Abstract

HomeCirculationVol. 116, No. 11Increased Endoplasmic Reticulum Stress in Atherosclerotic Plaques Associated With Acute Coronary Syndrome Free AccessEditorialPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessEditorialPDF/EPUBIncreased Endoplasmic Reticulum Stress in Atherosclerotic Plaques Associated With Acute Coronary SyndromeA Balancing Act Between Plaque Stability and Rupture Jeffrey G. Dickhout, Stephen M. Colgan, Šárka Lhoták and Richard C. Austin Jeffrey G. DickhoutJeffrey G. Dickhout From the Henderson Research Centre and McMaster University, Hamilton, Ontario, Canada. Search for more papers by this author , Stephen M. ColganStephen M. Colgan From the Henderson Research Centre and McMaster University, Hamilton, Ontario, Canada. Search for more papers by this author , Šárka LhotákŠárka Lhoták From the Henderson Research Centre and McMaster University, Hamilton, Ontario, Canada. Search for more papers by this author and Richard C. AustinRichard C. Austin From the Henderson Research Centre and McMaster University, Hamilton, Ontario, Canada. Search for more papers by this author Originally published11 Sep 2007https://doi.org/10.1161/CIRCULATIONAHA.107.728378Circulation. 2007;116:1214–1216Acute coronary syndrome (ACS) represents a series of indications of sudden cardiac ischemia that involve a variety of causes, including acute myocardial infarction and unstable angina.1 ACS usually is associated with thrombus formation in the coronary artery or coronary artery vasospasm, resulting in myocardial ischemia.2 Thrombus formation may involve platelet adhesion and degranulation on damaged or dysfunctional endothelium overlying the intact or ruptured atherosclerotic plaque, as well as microthrombi.3 Atherosclerotic plaque rupture represents a series of deleterious events linked to the breakdown of the fibrous cap. This process may occur as a result of the increased secretion of matrix metalloproteinases by lesion resident macrophages or enhanced apoptotic cell death within the fibrous cap. However, viable smooth muscle cells (SMCs) within the fibrous cap synthesize interstitial collagen fibers that enhance the structural integrity of the plaque and prevent its rupture.4Article p 1226SMCs within the vascular wall exist in predominantly 2 phenotypes: a contractile phenotype and a synthetic phenotype. Transformation from the quiescent contractile phenotype to the active synthetic phenotype can be stimulated by the products of coagulation, including platelet-derived growth factors.5 Indeed, it has been shown that aortic allografts undergoing atherosclerotic changes first experience endothelial disruption, then SMC activation and proliferation accompanied by increased synthesis of extracellular matrix proteins.6 The synthetic SMC phenotype is actively involved in de novo protein synthesis and as such may experience endoplasmic reticulum (ER) stress if the protein folding machinery of the ER is overwhelmed by the demand of newly synthesized proteins.7 As a consequence of this ER stress, the cell activates an evolutionarily conserved pathway called the unfolded protein response (UPR). Depending on the duration or severity of ER stress, activation of the UPR can increase cell survival or lead to apoptotic cell death.The primary function of the ER involves the synthesis and processing of correctly folded secretory or plasma membrane proteins.8 De novo protein synthesis in the ER is under a quality control mechanism that prevents the secretion or incorporation in the membrane of misfolded proteins.9 The correct tertiary structure of a protein is key to its function; thus, aberrantly folded proteins within the ER must be efficiently identified, retained, and disposed of to prevent pathological consequences. The recent emergence of misfolded proteins capable of propagating protein misfolding, namely the autocatalytic theory of prion propagation, represents a pathological condition