低强度运动对SHR大鼠血管内皮的作用及IL-33在动脉粥样硬化中的表达
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
高血压病的发病率有逐年增高的趋势。但是高血压病患者对该病的知晓率、治疗率、达标率均较低,这与治疗高血压病的药物存在多种副作用、常需联合用药、价格相对昂贵导致患者长期服用依从性差有关。重视非药物方法对治疗高血压有重要意义。
     低强度运动锻练能够在一定程度上降低血压。运动锻练可以降低心输出量,减低血管阻力,明显降低血管壁/血管腔的比值,运动可以使肌肉内静脉增生,静脉容量增大,运动导致参与运动的器官与组织毛细血管床增大,降低循环血容量与血管床容量比值,从而导致血压下降。
     胰岛素抵抗与高血压呈正相关,低强度运动锻练对胰岛素抵抗的胰岛素受体前水平、受体水平和受体后水平三个环节均有明显的改善作用。低强度运动还可以升高骨骼肌中PPARγ蛋白水平,PPARγ在心血管系统的表达和激活具有血管紧张素II阻断效应并降低血压。低强度运动锻练对胰岛素抵抗各个环节的改善,有效地增加了胰岛素敏感性,降低了过高的胰岛素水平,从而增加内皮细胞NO的合成与释放。这种作用主要是作用在NOS的转录和表达水平上。NO被认为是一种抗炎因子,其抗炎症的机制主要是抑制核因子-κB(NF-κB)活性。NF-κB与许多细胞因子的基因的启动子或增强子部位的κB位点发生特异性结合,启动和调节这些基因的转录。参与血管内皮功能紊乱的炎症因子的基因多为NF-κB的靶基因,通过NF-κB通路调节这些因子的转录与表达可以影响动脉粥样硬化形成。除NO外,诱生型热休克蛋白70也能够抑制激活的IKK和IκB的降解,从而抑制NF-κB的表达。运动能明显上调HSP70的表达,运动诱导HSP70增高与热休克转录因子(HSF1)激活有关。白介素(IL)33是近几年用计算机技术测序鉴定发现的一种因子,在很多组织中的大血管和小血管的内皮细胞核中都有结构性表达。IL-33对动脉粥样硬化起保护作用,诱导Th1细胞向Th2的转变,能显著对抗血管紧张素Ⅱ和去甲肾上腺素所诱导的NF-κB的产生,增加内皮细胞NO的生成,减低动脉斑块的炎症反应。本文采用临床常用的钙离子拮抗剂硝苯地平为阳性对照组,观察低强度运动对SHR大鼠降压作用,结果显示低强度运动锻练能明显降低SHR大鼠的血压,与硝苯地平单药治疗相类似,在降压的同时,低强度运动锻练还同时减轻胰岛素抵抗,从而增强血管内皮NOS的表达,明显增高血浆中的NO水平,增加血管内皮中HSP70和IL33的表达,抑制血管内皮NF-κB的表达,减轻血管内皮的病变程度,减少心血管并发症的发生。本实验还对30例人冠状动脉标本进行免疫组化观察IL-33的表达,结果显示IL-33在人冠状动脉内膜与斑块上表达,与粥样硬化斑块面积正相关。进一步阐明IL-33/ST2信号通道的细胞和核内的效应有助于开发新的心血管保护药物。
Hypertension is a chronic condition and characterized by high systemic circulation blood pressure.It is one of most common disorders and a risk factor for cardiovascular disease. The prevalence of hypertension is rising with age and considerable unmet medical need. Myocardial contractility, circulating blood volume and peripheral vascular resistance maintain the circulation blood pressure under the regulation of nerve-humoral system. Endothelium dysfunction plays a important role in the development of hypertension. Endothelium-dependent nitro oxide dysregulation is an important cause of the initiation hypertension. The NO subsequently trigger its bioactivity dependent of the following factors: (1) the activity of NOS in the endothelial cell, (2) the interaction with active oxidant, (3) the sensitivity of the vascular smooth cell.
     Insulin resistance is closely associated with hypertension. Diminished tissue sensitivity to the metabolic actions of insulin is a characteristic feature of insulin resistance, which leads to hyperinsulinism to meet the need of glucose metabolism. In normal concentration, insulin maintans vasodilatation depending on NO synthesis and realease, While hyperinsulinism stimulates ET expression and release. Insulin resistance reduces the ratio of NO/ET, which impair endothelial vasodilation and anti-embolism function .In the long run, insulin resistance results in vascular smooth cell proliferation and transposition.
     Less than 15% hypertension patients can be satisfiedly controlled by one type of anti-hypertension drug. The majority patients need more than one anti-hypertension medicines. The side-effects of anti-hypertension medicines have reduced their general application.While the new generation of medicines are effect with reduced side-effect, they are costly and can not meet the requirement for all people.Therefore, more effective and affordable stratagies are needed to control the prevalence of hypertention, in particularly non-drug treatment.
