1
|
Ugovšek S, Šebeštjen M. Lipoprotein(a)—The Crossroads of Atherosclerosis, Atherothrombosis and Inflammation. Biomolecules 2021; 12:biom12010026. [PMID: 35053174 PMCID: PMC8773759 DOI: 10.3390/biom12010026] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022] Open
Abstract
Increased lipoprotein(a) (Lp(a)) levels are an independent predictor of coronary artery disease (CAD), degenerative aortic stenosis (DAS), and heart failure independent of CAD and DAS. Lp(a) levels are genetically determinated in an autosomal dominant mode, with great intra- and inter-ethnic diversity. Most variations in Lp(a) levels arise from genetic variations of the gene that encodes the apolipoprotein(a) component of Lp(a), the LPA gene. LPA is located on the long arm of chromosome 6, within region 6q2.6–2.7. Lp(a) levels increase cardiovascular risk through several unrelated mechanisms. Lp(a) quantitatively carries all of the atherogenic risk of low-density lipoprotein cholesterol, although it is even more prone to oxidation and penetration through endothelia to promote the production of foam cells. The thrombogenic properties of Lp(a) result from the homology between apolipoprotein(a) and plasminogen, which compete for the same binding sites on endothelial cells to inhibit fibrinolysis and promote intravascular thrombosis. LPA has up to 70% homology with the human plasminogen gene. Oxidized phospholipids promote differentiation of pro-inflammatory macrophages that secrete pro-inflammatory cytokines (e. g., interleukin (IL)-1β, IL-6, IL-8, tumor necrosis factor-α). The aim of this review is to define which of these mechanisms of Lp(a) is predominant in different groups of patients.
Collapse
Affiliation(s)
- Sabina Ugovšek
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Miran Šebeštjen
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
- Department of Cardiology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Vascular Diseases, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Correspondence:
| |
Collapse
|
2
|
Schneider DJ, Hayes M, Wadsworth M, Taatjes H, Rincón M, Taatjes DJ, Sobel BE. Attenuation of Neointimal Vascular Smooth Muscle Cellularity in Atheroma by Plasminogen Activator Inhibitor Type 1 (PAI-1). J Histochem Cytochem 2016; 52:1091-9. [PMID: 15258185 DOI: 10.1369/jhc.4a6260.2004] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Rupture of vulnerable atheroma often underlies acute coronary syndromes. Vulnerable plaques exhibit a paucity of vascular smooth muscle cells (VSMCs) in the cap. Therefore, decreased VSMC migration into the neointima may predispose to vulnerability. The balance between cell surface plasminogen activator activity and its inhibition [mediated primarily by plasminogen activator inhibitor type 1 (PAI-1)] modulates migration of diverse types of cells. We sought to determine whether increased expression of PAI-1 would decrease migration of VSMCs in vitro and neointimal cellularity in vivo in apolipo-protein E knockout (ApoE−-/–) mice fed a high-fat diet. Increased vessel wall expression of PAI-1 in transgenic mice was induced with the SM22α promoter. VSMC migration through Matrigel in vitro was quantified with laser scanning cytometry. Expression of PAI-1 was increased threefold in the aortic wall of SM22-PAI transgene-positive mice. Neointimal cellularity of vascular lesions was decreased by 26% ( p=0.01; n=5 each) in ApoE−-/– mice with the SM22-PAI transgene compared with ApoE−-/– mice. VSMCs explanted from transgene-positive mice exhibited twofold greater expression of PAI-1 and their migration was attenuated by 27% ( p=0.03). Accordingly, increased expression of PAI-1 protein by VSMCs reduces their migration in vitro and their contribution to neointimal cellularity in vivo.