transmitted by protein misfolding10 and underscores the importance of a functional quality control system in the cell.Protein folding in the ER is aided by molecular chaperones that prevent the aggregation of misfolded proteins and assist proteins in overcoming the thermodynamic barriers that occur during the intermediate misfolded conformation to a final correctly folded and functional molecule. These ER resident molecular chaperones, including the 78-kDa glucose-regulated protein (GRP78) and protein disulfide isomerase, are induced by agents and/or conditions that cause protein misfolding in the lumen of the ER. Recent studies have identified several ER stress–inducing agents that are relevant to ACS, including homocysteine,11 peroxynitrite,12 and oxysterols.13 If an increase in molecular chaperones is unable to assist in the folding and/or clearance of misfolded proteins after exposure to these ER stress–inducing agents, activation of the UPR may trigger cellular apoptosis.14 It is well documented that the UPR signaling cascade contains late-phase proteins that are proapoptotic, including CHOP14 and TDAG51,15 whereas GRP78 has been shown to protect cells from apoptosis.16In this issue of Circulation, Myoishi and colleagues17 provide convincing evidence for the importance of ER stress in ACS. Coronary artery segments were obtained at autopsy (152 specimens from 71 patients) and tissue obtained by directional coronary atherectomy (40 patients). The autopsy specimens were classified into 5 groups: diffuse intimal thickening (normal), fibrous plaques (fibrous), thick-cap atheroma (thick), thin-cap atheroma (thin), and ruptured plaques (ruptured). The directional coronary atherectomy specimens were classified according to clinical manifestation as stable angina pectoris and unstable angina pectoris. Specifically, the authors demonstrated in the cap region the association between ER stress markers, including GRP78 and CHOP, and thin-walled or ruptured atherosclerotic plaques and unstable angina in coronary artery disease. Earlier studies performed in animal models of atherosclerosis showed that the ER stress markers GRP78 and CHOP are indeed upregulated in atherosclerotic lesions.18,19 However, Myoishi and colleagues have demonstrated for the first time in human coronary artery disease that thin-walled plaques prone to rupture and ruptured plaques show increased expression of ER stress markers, compared with stable plaques. Immunohistochemical studies showed increased staining with antibodies directed against the C-terminal KDEL retention sequence (known to recognize GRP78 and GRP94) or the proapoptotic gene CHOP were upregulated. KDEL upregulation was likely due to an increase in the ER resident molecular chaperone GRP78 as detected by increased mRNA expression with in situ hybridization in thin-walled plaques. In addition, in the atherectomy specimens, the number of KDEL- and CHOP-positive cells was significantly higher in unstable than in stable angina pectoris. From these findings, the authors propose a positive correlation between ER stress marker expression and plaque vulnerability.Thin-cap fibroatheroma is associated with a high risk of ACS and is postulated to be a precursor to plaque rupture. Thin-cap fibroatheromas and ruptured lesions are characterized by a thin (<65 μm) cap and, unlike stable thick-cap and fibrous lesions, contain a large number of macrophages in the cap region.20,21 This was indeed illustrated (but not commented on by the authors) in this study; the representative pictures of the normal, fibrous, thick, and stable angina pectoris groups showed almost no macrophages by CD68 immunostaining. However, in the thin, ruptured, and unstable angina pectoris groups, a large number of macrophages were identified by CD68 immunostaining. This indicates an active disease process in which macrophages/foam cells produce cytokines and matrix metalloproteinases and stimulate