501
|
Martínez-González J, Escudero I, Badimon L. Simvastatin potenciates PGI2 release induced by HDL in human VSMC: effect on Cox-2 up-regulation and MAPK signalling pathways activated by HDL. Atherosclerosis 2004; 174:305-13. [PMID: 15136060 DOI: 10.1016/j.atherosclerosis.2004.01.037] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2003] [Revised: 01/26/2004] [Accepted: 01/28/2004] [Indexed: 10/26/2022]
Abstract
High density lipoproteins (HDL) induce prostacyclin (PGI(2)) release in vascular smooth muscle cells (VSMC) by up-regulation of cyclooxygenase-2 (Cox-2). Our goal was to analyse the mechanisms underlying this effect, and its potential modulation by HMG-CoA reductase inhibition in human VSMC. The contribution of mitogen-activated protein kinase (MAPK) signalling pathways was assessed by Western blot analysis and using specific inhibitors [PD098059 for p42/44 MAPK kinase (MEK); SB203580 for p38 MAPK or L-JNKI1 for c-Jun N-terminal kinase-1 (JNK-1)]. HDL-induced PGI(2) release was inhibited by rofecoxib (a specific Cox-2 inhibitor, 5 microM). HDL induced the early activation of p42 MAPK, p38 MAPK and JNK-1. p42/44 MAPK was the major pathway involved in both Cox-2 up-regulation and PGI(2) synthesis; p38 MAPK was also involved in both processes while JNK inhibition only affected PGI(2) synthesis. Pertussis toxin (an inhibitor of Galphai/Galphao proteins) prevented MAPK activation and inhibited both Cox-2 up-regulation and PGI(2) release. Genistein (a tyrosine kinase inhibitor) inhibited PGI(2) release without affecting MAPK activation or Cox-2 up-regulation. Simvastatin (0.1-1 microM) increased HDL-induced PGI(2) release ( approximately 45% at 1 microM) but did not significantly modify early MAPK activation or Cox-2 expression. Simvastatin alone did not significantly affect PGI(2) release. Our results suggest that mechanisms associated with G protein-coupled receptor activation, trigger Cox-2 up-regulation and PGI(2) release via multiple MAPK signalling pathways in VSMC. The mechanism is independent of tyrosine kinase receptors, although cytosolic tyrosine kinases could activate Cox-2 post-translationally. The potential contribution of HDL to vascular homeostasis, via increases in PGI(2) synthesis, could be enhanced by HMG-CoA reductase inhibitors.
Collapse
MESH Headings
- Analysis of Variance
- Blotting, Northern
- Blotting, Western
- Cells, Cultured
- Cyclooxygenase 2
- Drug Interactions
- Enzyme Activation/drug effects
- Epoprostenol/metabolism
- Humans
- Isoenzymes/drug effects
- Isoenzymes/metabolism
- Lipoproteins, HDL/pharmacology
- Membrane Proteins
- Mitogen-Activated Protein Kinase 1/drug effects
- Mitogen-Activated Protein Kinase 1/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Prostaglandin-Endoperoxide Synthases/drug effects
- Prostaglandin-Endoperoxide Synthases/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sensitivity and Specificity
- Signal Transduction
- Simvastatin/pharmacology
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- José Martínez-González
- Centro de Investigación Cardiovascular, CSIC/ICCC-Hospital de la Santa Creu i Sant Pau, Avda. Sant Antoni Maria Claret #167, 08025 Barcelona, Spain
| | | | | |
Collapse
|
502
|
Abstract
The dyslipidemia typically found in subjects with the metabolic syndrome includes an elevated concentration of plasma triglyceride,a low-density lipoprotein fraction in which the particles are smaller and denser than normal, and a low concentration of highdensity lipoprotein (HDL) cholesterol. This article is concerned with the low HDL component. It provides an overview of HDL structure and metabolism and describes the functions of HDLs that may be cardioprotective. The article then outlines what is known about the concentration and subpopulation distribution of HDLs in the metabolic syndrome. Possible mechanisms responsible for the low HDL are discussed. The consequences of a low HDL concentration in this syndrome are addressed before the article concludes with a discussion of whether low HDL in the metabolic syndrome should be a therapeutic target.
Collapse
Affiliation(s)
- Philip Barter
- The Heart Research Institute, 145 Missenden Road, Camperdown, Sydney NSW 2050, Australia.
| |
Collapse
|
503
|
Connelly MA, Williams DL. Scavenger receptor BI: a scavenger receptor with a mission to transport high density lipoprotein lipids. Curr Opin Lipidol 2004; 15:287-95. [PMID: 15166784 DOI: 10.1097/00041433-200406000-00008] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW This review will survey recent findings on the cholesterol transport and scavenger functions of scavenger receptor BI. Although scavenger receptor BI and CD36 bind many of the same ligands, these two receptors have very specific lipid transport functions: CD36 facilitates the uptake of long chain fatty acids and SR-BI mediates the transport of cholesterol and cholesteryl ester from HDL particles. Scavenger receptor BI is a physiologically relevant HDL receptor that, along with HDL, is protective against cardiovascular disease. Its atheroprotective role has been hypothesized to be due to its function in the reverse cholesterol transport pathway. RECENT FINDINGS Recent studies suggest that scavenger receptor BI function is not only crucial for cholesterol delivery to the liver but is also important for cholesterol efflux at the vessel wall. Therefore, the receptor acts at both ends of the reverse cholesterol transport pathway. In addition, it stimulates nitric oxide production in endothelial cells, which may also contribute to its positive influence on the vasculature. Lastly, the glycoprotein was cloned as a scavenger receptor and in some cases is still thought to operate in this fashion. SUMMARY It will be interesting to follow future research on scavenger receptor BI that will delineate its functions in cholesterol transport as well as its scavenger functions. Additionally, we are only beginning to learn of the glycoprotein's effects on disease states besides atherosclerosis and cardiovascular disease.
Collapse
Affiliation(s)
- Margery A Connelly
- Department of Pharmacological Sciences, University Medical Center, State University of New York at Stony Brook, Stony Brook, NY 11794-8651, USA
| | | |
Collapse
|
504
|
Abstract
The concentration of high density lipoprotein cholesterol (HDL-C) has been found to be a powerful negative predictor of premature coronary heart disease (CHD) and stroke in human prospective population studies. Evidence of the protective properties of HDLs has also been documented in the elderly and their offspring. HDLs mediate several functions that provide an insight into their potential anti-atherogenic mechanisms. Intervention strategies to prevent CHD have generally focused on lowering low-density lipoprotein cholesterol (LDL-C). However, several lifestyle and pharmacological interventions have the capacity to raise the level of HDL-C. As data accumulate on the protective role of HDLs, there is growing support for interventions that act to raise HDL-C concentrations.
Collapse
Affiliation(s)
- Philip Barter
- The Heart Research Institute, 145 Missenden Road, Camperdown, Sydney, NSW 2050, Australia.
| |
Collapse
|
505
|
Drew BG, Fidge NH, Gallon-Beaumier G, Kemp BE, Kingwell BA. High-density lipoprotein and apolipoprotein AI increase endothelial NO synthase activity by protein association and multisite phosphorylation. Proc Natl Acad Sci U S A 2004; 101:6999-7004. [PMID: 15107497 PMCID: PMC406455 DOI: 10.1073/pnas.0306266101] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
NO propagates a number of antiatherogenic effects in the endothelium, and diminished availability has been associated with vascular disease. Recently it has been reported that phosphorylation of endothelial NO synthase (eNOS) at Ser-1179 is required to increase activity in response to stimuli, including high-density lipoprotein (HDL). The current study was undertaken to further examine the mechanism by which HDL activates eNOS and to specifically determine the role of the major apolipoprotein of HDL, apolipoprotein AI (ApoAI). Phosphorylation of eNOS residues Ser-116, Ser-617, Ser-635, Ser-1179, and Thr-497 after incubation with ApoAI and HDL was examined. There were significant increases in phosphorylation at Ser-116 in response to both HDL and ApoAI and similar magnitudes of dephosphorylation at Thr-497. Ser-1179 phosphorylation increased transiently but returned to basal level after 2.5 min. Data demonstrating activation of AMP activated protein kinase (AMPK) during HDL and ApoAI incubation suggests that AMPK may play a role in activation of eNOS. NO release in response to HDL and ApoAI stimulation in endothelial cells paralleled the time frames of phosphorylation, suggesting a causal relationship. Furthermore, ApoAI was found to associate with eNOS in endothelial cells and bind transfected eNOS in Chinese hamster ovary cells, whereas confocal data demonstrates colocalization of ApoAI and eNOS in the perinuclear region, suggesting a protein-protein interaction. Collectively, the results indicate that HDL and ApoAI increase eNOS activity by multisite phosphorylation changes, involving AMPK activation after protein association between ApoAI and eNOS.
Collapse
Affiliation(s)
- Brian G Drew
- Baker Heart Research Institute, P.O. Box 6492 St. Kilda Road Central, Melbourne, Victoria 8008, Australia
| | | | | | | | | |
Collapse
|
506
|
Cao WM, Murao K, Imachi H, Yu X, Abe H, Yamauchi A, Niimi M, Miyauchi A, Wong NCW, Ishida T. A mutant high-density lipoprotein receptor inhibits proliferation of human breast cancer cells. Cancer Res 2004; 64:1515-21. [PMID: 14973113 DOI: 10.1158/0008-5472.can-03-0675] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
High-density lipoprotein (HDL) stimulates the growth of many types of cells, including those of breast cancer. High levels of HDL are associated with an increased risk of breast cancer development. A scavenger receptor of the B class (SR-BI)/human homolog of SR-BI, CD36, and LIMPII analogous-1 (CLA-1) facilitates the cellular uptake of cholesterol from HDL and thus augments cell growth. Furthermore, HDL is also believed to have antiapoptotic effects on various cell types, and this feature adds to its ability to promote cell growth. These collaborative roles of HDL and CLA-1 prompted us to assess the function of these components on human breast cancer cells. In this study, we created a mutant CLA-1 (mCLA) that lacked the COOH-terminal tail to determine its potential role in breast cancer cell growth. Expression of mCLA inhibited the proliferation of breast cancer cell line MCF-7. This inhibitory action of mCLA required the transcriptional factor activator protein-1 (AP-1), and the mutant receptor also affected the antiapoptotic features of HDL. The effect of HDL on AP-1 activation and [(3)H]thymidine incorporation was abrogated by wortmannin, a specific inhibitor of phosphoinositide 3-kinase. Furthermore, the dominant negative mutant of Akt abolished the ability of HDL to activate AP-1. These findings raise the possibility that the inhibitors of the effects of HDL may be of therapeutic value for breast cancer.