     Exercise has different effects on blood pressure. Low intensity exercises can reduce the high blood pressure and improve a series of dysfunction. Low intensity exercises not only reduce cardiac output, vascular resistance and the ratio of vein wall/lumen, but also enlarge venous volume and capillary bed, which reduce the ratio of circulation volume/vascular bed volume and reduce the high blood pressure.
     Insulin resistance may represent a key mechanism of the spontaneous hytertension in rats. Low intensity exercises improve insulin sensitivity at the levels of pre-receptor, receptor and post-receptor. Typically, insulin-dependent glucose usage is partialy dependent on increased insulin in blood flow and substrate delivery to tissues, Low intensity exercises increase skeletal muscle fiber I and capillary quantity, which increase regional blood flow. Low intensity exercises increase insulin receptors expression in skeletal cell, while increase receptor tyrosine phosphorylation, insulin receptor substrate tyrosine phosphorylation and phosphatidylinositol kinase activation, which improve insulin metabolic signal pathway. Low intensity exercises also increase GLUT4 gene transcription, translocate GLUT4 to epicyte which enhance skeletal cell uptake glucose. Low intensity exercises reduce adipocyte size, increase oxidation of carbohydrate and fatty acids. It also increase PPARγprotein in skeletal cell, which increase the gene expression related to the fatty acids transportation and oxidation. Continouse exercises upregulate the PGC-1 gene expression, suggesting that exercises play a important role in PPARγpost-transcriptional regulation .The expression and activation of PPARγmay reduce the blood pressure like ACEI.
     In insulin resistance conditions, insulin stimulated NO bioactivity is decreased (decreased endothelial NO synthase activation and increased NO destruction), and ET release of endothelial cells increased. Low-intensity exercise improve all aspects of insulin resistance, increasing insulin sensitivity effectively, reducing the high insulin levels, thereby increase endothelial NO synthesis and release, through the effect on NOS transcription and expression, probably attribute to the PPARγstimulated by low intensity exercise, the same as PPARγ agonist rosiglitazon.
     Exercise effectively stimulate NO synthesis through increasing NOS expression; Exercise also reduce plasma ox-LDL levels, which then reduce the injury of vascular endothelial and the inhibition of NO generation; It also increases the expression of HSP90,which will improve anti-apoptosis and anti-oxidation capacity of cells, HSP90 can further regulate cell activity and control the information transmission pathway, which will promote eNOS’synthesis and secretion and play a protective effect on vascular endothelium.
     eNO is an endothelium-derived factor, the basic substrate of NO synthesis is L-arginine and oxygen, NOS is the key enzyme in this reaction. NO plays a pivotal role in vascular endothelial function.It blocks monocyte adhesion to endothelial cells by inhibiting platelet aggregation and expression of adhesion molecule,in the mean time, NO also inhibit LDL oxidation and vascular smooth muscle proliferation. NO is considered as an anti-inflammatory factor mainly by inhibiting nuclear factor-ΚB (NF-ΚB) activity. is mainly its anti-inflammatory mechanism Since ox-LDL can increase the vascular endothelial NF-κB activity, NO inhibits LDL oxidation to further suppress NF-κB activity. In addition, NO may also inhibit the activity of NF-κB by inhibiting the expression of macrophage colony-stimulating factor (M-CSF), monocyte chemoattractant protein -1 (MCP-1) expression and cell adhesion molecules .
     NF-κB binds to the promoter or enhancer of cytokines genes at the specific NF-Κb binding sites, initiates or regulates the transcription of these genes. Most of inflammatory cytokines promoters involved in endothelial dysfunction contain NF-κB sites, including IL-1β, IL-6, IL8, intercellular adhesion molecule (ICAM-1), vascular cell adhesion molecule (VCAM). The shear stress of high blood pressure and increased peroxide can trigger the NF-κB activation in vascular endothelial cells, promoting the expression of ICAM-1, VCAM-1 and monocyte chemotactic factor (MCP-1), leading to the proliferation and infiltration of mononuclear cells, damaging endothelia cells, prompting the formation of atherosclerosis.
     Heat shock proteins have attrached considerable attention in recent years by their endothelial protective function. When Organisms are stimulated by harmful stimuli in the environment, they rapidly adapt the changes by produce a series of short-term metabolism.Some protein synthesis is temporarily reduced and the expression of certain proteins are selectively up-regulated. It is a self-protective mechanism for the cells to responsed to the outside stimuli.which is called the heat shock response.