SMCs. Both macrophages and SMCs in these active lesions expressed KDEL and CHOP. These markers, however, were not observed in the normal, thick, and fibrous plaques or in directional coronary atherectomy specimens representing stable angina. This suggests that stable lesions were not undergoing ER stress.Although it is clear that ER stress is involved in the disease process leading to ACS, it is less clear how this ER stress is caused. To further explore a potential molecular mechanism for ER stress activation within the unstable lesions, coronary artery sections were immunostained with an antibody against the oxysterol, 7-ketocholesterol (7-KC). Increased staining for 7-KC was observed in the thin-cap atheromas, whereas no immunoreactivity was observed in thick-cap atheromas.The involvement of 7-KC in coronary artery disease was further examined through in vitro experimentation. Coronary artery SMCs or THP-1 cells, a human monocytic leukemia cell line, exposed to 7-KC showed increased steady-state mRNA levels of GRP78 and CHOP. The induction of ER stress marker expression after incubation with 7-KC was prevented with the addition of the antioxidant glutathione or its precursor N-acetylcysteine. The exposure of cells to 7-KC induced the intracellular production of reactive oxygen species (ROS), as shown by DCFA fluorescence, whereas the addition of glutathione decreased the DCFA signal. The effect of 7-KC on apoptotic cell death was examined in coronary artery SMCs and THP-1 cells with a terminal deoxynucleotidyl transferase–mediated dUTP nick-end labeling assay. Indeed, the addition of 7-KC induced CHOP expression and increased apoptotic cell death.It has been previously reported that 7-KC induces ROS, ER stress, and apoptosis in cultured human SMCs.13 This earlier study found that 7-KC induced apoptosis through the upregulation of the ROS-generating NAD(P)H oxidase subunit Nox-4. It was found that 7-KC induction of GRP78 and CHOP was dependent on Nox-4 expression. 7-KC–induced Nox-4 expression was found to be dependent on the ER stress sensor IRE-1. Interestingly, Nox-4 upregulation preceded ER stress because siRNA knockdown of Nox-4 inhibited CHOP and GRP78 expression and subsequent apoptosis. However, it remains unclear how Nox-4–driven ROS production results in protein misfolding in the ER. Previous studies that examined cholesterol loading in the ER membrane were able to determine an effect on ER Ca2+ depletion, leading to ER stress.19 Although it has been demonstrated that 7-KC specifically induces Ca2+ oscillation in SMCs, siRNA knockdown of Nox-4 had no effect on these Ca2+ perturbations. 7-KC–induced ROS production was shown to be detectable with DCFA, whereas polyethylene glycol/superoxide dismutase reduced this effect,13 implying that superoxide and hydrogen peroxide were generated. Determining the role of these ROS in ER stress induction, if any, might clarify the mechanism behind 7-KC–induced ER stress.Unresolved ER stress leads to apoptosis of SMCs and likely contributes to plaque rupture. To further the hypothesis of Myoishi and colleagues that ER stress causes plaque rupture, the ability to block ER stress–induced apoptosis in animal models of atherosclerosis may provide answers. Although established animal models of plaque rupture do not exist, studies in mouse models of atherosclerosis could examine surrogate measures of rupture, including plaque and necrotic core size or rate of SMC apoptosis. Knockout of ER stress proapoptotic genes CHOP or TDAG51 would provide at least partial answers to the role of ER stress–induced apoptosis as a cause of ACS. Ultimately, therapeutic strategies that resolve ER stress such as small chemical chaperones that mimic the effect of endogenous ER resident molecular chaperones22 and thus prevent ER stress–induced apoptosis may be of tremendous value in stabilizing thin-cap atheroma and preventing ACS.The initiation and progression