Collapse
Affiliation(s)
- David J Schneider
- University of Vermont, Colchester Research Facility, 208 S. Park Drive, Colchester, VT 05446, USA.
| | | | | | | | | | | | | |
Collapse
|
3
|
Shimizu T, Uematsu M, Yoshizaki T, Obata JE, Nakamura T, Fujioka D, Watanabe K, Watanabe Y, Kugiyama K. Myocardial Production of Plasminogen Activator Inhibitor-1 is Associated with Coronary Endothelial and Ventricular Dysfunction after Acute Myocardial Infarction. J Atheroscler Thromb 2015; 23:557-66. [PMID: 26632165 DOI: 10.5551/jat.32300] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Although plasminogen activator inhibitor-1 (PAI-1) is abundantly expressed in infarcted myocardium, the pathogenic role of myocardial PAI-1 remains unknown. This study examined whether PAI-1 in the infarcted lesion contributes to coronary endothelial dysfunction and left ventricular (LV) dysfunction in patients with acute myocardial infarction (AMI). METHODS Plasma levels of PAI-1 activity and tissue-plasminogen activator (tPA) antigen were measured 2 weeks and 6 months after MI by ELISA in plasma obtained from the aortic root (AO) and anterior interventricular vein (AIV) in 28 patients with a first AMI due to occlusion of the left anterior descending coronary artery (LAD). Coronary blood flow responses in LAD to intracoronary infusion of acetylcholine (ACh) and left ventriculography were measured at the same time points: 2 weeks and 6 months after MI. RESULTS The trans-myocardial gradient of PAI-1 from AO to AIV, reflecting production/release of PAI-1 in the infarcted lesion, was inversely correlated with the coronary blood flow response to ACh 6 months after MI (r=-0.43, p=0.02) and with the percentage change in LV regional motion in the LAD territory from 2 weeks to 6 months after MI (r=-0.38, p=0.04). The trans-myocardial gradient of tPA level showed no significant correlations. CONCLUSIONS PAI-1 produced in the infarcted myocardium and released into the coronary circulation is associated with endothelial dysfunction in resistance vessels of the infarct-related coronary arteries and with progressive dysfunction of the infarcted region of the left ventricle in AMI survivors.
Collapse
Affiliation(s)
- Takuya Shimizu
- University of Yamanashi, Department of Internal Medicine II
| | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Matsuura Y, Yamashita A, Iwakiri T, Sugita C, Okuyama N, Kitamura K, Asada Y. Vascular wall hypoxia promotes arterial thrombus formation via augmentation of vascular thrombogenicity. Thromb Haemost 2015; 114:158-72. [PMID: 25833755 DOI: 10.1160/th14-09-0794] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 01/30/2015] [Indexed: 12/15/2022]
Abstract
Atherosclerotic lesions represent a hypoxic milieu. However, the significance of this milieu in atherothrombosis has not been established. We aimed to assess the hypothesis that vascular wall hypoxia promotes arterial thrombus formation. We examined the relation between vascular wall hypoxia and arterial thrombus formation using a rabbit model in which arterial thrombosis was induced by 0.5 %-cholesterol diet and repeated balloon injury of femoral arteries. Vascular wall hypoxia was immunohistochemically detected by pimonidazole hydrochloride, a hypoxia marker. Rabbit neointima and THP-1 macrophages were cultured to analyse prothrombotic factor expression under hypoxic conditions (1 % O2). Prothrombotic factor expression and nuclear localisation of hypoxia-inducible factor (HIF)-1α and nuclear factor-kappa B (NF-κB) p65 were immunohistochemically assessed using human coronary atherectomy plaques. Hypoxic areas were localised in the macrophage-rich deep portion of rabbit neointima and positively correlated with the number of nuclei immunopositive for HIF-1α and NF-κB p65, and tissue factor (TF) expression. Immunopositive areas for glycoprotein IIb/IIIa and fibrin in thrombi were significantly correlated with hypoxic areas in arteries. TF and plasminogen activator inhibitor-1 (PAI-1) expression was increased in neointimal tissues and/or macrophages cultured under hypoxia, and both were suppressed by inhibitors of either HIF-1 or NF-κB. In human coronary plaques, the number of HIF-1α-immunopositive nuclei was positively correlated with that of NF-κB-immunopositive nuclei and TF-immunopositive and PAI-1-immunopositive area, and it was significantly higher in thrombotic plaques. Vascular wall hypoxia augments the thrombogenic potential of atherosclerotic plaque and thrombus formation on plaques via prothrombotic factor upregulation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yujiro Asada