     Heat shock proteins belong to the family of molecular chaperones and play an important role in the process of refolding misfolded protein and degradating protease. HSP70 is a kind of conserved protein, in addition to the above-mentioned molecular chaperone function, it also improve the cells tolerance to stress. HSP70 accumulas in the plaque and protects the vascular endothelium. Inducible heat shock protein 70 inhibits the activation of IKK and the degradation of IκB, thereby inhibits the expression of NF-κB, This may be the mechanisms underlying the endothelium-protective effect of HSP70. Exercise can significantly increase the expression of HSP70 by inducing heat shock transcription factor (HSF1) activation and there is a gender difference in exercise-induced increase in HSP70 protein.
     This project was to study the effect and mechanism of low-intensity exercise on reducing blood pressure while protecting the vascular endothelial. Using Nifedipine, the calcium channel blocker as a positive control drug, the effects of low-intensity exercise on blood pressure of SHR rats were studied, as well as the changes of insulin sensitivity, vascular endothelial function, NF-κB and HSP70.
     The results showed that:
     (1) low-intensity exercise significantly reduced blood pressure, SHR rats.
     (2) low-intensity exercise significantly reduced the level of insulin resistance in SHR rats.
     (3) It reduced high blood pressure in SHR rats while reducing the SHR rat vascular endothelial lesions.
     (4) It significantly increased the SHR rat aortic NOS expression and activity.
     (5) It significantly increased the SHR serum NO concentration.
     (6)It significantly reduced the SHR rat aortic expression level of NF-κB.
     (7) low-intensity exercise increased HSP70 expression in rat aortic SHR.
     (8) low-intensity exercise increased IL33 expression in rat aortic SHR.
     We found that low-intensity exercise training significantly reduced blood pressure in SHR rats, similar to nifedipine monotherapy.Low-intensity exercise training also reduced insulin resistance, thereby enhanced the expression of vascular endothelial NOS, significantly increased plasma NO levels, inhibited the expression of vascular endothelial NF-κB, reduced the degree of vascular endothelial lesions and reduced the incidence of cardiovascular complications. Therefore, physical exercise, an economic, convenient and effective method should be given particular attention both at the beginning of treating blood pressure also when using together with antihypertensive drugs.
     It was thought that atherosclerosis is resulted from large or medium-sized lipid deposition in arterial wall, now it has been realized that this is a process of inflammatiory process. A lot of circulation inflammatory markers are related to atherosclerosis. The inflammation in the atherosclerotic plaque leads to intravascular thrombosis and ischemic events in two ways. First, the endothelium covering the plaque shift from the anticoagulant state to the state of promoting thrombosision. This will induce the release of pro- inflammatory factors such as tumor necrosis factor (TNF), interleukin-1? (IL-1?) by the macrophages. Second, the release of MMP (including MMP2,3,9) mediated by T lymphocytes and macrophages results in the decay of the fibrous cap composed of elastin and collagen, leads to plaque instability and rupture. The expose of the internal necrotic lipid core and collagen to the blood leads to intravascular thrombosis and ischemic events.
     T cells recognize endougenous LDL antigen by T cell receptor before is one of the mechanisms leading to rapid progression of atherosclerotic plaque. It is well known that activated macrophages and ox -LDL are specific antigens which can stimulate the immune response and T cell proliferation. T cells recruit to the vessel wall via adhesion molecules and chemokines torecorganise the ox-LDL and HSP on the arterial wall, which will activate T cells in the local. The cytokines released by activated T cells stimulate macrophages, the key effect cells of atherosclerosis. Macrophages enter into the vessel wall with the facilitation of cell adhesion molecule which is up-secreted by the endothelial cells. Several interlukins and their receptors are associated with the atherosclerosis in this inflammatory process and play an important role, such as upregulating endothelial cell adhesion factor, activating macrophages and promoting smooth muscle cell proliferation. The experiments using gene knockout or transgenic mices found that some of the interleukins play a catalytic role in the artery atherosclerosis, while the other may serve as anti-atherosclerotic factors. interleukin-1 family is much important in this process.
     Interleukin33 was identified in 1995, It belongs to the IL1 family and is an anti-inflammatory factors in the process of atherosclerosis. IL-33 expresses in many tissues such as large or small vascular endothelial cells.IL-33 play a protective effect on atherosclerosis, by inducing the swithching from Th1 cells to Th2 , increase significantly serum levels of anti-atherosclerotic factors such as IL4, IL5, and IL13, increase the level of ox - LDL antibodies IgA, IgE, and IgG, which enhance anti-inflammatory effect, reduce the serum concentration of IFNγwhich can promote atherosclerosis.It also reduces the production of NF-KB induced by angiotensinⅡand phenylephrine to reduce arterial plaque inflammation. Recombinant IL-33 treatment can significantly reduce the atherosclerotic plaque. In this study the expression of IL-33 in coronary arterty specimens from 30 patients with different conditions were observed by immunohistochemistry, to further reveal the role of IL-33 in the process of human atherosclerosis. The results showed that IL-33 not only expressed in the human coronary artery intima and plaque, but also was correlated with the extent of atherosclerosis. Further study is needed to identify IL-33/ST2 signaling pathway inside the cells, which would contribute to the development of new cardiovascular protective drug.