Collapse
Affiliation(s)
- Wen M Cao
- First Department of Internal Medicine, Kagawa Medical University, 1750-1 Miki-cho, Kita-gun, Kagawa, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
507
|
|
508
|
Abstract
PURPOSE OF REVIEW We will discuss the diverse roles of lipoprotein receptors that contribute to the maintenance and integrity of the vascular wall. RECENT FINDINGS Lipoprotein receptors function not only as transporters for cholesterol and other lipids. They also act as sensors and signal transducers through which the endothelium, macrophages and smooth muscle cells communicate with their environment. SUMMARY Traditionally, lipoprotein receptors were thought of merely as transporters of cholesterol and triglycerides to specific target cells, either for the purpose of delivery and redistribution of nutrients, or for the destruction or clearance of modified (oxidized) lipids by macrophages. Only recently have we begun to appreciate that the same receptors engage in a much more sophisticated and multi-faceted interaction with their environment. Inasmuch, they not only act as mere transporters, but as surprisingly versatile and adaptive signal transducers and modulators throughout the vessel wall. These recent findings now begin to reshape our thinking of how such structurally different and evolutionarily unrelated lipoprotein receptors orchestrate the response of the vessel wall to mechanical or metabolic damage.
Collapse
Affiliation(s)
- Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9046, USA.
| | | |
Collapse
|
509
|
Zannis VI, Chroni A, Kypreos KE, Kan HY, Cesar TB, Zanni EE, Kardassis D. Probing the pathways of chylomicron and HDL metabolism using adenovirus-mediated gene transfer. Curr Opin Lipidol 2004; 15:151-66. [PMID: 15017358 DOI: 10.1097/00041433-200404000-00008] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF THE REVIEW This review clarifies the functions of key proteins of the chylomicron and the HDL pathways. RECENT FINDINGS Adenovirus-mediated gene transfer of several apolipoprotein (apo)E forms in mice showed that the amino-terminal 1-185 domain of apoE can direct receptor-mediated lipoprotein clearance in vivo. Clearance is mediated mainly by the LDL receptor. The carboxyl-terminal 261-299 domain of apoE induces hypertriglyceridemia, because of increased VLDL secretion, diminished lipolysis and inefficient VLDL clearance. Truncated apoE forms, including apoE2-202, have a dominant effect in remnant clearance and may have future therapeutic applications for the correction of remnant removal disorders. Permanent expression of apoE and apoA-I following adenoviral gene transfer protected mice from atherosclerosis. Functional assays, protein cross-linking, and adenovirus-mediated gene transfer of apoA-I mutants in apoA-I deficient mice showed that residues 220-231, as well as the central helices of apoA-I, participate in ATP-binding cassette transporter A1-mediated lipid efflux and HDL biogenesis. Following apoA-I gene transfer, an amino-terminal deletion mutant formed spherical alpha-HDL, a double amino- and carboxyl-terminal deletion mutant formed discoidal HDL, and a carboxyl-terminal deletion mutant formed only pre-beta-HDL. The findings support a model of cholesterol efflux that requires direct physical interactions between apoA-I and ATP-binding cassette transporter A1, and can explain Tangier disease and other HDL deficiencies. SUMMARY New insights are provided into the role of apoE in cholesterol and triglyceride homeostasis, and of apoA-I in the biogenesis of HDL. Clearance of the lipoprotein remnants and increase in HDL synthesis are obvious targets for therapeutic interventions.
Collapse
Affiliation(s)
- Vassilis I Zannis
- Molecular Genetics, Boston University School of Medicine, Boston, Massachusetts 02118-2394, USA. vzannis!bu.edu
| | | | | | | | | | | | | |
Collapse
|
510
|
Nofer JR, van der Giet M, Tölle M, Wolinska I, von Wnuck Lipinski K, Baba HA, Tietge UJ, Gödecke A, Ishii I, Kleuser B, Schäfers M, Fobker M, Zidek W, Assmann G, Chun J, Levkau B. HDL induces NO-dependent vasorelaxation via the lysophospholipid receptor S1P3. J Clin Invest 2004; 113:569-81. [PMID: 14966566 PMCID: PMC338256 DOI: 10.1172/jci18004] [Citation(s) in RCA: 193] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2003] [Accepted: 11/25/2003] [Indexed: 11/17/2022] Open
Abstract
HDL is a major atheroprotective factor, but the mechanisms underlying this effect are still obscure. HDL binding to scavenger receptor-BI has been shown to activate eNOS, although the responsible HDL entities and signaling pathways have remained enigmatic. Here we show that HDL stimulates NO release in human endothelial cells and induces vasodilation in isolated aortae via intracellular Ca2+ mobilization and Akt-mediated eNOS phosphorylation. The vasoactive effects of HDL could be mimicked by three lysophospholipids present in HDL: sphingosylphosphorylcholine (SPC), sphingosine-1-phosphate (S1P), and lysosulfatide (LSF). All three elevated intracellular Ca2+ concentration and activated Akt and eNOS, which resulted in NO release and vasodilation. Deficiency of the lysophospholipid receptor S1P3 (also known as LPB3 and EDG3) abolished the vasodilatory effects of SPC, S1P, and LSF and reduced the effect of HDL by approximately 60%. In endothelial cells from S1P3-deficient mice, Akt phosphorylation and Ca2+ increase in response to HDL and lysophospholipids were severely reduced. In vivo, intra-arterial administration of HDL or lysophospholipids lowered mean arterial blood pressure in rats. In conclusion, we identify HDL as a carrier of bioactive lysophospholipids that regulate vascular tone via S1P3-mediated NO release. This mechanism may contribute to the vasoactive effect of HDL and represent a novel aspect of its antiatherogenic function.
Collapse
MESH Headings
- Animals
- Aorta/anatomy & histology
- Aorta/metabolism
- Calcium/metabolism
- Cells, Cultured
- Endothelium, Vascular/cytology
- Endothelium, Vascular/metabolism
- Enzyme Activation
- Enzyme Inhibitors/metabolism
- Female
- Humans
- In Vitro Techniques
- Lipoproteins, HDL/metabolism
- Lysophospholipids/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Nitric Oxide/metabolism
- Nitric Oxide Synthase/metabolism
- Nitric Oxide Synthase Type II
- Nitric Oxide Synthase Type III
- Phosphorylation
- Protein Serine-Threonine Kinases
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-akt
- Rats
- Rats, Inbred WKY
- Rats, Wistar
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Lysophospholipid
- Vasodilation/physiology
Collapse
Affiliation(s)
- Jerzy-Roch Nofer
- Institut für Klinische Chemie und Laboratoriumsmedizin, Westfälische Wilhelms-Universität, Münster, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
511
|
Abstract
Circulating levels of HDL cholesterol are inversely related to the risk of atherosclerosis, and therapeutic increases in HDL reduce the incidence of cardiovascular events. A new study shows that HDL-associated lysophospholipids stimulate the production of the potent antiatherogenic signaling molecule NO by the vascular endothelium.
Collapse
Affiliation(s)
- Philip W Shaul
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | | |
Collapse
|
512
|
Abstract
There is a large body of evidence demonstrating an inverse correlation between circulating levels of high-density lipoprotein (HDL) cholesterol and cardiovascular disease risk. For every 1-mg/dL increase in HDL, it is estimated that the risk of cardiovascular events decreases by 2% to 3%. HDL is one of many factors that contribute to the regulation of the atherosclerotic process. HDL mediates reverse cholesterol transport and exhibits numerous beneficial properties, including antioxidant, antiinflammatory, and antithrombotic effects on the vasculature. Recent studies have expanded our understanding of the vasoprotective mechanisms of HDL to include enhanced nitric oxide production and improved endothelium-dependent relaxation. Progress has also been made in determining the molecular mechanisms that mediate reverse cholesterol transport. Recently published National Cholesterol Education Program Adult Treatment Panel guidelines have broadened the definition of low levels of HDL and encourage more aggressive screening and treatment of lipid abnormalities. Several therapeutic interventions can augment HDL concentrations, and there is increasing evidence that these interventions improve cardiovascular outcomes. Research focusing on defining the molecular roles of HDL will likely identify potential therapeutic targets for decreasing the incidence and progression of coronary heart disease. This review highlights the role of HDL in coronary heart disease, from basic mechanisms of action to recent clinical trial results.
Collapse
Affiliation(s)
- Christine E Young
- Department of Medicine/Division of Cardiology, Tufts-New England Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
| | | | | |
Collapse
|
513
|
Viswambharan H, Ming XF, Zhu S, Hubsch A, Lerch P, Vergères G, Rusconi S, Yang Z. Reconstituted high-density lipoprotein inhibits thrombin-induced endothelial tissue factor expression through inhibition of RhoA and stimulation of phosphatidylinositol 3-kinase but not Akt/endothelial nitric oxide synthase. Circ Res 2004; 94:918-25. [PMID: 14988229 DOI: 10.1161/01.res.0000124302.20396.b7] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Endothelial cells express negligible amounts of tissue factor (TF) that can be induced by thrombin, which is important for acute coronary syndromes. Recent research suggests that endothelial TF expression is positively regulated by RhoA and p38mapk, but negatively by Akt/endothelial nitric oxide synthase (eNOS) pathway. High-density lipoprotein (HDL) is atheroprotective and exerts antiatherothrombotic effect. This study investigated the effect of a reconstituted HDL (rHDL) on endothelial TF expression induced by thrombin and the underlying mechanisms. In cultured human umbilical vein and aortic endothelial cells, thrombin (4 U/mL, 4 hours) increased TF protein level, which was reduced by rHDL (0.1 mg/mL, 43% inhibition, n=3 to 7, P<0.01). Activation of RhoA but not p38mapk by thrombin was prevented by rHDL. rHDL stimulated Akt/eNOS pathway. The phosphatidylinositol 3-kinase (PI3K) inhibitors wortmannin or LY294002 abolished the activation of Akt/eNOS and reversed the inhibitory effect of rHDL on TF expression. Adenoviral expression of the active PI3K mutant (p110) reduced TF expression stimulated by thrombin without inhibiting RhoA activation, whereas expression of the active Akt mutant (m/p) further facilitated TF upregulation by thrombin. Moreover, a dominant-negative Akt mutant (KA) reduced thrombin's effect and did not reverse the rHDL's inhibitory effect on TF expression. Inhibition of eNOS by N(omega)-nitro-L-arginine methyl ester (100 micromol/L) did not affect the rHDL's effect. In conclusion, rHDL inhibits thrombin-induced human endothelial TF expression through inhibition of RhoA and activation of PI3K but not Akt/eNOS. These findings implicate a novel mechanism of antiatherothrombotic effects of HDL.