- Yujiro Asada, MD, Department of Pathology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan, Tel.: + 81 985 85 2810, Fax: + 81 985 85 7614, E-mail:
| |
Collapse
|
5
|
Takaoka N, Tsujita K, Kaikita K, Hokimoto S, Mizobe M, Nagano M, Horio E, Sato K, Nakayama N, Yoshimura H, Yamanaga K, Komura N, Kojima S, Tayama S, Nakamura S, Ogawa H. Comprehensive analysis of intravascular ultrasound and angiographic morphology of culprit lesions between ST-segment elevation myocardial infarction and non-ST-segment elevation acute coronary syndrome. Int J Cardiol 2014; 171:423-30. [DOI: 10.1016/j.ijcard.2013.12.094] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 11/26/2013] [Accepted: 12/21/2013] [Indexed: 11/28/2022]
|
6
|
Abstract
Recently published epidemiological and genetic studies strongly suggest a causal relationship of elevated concentrations of lipoprotein (a) [Lp(a)] with cardiovascular disease (CVD), independent of low-density lipoproteins (LDLs), reduced high density lipoproteins (HDL), and other traditional CVD risk factors. The atherogenicity of Lp(a) at a molecular and cellular level is caused by interference with the fibrinolytic system, the affinity to secretory phospholipase A2, the interaction with extracellular matrix glycoproteins, and the binding to scavenger receptors on macrophages. Lipoprotein (a) plasma concentrations correlate significantly with the synthetic rate of apo(a) and recent studies demonstrate that apo(a) expression is inhibited by ligands for farnesoid X receptor. Numerous gaps in our knowledge on Lp(a) function, biosynthesis, and the site of catabolism still exist. Nevertheless, new classes of therapeutic agents that have a significant Lp(a)-lowering effect such as apoB antisense oligonucleotides, microsomal triglyceride transfer protein inhibitors, cholesterol ester transfer protein inhibitors, and PCSK-9 inhibitors are currently in trials. Consensus reports of scientific societies are still prudent in recommending the measurement of Lp(a) routinely for assessing CVD risk. This is mainly caused by the lack of definite intervention studies demonstrating that lowering Lp(a) reduces hard CVD endpoints, a lack of effective medications for lowering Lp(a), the highly variable Lp(a) concentrations among different ethnic groups and the challenges associated with Lp(a) measurement. Here, we present our view on when to measure Lp(a) and how to deal with elevated Lp(a) levels in moderate and high-risk individuals.
Collapse
Affiliation(s)
- Karam M Kostner
- Associate Professor of Medicine, Mater Hospital, University of Queensland, St Lucia, QLD, Australia
| | | | | |
Collapse
|
7
|
Circulating thrombotic and haemostatic components in patients with coronary artery disease. Indian J Clin Biochem 2010; 25:20-8. [PMID: 23105879 DOI: 10.1007/s12291-010-0005-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The study aimed to analyze the circulating levels of thrombotic and haemostatic components; tissue factor, tissue factor pathway inhibitor, tissue plasminogen activator and plasminogen activator inhibitor-1 in patients with acute myocardial infarction at presentation (Group 1, n=49), unstable angina and Non-ST elevated MI after treatment (Group 2, n=22), stable angina (Group 3, n=18) and healthy individuals (Group 4, n=31). Significant finding was increase in tissue factor not only in Group 1 (2.0 fold, P=0.001), Group 2 (2.2 fold, P=0.015) but also in Group 3 (1.8 fold, P=0.018) as compared to controls. In Group 1 Plasminogen activator inhibitor-1 increased significantly (35.8%, P=0.02). Tissue factor pathway inhibitor and tissue plasminogen activator demonstrated increase in Group 1 of age<40 years while insignificant changes in elder patients. Increased thrombotic and decreased fibrinolytic conditions in acute myocardial infarction patients were observed. Increase TF in stable angina demonstrates procoagulant status in these patients as well.