引文
[1] Veras-Silva AS, Mattos KC, Gava NS, et al. Low-intensity exercise training decreases cardiac output and hypertension in spontaneously hypertensive rats. Am J Physiol JT - The American journal of physiology, 1997, 273(6 Pt 2):H2627-31.
    [2] Mac Donnell SM, Kubo H, Crabbe DL, et al. Improved myocardial beta-adrenergic responsiveness and signaling with exercise training in hypertension. Circulation JT - Circulation, 2005, 111(25):3420-8.
    [3] Urata H, Tanabe Y, Kiyonaga A, et al. Antihypertensive and volume-depleting effects of mild exercise on essential hypertension. Hypertension JT-Hypertension, 1987, 9(3): 245-52.
    [4] Brum PC, Da Silva GJ, Moreira ED, et al. Exercise training increases baroreceptor gain sensitivity in normal and hypertensive rats. Hypertension JT-Hypertension, 2000, 36(6): 1018-22.
    [5] Evenwel R, Struyker-Boudier H. Effect of physical training on the development of hypertension in the spontaneously hypertensive rat. Pflugers Arch JT-Pflugers Archiv: European journal of physiology, 1979, 381(1):19-24.
    [6]蔡恒,王凤芝,张薇,等.原发性高血压的运动治疗研究.中国运动医学杂志, 2003(03):247-250.
    [7] Amaral SL, Silveira NP, Zorn TM, et al. Exercise training causes skeletal muscle venular growth and alters hemodynamic responses in spontaneously hypertensive rats. J Hypertens JT-Journal of hypertension, 2001, 19(5):931-40.
    [8] Amaral SL, Zorn TM, Michelini LC. Exercise training normalizes wall-to-lumen ratio of the gracilis muscle arterioles and reduces pressure in spontaneously hypertensive rats. J Hypertens JT-Journal of hypertension, 2000, 18(11):1563-72.
    [9] Melo RM, Martinho E Jr, Michelini LC. Training-induced, pressure-lowering effect in SHR: wide effects on circulatory profile of exercised and nonexercised muscles. Hypertension JT-Hypertension, 2003, 42(4):851-7.
    [10] Saito Y, Eraslan A, Lockard V, et al. Role of venular endothelium in control of arteriolar diameter during functional hyperemia. Am J Physiol JT-The American journal of physiology, 1994, 267(3 Pt 2):H1227-31.
    [11] Sangaleti CT, Crescenzi A, Michelini LC. Endogenous angiotensin and pressure modulate brain angiotensinogen and AT1A mRNA expression. Hypertension JT- Hypertension, 2004, 43(2):317-23.
    [12] Felix JV, Michelini LC. Training-induced pressure fall in spontaneously hypertensive rats is associated with reduced angiotensinogen mRNA expression within the nucleus tractus solitarii. Hypertension JT-Hypertension, 2007, 50(4):780-5.
    [13] Klett CP, Anderson D, Sholook M, et al. Antisense oligodeoxynucleotides directed against a novel angiotensinogen mRNA-stabilizing protein reduce blood pressure in spontaneously hypertensive rats. Am J Physiol Regul Integr Comp Physiol JT- American journal of physiology. Regulatory, integrative and comparative physiology, 2004, 287(3):R619-26.
    [14]仇大勇,张燕,张钧.运动对SHR大鼠心肌AngⅡ及AT-1mRNA表达的影响.南京体育学院学报(自然科学版), 2009(01):16-19.
    [15] Michelini LC, Morris M. Endogenous vasopressin modulates the cardiovascular responses to exercise. Ann N Y Acad Sci JT-Annals of the New York Academy of Sciences, 1999, 897:198-211.
    [16] Higa KT, Mori E, Viana FF, et al. Baroreflex control of heart rate by oxytocin in the solitary-vagal complex. Am J Physiol Regul Integr Comp Physiol JT-American journal of physiology. Regulatory, integrative and comparative physiology, 2002, 282(2):R537-45.
    [17] Mohr E, Richter D. Molecular determinants and physiological relevance of extrasomatic RNA localization in neurons. Front Neuroendocrinol JT-Frontiers in neuroendocri- nology, 2003, 24(2):128-39.
    [18] Martins AS, Crescenzi A, Stern JE, et al. Hypertension and exercise training differentially affect oxytocin and oxytocin receptor expression in the brain. Hypertension JT-Hypertension, 2005, 46(4):1004-9.