of atherosclerotic disease appear to involve ER stress/UPR activation, as observed in apolipoprotein E–deficient mice fed a control chow diet (Figure).18 The resolution of this cellular stress response over time, be it increased protein synthetic capacity of lesion-resident SMCs or the induction of apoptotic cell death resulting from conditions or agents that cause ER stress, may ultimately determine the fate of the fibrous cap and the risk of plaque rupture in ACS. Download figureDownload PowerPointSchematic representation of the role of ER stress/UPR activation on atherosclerotic lesion development and its influence on ACS. Aortic root sections from early, intermediate, or advanced atherosclerotic lesions from apolipoprotein E–deficient mice fed control chow diet were immunostained for the ER stress marker, phospho-PERK. A, Different stages of atherosclerotic lesion development and whole vascular wall involvement illustrate the extent of ER stress/UPR activation in the lesion. Relevant pathophysiological factors that contribute to ACS may be responsible for the initiation of ER stress. B, ER stress/UPR activation seems to progress after the appearance of lesion-resident macrophage foam cells. C, This progression leads to the involvement of the whole vascular wall, including macrophages and smooth muscle cells. How this ER stress response is resolved can influence the balance between increasing protein folding capacity within the smooth muscle layer and induction of SMC apoptosis. This may ultimately contribute to the formation of a stable fibrotic plaque or lead to plaque rupture and ACS.The opinions expressed in this article are not necessarily those of the editors or of the American Heart Association.Sources of FundingResearch in the laboratory of Dr Austin is supported by the Heart and Stroke Foundation of Ontario (T-6146, NA-6024) and the Canadian Institutes of Health Research (MOP-67116, MOP-74477). Dr Austin is a Career Investigator of the Heart and Stroke Foundation of Ontario. Stephen M. Colgan is a recipient of a Graduate Studentship from the Canadian Liver Foundation.DisclosuresNone.FootnotesCorrespondence to Richard C. Austin, PhD, Henderson Research Centre, 711 Concession St, Hamilton, Ontario, Canada L8V 1C3. E-mail [email protected] References 1 Bertoni AG, Bonds DE, Thom T, Chen GJ, Goff DC Jr. Acute coronary syndrome national statistics: challenges in definitions. Am Heart J. 2005; 149: 1055–1061.CrossrefMedlineGoogle Scholar2 Achar SA, Kundu S, Norcross WA. Diagnosis of acute coronary syndrome. Am Fam Physician. 2005; 72: 119–126.MedlineGoogle Scholar3 Grech ED, Ramsdale DR. Acute coronary syndrome: unstable angina and non-ST segment elevation myocardial infarction. BMJ. 2003; 326: 1259–1261.CrossrefMedlineGoogle Scholar4 Kockx MM, Knaapen MW. Pathological changes in the coronary arteries in the acute coronary syndromes. Heart. 2006; 92: 1557–1558.CrossrefMedlineGoogle Scholar5 Rensen SS, Doevendans PA, van Eys GJ. Regulation and characteristics of vascular smooth muscle cell phenotypic diversity. Neth Heart J. 2007; 15: 100–108.CrossrefMedlineGoogle Scholar6 Religa P, Bojakowski K, Gaciong Z, Thyberg J, Hedin U. Arteriosclerosis in rat aortic allografts: dynamics of cell growth, apoptosis and expression of extracellular matrix proteins. Mol Cell Biochem. 2003; 249: 75–83.CrossrefMedlineGoogle Scholar7 Ron D. Translational control in the endoplasmic reticulum stress response. J Clin Invest. 