Collapse
|
8
|
Isordia-Salas I, Leaños-Miranda A, Sainz IM, Reyes-Maldonado E, Borrayo-Sánchez G. Association of the plasminogen activator inhibitor-1 gene 4G/5G polymorphism with ST elevation acute myocardial infarction in young patients. Rev Esp Cardiol 2009; 62:365-72. [PMID: 19401121 DOI: 10.1016/s1885-5857(09)71663-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION AND OBJECTIVES To investigate the role of the 4G/5G polymorphism in the plasminogen activator inhibitor-1 (PAI-1) gene in patients with ST-elevation myocardial infarction (STEMI) aged < or =45 years and its influence on regulation of the plasma PAI-1 concentration. METHODS This case-control study included 127 consecutive patients aged < or =45 years with a diagnosis of STEMI who were admitted to a cardiovascular intensive care unit and 127 controls recruited between January 2006 and March 2007. Participants were genotyped for the 4G/5G polymorphism using the polymerase chain reaction and restriction fragment length polymorphism analysis, and their plasma PAI-1 concentrations were measured. Informed consent was obtained from all participants. RESULTS There was a significant difference in genotype distribution between the two groups (P< .002). The 4G allele occurred more frequently in the patient group (P=.032). In addition, there were significant independent associations between STEMI and the 4G allele (i.e., 4G/4G plus 4G/5G; odds ratio [OR]=2.29; 95% confidence interval [CI], 1.12-4.68; P=.022), smoking (OR=23.23; 95% CI, 8.92-60.47; P< .001), a family history of cardiovascular disease (OR=4.66; 95% CI, 2.06-10.52; P=.001) and hypertension (OR=5.42; 95% CI, 1.67-17.56; P=.005). The plasma PAI-1 concentration was higher in individuals who were homozygous for the 4G allele (P< .001). CONCLUSIONS The study findings indicate that the 4G allele is an independent risk factor for acute myocardial infarction in young patients, as are smoking, hypertension and a family history of inherited cardiovascular disease.
Collapse
Affiliation(s)
- Irma Isordia-Salas
- Unidad de Investigación Médica en Trombosis, Hemostasia y Aterogénesis, Instituto Mexicano del Seguro Social, México DF, México.
| | | | | | | | | |
Collapse
|
9
|
Isordia-Salas I, Leaños-Miranda A, Sainz IM, Reyes-Maldonado E, Borrayo-Sánchez G. Asociación entre el polimorfismo 4G/5G en el gen del inhibidor del activador del plasminógeno-1 (PAI-1) y el infarto agudo de miocardio con elevación del ST en pacientes jóvenes. Rev Esp Cardiol 2009. [DOI: 10.1016/s0300-8932(09)70893-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
10
|
Onalan O, Balta G, Oto A, Kabakci G, Tokgozoglu L, Aytemir K, Altay C, Gurgey A, Nazli N. Plasminogen activator inhibitor-1 4G4G genotype is associated with myocardial infarction but not with stable coronary artery disease. J Thromb Thrombolysis 2007; 26:211-7. [PMID: 17721742 DOI: 10.1007/s11239-007-0083-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2006] [Accepted: 02/01/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND A case control study was conducted to test the hypothesis that plasminogen activator inhibitor type-1 (PAI-1) 4G/5G gene polymorphism confers an increased risk for myocardial infarction (MI) in patients with known coronary atherosclerosis. METHODS One hundred fifty-six consecutive patients who presented with acute MI and 111 stable coronary artery disease (SCAD) patients with documented critical coronary artery stenoses were prospectively enrolled. PAI-1 4G/5G gene polymorphism and conventional atherosclerotic risk factors were studied in all patients. PAI-1 4G/5G gene polymorphism was studied in another 281 healthy blood bank donors. RESULTS The frequency 4G4G genotype was significantly higher in the MI group as compared to SCAD group (32.7% vs. 15.3%, P = 0.001) while it was not statistically significant between MI and healthy control groups (32.7% vs. 26.0%, P = 0.136). Comparing with healthy controls SCAD group had significantly lower frequency of 4G4G genotype (P = 0.024). In comparison with SCAD group PAI-1 4G/4G genotype, male sex and smoking habits favored to MI in univariate analysis with a P value of less than 0.2. These variables were included in multivariate regression model to estimate the associated risk for MI. PAI-1 4G/4G genotype was the only independent variable (OR 2.67, 95%CI 1.43-4.96, P = 0.002) associated with MI in this regression model. Comparing with healthy control group 4G4G genotype was not associated with MI (OR 1.38, 95%CI 0.90-2.12). However, presence of 4G4G genotype had a protective effect against development of SCAD (OR 0.52, 96%CI 0.29-0.92). CONCLUSION Compared to patients with critical coronary stenoses, PAI-1 4G/4G genotype was found to be an independent predictor for development of MI in this population. PAI-1 4G4G genotype have a protective effect against development of high grade stable coronary stenoses.