    [19] Braga DC, Mori E, Higa KT, et al. Central oxytocin modulates exercise-inducedtachycardia. Am J Physiol Regul Integr Comp Physiol JT-American journal of physiology. Regulatory, integrative and comparative physiology, 2000, 278(6):R1474-82.
    [20] Ulker S, McKeown PP, Bayraktutan U. Vitamins reverse endothelial dysfunction through regulation of eNOS and NAD(P)H oxidase activities. Hypertension JT- Hypertension, 2003, 41(3):534-9.
    [21]窦丽,陈华卫,张钧. 10周游泳运动对高血压大鼠血小板NO及自由基代谢的影响.中国运动医学杂志, 2008(01):88-89, 83.
    [22] Di Massimo C, Scarpelli P, Penco M, et al. Possible involvement of plasma antioxidant defences in training-associated decrease of platelet responsiveness in humans. Eur J Appl Physiol JT-European journal of applied physiology, 2004, 91(4):406-12.
    [23] Tozzi-Ciancarelli MG, Penco M, Di Massimo C. Influence of acute exercise on human platelet responsiveness: possible involvement of exercise-induced oxidative stress. Eur J Appl Physiol JT-European journal of applied physiology, 2002, 86(3):266-72.
    [24] Rush JW, Turk JR, Laughlin MH. Exercise training regulates SOD-1 and oxidative stress in porcine aortic endothelium. Am J Physiol Heart Circ Physiol JT-American journal of physiology. Heart and circulatory physiology, 2003, 284(4):H1378-87.
    [25] Graham DA, Rush JW. Exercise training improves aortic endothelium-dependent vasorelaxation and determinants of nitric oxide bioavailability in spontaneously hypertensive rats. J Appl Physiol JT-Journal of applied physiology (Bethesda, Md: 1985), 2004, 96(6):2088-96.
    [26]邵兰真.有氧运动对原发性高血压患者血压及血浆内皮素的影响.中国临床康复, 2003, 7(12):1854-1855.
    [27]吴红群.运动对高血压大鼠血压和内皮素的影响.中国临床康复, 2005, (40):111-113.
    [28] Maeda S, Tanabe T, Miyauchi T, et al. Aerobic exercise training reduces plasma endothelin-1 concentration in older women. J Appl Physiol JT-Journal of applied physiology (Bethesda, Md: 1985), 2003, 95(1):336-41.
    [29]窦丽,张钧.有氧运动对高血压大鼠血浆6-酮-前列腺素F1α、血栓素B-2含量和PGI-2/TXA-2系统的影响.中国康复医学杂志, 2008, (03): 251-253.
    [30] Poli KA, Tofler GH, Larson MG, et al. Association of blood pressure with fibrinolyticpotential in the Framingham offspring population. Circulation JT-Circulation, 2000, 101(3):264-9.
    [31]吴红群.运动对高血压大鼠纤溶功能的影响.中国临床康复, 2005, (43): 41-43.
    [32] Sakata K, Pawlak R, Urano T, et al. Effects of a long-term pharmacological interruption of the renin-angiotensin system on the fibrinolytic system in essential hypertension. Pathophysiol Haemost Thromb JT - Pathophysiology of haemostasis and thrombosis, 2002, 32(2):67-75.
    [33]卢开信,黄叔怀.不同运动负荷对大鼠主动脉平滑肌HO-CO系统的影响.中国运动医学杂志, 2002(06):570-573.
    [34]任彩玲,张钧.血红素氧合酶-1在心血管及运动中的作用.中国康复医学杂志, 2005(01):79-81.
    [35] Douglas SA, Tayara L, Ohlstein EH, et al. Congestive heart failure and expression of myocardial urotensin II. Lancet JT-Lancet, 2002, 359(9322):1990-7.
    [36] Cheung BM, Leung R, Man YB, et al. Plasma concentration of urotensin II is raised in hypertension. J Hypertens JT-Journal of hypertension, 2004, 22(7):1341-4.
    [37]朱桂平,周万兴,曾智桓,等.幼龄自发性高血压大鼠血浆尾加压素Ⅱ的变化及负荷运动对它的影响.中国心血管病研究杂志, 2004(09):731-733.
    [38]于明月,边树怀,张扬,等.自发性高血压大鼠血浆尾加压素Ⅱ的变化及运动干预的影响.河北医科大学学报, 2006(01):28-29.
    [39] Davignon J, Ganz P. Role of endothelial dysfunction in atherosclerosis. Circulation JT - Circulation, 2004, 109(23 Suppl 1):III27-32.
    [40] Zhou X, Stemme S, Hansson GK. Evidence for a local immune response in atherosclerosis. CD4+ T cells infiltrate lesions of apolipoprotein-E-deficient mice. Am J Pathol JT-The American journal of pathology, 1996, 149(2):359-66.