Collapse
Affiliation(s)
- Hema Viswambharan
- Department of Medicine, Divisions of Physiology, University of Fribourg, Fribourg, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
514
|
Cuchel M, Rader DJ. Genetics of increased HDL cholesterol levels: insights into the relationship between HDL metabolism and atherosclerosis. Arterioscler Thromb Vasc Biol 2004; 23:1710-2. [PMID: 14555638 DOI: 10.1161/01.atv.0000092947.15939.93] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
515
|
|
516
|
Nofer JR, van der Giet M, Tölle M, Wolinska I, von Wnuck Lipinski K, Baba HA, Tietge UJ, Gödecke A, Ishii I, Kleuser B, Schäfers M, Fobker M, Zidek W, Assmann G, Chun J, Levkau B. HDL induces NO-dependent vasorelaxation via the lysophospholipid receptor S1P3. J Clin Invest 2004. [DOI: 10.1172/jci200418004] [Citation(s) in RCA: 510] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
517
|
de Beer MC, Castellani LW, Cai L, Stromberg AJ, de Beer FC, van der Westhuyzen DR. ApoA-II modulates the association of HDL with class B scavenger receptors SR-BI and CD36. J Lipid Res 2004; 45:706-15. [PMID: 14729860 DOI: 10.1194/jlr.m300417-jlr200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The class B scavenger receptors SR-BI and CD36 exhibit a broad ligand binding specificity. SR-BI is well characterized as a HDL receptor that mediates selective cholesteryl ester uptake from HDL. CD36, a receptor for oxidized LDL, also binds HDL and mediates selective cholesteryl ester uptake, although much less efficiently than SR-BI. Apolipoprotein A-II (apoA-II), the second most abundant HDL protein, is considered to be proatherogenic, but the underlying mechanisms are unclear. We previously showed that apoA-II modulates SR-BI-dependent binding and selective uptake of cholesteryl ester from reconstituted HDL. To investigate the effect of apoA-II in naturally occurring HDL on these processes, we compared HDL without apoA-II (from apoA-II null mice) with HDLs containing differing amounts of apoA-II (from C57BL/6 mice and transgenic mice expressing a mouse apoA-II transgene). The level of apoA-II in HDL was inversely correlated with HDL binding and selective cholesteryl ester uptake by both scavenger receptors, particularly CD36. Interestingly, for HDL lacking apoA-II, the efficiency with which CD36 mediated selective uptake reached a level similar to that of SR-BI. These results demonstrate that apoA-II exerts a marked effect on HDL binding and selective lipid uptake by the class B scavenger receptors and establishes a potentially important relationship between apoA-II and CD36.
Collapse
Affiliation(s)
- Maria C de Beer
- Department of Internal Medicine, University of Kentucky Medical Center, Lexington, KY 40536, USA
| | | | | | | | | | | |
Collapse
|
518
|
Hersberger M, von Eckardstein A. Low high-density lipoprotein cholesterol: physiological background, clinical importance and drug treatment. Drugs 2004; 63:1907-45. [PMID: 12930163 DOI: 10.2165/00003495-200363180-00003] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Low high-density lipoprotein (HDL) cholesterol is an important risk factor for coronary heart disease (CHD). In vitro, HDL exerts several potentially anti-atherogenic activities. HDLs mediate the reverse cholesterol transport (RCT) from peripheral cells to the liver, inhibit oxidation of low-density lipoprotein (LDL), adhesion of monocytes to the endothelium, apoptosis of vascular endothelial and smooth muscle cells and platelet activation, and stimulate the endothelial secretion of vasoactive substances as well as smooth muscle cell proliferation. Hence, raising HDL-cholesterol levels has become an interesting target for anti-atherosclerotic drug therapy. Levels of HDL cholesterol and the composition of HDL subclasses in plasma are regulated by apolipoproteins, lipolytic enzymes, lipid transfer proteins, receptors and cellular transporters. The interplay of these factors leads to RCT and determines the composition and, thereby, the anti-atherogenic properties of HDL. Several inborn errors of metabolism, as well as genetic animal models, are characterised by both elevated HDL cholesterol and increased rather than decreased cardiovascular risk. These findings suggest that the mechanism of HDL modification rather than simply increasing HDL cholesterol determine the efficacy of anti-atherosclerotic drug therapy. In several controlled and prospective intervention studies, patients with low HDL cholesterol and additional risk factors benefited from treatment with fibric acid derivatives (fibrates) or HMG-CoA reductase inhibitors (statins). However, only in some trials was prevention of coronary events in patients with low HDL cholesterol and hypertriglyceridaemia related to an increase in HDL cholesterol. We discuss the clinical and metabolic effects of fibrates, statins, nicotinic acid and sex steroids, and present novel therapeutic strategies that show promise in modifying HDL metabolism. In conclusion, HDL-cholesterol levels increase only moderately after treatment with currently available drugs and do not necessarily correlate with the functionality of HDL. Therefore, the anti-atherosclerotic therapy of high-risk cardiovascular patients should currently be focused on the correction of other risk factors present besides low HDL cholesterol. However, modification of HDL metabolism and improvement of RCT remain an attractive target for the development of new regimens of anti-atherogenic drug therapy.
Collapse
Affiliation(s)
- Martin Hersberger
- Institute of Clinical Chemistry, University Hospital Zurich, Zurich, Switzerland
| | | |
Collapse
|
519
|
Frank PG, Lee H, Park DS, Tandon NN, Scherer PE, Lisanti MP. Genetic Ablation of Caveolin-1 Confers Protection Against Atherosclerosis. Arterioscler Thromb Vasc Biol 2004; 24:98-105. [PMID: 14563650 DOI: 10.1161/01.atv.0000101182.89118.e5] [Citation(s) in RCA: 175] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
The development of atherosclerosis is a process characterized by the accumulation of lipids in the form of modified lipoproteins in the subendothelial space. This initiating step is followed by the subsequent recruitment and proliferation of other cell types, including monocytes/macrophages and smooth muscle cells. Here, we evaluate the potential role of caveolae membrane domains in the pathogenesis of atherosclerosis by using apolipoprotein E-deficient (ApoE−/−) mice as a model system.
Methods and Results—
Caveolin-1 (Cav-1) is a principal structural protein component of caveolae membrane domains. To directly assess the in vivo role of caveolae and Cav-1 in atherosclerosis, we interbred Cav-1−/− mice with ApoE−/− mice. Interestingly, loss of Cav-1 resulted in a dramatic >2-fold increase in non-HDL plasma cholesterol levels in the ApoE−/− background. However, despite this hypercholesterolemia, we found that loss of Cav-1 gene expression was clearly protective against the development of aortic atheromas, with up to an ≈70% reduction in atherosclerotic lesion area. Mechanistically, we demonstrated that loss of Cav-1 resulted in the dramatic downregulation of certain proatherogenic molecules, namely, CD36 and vascular cell adhesion molecule-1.
Conclusions—
Taken together, our results indicate that loss of Cav-1 can counteract the detrimental effects of atherogenic lipoproteins. Thus, Cav-1 is a novel target for drug development in the pharmacologic prevention of atheroma formation. Our current data also provide the first molecular genetic evidence to support the hypothesis that caveolar transcytosis of modified lipoproteins (from the blood to the sub-endothelial space) is a critical initiating step in atherosclerosis.
Collapse
Affiliation(s)
- Philippe G Frank
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Golding 202, Bronx, NY 10461, USA
| | | | | | | | | | | |
Collapse
|
520
|
Thorngate FE, Yancey PG, Kellner-Weibel G, Rudel LL, Rothblat GH, Williams DL. Testing the role of apoA-I, HDL, and cholesterol efflux in the atheroprotective action of low-level apoE expression. J Lipid Res 2003; 44:2331-8. [PMID: 12951361 DOI: 10.1194/jlr.m300224-jlr200] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Low levels of transgenic mouse apolipoprotein E (apoE) suppress atherosclerosis in apoE knockout (apoE-/-) mice without normalizing plasma cholesterol. To test whether this is due to facilitation of cholesterol efflux from the vessel wall, we produced apoA-I-/-/apoE-/- mice with or without the transgene. Even without apoA-I and HDL, apoA-I-/-/apoE-/- mice had the same amount of aorta cholesteryl ester as apoE-/- mice. Low apoE in the apoA-I-/-/apoE-/- transgenic mice reduced aortic lesions by 70% versus their apoA-I-/-/apoE-/- siblings. To define the free cholesterol (FC) efflux capacity of lipoproteins from the various genotypes, sera were assayed on macrophages expressing ATP-binding cassette transporter A1 (ABCA1). Surprisingly, ABCA1 FC efflux was twice as high to sera from the apoA-I-/-/apoE-/- or apoE-/- mice compared with wild-type mice, and this activity correlated with serum apoA-IV. Immunodepletion of apoA-IV from apoA-I-/-/apoE-/- serum abolished ABCA1 FC efflux, indicating that apoAI-V serves as a potent acceptor for FC efflux via ABCA1. With increasing apoE expression, apoA-IV and FC acceptor capacity decreased, indicating a reciprocal relationship between plasma apoE and apoA-IV. Low plasma apoE (1-3 x 10(-8) M) suppresses atherosclerosis by as yet undefined mechanisms, not dependent on the presence of apoA-I or HDL or an increased capacity of serum acceptors for FC efflux.
Collapse
Affiliation(s)
- Fayanne E Thorngate
- Department of Pharmacological Sciences, University Medical Center, State University of New York at Stony Brook, Stony Brook, NY 11794, USA
| | | | | | | | | | | |
Collapse
|
521
|
Maas R, Böger RH. Old and new cardiovascular risk factors: from unresolved issues to new opportunities. ATHEROSCLEROSIS SUPP 2003; 4:5-17. [PMID: 14664897 DOI: 10.1016/s1567-5688(03)00028-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
With the aim of identifying areas that may deserve some further thinking the present review deliberately points out controversial issues in cardiovascular research and risk assessment. In the first part of the review general aspects are addressed regarding the evaluation of risk factors. A first point of concern is the frequent practice of combining different vascular events and effects in different vascular beds into a single endpoint. Furthermore, verification of vascular events in clinical reality may be surprisingly inaccurate. Problems in risk assessment also arise from overlapping properties (shared pathophysiological pathways) of traditional risk factors like hypertension, obesity and diabetes. In the second part of the review unresolved issues concerning selected established and emerging risk factors are discussed. The difficulty of establishing causality in cardiovascular disease is addressed, using modification of LDL cholesterol and accumulating evidence for pleiotropic effects of the LDL cholesterol-lowering statins as an example. As an alternative or supplement to the notion of LDL-related cardiovascular risk it is proposed to distinguish between statin-sensitive and statin-insensitive cardiovascular risk. Finally the future prospects of selected new and emerging risk factors like CRP, homocysteine, asymmetrical dimethylarginine (ADMA), oxLDL, and isoprostanes are evaluated. In summary, imprecise terminology and varying definitions of "cardiovascular risk" may lead to a considerable blurring of our current risk estimates, which may also explain some presently controversial issues. With several new risk factors and substantial changes in lifestyle and treatment patterns on the horizon major changes in the hierarchy of risk factors may be inevitable.
Collapse
Affiliation(s)
- Renke Maas
- Institut für Experimentelle und Klinische Pharmakologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | | |
Collapse
|
522
|
Calabresi L, Gomaraschi M, Franceschini G. Endothelial protection by high-density lipoproteins: from bench to bedside. Arterioscler Thromb Vasc Biol 2003; 23:1724-31. [PMID: 12969988 DOI: 10.1161/01.atv.0000094961.74697.54] [Citation(s) in RCA: 186] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
There are several potential mechanisms by which HDLs protect against the development of vascular disease. One relates to the unique ability of these lipoproteins to remove cholesterol from the arterial wall. Another is the ability of HDL to prevent and eventually correct endothelial dysfunction, a key variable in the pathogenesis of atherosclerosis and its complications. HDLs help maintain endothelial integrity, facilitate vascular relaxation, inhibit blood cell adhesion to vascular endothelium, reduce platelet aggregability and coagulation, and may favor fibrinolysis. These functions of HDLs complement their activity in arterial cholesterol removal by providing an excellent rationale for favorably influencing pathological processes underlying a variety of clinical conditions, such as accelerated atherosclerosis, acute coronary syndromes, and restenosis after coronary angioplasty, through a chronic or acute elevation of plasma HDL concentration.