Collapse
Affiliation(s)
- Orhan Onalan
- Faculty of Medicine, Department of Cardiology, Hacettepe University, Ankara, Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Komissarov AA, Zhou A, Declerck PJ. Modulation of serpin reaction through stabilization of transient intermediate by ligands bound to alpha-helix F. J Biol Chem 2007; 282:26306-15. [PMID: 17613529 DOI: 10.1074/jbc.m702089200] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mechanism-based inhibition of proteinases by serpins involves enzyme acylation and fast insertion of the reactive center loop (RCL) into the central beta-sheet of the serpin, resulting in mechanical inactivation of the proteinase. We examined the effects of ligands specific to alpha-helix F (alphaHF) of plasminogen activator inhibitor-1 (PAI-1) on the stoichiometry of inhibition (SI) and limiting rate constant (k(lim)) of RCL insertion for reactions with beta-trypsin, tissue-type plasminogen activator (tPA), and urokinase. The somatomedin B domain of vitronectin (SMBD) did not affect SI for any proteinase or k(lim) for tPA but decreased the k(lim) for beta-trypsin. In contrast to SMBD, monoclonal antibodies MA-55F4C12 and MA-33H1F7, the epitopes of which are located at the opposite side of alphaHF, decreased k(lim) and increased SI for every enzyme. These effects were enhanced in the presence of SMBD. RCL insertion for beta-trypsin and tPA is limited by different subsequent steps of PAI-1 mechanism as follows: enzyme acylation and formation of a loop-displaced acyl complex (LDA), respectively. Stabilization of LDA through the disruption of the exosite interactions between PAI-1 and tPA induced an increase in the k(lim) but did not affect the SI. Thus it is unlikely that LDA contributes significantly to the outcome of the serpin reaction. These results demonstrate that the rate of RCL insertion is not necessarily correlated with SI and indicate that an intermediate, different from LDA, which forms during the late steps of PAI-1 mechanism, and could be stabilized by ligands specific to alphaHF, controls bifurcation between the inhibitory and the substrate pathways.
Collapse
Affiliation(s)
- Andrey A Komissarov
- Department of Chemistry, Portland State University, Portland, Oregon 97207-0751, USA.
| | | | | |
Collapse
|
12
|
Sakamoto T, Ishibashi T, Sugimoto K, Sakamoto N, Ohkawara H, Niinuma M, Nagata K, Kamioka M, Sugimoto N, Watanabe A, Kurabayashi M, Takuwa Y, Maruyama Y. RhoA-dependent PAI-1 gene expression induced in endothelial cells by monocyte adhesion mediates geranylgeranyl transferase I and Ca2+ signaling. Atherosclerosis 2007; 193:44-54. [PMID: 16973169 DOI: 10.1016/j.atherosclerosis.2006.08.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2005] [Revised: 08/07/2006] [Accepted: 08/11/2006] [Indexed: 11/29/2022]
Abstract
We investigated the role of RhoA activation and its mechanism in plasminogen activator inhibitor-1 (PAI-1) gene expression induced in endothelial cells by monocyte adhesion. Isolated human peripheral blood monocytes were added to cultured human coronary endothelial cells. Monocyte adhesion to endothelial cells increased PAI-1 expression at the transcriptional level and activated RhoA which was accompanied by an increase in the activity of geranylgeranyl transferase I (GGTase I), an enzyme responsible for geranylgeranylation, and actin stress fiber formation. Inhibition of RhoA by C3 exoenzyme or by adenovirus-mediated expression of N19RhoA, as well as by pravastatin, prevented the upregulation of PAI-1 induced by monocyte adhesion. GGTI-286, an inhibitor of GGTase I, prevented the monocyte-induced RhoA activation and PAI-1 expression in endothelial cells. Monocyte attachment induced an increase in intracellular Ca(2+) concentration ([Ca(2+)](i)) in endothelial cells and Ca(2+) chelation prevented the increased promoter activity and expression of PAI-1 induced by monocyte adhesion. C3 exoenzyme and GGTI-286 also suppressed endothelial intracellular Ca(2+) mobilization and Ca(2+) entry induced by monocytes. The present study shows that GGTase I plays a role in the RhoA activation in endothelial cells induced by monocyte adhesion and that GGTase I-mediated Ca(2+) signaling may contribute to RhoA-dependent PAI-1 gene expression.