    [41] Toole JF. ACAS recommendations for carotid endarterectomy. ACAS Executive Committee. Lancet JT- Lancet, 1996, 347(8994):121.
    [42] Libby P. Inflammation in atherosclerosis. Nature JT - Nature, 2002,420(6917):868-74.
    [43] Nawroth PP, Handley DA, Esmon CT, et al. Interleukin 1 induces endothelial cell procoagulant while suppressing cell-surface anticoagulant activity. Proc Natl Acad SciUSA JT-Proceedings of the National Academy of Sciences of the United States of America, 1986, 83(10):3460-4.
    [44] Lutgens E, Daemen MJ. CD40-CD40L interactions in atherosclerosis. Trends Cardiovasc Med JT-Trends in cardiovascular medicine, 2002, 12(1):27-32.
    [45] Jander S, Sitzer M, Wendt A, et al. Expression of tissue factor in high-grade carotid artery stenosis: association with plaque destabilization. Stroke JT-Stroke, a journal of cerebral circulation, 2001, 32(4):850-4.
    [46] Hwang SJ, Ballantyne CM, Sharrett AR, et al. Circulating adhesion molecules VCAM-1, ICAM-1, and E-selectin in carotid atherosclerosis and incident coronary heart disease cases: the Atherosclerosis Risk In Communities (ARIC) study. Circulation JT- Circulation, 1997, 96(12):4219-25.
    [47] Ridker PM, Rifai N, Pfeffer M, et al. Elevation of tumor necrosis factor-alpha and increased risk of recurrent coronary events after myocardial infarction. Circulation JT - Circulation, 2000, 101(18):2149-53.
    [48] Ridker PM. Clinical application of C-reactive protein for cardiovascular disease detection and prevention. Circulation JT-Circulation, 2003, 107(3):363-9.
    [49] Conde ID, Kleiman NS. Soluble CD40 ligand in acute coronary syndromes. N Engl J Med JT-The New England journal of medicine, 2003, 348(25):2575-7.
    [50] Gadina M, Jefferies CA. IL-33: a sheep in wolf's clothing?. Sci STKE JT-Science's STKE: signal transduction knowledge environment, 2007, 2007(390):pe31.
    [51] Curry AJ, Portig I, Goodall JC, et al. T lymphocyte lines isolated from atheromatous plaque contain cells capable of responding to Chlamydia antigens. Clin Exp Immunol JT - Clinical and experimental immunology, 2000, 121(2):261-9.
    [52] Gretarsdottir S, Thorleifsson G, Reynisdottir ST, et al. The gene encoding phosphod- iesterase 4D confers risk of ischemic stroke. Nat Genet JT-Nature genetics, 2003, 35(2): 131-8.
    [53] DeGraba TJ. Immunogenetic susceptibility of atherosclerotic stroke: implications on current and future treatment of vascular inflammation. Stroke JT-Stroke; a journal of cerebral circulation, 2004, 35(11 Suppl 1):2712-9.
    [54] Pola R, Flex A, Gaetani E, et al. Synergistic effect of -174 G/C polymorphism of the interleukin-6 gene promoter and 469 E/K polymorphism of the intercellular adhesion molecule-1 gene in Italian patients with history of ischemic stroke. Stroke JT-Stroke; a journal of cerebral circulation, 2003, 34(4):881-5.
    [55] Worrall BB, Azhar S, Nyquist PA, et al. Interleukin-1 receptor antagonist gene polymorphisms in carotid atherosclerosis. Stroke JT-Stroke; a journal of cerebral circulation, 2003, 34(3):790-3.
    [56] Mason JC. Statins and their role in vascular protection. Clin Sci (Lond) JT-Clinical science (London, England: 1979), 2003, 105(3):251-66.
    [57] Moghadasian MH, McManus BM, Nguyen LB, et al. Pathophysiology of apolipoprotein E deficiency in mice: relevance to apo E-related disorders in humans. FASEB J JT-The FASEB journal: official publication of the Federation of American Societies for Experimental Biology, 2001, 15(14):2623-30.
    [58] Stemme S, Faber B, Holm J, et al. T lymphocytes from human atherosclerotic plaques recognize oxidized low density lipoprotein. Proc Natl Acad Sci USA JT-Proceedings of the National Academy of Sciences of the United States of America, 1995, 92(9):3893-7.
    [59] Benagiano M, D'Elios MM, Amedei A, et al. Human 60-kDa heat shock protein is a target autoantigen of T cells derived from atherosclerotic plaques. J Immunol JT-Journal of immunology (Baltimore, Md: 1950), 2005, 174(10):6509-17.