Collapse
Affiliation(s)
- Laura Calabresi
- Center E. Grossi Paoletti, Department of Pharmacological Sciences, University of Milano, Italy
| | | | | |
Collapse
|
523
|
Trigatti BL, Krieger M, Rigotti A. Influence of the HDL receptor SR-BI on lipoprotein metabolism and atherosclerosis. Arterioscler Thromb Vasc Biol 2003; 23:1732-8. [PMID: 12920050 DOI: 10.1161/01.atv.0000091363.28501.84] [Citation(s) in RCA: 185] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The scavenger receptor class B type I (SR-BI) was the first molecularly well-defined cell-surface HDL receptor to be described. SR-BI mediates selective HDL cholesterol uptake by formation of a productive lipoprotein/receptor complex, which requires specific structural domains and conformation states of apolipoprotein A-I present in HDL particles. SR-BI is abundantly expressed in several tissues, including the liver, where its expression is regulated by various mechanisms, including the transcriptional activity of nuclear receptors. The importance of SR-BI in overall HDL cholesterol metabolism and its antiatherogenic activity in vivo has been definitively established by SR-BI gene manipulation in mice. Remarkably, SR-BI/apolipoprotein E double-knockout mice develop complex coronary artery disease, myocardial infarction, and heart failure. Additional studies should help to define the importance of SR-BI in human health and disease.
Collapse
|
524
|
Escudero I, Martínez-González J, Alonso R, Mata P, Badimon L. Experimental and interventional dietary study in humans on the role of HDL fatty acid composition in PGI2 release and Cox-2 expression by VSMC. Eur J Clin Invest 2003; 33:779-86. [PMID: 12925037 DOI: 10.1046/j.1365-2362.2003.01221.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND High-density lipoproteins (HDLs) induce prostacyclin (PGI2) release in vascular smooth muscle cells (VSMCs) by up-regulation of cyclooxygenase-2 (Cox-2). Our goal was to analyze the role of human HDL lipid moiety on Cox-2-dependent PGI2 synthesis in human VSMCs and to assess the impact that the intake of diets with different fatty acid composition exert on HDL-induced PGI2 release. MATERIALS AND METHODS Human VSMCs were treated with HDL or fatty acids in the presence or absence of different cell signalling inhibitors and PGI2 (by enzyme immunoassay) and Cox-2 protein levels (by Western blot) were analyzed. High-density lipoproteins were obtained from a plasma pool or from plasma of 12 volunteers subjected to a longitudinal dietary interventional study of three consecutive diets periods enriched in monounsaturated fatty acids (MUFAs), polyunsaturated fatty acids n-6 (PUFA n-6) or n-3 (PUFA n-3). RESULTS High-density lipoprotein delipidation attenuated the effect of HDL on both PGI2 synthesis and Cox-2 up-regulation, while arachidonic acid (AA) but not other fatty acids mimicked the effects of HDL. Arachidonic acid induced PGI2 synthesis and Cox-2 expression through similar mechanisms to those activated by HDL [pertussis toxin-sensitive G proteins, p42/44 mitogen-activated protein kinase (MAPK), p38MAPK, and c-Jun N-terminal kinase-1 (JNK-1) pathways]. Finally, we observed that HDL from the PUFA n-3 dietary period induced lower PGI2 release than that from the PUFA n-6 period (64% vs. 100%). CONCLUSIONS Our results suggest that lipid moiety modulates HDL-induced PGI2 release/Cox-2 up-regulation in human VSMCs, and that changes in fatty acids as accomplished with the diet can modulate vascular PGI2 homeostasis.
Collapse
Affiliation(s)
- I Escudero
- Centro de Investigación Cardiovascular, CSIC/ICCC-Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | | | | | | |
Collapse
|
525
|
Covey SD, Krieger M, Wang W, Penman M, Trigatti BL. Scavenger receptor class B type I-mediated protection against atherosclerosis in LDL receptor-negative mice involves its expression in bone marrow-derived cells. Arterioscler Thromb Vasc Biol 2003; 23:1589-94. [PMID: 12829524 DOI: 10.1161/01.atv.0000083343.19940.a0] [Citation(s) in RCA: 177] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Scavenger receptor class B type I (SR-BI) is a cell-surface HDL receptor that is implicated in reverse cholesterol transport and protection against atherosclerosis. We have previously demonstrated that SR-BI/apolipoprotein E double-knockout mice develop severe occlusive coronary artery disease and myocardial infarction and die at approximately 6 weeks of age. To determine if this is a general effect of a lack of SR-BI, we generated mice deficient in both SR-BI and the LDL receptor. METHODS AND RESULTS Complete ablation of SR-BI expression in LDL receptor knockout mice resulted in increased plasma cholesterol associated with HDL particles of abnormally large size and a 6-fold increase in diet-induced aortic atherosclerosis but no macroscopic evidence of early-onset coronary artery disease, cardiac pathology, or early death. Furthermore, selective elimination of SR-BI expression in bone marrow-derived cells resulted in increased diet-induced atherosclerosis in LDL receptor knockout mice without concomitant alterations in the distributions of plasma lipoprotein cholesterol. CONCLUSIONS SR-BI expression protects against atherosclerosis in LDL receptor-deficient as well as apolipoprotein E-deficient mice, and its expression in bone marrow-derived cells contributes to this protection.
Collapse
MESH Headings
- Animals
- Arteriosclerosis/prevention & control
- Bone Marrow Cells/metabolism
- Bone Marrow Transplantation
- CD36 Antigens/biosynthesis
- CD36 Antigens/genetics
- CD36 Antigens/physiology
- Diet, Atherogenic
- Membrane Proteins
- Mice
- Mice, Inbred C57BL
- Mice, Inbred Strains
- Mice, Knockout
- Receptors, Immunologic
- Receptors, LDL/deficiency
- Receptors, Lipoprotein/biosynthesis
- Receptors, Lipoprotein/genetics
- Receptors, Lipoprotein/physiology
- Receptors, Scavenger
- Scavenger Receptors, Class B
Collapse
Affiliation(s)
- Scott D Covey
- Department of Biochemistry, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | |
Collapse
|
526
|
Abstract
Plasma levels of high-density lipoprotein (HDL) cholesterol and its major protein, apolipoprotein A-I, are inversely correlated with the incidence of atherosclerotic cardiovascular disease. Low HDL cholesterol and apolipoprotein A-I levels often are found in association with other cardiovascular risk factors, including the metabolic syndrome, insulin resistance, and type 2 diabetes mellitus. However, overexpression of apolipoprotein A-I in animals has been shown to reduce progression and even induce regression of atherosclerosis, indicating that apolipoprotein A-I is directly protective against atherosclerosis. A major mechanism by which apolipoprotein A-I inhibits atherosclerosis may be by promoting cholesterol efflux from macrophages and returning it to the liver for excretion, a process termed reverse cholesterol transport. This article focuses on new developments in the regulation of reverse cholesterol transport and the clinical implications of those developments.
Collapse
Affiliation(s)
- Daniel J Rader
- Preventive Cardiology/Lipid Research Center, University of Pennsylvania Health System, Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
527
|
Kuvin JT, Patel AR, Sidhu M, Rand WM, Sliney KA, Pandian NG, Karas RH. Relation between high-density lipoprotein cholesterol and peripheral vasomotor function. Am J Cardiol 2003; 92:275-9. [PMID: 12888130 DOI: 10.1016/s0002-9149(03)00623-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Low levels of high-density lipoprotein (HDL) cholesterol are one of the most common lipid abnormalities in patients with coronary artery disease. Endothelial dysfunction is also highly prevalent in patients with coronary artery disease. We sought to determine whether HDL cholesterol levels are correlated with endothelium-dependent vasomotion in patients being evaluated for atherosclerosis. Peripheral vascular endothelial function was assessed by high-resolution brachial artery ultrasound. Flow-mediated dilation (FMD) during reactive hyperemia was defined as the percent change in arterial diameter following 5-minute arterial occlusion. All patients underwent stress testing with nuclear single-photon emission computed tomographic imaging to determine percent left ventricular ejection fraction and define the presence or absence of coronary artery disease. One hundred fifty-one subjects (87 men, 64 women) were enrolled (average age 58 +/- 11 years). Total cholesterol, HDL cholesterol, low-density lipoprotein cholesterol, and triglyceride levels were 188 +/- 48, 47 +/- 13, 108 +/- 37 and 154 +/- 88 mg/dl, respectively. The mean FMD for the entire group was 9.9 +/- 5.2%. Subjects with an HDL cholesterol of <40 mg/dl (n = 39) had lower FMD (7.4 +/- 3.6%) compared with those with an HDL cholesterol >/=40 mg/dl (11.0 +/- 5.5%, p <0.001). There was a significant correlation between FMD and HDL cholesterol level (linear regression, p <0.001), and in multivariate analysis, HDL cholesterol was an independent predictor of FMD. Peripheral endothelial function was abnormal in subjects with low HDL cholesterol and well-preserved in those with high HDL cholesterol. These data suggest that impaired endothelial function associated with low HDL cholesterol may be an additional, previously unrecognized mechanism contributing to the increased risk of atherosclerosis in these patients.
Collapse
Affiliation(s)
- Jeffrey T Kuvin
- Department of Medicine/Division of Cardiology, Tufts-New England Medical Center, Boston, Massachusetts 02111, USA.
| | | | | | | | | | | | | |
Collapse
|
528
|
Silver DL, Wang N, Vogel S. Identification of small PDZK1-associated protein, DD96/MAP17, as a regulator of PDZK1 and plasma high density lipoprotein levels. J Biol Chem 2003; 278:28528-32. [PMID: 12754212 DOI: 10.1074/jbc.m304109200] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Scavenger receptor class B, type I (SR-BI) is the high density lipoprotein (HDL) receptor essential for hepatic uptake of HDL cholesterol. SR-BI was shown to impact plasma HDL levels and be anti-atherogenic. Thus, the ability to regulate hepatic SR-BI may allow for the modulation of plasma HDL cholesterol and progression of atherosclerosis. However, regulation of SR-BI in liver is not well understood. Recently, the PDZ domain containing protein PDZK1 was shown to interact with SR-BI and may serve an essential role in SR-BI cell surface expression. Here we identify an in vivo PDZK1-interacting protein that we named small PDZK1-associated protein (SPAP; also known as DD96/MAP17). Unexpectedly, we found that hepatic overexpression of SPAP in mice resulted in liver deficiency of PDZK1. The absence of PDZK1 in SPAP transgenic mice resulted in a deficiency of SR-BI in liver and markedly increased plasma HDL. Metabolic labeling experiments showed that the proteasome plays a role in the turnover of newly synthesized PDZK1, but that SPAP overexpression in liver increased PDZK1 turnover in an alternate, proteasome-independent pathway. Thus, SPAP may be an endogenous regulator of cellular PDZK1 levels by regulating PDZK1 turnover.