Collapse
Affiliation(s)
- Takayuki Sakamoto
- First Department of Internal Medicine, Fukushima Medical University, Fukushima, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Blaschke F, Spanheimer R, Khan M, Law RE. Vascular effects of TZDs: New implications. Vascul Pharmacol 2006; 45:3-18. [PMID: 16740417 DOI: 10.1016/j.vph.2005.11.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2005] [Revised: 11/01/2005] [Accepted: 11/01/2005] [Indexed: 12/18/2022]
Abstract
The incidence of diabetes, now affecting more than 170 million individuals is growing rapidly. Type 2 diabetes, which accounts for 90% of all diabetes cases, is associated with increased cardiovascular morbidity and mortality. Thiazolidinediones (TZDs), used for the treatment of patients with type 2 diabetes improve insulin sensitivity and endothelial dysfunction and exert beneficial effects on the lipid profile by activating the peroxisome proliferator-activated receptor gamma (PPAR-gamma). Moreover, a large body of evidence indicates that TZDs exhibit antiatherogenic effects independent of their antidiabetic and lipid-lowering properties by modulating inflammatory processes. This review will focus on the role of PPAR-gamma agonists in the vessel wall and summarize their effects on C-reactive protein (CRP), plasminogen activator inhibitor type-1 (PAI-1), matrix metalloproteinase-9 (MMP-9), adiponectin and ATP-binding cassette transporter A1 (ABCA1) and their implications for treatment of advanced stages of atherosclerosis, particularly in a setting of type 2 diabetes.
Collapse
Affiliation(s)
- Florian Blaschke
- Division of Endocrinology, Diabetes and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
14
|
Abstract
Lipoprotein (a) [Lp(a)] appears to be one of the most atherogenic lipoproteins. It consists of a low-density lipoprotein (LDL) core in addition to a covalently bound glycoprotein, apolipoprotein (a) [apo(a)]. Apo(a) exists in numerous polymorphic forms. The size polymorphism is mediated by the variable number of kringle-4 Type-II repeats found in apo(a). Plasma Lp(a) levels are determined to more than 90% by genetic factors. Plasma Lp(a) levels in healthy individuals correlate significantly high with apo(a) biosynthesis and not with its catabolism. There are several hormones known to have a strong impact on Lp(a) metabolism. In certain diseases, such as kidney disease, Lp(a) catabolism is impaired leading to up to fivefold elevations. Lp(a) levels rise with age but are otherwise influenced only little by diet and lifestyle. There is no safe and efficient way of treating individuals with elevated plasma Lp(a) concentrations. Most of the lipid-lowering drugs have either no significant influence on Lp(a) or exhibit a variable effect in patients with different forms of primary and secondary hyperlipoproteinemia. There is without doubt a strong need to concentrate on the development of specific medications to selectively target Lp(a) biosynthesis, Lp(a) assembly and Lp(a) catabolism. So far only anabolic steroids were found to drastically reduce Lp(a) plasma levels. This class of substance cannot, of course, be used for treatment of patients with hyper-Lp(a). We recommend that the mechanism of action of these drugs be studied in more detail and that the possibility of synthesizing derivatives which may have a more specific effect on Lp(a) without having any side effects be pursued. Other strategies that may be of use in the development of drugs for treatment of patients with hyper-Lp(a) are discussed in this review.
Collapse
Affiliation(s)
- K M Kostner
- Research Wing Level 3, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia.