    [60] Zhou X, Nicoletti A, Elhage R, et al. Transfer of CD4(+) T cells aggravates atherosclerosis in immunodeficient apolipoprotein E knockout mice. Circulation JT- Circulation, 2000, 102(24):2919-22.
    [61] Whitman SC, Ravisankar P, Elam H, et al. Exogenous interferon-gamma enhances atherosclerosis in apolipoprotein E-/- mice. Am J Pathol JT-The American journal of pathology, 2000, 157(6):1819-24.
    [62] Davenport P, Tipping PG. The role of interleukin-4 and interleukin-12 in the progression of atherosclerosis in apolipoprotein E-deficient mice. Am J Pathol JT-The American journal of pathology, 2003, 163(3):1117-25.
    [63] Whitman SC, Ravisankar P, Daugherty A. Interleukin-18 enhances atherosclerosis inapolipoprotein E(-/-) mice through release of interferon-gamma. Circ Res JT- Circul- ation research, 2002, 90(2):E34-8.
    [64] Tenger C, Sundborger A, Jawien J, et al. IL-18 accelerates atherosclerosis accompanied by elevation of IFN-gamma and CXCL16 expression independently of T cells. Arterioscler Thromb Vasc Biol JT-Arteriosclerosis, thrombosis, and vascular biology, 2005, 25(4):791-6.
    [65] Rus HG, Niculescu F, Vlaicu R. Tumor necrosis factor-alpha in human arterial wall with atherosclerosis. Atherosclerosis JT-Atherosclerosis, 1991, 89(2-3):247-54.
    [66] Ohta H, Wada H, Niwa T, et al. Disruption of tumor necrosis factor-alpha gene diminishes the development of atherosclerosis in ApoE-deficient mice. Atherosclerosis JT-Atherosclerosis, 2005, 180(1):11-7.
    [67] Branen L, Hovgaard L, Nitulescu M, et al. Inhibition of tumor necrosis factor-alpha reduces atherosclerosis in apolipoprotein E knockout mice. Arterioscler Thromb Vasc Biol JT-Arteriosclerosis, thrombosis, and vascular biology, 2004, 24(11):2137-42.
    [68] Poston RN, Haskard DO, Coucher JR, et al. Expression of intercellular adhesion molecule-1 in atherosclerotic plaques. Am J Pathol JT-The American journal of pathology, 1992, 140(3):665-73.
    [69] Lessner SM, Prado HL, Waller EK, et al. Atherosclerotic lesions grow through recruitment and proliferation of circulating monocytes in a murine model. Am J Pathol JT-The American journal of pathology, 2002, 160(6):2145-55.
    [70] Li AC, Glass CK. The macrophage foam cell as a target for therapeutic intervention. Nat Med JT-Nature medicine, 2002, 8(11):1235-42.
    [71] Lutgens E, Faber B, Schapira K, et al. Gene profiling in atherosclerosis reveals a key role for small inducible cytokines: validation using a novel monocyte chemoattractant protein monoclonal antibody. Circulation JT - Circulation, 2005, 111(25):3443-52.
    [72] Smith JD, Trogan E, Ginsberg M, et al. Decreased atherosclerosis in mice deficient in both macrophage colony-stimulating factor (op) and apolipoprotein E. Proc Natl Acad Sci USA JT-Proceedings of the National Academy of Sciences of the United States of America, 1995, 92(18):8264-8.
    [73] Schonbeck U, Libby P. Inflammation, immunity, and HMG-CoA reductase inhibitors: statins as antiinflammatory agents?. Circulation JT-Circulation, 2004,109(21 Suppl 1):II18-26.
    [74] Edelberg JM, Christie PD, Rosenberg RD. Regulation of vascular bed-specific prothrombotic potential. Circ Res JT-Circulation research, 2001, 89(2):117-24.
    [75] Schrijver HM, Crusius JB, Uitdehaag BM, et al. Association of interleukin-1beta and interleukin-1 receptor antagonist genes with disease severity in MS. Neurology JT- Neurology, 1999, 52(3):595-9.
    [76] Liao F, Andalibi A, Qiao JH, et al. Genetic evidence for a common pathway mediating oxidative stress, inflammatory gene induction, and aortic fatty streak formation in mice. J Clin Invest JT-The Journal of clinical investigation, 1994, 94(2):877-84.
    [77] Aikawa M, Libby P. Atherosclerotic plaque inflammation: the final frontier?. Can J Cardiol JT-The Canadian journal of cardiology, 2004, 20(6):631-4.
    [78] Skaro AI, Liwski RS, Zhou J, et al. CD8+ T cells mediate aortic allograft vasculopathy by direct killing and an interferon-gamma-dependent indirect pathway. Cardiovasc Res JT-Cardiovascular research, 2005, 65(1):283-91.