Collapse
MESH Headings
- Acetylcysteine/analogs & derivatives
- Acetylcysteine/pharmacology
- Amino Acid Sequence
- Animals
- CD36 Antigens/genetics
- CD36 Antigens/physiology
- Cholesterol, HDL/blood
- Chromatography, High Pressure Liquid
- Cysteine Endopeptidases
- Electrophoresis, Polyacrylamide Gel
- Enzyme Inhibitors/pharmacology
- Gene Expression
- Gene Expression Regulation
- Humans
- Liver/chemistry
- Liver/metabolism
- Membrane Proteins/analysis
- Membrane Proteins/chemistry
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Membrane Proteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Molecular Sequence Data
- Multienzyme Complexes/antagonists & inhibitors
- Neoplasm Proteins
- Proteasome Endopeptidase Complex
- Receptors, Immunologic
- Receptors, Lipoprotein
- Receptors, Scavenger
- Scavenger Receptors, Class B
- Transfection
Collapse
Affiliation(s)
- David L Silver
- Department of Medicine, Division of Molecular Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA.
| | | | | |
Collapse
|
529
|
Kimura T, Sato K, Malchinkhuu E, Tomura H, Tamama K, Kuwabara A, Murakami M, Okajima F. High-density lipoprotein stimulates endothelial cell migration and survival through sphingosine 1-phosphate and its receptors. Arterioscler Thromb Vasc Biol 2003; 23:1283-8. [PMID: 12775579 DOI: 10.1161/01.atv.0000079011.67194.5a] [Citation(s) in RCA: 181] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Plasma high-density lipoprotein (HDL) level is inversely correlated with the risk of atherosclerosis. However, the cellular mechanism by which HDL exerts antiatherogenic actions is not well understood. In this study, we focus on the lipid components of HDL as mediators of the lipoprotein-induced antiatherogenic actions. METHODS AND RESULTS HDL and sphingosine 1-phosphate (S1P) stimulated the migration and survival of human umbilical vein endothelial cells. These responses to HDL and S1P were almost completely inhibited by pertussis toxin and other specific inhibitors for intracellular signaling pathways, although the inhibition profiles of migration and survival were different. The HDL-stimulated migration and survival of the cells were markedly inhibited by antisense oligonucleotides against the S1P receptors EDG-1/S1P1 and EDG-3/S1P3. Cell migration was sensitive to both receptors, but cell survival was exclusively sensitive to S1P1. The S1P-rich fraction and chromatographically purified S1P from HDL stimulated cell migration, but the rest of the fraction did not, as was the case of the cell survival. CONCLUSIONS HDL-induced endothelial cell migration and survival may be mediated by the lipoprotein component S1P and the lipid receptors S1P1 and S1P3.
Collapse
Affiliation(s)
- Takao Kimura
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | | | | | | | | | | | | | | |
Collapse
|
530
|
Thiemermann C, Patel NSA, Kvale EO, Cockerill GW, Brown PAJ, Stewart KN, Cuzzocrea S, Britti D, Mota-Filipe H, Chatterjee PK. High density lipoprotein (HDL) reduces renal ischemia/reperfusion injury. J Am Soc Nephrol 2003; 14:1833-43. [PMID: 12819243 DOI: 10.1097/01.asn.0000075552.97794.8c] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
High-density lipoproteins (HDL) have been shown to reduce organ injury and mortality in animal models of shock via modulation of the expression of adhesion molecules and pro-inflammatory enzymes. As renal inflammation plays an important role in the development of ischemia/reperfusion (I/R) injury of the kidney, the aim of this study was to investigate the ability of HDL to alleviate renal dysfunction and injury in a rat model of renal I/R. HDL (80 mg/kg, intravenous) was administered to male Wistar rats 30 min before bilateral renal ischemia for 45 min followed by reperfusion for up to 48 h. After 6-h reperfusion, HDL significantly reduced (1) renal and tubular dysfunction, (2) tubular and reperfusion-injury, and (3) histologic evidence of renal injury. HDL also improved renal function (after 24-h and 48-h reperfusion) and reduced histologic signs of renal injury (after 48-h reperfusion). Administration of HDL significantly reduced the numbers of polymorphonuclear leukocytes (PMN) infiltrating into renal tissues during reperfusion, which was reflected by an attenuation of the increase in renal myeloperoxidase activity caused by I/R. Furthermore, HDL markedly reduced expression of the adhesion molecules, intercellular adhesion molecule-1 (ICAM-1), and P-selectin during reperfusion. The increase in renal malondialdehyde levels caused by renal I/R was also significantly reduced by HDL, suggesting attenuation of lipid peroxidation subsequent to oxidative stress. These results demonstrate that HDL significantly reduces renal I/R injury and severity of ischemic acute renal failure. It is proposed that the mechanism of protection involves reduction of the expression of adhesion molecules, resulting in reduction of PMN infiltration and oxidative stress.
Collapse
Affiliation(s)
- Christoph Thiemermann
- Department of Experimental Medicine & Nephrology, William Harvey Research Institute, Queen Mary-University of London, Charterhouse Square, London EC1M 6BQ, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
531
|
Frank PG, Woodman SE, Park DS, Lisanti MP. Caveolin, caveolae, and endothelial cell function. Arterioscler Thromb Vasc Biol 2003; 23:1161-8. [PMID: 12689915 DOI: 10.1161/01.atv.0000070546.16946.3a] [Citation(s) in RCA: 281] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Caveolae are 50- to 100-nm cell-surface plasma membrane invaginations observed in terminally differentiated cells. They are particularly abundant in endothelial cells, where they are believed to play a major role in the regulation of endothelial vesicular trafficking and signal transduction. The use of caveolin-1-deficient mice has provided many new insights into the roles of caveolae and caveolin-1 in the regulation of endothelial cell function. These novel findings suggest an important role for caveolin-1 in the pathogenesis of cancer, atherosclerosis, and vascular disease.
Collapse
Affiliation(s)
- Philippe G Frank
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | |
Collapse
|
532
|
Rämet ME, Rämet M, Lu Q, Nickerson M, Savolainen MJ, Malzone A, Karas RH. High-density lipoprotein increases the abundance of eNOS protein in human vascular endothelial cells by increasing its half-life. J Am Coll Cardiol 2003; 41:2288-97. [PMID: 12821261 DOI: 10.1016/s0735-1097(03)00481-9] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Given the importance of endothelial nitric oxide synthase (eNOS) in regulating endothelium-dependent vasorelaxation, we investigated the effects of high-density lipoprotein in (HDL) on eNOS protein abundance in cultured human vascular endothelial cells. BACKGROUND Endothelial dysfunction, characterized by decreased nitric oxide production, is one of the early features in the development of atherosclerosis. We have recently shown in vivo that niacin therapy increases plasma HDL concentration and improves endothelium-dependent vasorelaxation in patients with coronary artery disease. METHODS Human vascular endothelial cells were cultured in the presence or absence of HDL or apolipoprotein (apo)A-I. The eNOS protein abundance was assessed by immunoblotting, and protein half-life was assessed by pulse-chase techniques. The eNOS messenger ribonucleic acid (mRNA) abundance was measured using real-time quantitative polymerase chain reaction. RESULTS High density lipoprotein, or apoA-I alone, increased eNOS protein abundance by 3.5 +/- 0.7 and 2.7 +/- 0.5-fold, respectively (p < 0.05 for both). However, neither HDL nor apoA-I increased eNOS mRNA abundance. It was shown that HDL increased eNOS protein half-life up to 3.3 +/- 0.2-fold (p = 0.001). Both HDL and apoA-I activated mitogen-activated protein-kinase and phosphatidylinositol 3-kinase (PI3K) Akt-pathways in human arterial endothelial cells, and inhibition of either of these pathways by specific pharmacologic inhibitors abolished the effect of HDL on eNOS. CONCLUSIONS We demonstrate that HDL activates both extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt, resulting in enhanced eNOS protein stability and subsequent accumulation of eNOS protein. This posttranslational regulation represents a previously unrecognized mechanism for regulating eNOS.
Collapse
Affiliation(s)
- Maria E Rämet
- Molecular Cardiology Research Institute and Department of Medicine, New England Medical Center Hospitals, Inc., Tufts University School of Medicine, Boston, Massachusetts 02111, USA
| | | | | | | | | | | | | |
Collapse
|
533
|
Bisoendial RJ, Hovingh GK, Levels JHM, Lerch PG, Andresen I, Hayden MR, Kastelein JJP, Stroes ESG. Restoration of endothelial function by increasing high-density lipoprotein in subjects with isolated low high-density lipoprotein. Circulation 2003; 107:2944-8. [PMID: 12771001 DOI: 10.1161/01.cir.0000070934.69310.1a] [Citation(s) in RCA: 217] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Loss-of-function mutations in the ATP-binding cassette (ABCA)-1 gene locus are the underlying cause for familial hypoalphalipoproteinemia, providing a human isolated low-HDL model. In these familial hypoalphalipoproteinemia subjects, we evaluated the impact of isolated low HDL on endothelial function and the vascular effects of an acute increase in HDL. METHODS AND RESULTS In 9 ABCA1 heterozygotes and 9 control subjects, vascular function was assessed by venous occlusion plethysmography. Forearm blood flow responses to the endothelium-dependent and -independent vasodilators serotonin (5HT) and sodium nitroprusside, respectively, and the inhibitor of nitric oxide synthase NG-monomethyl-l-arginine (L-NMMA) were measured. Dose-response curves were repeated after systemic infusion of apolipoprotein A-I/phosphatidylcholine (apoA-I/PC) disks. At baseline, ABCA1 heterozygotes had decreased HDL levels (0.4+/-0.2 mmol/L; P<0.05), and their forearm blood flow responses to both 5HT (maximum, 49.0+/-10.4%) and L-NMMA (maximum, -22.8+/-22.9%) were blunted compared with control subjects (both P< or =0.005). Infusion of apoA-I/PC disks increased plasma HDL to 1.3+/-0.4 mmol/L in ABCA1 heterozygotes, which resulted in complete restoration of vasomotor responses to both 5HT and L-NMMA (both P</=0.001). Endothelium-independent vasodilation remained unaltered throughout the protocol. CONCLUSIONS In ABCA1 heterozygotes, isolated low HDL is associated with endothelial dysfunction, attested to by impaired basal and stimulated NO bioactivity. Strikingly, both parameters were completely restored after a single, rapid infusion of apoA-I/PC. These findings indicate that in addition to its long-term role within reverse cholesterol transport, HDL per se also exerts direct beneficial effects on the arterial wall.