| | | |
Collapse
|
15
|
Furumoto T, Fujii S, Nishihara K, Yamada S, Komuro K, Goto K, Onozuka H, Mikami T, Kitabatake A, Sobel BE. Maladaptive arterial remodeling with systemic hypertension associated with increased concentrations in blood of plasminogen activator inhibitor type-1 (PAI-1). Am J Cardiol 2004; 93:997-1001. [PMID: 15081442 DOI: 10.1016/j.amjcard.2003.12.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2003] [Revised: 12/24/2003] [Accepted: 12/24/2003] [Indexed: 11/28/2022]
Abstract
Increased carotid artery intima-media thickness (IMT), but not necessarily peripheral vessel IMT, accompanies atherosclerosis. We hypothesized that IMT in a peripheral, muscular artery known to be resistant to atherosclerotic changes would increase with hypertension, thereby limiting increases in wall stress and potentially preserving endothelial cell function reflected by flow-mediated dilation (FMD). Plasminogen activator inhibitor type-1 (PAI-1) can inhibit vascular smooth muscle cell migration contributing to increased IMT. Thus, increased PAI-1 may attenuate the mural adaptive response. A high-resolution scanner designed to delineate brachial artery FMD and IMT was used in studies of previously untreated patients with essential hypertension (n = 18) and age- and gender-matched normotensive subjects (n = 15). Brachial IMT was increased with hypertension (0.36 +/- 0.07 vs 0.27 +/- 0.03 mm in controls, p <0.01), and FMD was lower (3.6 +/- 1.5% vs 7.8 +/- 3.6, p <0.01). PAI-1 antigen in blood was increased (40.5 +/- 31.8 vs 26.3 +/- 11.6 ng/ml, p <0.05). IMT and FMD correlated positively (r = 0.63, p <0.05) in hypertensive patients. FMD correlated inversely with wall stress (r = -0.57, p <0.05). IMT correlated inversely with PAI-1 (r = -0.61, p <0.05). These observations support the hypothesis that increased PAI-1 attenuated increases in neointimal vascular smooth muscle cell cellularity. Thus, increased PAI-1 may attenuate a mural, adaptive response to hypertension associated with preservation of endothelial cell function.
Collapse
Affiliation(s)
- Tomoo Furumoto
- Department of Cardiovascular Medicine, Graduate School of Medicine and College of Medical Technology, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Fujii K, Kobayashi Y, Mintz GS, Takebayashi H, Dangas G, Moussa I, Mehran R, Lansky AJ, Kreps E, Collins M, Colombo A, Stone GW, Leon MB, Moses JW. Intravascular Ultrasound Assessment of Ulcerated Ruptured Plaques. Circulation 2003; 108:2473-8. [PMID: 14610010 DOI: 10.1161/01.cir.0000097121.95451.39] [Citation(s) in RCA: 192] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
It is not clear why some plaque ruptures lead to acute coronary syndromes (ACS) but others do not.
Methods and Results—
We analyzed 80 plaque ruptures in 74 patients and compared culprit lesions of ACS patients with nonculprit lesions of ACS patients and lesions of non-ACS patients; both culprit and nonculprit plaque ruptures were studied in 6 of 54 ACS patients. Intravascular ultrasound findings suggesting thrombus were observed more frequently in culprit lesions of ACS patients (n=35) compared with nonculprit lesions of ACS patients (n=19) and lesions of non-ACS patients (n=26): 60% versus 32% versus 8% (
P
<0.001). At the minimal lumen site, smaller lumen areas (3.3±1.5 versus 5.4±2.6 versus 6.1±2.0 mm
2
,
P
<0.001) and greater area stenosis (61±15% versus 50±14% versus 46±18%,
P
=0.002) and plaque burden (80±8% versus 71±8% versus 69±10%,
P
<0.001) were observed in culprit lesions of ACS patients compared with nonculprit lesions of ACS patients and lesions of non-ACS patients. Lesions were longer (18.7±6.4 versus 154.9±6.1 versus 12.0±4.9 mm,
P
<0.001) and rupture site remodeling indices were greater (1.26±0.21 versus 1.24±0.21 versus 1.09±0.05,
P
=0.002). Independent predictors of culprit plaque ruptures in ACS patients were smaller minimum lumen areas (
P
=0.02) and presence of thrombus (
P
=0.01).
Conclusions—
Ruptured plaques in culprit lesions of ACS patients have smaller lumens; greater plaque burdens, area stenosis, and remodeling indices; and more thrombus. Plaque rupture itself does not lead to symptoms. The association of plaque rupture with a smaller lumen area and/or thrombus formation causes lumen compromise and leads to symptoms.
Collapse
Affiliation(s)
- Kenichi Fujii
- Cardiovascular Research Foundation, Lenox Hill Heart and Vascular Institute, 130 East 77th St, 9th Floor, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Sobel BE, Taatjes DJ, Schneider DJ. Intramural plasminogen activator inhibitor type-1 and coronary atherosclerosis. Arterioscler Thromb Vasc Biol 2003; 23:1979-89. [PMID: 12920048 DOI: 10.1161/01.atv.0000091250.53231.4d] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Altered expression of plasminogen activator inhibitor type-1 in vessel walls, reviewed here, might affect coronary atherogenesis. Upregulation might exacerbate vasculopathy by potentiating thrombosis and by inhibiting vascular smooth muscle cell migration, resulting in attenuation of thickness of elaborated fibrous caps implicated in the vulnerability of atheroma to rupture.