2002; 110: 1383–1388.CrossrefMedlineGoogle Scholar8 Lee AS. The glucose-regulated proteins: stress induction and clinical applications. Trends Biochem Sci. 2001; 26: 504–510.CrossrefMedlineGoogle Scholar9 Klausner RD, Sitia R. Protein degradation in the endoplasmic reticulum. Cell. 1990; 62: 611–614.CrossrefMedlineGoogle Scholar10 Malolepsza E, Boniecki M, Kolinski A, Piela L. Theoretical model of prion propagation: a misfolded protein induces misfolding. Proc Natl Acad Sci U S A. 2005; 102: 7835–7840.CrossrefMedlineGoogle Scholar11 Outinen PA, Sood SK, Liaw PC, Sarge KD, Maeda N, Hirsh J, Ribau J, Podor TJ, Weitz JI, Austin RC. Characterization of the stress-inducing effects of homocysteine. Biochem J. 1998; 332: 213–221.CrossrefMedlineGoogle Scholar12 Dickhout JG, Hossain GS, Pozza LM, Zhou J, Lhotak S, Austin RC. Peroxynitrite causes endoplasmic reticulum stress and apoptosis in human vascular endothelium: implications in atherogenesis. Arterioscler Thromb Vasc Biol. 2005; 25: 2623–2629.LinkGoogle Scholar13 Pedruzzi E, Guichard C, Ollivier V, Driss F, Fay M, Prunet C, Marie JC, Pouzet C, Samadi M, Elbim C, O’dowd Y, Bens M, Vandewalle A, Gougerot-Pocidalo MA, Lizard G, Ogier-Denis E. NAD(P)H oxidase Nox-4 mediates 7-ketocholesterol-induced endoplasmic reticulum stress and apoptosis in human aortic smooth muscle cells. Mol Cell Biol. 2004; 24: 10703–10717.CrossrefMedlineGoogle Scholar14 Kaufman RJ. Orchestrating the unfolded protein response in health and disease. J Clin Invest. 2002; 110: 1389–1398.CrossrefMedlineGoogle Scholar15 Hossain GS, van Thienen JV, Werstuck GH, Zhou J, Sood SK, Dickhout JG, de Koning AB, Tang D, Wu D, Falk E, Poddar R, Jacobsen DW, Zhang K, Kaufman RJ, Austin RC. TDAG51 is induced by homocysteine, promotes detachment-mediated programmed cell death, and contributes to the development of atherosclerosis in hyperhomocysteinemia. J Biol Chem. 2003; 278: 30317–30327.CrossrefMedlineGoogle Scholar16 Reddy RK, Mao C, Baumeister P, Austin RC, Kaufman RJ, Lee AS. Endoplasmic reticulum chaperone protein GRP78 protects cells from apoptosis induced by topoisomerase inhibitors: role of ATP binding site in suppression of caspase-7 activation. J Biol Chem. 2003; 278: 20915–20924.CrossrefMedlineGoogle Scholar17 Myoishi M, Hao H, Minamino T, Watanabe K, Nishihira K, Hatakeyama K, Asada Y, Okada K, Ishibashi-Ueda H, Gabbiani G, Bochaton-Piallat M, Mochizuki N, Kitakaze M. Increased endoplasmic reticulum stress in atherosclerotic plaques associated with acute coronary syndrome. Circulation. 2007; 116: 1226–1233.LinkGoogle Scholar18 Zhou J, Lhotak S, Hilditch BA, Austin RC. Activation of the unfolded protein response occurs at all stages of atherosclerotic lesion development in apolipoprotein E–deficient mice. Circulation. 2005; 111: 1814–1821.LinkGoogle Scholar19 Feng B, Yao PM, Li Y, Devlin CM, Zhang D, Harding HP, Sweeney M, Rong JX, Kuriakose G, Fisher EA, Marks AR, Ron D, Tabas I. The endoplasmic reticulum is the site of cholesterol-induced cytotoxicity in macrophages. Nat Cell Biol. 2003; 5: 781–792.CrossrefMedlineGoogle Scholar20 Virmani R, Burke AP, Farb A, Kolodgie FD. Pathology of the vulnerable plaque. J Am Coll Cardiol. 2006; 47: C13–C18.CrossrefMedlineGoogle Scholar21 Fuster V, Moreno PR, Fayad ZA, Corti R, Badimon JJ. Atherothrombosis and high-risk plaque, part I: evolving concepts. J Am Coll Cardiol. 2005; 46: 937–954.CrossrefMedlineGoogle Scholar22 Özcan U, Yilmaz E, Özcan L, Furuhashi M, Vaillancourt E, Smith RO, Görgün CZ, Hotamisligil GS. Chemical chaperones reduce ER stress and restore glucose homeostasis in a mouse model of type 2 diabetes. Science. 2006; 313: 1137–1140.CrossrefMedlineGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetailsCited By Tian H, Wang T, Zhang Y, Pan T, Yao S, Yu H, Ma K and Wang S (2022) Astragaloside IV protects against C/EBP homologous protein-mediated apoptosis in oxidized low-density lipoprotein-treated macrophages by promoting autophagy, European Journal of Pharmacology, 10.1016/j.ejphar.2022.174912, 923, (174912), Online publication date: 1-May-2022. Tian H, Zhang Z, Han X, Pan T, Tao G, Jiao P, Zhai L, Yang L, Wang X, Yao Y, Qin S and Yao S (2021) D4F alleviates the C/EBP homologous protein-mediated apoptosis in glycated high-density lipoprotein-treated macrophages by facilitating autophagy, Experimental Biology and Medicine, 10.1177/15353702211045323, 246:24, (2595-2609), Online publication