Collapse
Affiliation(s)
- Radjesh J Bisoendial
- Department of Vascular Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
534
|
Braun A, Zhang S, Miettinen HE, Ebrahim S, Holm TM, Vasile E, Post MJ, Yoerger DM, Picard MH, Krieger JL, Andrews NC, Simons M, Krieger M. Probucol prevents early coronary heart disease and death in the high-density lipoprotein receptor SR-BI/apolipoprotein E double knockout mouse. Proc Natl Acad Sci U S A 2003; 100:7283-8. [PMID: 12771386 PMCID: PMC165867 DOI: 10.1073/pnas.1237725100] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2002] [Accepted: 04/17/2003] [Indexed: 12/20/2022] Open
Abstract
Mice with homozygous null mutations in the high-density lipoprotein receptor SR-BI (scavenger receptor class B, type I) and apolipoprotein E genes fed a low-fat diet exhibit a constellation of pathologies shared with human atherosclerotic coronary heart disease (CHD): hypercholesterolemia, occlusive coronary atherosclerosis, myocardial infarctions, cardiac dysfunction (heart enlargement, reduced systolic function and ejection fraction, and ECG abnormalities), and premature death (mean age 6 weeks). They also exhibit a block in RBC maturation and abnormally high plasma unesterified-to-total cholesterol ratio (0.8) with associated abnormal lipoprotein morphology (lamellar/vesicular and stacked discoidal particles reminiscent of those in lecithin/cholesterol acyltransferase deficiency and cholestasis). Treatment with the lipid-lowering, antiatherosclerosis, and antioxidation drug probucol extended life to as long as 60 weeks (mean 36 weeks), and at 5-6 weeks of age, virtually completely reversed the cardiac and most RBC pathologies and corrected the unesterified to total cholesterol ratio (0.3) and associated distinctive abnormal lipoprotein morphologies. Manipulation of the timing of administration and withdrawal of probucol could control the onset of death and suggested that critical pathological changes usually occurred in untreated double knockout mice between approximately 3 (weaning) and 5 weeks of age and that probucol delayed heart failure even after development of substantial CHD. The ability of probucol treatment to modulate pathophysiology in the double knockout mice enhances the potential of this murine system for analysis of the pathophysiology of CHD and preclinical testing of new approaches for the prevention and treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Anne Braun
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
535
|
Abstract
Observational studies provide overwhelming evidence that a low high-density lipoprotein (HDL)-cholesterol level increases the risk of coronary events, both in healthy subjects and in patients with coronary heart disease. Based on in vitro experiments, several mechanistic explanations for the atheroprotective function of HDL have been suggested. However, few of these were verified in vivo in humans or in experiments with transgenic animals. The HDL functions currently most widely held to account for the antiatherogenic effect include participation in reverse cholesterol transport, protection against endothelial dysfunction, and inhibition of oxidative stress. This review summarizes current views on the molecular mechanism underlying these atheroprotective effects of HDL.
Collapse
Affiliation(s)
- Gerd Assmann
- Institut für Klinische Chemie und Laboratoriumsmedizin, Westfälische Wilhelms-Universität, Münster, Germany.
| | | |
Collapse
|
536
|
Rigotti A, Miettinen HE, Krieger M. The role of the high-density lipoprotein receptor SR-BI in the lipid metabolism of endocrine and other tissues. Endocr Rev 2003; 24:357-87. [PMID: 12788804 DOI: 10.1210/er.2001-0037] [Citation(s) in RCA: 324] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Because cholesterol is a precursor for the synthesis of steroid hormones, steroidogenic tissues have evolved multiple pathways to ensure adequate supplies of cholesterol. These include synthesis, storage as cholesteryl esters, and import from lipoproteins. In addition to endocytosis via members of the low-density lipoprotein receptor superfamily, steroidogenic cells acquire cholesterol from lipoproteins by selective lipid uptake. This pathway, which does not involve lysosomal degradation of the lipoprotein, is mediated by the scavenger receptor class B type I (SR-BI). SR-BI is highly expressed in steroidogenic cells, where its expression is regulated by various trophic hormones, as well as in the liver. Studies of genetically manipulated strains of mice have established that SR-BI plays a key role in regulating lipoprotein metabolism and cholesterol transport to steroidogenic tissues and to the liver for biliary secretion. In addition, analysis of SR-BI-deficient mice has shown that SR-BI expression is important for alpha-tocopherol and nitric oxide metabolism, as well as normal red blood cell maturation and female fertility. These mouse models have also revealed that SR-BI can protect against atherosclerosis. If SR-BI plays similar physiological and pathophysiological roles in humans, it may be an attractive target for therapeutic intervention in cardiovascular and reproductive diseases.
Collapse
Affiliation(s)
- Attilio Rigotti
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica, Santiago, Chile
| | | | | |
Collapse
|
537
|
Barter P, Kastelein J, Nunn A, Hobbs R. High density lipoproteins (HDLs) and atherosclerosis; the unanswered questions. Atherosclerosis 2003; 168:195-211. [PMID: 12801602 DOI: 10.1016/s0021-9150(03)00006-6] [Citation(s) in RCA: 148] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The concentration of high density lipoprotein-cholesterol (HDL-C) has been found consistently to be a powerful negative predictor of premature coronary heart disease (CHD) in human prospective population studies. There is also circumstantial evidence from human intervention studies and direct evidence from animal intervention studies that HDLs protect against the development of atherosclerosis. HDLs have several documented functions, although the precise mechanism by which they prevent atherosclerosis remains uncertain. Nor is it known whether the cardioprotective properties of HDL are specific to one or more of the many HDL subpopulations that comprise the HDL fraction in human plasma. Several lifestyle and pharmacological interventions have the capacity to raise the level of HDL-C, although it is not known whether all are equally protective. Indeed, despite the large body of information identifying HDLs as potential therapeutic targets for the prevention of atherosclerosis, there remain many unanswered questions that must be addressed as a matter of urgency before embarking wholesale on HDL-C-raising therapies as strategies to prevent CHD. This review summarises what is known and highlights what we still need to know.
Collapse
Affiliation(s)
- Philip Barter
- Cardiovascular Investigation Unit, Royal Adelaide Hospital, North Terrace, Adelaide, SA 5000, Australia.
| | | | | | | |
Collapse
|
538
|
Gong M, Wilson M, Kelly T, Su W, Dressman J, Kincer J, Matveev SV, Guo L, Guerin T, Li XA, Zhu W, Uittenbogaard A, Smart EJ. HDL-associated estradiol stimulates endothelial NO synthase and vasodilation in an SR-BI-dependent manner. J Clin Invest 2003; 111:1579-87. [PMID: 12750408 PMCID: PMC155043 DOI: 10.1172/jci16777] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular diseases remain the leading cause of death in the United States. Two factors associated with a decreased risk of developing cardiovascular disease are elevated HDL levels and sex - specifically, a decreased risk is found in premenopausal women. HDL and estrogen stimulate eNOS and the production of nitric oxide, which has numerous protective effects in the vascular system including vasodilation, antiadhesion, and anti-inflammatory effects. We tested the hypothesis that HDL binds to its receptor, scavenger receptor class B type I (SR-BI), and delivers estrogen to eNOS, thereby stimulating the enzyme. HDL isolated from women stimulated eNOS, whereas HDL isolated from men had minimal activity. Studies with ovariectomized and ovariectomized/estrogen replacement mouse models demonstrated that HDL-associated estradiol stimulation of eNOS is SR-BI dependent. Furthermore, female HDL, but not male HDL, promoted the relaxation of muscle strips isolated from C57BL/6 mice but not SR-BI null mice. Finally, HDL isolated from premenopausal women or postmenopausal women receiving estradiol replacement therapy stimulated eNOS, whereas HDL isolated from postmenopausal women did not stimulate eNOS. We conclude that HDL-associated estrodial is capable of the stimulating eNOS. These studies establish a new paradigm for examining the cardiovascular effects of HDL and estrogen.
Collapse
MESH Headings
- Animals
- CD36 Antigens/genetics
- CD36 Antigens/metabolism
- Cells, Cultured
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Estradiol/metabolism
- Estradiol/pharmacology
- Female
- Femoral Artery/drug effects
- Femoral Artery/physiology
- Hormone Replacement Therapy
- Humans
- In Vitro Techniques
- Lipoproteins, HDL/metabolism
- Lipoproteins, HDL/pharmacology
- Male
- Membrane Proteins
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Nitric Oxide Synthase/drug effects
- Nitric Oxide Synthase/metabolism
- Nitric Oxide Synthase Type II
- Nitric Oxide Synthase Type III
- Ovariectomy
- Receptors, Immunologic
- Receptors, Lipoprotein
- Receptors, Scavenger
- Scavenger Receptors, Class B
- Sex Factors
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Ming Gong
- Department of Pediatrics, University of Kentucky Medical School, Lexington, Kentucky, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
539
|
Grewal T, de Diego I, Kirchhoff MF, Tebar F, Heeren J, Rinninger F, Enrich C. High density lipoprotein-induced signaling of the MAPK pathway involves scavenger receptor type BI-mediated activation of Ras. J Biol Chem 2003; 278:16478-81. [PMID: 12637559 DOI: 10.1074/jbc.c300085200] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
High density lipoprotein (HDL) stimulates multiple signaling pathways. HDL-induced activation of the mitogen-activated protein kinase (MAPK) pathway can be mediated by protein kinase C (PKC) and/or pertussis toxin-sensitive G-proteins. Although HDL-induced activation of MAPK involves Raf-1, Mek, and Erk1/2, the upstream contribution of p21(ras) (Ras) on the activation of Raf-1 and MAPK remains elusive. Here we examine the effect of HDL on Ras activity and demonstrate that HDL induces PKC-independent activation of Ras that is completely blocked by pertussis toxin, thus implicating heterotrimeric G-proteins. In addition, the HDL-induced activation of Ras is inhibited by a neutralizing antibody against scavenger receptor type BI. We conclude that the binding of HDL to scavenger receptor type BI activates Ras in a PKC-independent manner with subsequent induction of the MAPK signaling cascade.
Collapse
Affiliation(s)
- Thomas Grewal
- Departament de Biologia Cellular, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
540
|
Arking DE, Becker DM, Yanek LR, Fallin D, Judge DP, Moy TF, Becker LC, Dietz HC. KLOTHO allele status and the risk of early-onset occult coronary artery disease. Am J Hum Genet 2003; 72:1154-61. [PMID: 12669274 PMCID: PMC1180268 DOI: 10.1086/375035] [Citation(s) in RCA: 188] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2003] [Accepted: 02/07/2003] [Indexed: 11/04/2022] Open
Abstract
We previously identified a functional variant of KLOTHO (termed "KL-VS"), which harbors two amino acid substitutions in complete linkage disequilibrium and is associated with reduced human longevity when in homozygosity. Klotho-deficient mice display extensive arteriosclerosis when fed a normal diet, suggesting a potent genetic predisposition. To determine whether klotho influences atherosclerotic risk in humans, we performed cross-sectional studies to assess the association between the KL-VS allele and occult coronary artery disease (CAD) in two independent samples of apparently healthy siblings of individuals with early-onset (age <60 years) CAD (SIBS-I [N=520] and SIBS-II [N=436]). Occult CAD was defined as the occurrence of a reversible perfusion defect during exercise thallium scintigraphy and/or as an abnormal result of an exercise electrocardiogram (SIBS-I, n=97; SIBS-II, n=56). In SIBS-I, the KL-VS allele conferred a relative odds of 1.90 (95% confidence interval 1.21-2.98) for occult CAD, after adjusting for familial intraclass correlations (P<.005). Logistic regression modeling, incorporating known CAD risk factors, demonstrated that the KL-VS allele is an independent risk factor (P<.019) and that the imposed risk of KL-VS allele status is influenced by modifiable risk factors. Hypertension (P<.022) and increasing high-density lipoprotein cholesterol (HDL-C) levels (P<.022) mask or reduce the risk conferred by the KL-VS allele, respectively, whereas current smoking (P<.004) increases the risk. Remarkably concordant effects of the KL-VS allele and modifying factors on the risk of occult CAD were seen in SIBS-II. These results demonstrate that the KL-VS allele is an independent risk factor for occult CAD in two independent high-risk samples. Modifiable risk factors, including hypertension, smoking status, and HDL-C level, appear to influence the risk imposed by this allele.
