathology , Endothelium, Vascular/metabolism , Humans , Hyperlipidemias/metabolism , Macrophages/metabolism , Matrix Metalloproteinase 11 , Metalloendopeptidases/isolation & purification , Mice , Mice, Mutant Strains , Muscle, Smooth, Vascular/metabolism , Receptors, LDL/genetics , Signal Transduction
18.
Arterioscler Thromb Vasc Biol ; 19(3): 546-51, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10073956

ABSTRACT

Plasminogen activator inhibitor type-1 (PAI-1) is a major physiological inhibitor of fibrinolysis, with its plasma levels correlating with the risk for myocardial infarction and venous thrombosis. The regulation of PAI-1 transcription by endothelial cells (ECs), a major source of PAI-1, remains incompletely understood. Adipocytes also produce PAI-1, suggesting possible common regulatory pathways between adipocytes and ECs. Peroxisomal proliferator-activated receptor-gamma (PPAR)gamma is a ligand-activated transcription factor that regulates gene expression in response to various mediators such as 15-deoxy-Delta12, 14-prostaglandin J2 (15d-PGJ2) and oxidized linoleic acid (9- and 13-HODE). The present study tested the hypotheses that human ECs express PPARgamma and that this transcriptional activator regulates PAI-1 expression in this cell type. We found that human ECs contain both PPARgamma mRNA and protein. Immunohistochemistry of human carotid arteries also revealed the presence of PPARgamma in ECs. Bovine ECs transfected with a PPAR response element (PPRE)-luciferase construct responded to stimulation by the PPARgamma agonist 15d-PGJ2 in a concentration-dependent manner, suggesting a functional PPARgamma in ECs. Treatment of human ECs with 15d-PGJ2, 9(S)-HODE, or 13(S)-HODE augmented PAI-1 mRNA and protein expression, whereas multiple PPARalpha activators did not change PAI-1 levels. Introduction of increasing amounts of a PPARgamma expression construct in human fibroblasts enhanced PAI-1 secretion from these cells in proportion to the amount of transfected DNA. Thus, ECs express functionally active PPARgamma that regulates PAI-1 expression in ECs. Our results establish a role for PPARgamma in the regulation of EC gene expression, with important implications for the clinical links between obesity and atherosclerosis.


Subject(s)
Arteriosclerosis/metabolism , Plasminogen Activator Inhibitor 1/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Adipose Tissue/blood supply , Adipose Tissue/metabolism , Animals , Arteriosclerosis/genetics , Blood Coagulation , Carotid Arteries/cytology , Carotid Arteries/metabolism , Cattle , Cells, Cultured , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme Activation/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression/physiology , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/pharmacology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Saphenous Vein/cytology , Signal Transduction/physiology , Transfection
19.
Circulation ; 99(1): 96-104, 1999.
Article in English | MEDLINE | ID: mdl-9884385

ABSTRACT

BACKGROUND: Thinning of the tunica media and rarefaction of smooth muscle cells (SMCs) characterize aneurysmal aortas. Apoptosis determines the cellularity and morphogenesis of tissue. Macrophages and T lymphocytes infiltrate the wall of abdominal aortic aneurysms (AAAs) and produce death-promoting proteins (perforin, Fas, and FasL). This study investigated whether apoptosis occurs in association with the expression of these proteins. METHODS AND RESULTS: We examined signs of apoptosis and expression of death-promoting mediators in segments of AAAs from patients undergoing elective repair (n=20). Anti-alpha-actin immunostaining showed a reduced number of SMCs in AAAs. In situ terminal transferase-mediated dUTP nick end-labeling (TUNEL) showed higher levels of DNA fragmentation in AAAs than in controls (n=5). The AAA walls contained more cells bearing markers of apoptosis than normal aorta (P<0.05, Student's t test). Double immunostaining identified SMCs and macrophages as the principal cell types displaying fragmented DNA. Immunohistochemistry revealed that AAAs but not normal aorta contained CD4(+) and CD8(+) T cells that expressed well-characterized cytotoxic mediators: perforin, which produces membrane damage, and Fas, which acts by ligand-receptor interaction. Double immunostaining also identified SMCs that expressed Fas. Immunoblotting confirmed the presence and, in the case of Fas, activation of these proteins in aneurysmal tissue. CONCLUSIONS: Many medial SMCs in AAAs bear markers of apoptosis and signals capable of initiating cell death. Apoptotic death may contribute to the reduction of cellularity and to the impaired repair and maintenance of the arterial extracellular matrix in AAAs. Macrophages and T lymphocytes infiltrate the wall of AAAs, where they can produce cytotoxic mediators such as cytokines, perforin, and Fas/FasL. These death-promoting products of activated immune cells may contribute to elimination of SMCs, a source of elastin and collagen, during the pathogenesis of AAAs.


Subject(s)
Aortic Aneurysm, Abdominal/pathology , Apoptosis/physiology , Muscle, Smooth, Vascular/pathology , T-Lymphocytes/pathology , Aged , Aged, 80 and over , Aortic Aneurysm, Abdominal/immunology , Biomarkers , Case-Control Studies , Cell Death/physiology , Female , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Leukocytes/pathology , Male , Middle Aged
20.
Mol Cell ; 2(2): 275-81, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9734366

ABSTRACT

Recruitment of blood monocytes into the arterial subendothelium is one of the earliest steps in atherogenesis. Monocyte chemoattractant protein-1 (MCP-1), a CC chemokine, is one likely signal involved in this process. To test MCP-1's role in atherogenesis, low density lipoprotein (LDL) receptor-deficient mice were made genetically deficient for MCP-1 and fed a high cholesterol diet. Despite having the same amount of total and fractionated serum cholesterol as LDL receptor-deficient mice with wild-type MCP-1 alleles, LDL receptor/MCP-1-deficient mice had 83% less lipid deposition throughout their aortas. Consistent with MCP-1 's monocyte chemoattractant properties, compound-deficient mice also had fewer macrophages in their aortic walls. Thus, MCP-1 plays a unique and crucial role in the initiation of atherosclerosis and may provide a new therapeutic target in this disorder.


Subject(s)
Arteriosclerosis/prevention & control , Chemokine CCL2/deficiency , Receptors, LDL/deficiency , Animals , Aorta/metabolism , Aorta/pathology , Arteriosclerosis/etiology , Arteriosclerosis/physiopathology , Base Sequence , Cell Movement , Chemokine CCL2/genetics , Chemokine CCL2/physiology , DNA Primers/genetics , Diet, Atherogenic , Female , Lipid Metabolism , Lipids/blood , Macrophages/pathology , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/physiology , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...