date: 1-Dec-2021. Che X, Xiao Q, Song W, Zhang H, Sun B, Geng N, Tao Z, Shao Q and Pu J (2021) Protective Functions of Liver X Receptor α in Established Vulnerable Plaques: Involvement of Regulating Endoplasmic Reticulum–Mediated Macrophage Apoptosis and Efferocytosis, Journal of the American Heart Association, 10:10, Online publication date: 18-May-2021. Ren J, Chen Y, Zhang L, Zhang Y, Liu S, Yu Y, Jia M, Tang C, Qi Y and Lu W (2021) Intermedin1-53 attenuates atherosclerotic plaque vulnerability by inhibiting CHOP-mediated apoptosis and inflammasome in macrophages, Cell Death & Disease, 10.1038/s41419-021-03712-w, 12:5 Yang D, Wang Q, Wei G, Wu J, Zhu Y, Zhu Q, Ni T, Liu X and Zhu Y (2020) Smyd3-PARP16 axis accelerates unfolded protein response and vascular aging, Aging, 10.18632/aging.103895, 12:21, (21423-21445), Online publication date: 3-Nov-2020. Vaisar T, Hu J, Airhart N, Fox K, Heinecke J, Nicosia R, Kohler T, Potter Z, Simon G, Dix M, Cravatt B, Gharib S and Dichek D (2020) Parallel Murine and Human Plaque Proteomics Reveals Pathways of Plaque Rupture, Circulation Research, 127:8, (997-1022), Online publication date: 25-Sep-2020. Yang S, Wu M, Li X, Zhao R, Zhao Y, Liu L and Wang S (2020) Role of Endoplasmic Reticulum Stress in Atherosclerosis and Its Potential as a Therapeutic Target, Oxidative Medicine and Cellular Longevity, 10.1155/2020/9270107, 2020, (1-15), Online publication date: 9-Sep-2020. Lynn E, Lhoták Š, Lebeau P, Byun J, Chen J, Platko K, Shi C, O'Brien R and Austin R (2019) 4‐Phenylbutyrate protects against atherosclerotic lesion growth by increasing the expression of HSP25 in macrophages and in the circulation of Apoe −/− mice , The FASEB Journal, 10.1096/fj.201802293RR, 33:7, (8406-8422), Online publication date: 1-Jul-2019. Tian H, Li Y, Kang P, Wang Z, Yue F, Jiao P, Yang N, Qin S and Yao S (2019) Endoplasmic reticulum stress‐dependent autophagy inhibits glycated high‐density lipoprotein‐induced macrophage apoptosis by inhibiting CHOP pathway , Journal of Cellular and Molecular Medicine, 10.1111/jcmm.14203, 23:4, (2954-2969), Online publication date: 1-Apr-2019. Hoseini Z, Sepahvand F, Rashidi B, Sahebkar A, Masoudifar A and Mirzaei H (2017) NLRP3 inflammasome: Its regulation and involvement in atherosclerosis, Journal of Cellular Physiology, 10.1002/jcp.25930, 233:3, (2116-2132), Online publication date: 1-Mar-2018. Grechowa I, Horke S, Wallrath A, Vahl C and Dorweiler B (2017) Human neutrophil elastase induces endothelial cell apoptosis by activating the PERK‐CHOP branch of the unfolded protein response, The FASEB Journal, 10.1096/fj.201700012R, 31:9, (3868-3881), Online publication date: 1-Sep-2017. Woo M, Kim M, Noh J, Park C and Song Y (2017) Kimchi attenuates fatty streak formation in the aorta of low-density lipoprotein receptor knockout mice via inhibition of endoplasmic reticulum stress and apoptosis, Nutrition Research and Practice, 10.4162/nrp.2017.11.6.445, 11:6, (445), . Carlisle R, Werner K, Yum V, Lu C, Tat V, Memon M, No Y, Ask K and Dickhout J (2016) Endoplasmic reticulum stress inhibition reduces hypertension through the preservation of resistance blood vessel structure and function, Journal of Hypertension, 10.1097/HJH.0000000000000943, 34:8, (1556-1569), Online publication date: 1-Aug-2016. de Pedro-Cuesta J, Martínez-Martín P, Rábano A, Ruiz-Tovar M, Alcalde-Cabero E and Calero M (2016) Etiologic Framework for the Study of Neurodegenerative Disorders as Well as Vascular and Metabolic Comorbidities on the Grounds of Shared Epidemiologic and Biologic Features, Frontiers in Aging Neuroscience, 10.3389/fnagi.2016.00138, 8 Liu M, Chen Z and Chen L (2016) Endoplasmic reticulum stress: a novel mechanism and therapeutic target for cardiovascular diseases, Acta Pharmacologica Sinica, 10.1038/aps.2015.145, 37:4, (425-443), Online publication date: 1-Apr-2016. Ma M, Song L, Yan H, Liu M, Zhang L, Ma Y, Yuan J, Hu J, Ji Z, Zhang R, Li