Collapse
Affiliation(s)
- Dan E. Arking
- McKusick-Nathans Institute of Genetic Medicine, Center for Health Promotion, Division of Internal Medicine, and Division of Cardiology, Johns Hopkins University School of Medicine, Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, and Howard Hughes Medical Institute, Baltimore
| | - Diane M. Becker
- McKusick-Nathans Institute of Genetic Medicine, Center for Health Promotion, Division of Internal Medicine, and Division of Cardiology, Johns Hopkins University School of Medicine, Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, and Howard Hughes Medical Institute, Baltimore
| | - Lisa R. Yanek
- McKusick-Nathans Institute of Genetic Medicine, Center for Health Promotion, Division of Internal Medicine, and Division of Cardiology, Johns Hopkins University School of Medicine, Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, and Howard Hughes Medical Institute, Baltimore
| | - Daniele Fallin
- McKusick-Nathans Institute of Genetic Medicine, Center for Health Promotion, Division of Internal Medicine, and Division of Cardiology, Johns Hopkins University School of Medicine, Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, and Howard Hughes Medical Institute, Baltimore
| | - Daniel P. Judge
- McKusick-Nathans Institute of Genetic Medicine, Center for Health Promotion, Division of Internal Medicine, and Division of Cardiology, Johns Hopkins University School of Medicine, Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, and Howard Hughes Medical Institute, Baltimore
| | - Taryn F. Moy
- McKusick-Nathans Institute of Genetic Medicine, Center for Health Promotion, Division of Internal Medicine, and Division of Cardiology, Johns Hopkins University School of Medicine, Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, and Howard Hughes Medical Institute, Baltimore
| | - Lewis C. Becker
- McKusick-Nathans Institute of Genetic Medicine, Center for Health Promotion, Division of Internal Medicine, and Division of Cardiology, Johns Hopkins University School of Medicine, Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, and Howard Hughes Medical Institute, Baltimore
| | - Harry C. Dietz
- McKusick-Nathans Institute of Genetic Medicine, Center for Health Promotion, Division of Internal Medicine, and Division of Cardiology, Johns Hopkins University School of Medicine, Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, and Howard Hughes Medical Institute, Baltimore
| |
Collapse
|
541
|
Okamoto H, Iwamoto Y, Maki M, Sotani T, Yonemori F, Wakitani K. Effect of JTT-705 on cholesteryl ester transfer protein and plasma lipid levels in normolipidemic animals. Eur J Pharmacol 2003; 466:147-54. [PMID: 12679151 DOI: 10.1016/s0014-2999(03)01548-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
This study evaluated JTT-705, S-[2-([[1-(2-ethylbutyl)cyclohexyl]carbonyl]amino)phenyl]2-methylpropanethioate, as a cholesteryl ester transfer protein (CETP) inhibitor in several animal species. In vitro, JTT-705 inhibited plasma CETP activities of humans, rabbits, hamsters, cynomolgus monkeys and marmosets with IC(50) values of 5.5, 1.0, 11.7, 2.4 and 6.3 microM, respectively. The thiol form (JTP-25203) also inhibited those activities with IC(50) values of 2.8, 0.44, 0.52, 1.3 and 1.1 microM, respectively. Following oral administration to normolipidemic animals (rabbits, hamsters and marmosets), JTT-705 reduced plasma CETP activity, increased high density lipoprotein cholesterol (HDL-cholesterol), and decreased the ratio of non-HDL-cholesterol to HDL-cholesterol (atherogenic index) in all species. In marmosets, JTT-705 increased slow alpha-migrating lipoprotein (apolipoprotein E-rich HDL) in agarose gel electrophoresis, indicating that HDL metabolism in JTT-705-treated marmosets is similar to that in CETP-deficient humans. These results indicate that JTT-705 can be expected to inhibit plasma CETP activity and improve plasma lipoprotein profiles in a wide range of animal species, including humans.
Collapse
Affiliation(s)
- Hiroshi Okamoto
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, 569-1125, Osaka, Japan.
| | | | | | | | | | | |
Collapse
|
542
|
Kosaka H, Yoneyama H, Zhang L, Fujii S, Yamamoto A, Igarashi J. Induction of LOX-1 and iNOS expressions by ischemia-reperfusion of rat kidney and the opposing effect of L-arginine. FASEB J 2003; 17:636-43. [PMID: 12665476 DOI: 10.1096/fj.02-0585com] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Lectin-like oxidized low-density lipoprotein receptor (LOX-1) is a newly identified endothelial cell surface major receptor for oxidatively modified low-density lipoprotein. Progression of arthrosclerosis in the donor organ after organ transplantation is a major problem. We hypothesized that ischemia-reperfusion induces LOX-1. After 1 h ischemia of bilateral kidneys plus 3, 6, or 12 h reperfusion, we first revealed that LOX-1 mRNA expression was increased in renal cortex and medulla at 6 h after reperfusion, which was decreased by L-arginine supplement. Plasma nitric oxide (NO) end-product nitrite plus nitrate and inducible nitric oxide synthase (NOS) expression were increased after reperfusion of 6 h. However, NOS substrate L-arginine did not augment but markedly decreased plasma NO end product, because L-arginine supplement suppressed inducible NOS expression in kidney. We hypothesized that available L-arginine is depleted by ischemia-reperfusion, leading to inducible NOS induction. Ischemia decreased L-arginine levels in kidney and L-arginine supplement increased NO end products in renal cortex in the earliest phase of reperfusion. These results disclosed for the first time that a deficiency in L-arginine by ischemia reperfusion causes uncoupling of constitutive NOS, which induces inducible NOS and LOX-1, implying why L-arginine is effective for stroke or transplantation in preventing atherosclerotic progress.
Collapse
Affiliation(s)
- Hiroaki Kosaka
- The 2nd Department of Physiology, Kagawa Medical University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | | | | | | | | | | |
Collapse
|
543
|
Canizales-Quinteros S, Aguilar-Salinas CA, Reyes-Rodríguez E, Riba L, Rodríguez-Torres M, Ramírez-Jiménez S, Huertas-Vázquez A, Fragoso-Ontiveros V, Zentella-Dehesa A, Ventura-Gallegos JL, Vega-Hernández G, López-Estrada A, Aurón-Gómez M, Gómez-Pérez F, Rull J, Cox NJ, Bell GI, Tusié-Luna MT. Locus on chromosome 6p linked to elevated HDL cholesterol serum levels and to protection against premature atherosclerosis in a kindred with familial hypercholesterolemia. Circ Res 2003; 92:569-76. [PMID: 12609970 DOI: 10.1161/01.res.0000064174.69165.66] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Heterozygous familial hypercholesterolemia (FH) is a highly atherogenic genetic disorder leading to premature coronary heart disease (CHD), usually before 60 years of age. We studied an extended multigenerational kindred with FH linked to chromosome 1p32 in which atherosclerotic complications were either delayed or prevented in individuals with elevated HDL cholesterol (HDL-C) levels or hyperalphalipoproteinemia (HA). Premature CHD was observed in FH individuals without HA. The study of this family established that the HA trait in the family also followed an autosomal dominant mode of inheritance with a pattern of segregation independent from FH. We identified a locus on chromosome 6 linked to elevated HDL-C levels (HA) in this family. Haplotype analysis refined the localization to a 7.32-cM interval (73 to 80 cM from pter) flanked by markers D6S1280 and D6S1275. Parametric 2-point and multipoint analyses yielded maximum LOD scores of 3.05 and 3.17, respectively. This finding was confirmed with a nonparametric multipoint score of 3.78 (P=0.0009). We propose that this locus, linked to elevated HDL-C levels, confers protection against premature CHD within an FH context.
Collapse
Affiliation(s)
- Samuel Canizales-Quinteros
- Unidad de Biología Molecular y Medicina Genómica, Instituto de Investigaciones Biomédicas de la Universidad Nacional Autónoma de México, México City, Mexico
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
544
|
Mineo C, Yuhanna IS, Quon MJ, Shaul PW. High density lipoprotein-induced endothelial nitric-oxide synthase activation is mediated by Akt and MAP kinases. J Biol Chem 2003; 278:9142-9. [PMID: 12511559 DOI: 10.1074/jbc.m211394200] [Citation(s) in RCA: 296] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
High density lipoprotein (HDL) activates endothelial nitric-oxide synthase (eNOS), leading to increased production of the antiatherogenic molecule NO. A variety of stimuli regulate eNOS activity through signaling pathways involving Akt kinase and/or mitogen-activated protein (MAP) kinase. In the present study, we investigated the role of kinase cascades in HDL-induced eNOS stimulation in cultured endothelial cells and COS M6 cells transfected with eNOS and the HDL receptor, scavenger receptor B-I. HDL (10-50 microg/ml, 20 min) caused eNOS phosphorylation at Ser-1179, and dominant negative Akt inhibited both HDL-mediated phosphorylation and activation of the enzyme. Phosphoinositide 3-kinase (PI3 kinase) inhibition or dominant negative PI3 kinase also blocked the phosphorylation and activation of eNOS by HDL. Studies with genistein and PP2 showed that the nonreceptor tyrosine kinase, Src, is an upstream stimulator of the PI3 kinase-Akt pathway in this paradigm. In addition, HDL activated MAP kinase through PI3 kinase, and mitogen-activated protein kinase/extracellular signal-regulated kinase kinase inhibition fully attenuated eNOS stimulation by HDL without affecting Akt or eNOS Ser-1179 phosphorylation. Conversely, dominant negative Akt did not alter HDL-induced MAP kinase activation. These results indicate that HDL stimulates eNOS through common upstream, Src-mediated signaling, which leads to parallel activation of Akt and MAP kinases and their resultant independent modulation of the enzyme.