Collapse
Affiliation(s)
- Burton E Sobel
- Department of Medicine, University of Vermont, Burlington, USA.
| | | | | |
Collapse
|
18
|
Affiliation(s)
- Antoine Lafont
- Cardiology Department, Hôpital Européen Georges Pompidou, 20 rue Leblanc, Paris 75015, France.
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Lipoprotein(a) belongs to the class of the most atherogenic lipoproteins. Despite intensive research - in the last year more than 80 papers have been published on this topic - information is still lacking on the physiological function of lipoprotein(a) and the site of its catabolism. Important advances have been made in the knowledge of these points, which may have some therapeutic implications. RECENT FINDINGS The association of high lipoprotein(a) values with an increase in risk for coronary events has been documented in further prospective studies. This increased risk may relate to recent findings that apolipoprotein(a) is produced in situ within the vessel wall. In addition, lipoprotein(a) binds and inactivates the tissue factor pathway inhibitor and induces plasminogen activator inhibitor type 2 expression in monocytes. A new antisense oligonucleotide strategy has been proposed which efficiently inhibits apolipoprotein(a) expression in vitro and in vivo. Apolipoprotein(a), however, suppresses angiogenesis and thus may interfere with the infiltration of tumor cells. Finally, the enzymatic activity leading to the formation of apolipoprotein(a) fragments in plasma and their catabolism have been further elucidated. SUMMARY We are still far away from understanding the pathways involved in lipoprotein(a) catabolism, and the physiological function of this lipoprotein. Recent findings, however, provide new insight into pathomechanisms in patients with increased lipoprotein(a) related to hemostasis, which may serve as a basis for designing new treatment strategies.
Collapse
Affiliation(s)
- Karam M Kostner
- Department of Cardiology, University Hospital of Vienna, Austria
| | | |
Collapse
|
20
|
Ishibashi T, Nagata K, Ohkawara H, Sakamoto T, Yokoyama K, Shindo J, Sugimoto K, Sakurada S, Takuwa Y, Teramoto T, Maruyama Y. Inhibition of Rho/Rho-kinase signaling downregulates plasminogen activator inhibitor-1 synthesis in cultured human monocytes. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1590:123-30. [PMID: 12063175 DOI: 10.1016/s0167-4889(02)00201-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Increased production of plasminogen activator inhibitor-1 (PAI-1) in plaques plays a role in the pathogenesis of atherosclerosis. This study was conducted to investigate the effect of blockade of Rho/Rho-kinase signaling on the synthesis of PAI-1 in cultured human peripheral blood monocytes. HMG-CoA reductase inhibitors (statins) and inhibitors of Rho and Rho-kinase were added to monocyte cultures. The levels of PAI antigen and mRNA were determined by Western blotting and RT-PCR, respectively, and PAI-1 expression was assessed by immunohistochemistry. We performed pull-down assays to determine the activity of Rho by measuring the GTP-bound form of Rho A. In unstimulated and lipopolysaccharide (LPS)-stimulated cultured monocytes, statins reduced the levels of PAI-1 antigen and mRNA. The suppressive effects of statins on PAI-1 synthesis were reversed by geranylgeranylpyrophosphate (GGPP) and were mimicked by C3 exoenzyme. Immunohistochemistry confirmed the role of lipid modification by GGPP in suppressive effect of statins in PAI-1 synthesis. Pull-down assays demonstrated that statins decreased the levels of the GTP-bound form of Rho A. Our findings suggest that statins decrease the activity of Rho by inhibiting geranylgeranylation. Moreover, Rho-kinase inhibitors, Y-27632 and fasudil, suppressed the synthesis of PAI-1 in this culture system. We show that inhibition of Rho/Rho-kinase signaling downregulates the synthesis of PAI-1 in human monocytes.
Collapse
Affiliation(s)
- Toshiyuki Ishibashi
- First Department of Internal Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|