C, Wang H, Tao L, Zhang Y and Li Y (2016) Low dose tunicamycin enhances atherosclerotic plaque stability by inducing autophagy, Biochemical Pharmacology, 10.1016/j.bcp.2015.11.020, 100, (51-60), Online publication date: 1-Jan-2016. Tomita K, Teratani T, Suzuki T, Shimizu M, Sato H, Narimatsu K, Usui S, Furuhashi H, Kimura A, Nishiyama K, Maejima T, Okada Y, Kurihara C, Shimamura K, Ebinuma H, Saito H, Yokoyama H, Watanabe C, Komoto S, Nagao S, Sugiyama K, Aosasa S, Hatsuse K, Yamamoto J, Hibi T, Miura S, Hokari R and Kanai T (2014) Acyl-CoA:cholesterol acyltransferase 1 mediates liver fibrosis by regulating free cholesterol accumulation in hepatic stellate cells, Journal of Hepatology, 10.1016/j.jhep.2014.03.018, 61:1, (98-106), Online publication date: 1-Jul-2014. Liu M, Wang X, Wang C, Song D, Liu X and Shi D (2013) Panax Quinquefolium Saponin Attenuates Ventricular Remodeling After Acute Myocardial Infarction by Inhibiting Chop-Mediated Apoptosis, Shock, 10.1097/SHK.0b013e3182a3f9e5, 40:4, (339-344), Online publication date: 1-Oct-2013. Rong J, Blachford C, Feig J, Bander I, Mayne J, Kusunoki J, Miller C, Davis M, Wilson M, Dehn S, Thorp E, Tabas I, Taubman M, Rudel L and Fisher E (2012) ACAT Inhibition Reduces the Progression of Preexisting, Advanced Atherosclerotic Mouse Lesions Without Plaque or Systemic Toxicity, Arteriosclerosis, Thrombosis, and Vascular Biology, 33:1, (4-12), Online publication date: 1-Jan-2013. Carlisle R, Heffernan A, Brimble E, Liu L, Jerome D, Collins C, Mohammed-Ali Z, Margetts P, Austin R and Dickhout J (2012) TDAG51 mediates epithelial-to-mesenchymal transition in human proximal tubular epithelium, American Journal of Physiology-Renal Physiology, 10.1152/ajprenal.00481.2011, 303:3, (F467-F481), Online publication date: 1-Aug-2012. Zhou G, Isoe J, Day W, Miesfeld R and Uversky V (2011) Alpha-COPI Coatomer Protein Is Required for Rough Endoplasmic Reticulum Whorl Formation in Mosquito Midgut Epithelial Cells, PLoS ONE, 10.1371/journal.pone.0018150, 6:3, (e18150) Dickhout J, Carlisle R, Austin R and Kitakaze M (2011) Interrelationship Between Cardiac Hypertrophy, Heart Failure, and Chronic Kidney Disease, Circulation Research, 108:5, (629-642), Online publication date: 4-Mar-2011. Dickhout J, Lhoták Š, Hilditch B, Basseri S, Colgan S, Lynn E, Carlisle R, Zhou J, Sood S, Ingram A and Austin R (2010) Induction of the unfolded protein response after monocyte to macrophage differentiation augments cell survival in early atherosclerotic lesions, The FASEB Journal, 10.1096/fj.10-159319, 25:2, (576-589), Online publication date: 1-Feb-2011. Lhoták Š, Zhou J and Austin R (2011) Immunohistochemical Detection of the Unfolded Protein Response in Atherosclerotic Plaques The Unfolded Protein Response and Cellular Stress, Part A, 10.1016/B978-0-12-385116-1.00002-9, (23-46), . Engin F and Hotamisligil G (2010) Restoring endoplasmic reticulum function by chemical chaperones: an emerging therapeutic approach for metabolic diseases, Diabetes, Obesity and Metabolism, 10.1111/j.1463-1326.2010.01282.x, 12, (108-115), Online publication date: 1-Oct-2010. Santos C, Tanaka L, Wosniak J and Laurindo F (2009) Mechanisms and Implications of Reactive Oxygen Species Generation During the Unfolded Protein Response: Roles of Endoplasmic Reticulum Oxidoreductases, Mitochondrial Electron Transport, and NADPH Oxidase, Antioxidants & Redox Signaling, 10.1089/ars.2009.2625, 11:10, (2409-2427), Online publication date: 1-Oct-2009. September 11, 2007Vol 116, Issue 11 Advertisement Article InformationMetrics https://doi.org/10.1161/CIRCULATIONAHA.107.728378PMID: 17846339 Originally publishedSeptember 11, 2007 Keywordsapoptosisplaqueatherosclerosiscoronary diseaseEditorialsPDF download Advertisement SubjectsAcute Coronary SyndromesCell Signaling/Signal TransductionEpidemiologyPathophysiologySmooth Muscle Proliferation and Differentiation

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call