Collapse
Affiliation(s)
- Chieko Mineo
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | | | | | | |
Collapse
|
545
|
Vaudo G, Marchesi S, Lupattelli G, Pirro M, Pasqualini L, Roscini AR, Siepi D, Schillaci G, Mannarino E. Early vascular damage in primary hypoalphalipoproteinemia. Metabolism 2003; 52:328-32. [PMID: 12647271 DOI: 10.1053/meta.2003.50070] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The relationship between hypoalphalipoproteinemia (hypoalpha), a metabolic disorder characterized by reduced high-density lipoprotein (HDL) cholesterol levels, and atherosclerotic disease is not completely understood. We investigated arterial functional and structural changes in 19 subjects with hypoalpha (HDL cholesterol < or = 0.7 mmol/L for men and < or = 0.8 mmol/L for women; 13 men; 47 +/- 7 years) and in 21 healthy control subjects (11 men; 46 +/- 13 years). Brachial-artery flow-mediated vasodilation (FMV) and intima-media thickness (IMT) of the carotid and femoral arteries were determined in all subjects. FMV was significantly lower in hypoalpha than in controls (5.6% +/- 4.3% v 8.2% +/- 2.7%; P <.05). IMT was greater in hypoalpha than in controls at both the internal carotid (0.83 +/- 0.1 mm v 0.69 +/- 0.1 mm) and superficial femoral level (0.83 +/- 0.2 mm v 0.68 +/- 0.1 mm; both P <.05). FMV had a positive correlation with HDL cholesterol (r =.42, P =.06) and a negative one with triglycerides (r = -0.38, P =.01). An inverse relationship was found between HDL cholesterol and internal carotid and superficial femoral IMT (r = -0.64 and r = -0.60, respectively; P <.01 for both) and a positive one between triglycerides and internal carotid and superficial femoral IMT (r =.53 and r =.47, P <.05). In a multivariate regression analysis, brachial FMV was predicted by HDL cholesterol and brachial diameter (beta =.42 and -0.43, respectively; both P <.05). HDL cholesterol was the only significant predictor of internal carotid and superficial femoral IMT (beta = -0.45 and -0.49, respectively; both P <.05). In conclusion, subjects with primary hypoalpha, without overt cardiovascular disease, are characterized by an impaired endothelial function and by an increase in large-artery IMT.
Collapse
Affiliation(s)
- Gaetano Vaudo
- Department of Internal Medicine, Angiology, and Atherosclerosis, University of Perugia, Perugia, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
546
|
Briand O, Lestavel S, Pilon A, Torpier G, Fruchart JC, Clavey V. SR-BI does not require raft/caveola localisation for cholesteryl ester selective uptake in the human adrenal cell line NCI-H295R. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1631:42-50. [PMID: 12573448 DOI: 10.1016/s1388-1981(02)00354-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Class B type I scavenger receptor (SR-BI) mediates the selective uptake of high-density lipoprotein (HDL)-derived cholesteryl esters (HDL-CE) in steroidogenic cells and hepatocytes. SR-BI is enriched in the caveolae of some cell types, genetically modified or not, and these domains have already been shown to constitute primary acceptors for HDL-CE. Nevertheless, the fate of caveola-free cell types has not yet been discussed.NCI-H295R, a human adrenal cell line, highly active in HDL-CE uptake via SR-BI, does not display any morphologically defined caveolae and expresses caveolin at a very low level. Using two different fractionation protocols, we have shown, in this cell type, that SR-BI is homogeneously distributed along the plasma membrane and consists principally of a non-raft membrane-associated pool. Raft destabilisation and caveolin-1 displacement from plasma membrane did not modify the SR-BI-mediated HDL-CE selective uptake. Moreover, the induction of SR-BI expression that is associated with increased CE selective uptake was not associated with any modification in caveolin-1 expression or any raft-targeting mechanism of SR-BI in NCI-H295R. In conclusion, we provide evidence that SR-BI does not require raft/caveola localisation to be implicated in CE selective uptake either in basal or in induced conditions.
Collapse
Affiliation(s)
- Olivier Briand
- Inserm UR545, Institut Pasteur de Lille and Faculté des Sciences Pharmaceutiques et Biologiques, Université de Lille 2, 1 rue du Professeur Calmette BP245, 59019, Lille, France
| | | | | | | | | | | |
Collapse
|
547
|
Abstract
Early hypercholesterolaemia-induced vascular disease is characterized by an attenuated capacity for endothelial production of the antiatherogenic molecule nitric oxide (NO), which is generated by endothelial NO synthase (eNOS). In recent studies we have determined the impact of lipoproteins on eNOS subcellular localization and action, thereby providing a causal link between cholesterol status and initial abnormalities in endothelial function. We have demonstrated that eNOS is normally targeted to cholesterol-enriched caveolae where it resides in a signalling module. Oxidized low density lipoprotein (LDL; oxLDL) causes displacement of eNOS from caveolae by binding to endothelial cell CD36 receptors and by depleting caveolae cholesterol content, resulting in the disruption of eNOS activation. The adverse effects of oxLDL are fully prevented by high density lipoprotein (HDL) via binding to scavenger receptor BI (SR-BI), which is colocalized with eNOS in endothelial caveolae. This occurs through the maintenance of caveolae cholesterol content by cholesterol ester uptake from HDL. As importantly, HDL binding to SR-BI causes robust stimulation of eNOS activity in endothelial cells, and this process is further demonstrable in isolated endothelial cell caveolae. HDL also enhances endothelium- and NO-dependent relaxation in aortae from wild-type mice, but not in aortae from homozygous null SR-BI knockout mice. Thus, lipoproteins have potent effects on eNOS function in caveolae via actions on both membrane cholesterol homeostasis and the level of activation of the enzyme. These processes may be critically involved in the earliest phases of atherogenesis, which recent studies suggest may occur during fetal life.
Collapse
Affiliation(s)
- Philip W Shaul
- Department of Pediatrics, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, TX 75390-9063, USA.
| |
Collapse
|
548
|
Abstract
The low-density-lipoprotein (LDL) receptor family is an evolutionarily ancient gene family of structurally closely related cell-surface receptors. Members of the family are involved in the cellular uptake of extracellular ligands and regulate diverse biological processes including lipid and vitamin metabolism and cell-surface protease activity. Some members of the family also participate in cellular signaling and regulate the development and functional maintenance of the nervous system. Here we review the roles of this family of multifunctional receptors in the nervous system and focus on recent advances toward the understanding of the mechanisms by which lipoprotein receptors and their ligands transmit and modulate signals in the brain.
Collapse
Affiliation(s)
- Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA.
| | | |
Collapse
|
549
|
Husemann J, Loike JD, Anankov R, Febbraio M, Silverstein SC. Scavenger receptors in neurobiology and neuropathology: their role on microglia and other cells of the nervous system. Glia 2002; 40:195-205. [PMID: 12379907 DOI: 10.1002/glia.10148] [Citation(s) in RCA: 272] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Scavenger receptor class A (SR-A, CD204), scavenger receptor-BI (SR-BI), and CD36 are cell surface proteins that mediate cell adhesion to, and endocytosis of, various native and pathologically modified substances, and participate in intracellular signaling, lipid metabolism, and host defense against bacterial pathogens. Microglia, Mato cells, astrocytes, cerebral microvascular endothelial cells, cerebral arterial smooth muscle cells, and retinal pigment epithelial cells express one or more of these SR. Expression of SR-A and SR-BI by microglia is developmentally regulated. Neonatal microglia express SR-A and SR-BI, while microglia in normal mouse and human adult brain express neither. Astrocytes in adult brain express SR-BI. In Alzheimer's disease, microglial expression of SR-A is increased. Such findings, and evidence that SR-A and SR-BI mediate adhesion and endocytosis of fibrillar beta-amyloid by microglia and astrocytes, respectively, and that SR-A, SR-BI, and CD36 participate in secretion of reactive oxygen species by microglia, suggest roles for these receptors in homeostasis and neuropathology.
Collapse
Affiliation(s)
- Jens Husemann
- Department of Physiology and Cellular Biophysics, Columbia University, New York, New York
| | - John D Loike
- Department of Physiology and Cellular Biophysics, Columbia University, New York, New York
| | - Roman Anankov
- Department of Physiology and Cellular Biophysics, Columbia University, New York, New York
| | - Maria Febbraio
- Department of Medicine, Division of Hematology and Medical Oncology, Center of Vascular Biology, Weill Medical College of Cornell University, New York, New York
| | - Samuel C Silverstein
- Department of Physiology and Cellular Biophysics, Columbia University, New York, New York
| |
Collapse
|
550
|
Ishii J, Adachi H, Aoki J, Koizumi H, Tomita S, Suzuki T, Tsujimoto M, Inoue K, Arai H. SREC-II, a new member of the scavenger receptor type F family, trans-interacts with SREC-I through its extracellular domain. J Biol Chem 2002; 277:39696-702. [PMID: 12154095 DOI: 10.1074/jbc.m206140200] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The scavenger receptor expressed by endothelial cells (SREC) with an extremely large cytoplasmic domain, was originally identified in a human endothelial cell line. In this study, we have cloned a second isoform named SREC-II and shown that there is a heterophilic interaction between SREC-I and -II at their extracellular domains. The cDNA for murine SREC-II encodes an 834-amino acid protein with 35% homology to SREC-I. Similar to SREC-I, SREC-II contains multiple epidermal growth factor-like repeats in its extracellular domain. However, in contrast to SREC-I, SREC-II had little activity to internalize modified low density lipoproteins (LDL). A Northern blot analysis revealed a tissue expression pattern of SREC-II similar to that of SREC-I with predominant expression in human heart, lung, ovary, and placenta. Mouse fibroblast L cells with no tendency to associate showed noticeable aggregation when SREC-I was overexpressed in these cells, whereas overexpression of SREC-II caused only slight aggregation. Remarkably, intense aggregation was observed when SREC-I-expressing cells were mixed with those expressing SREC-II. Deletion of almost all of the cytoplasmic receptor domain had no effect on the receptor expression and cell aggregation, indicating that solely the extracellular domain is involved in cell aggregation. The association of SREC-I and -II was effectively suppressed by the presence of scavenger receptor ligands such as acetylated LDL and oxidized LDL. These findings suggest that SREC-I and -II show weak cell-cell interaction by their extracellular domains (termed homophilic trans-interaction) but display strong heterophilic trans-interaction through the extracellular epidermal growth factor-like repeat domains.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Blotting, Northern
- CHO Cells
- Cell Adhesion Molecules
- Cell Division
- Cloning, Molecular
- Cricetinae
- DNA, Complementary/metabolism
- Databases as Topic
- Expressed Sequence Tags
- Gene Deletion
- Humans
- Leukocytes/metabolism
- Lipoproteins/metabolism
- Mice
- Molecular Sequence Data
- Oxygen/metabolism
- Phosphorylation
- Protein Binding
- Protein Structure, Tertiary
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/metabolism
- Receptors, LDL/chemistry
- Receptors, LDL/metabolism
- Receptors, Scavenger
- Scavenger Receptors, Class F
- Sequence Analysis, DNA
- Sequence Homology, Amino Acid
- Tissue Distribution
- Transfection
Collapse
Affiliation(s)
- Junko Ishii
- Laboratory of Cellular Biochemistry, RIKEN (the Institute of Physical and Chemical Research), 2-1 Wako-shi, Saitama 351-0198, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|