    [79] Freigang S, Horkko S, Miller E, et al. Immunization of LDL receptor-deficient mice with homologous malondialdehyde-modified and native LDL reduces progression of atherosclerosis by mechanisms other than induction of high titers of antibodies to oxidative neoepitopes. Arterioscler Thromb Vasc Biol JT-Arteriosclerosis, thrombosis, and vascular biology, 1998, 18(12):1972-82.
    [80] Chyu KY, Reyes OS, Zhao X, et al. Timing affects the efficacy of LDL immunization on atherosclerotic lesions in apo E (-/-) mice. Atherosclerosis JT-Atherosclerosis, 2004, 176(1):27-35.
    [81] Caligiuri G, Nicoletti A, Poirier B, et al. Protective immunity against atherosclerosis carried by B cells of hypercholesterolemic mice. J Clin Invest JT-The Journal of clinical investigation, 2002, 109(6):745-53.
    [82] Major AS, Fazio S, Linton MF. B-lymphocyte deficiency increases atherosclerosis in LDL receptor-null mice. Arterioscler Thromb Vasc Biol JT-Arteriosclerosis, thrombosis,and vascular biology, 2002, 22(11):1892-8.
    [83] Goncalves I, Gronholdt ML, Soderberg I, et al. Humoral immune response against defined oxidized low-density lipoprotein antigens reflects structure and disease activity of carotid plaques. Arterioscler Thromb Vasc Biol JT-Arteriosclerosis, thrombosis, and vascular biology, 2005, 25(6):1250-5.
    [84] Di Napoli M. HMG-CoA reductase inhibitors (statins): a promising approach to stroke prevention. Neurology JT-Neurology, 2000, 55(7):1066-7.
    [85] Weber MS, Prod'homme T, Steinman L, et al. Drug Insight: using statins to treat neuroinflammatory disease. Nat Clin Pract Neurol JT-Nature clinical practice. Neur- ology, 2005, 1(2):106-12.
    [86] Martin-Ventura JL, Blanco-Colio LM, Gomez-Hernandez A, et al. Intensive treatment with atorvastatin reduces inflammation in mononuclear cells and human atherosclerotic lesions in one month. Stroke JT-Stroke; a journal of cerebral circulation, 2005, 36(8): 1796-800.
    [87] Aikawa M, Rabkin E, Sugiyama S, et al. An HMG-CoA reductase inhibitor, cerivastatin, suppresses growth of macrophages expressing matrix metalloproteinases and tissue factor in vivo and in vitro. Circulation JT-Circulation, 2001, 103(2):276-83.
    [88] Kennedy J, Quan H, Buchan AM, et al. Statins are associated with better outcomes after carotid endarterectomy in symptomatic patients. Stroke JT-Stroke; a journal of cerebral circulation, 2005, 36(10):2072-6.
    [89] Crisby M, Nordin-Fredriksson G, Shah PK, et al. Pravastatin treatment increases collagen content and decreases lipid content, inflammation, metalloproteinases, and cell death in human carotid plaques: implications for plaque stabilization. Circulation JT- Circulation, 2001, 103(7):926-33.
    [90] Paoletti R, Gotto AM Jr, Hajjar DP. Inflammation in atherosclerosis and implications for therapy. Circulation JT-Circulation, 2004, 109(23 Suppl 1):III20-6.
    [91] Cannon CP, Braunwald E, McCabe CH, et al. Intensive versus moderate lipid lowering with statins after acute coronary syndromes. N Engl J Med JT-The New England journal of medicine, 2004, 350(15):1495-504.
    [92] Khan Q, Mehta JL. Relevance of platelet-independent effects of aspirin to its salutary effect in atherosclerosis-related events. J Atheroscler Thromb JT-Journal of atherosclerosis and thrombosis, 2005, 12(4):185-90.
    [93] Cipollone F, Fazia M, Mezzetti A. Novel determinants of plaque instability. J Thromb Haemost JT-Journal of thrombosis and haemostasis: JTH, 2005, 3(9):1962-75.
    [94] Hachicha M, Pouliot M, Petasis NA, et al. Lipoxin (LX)A4 and aspirin-triggered 15-epi- LXA4 inhibit tumor necrosis factor 1alpha-initiated neutrophil responses and trafficking: regulators of a cytokine-chemokine axis. J Exp Med JT-The Journal of experimental medicine, 1999, 189(12):1923-30.
    [95] Halkin A, Keren G. Potential indications for angiotensin-converting enzyme inhibitors in atherosclerotic vascular disease. Am J Med JT-The American journal of medicine, 2002, 112(2):126-34.
    [96] Laurat E, Poirier B, Tupin E, et al. In vivo downregulation of T helper cell 1 immune responses reduces atherogenesis in apolipoprotein E-knockout mice. Circulation JT- Circulation, 2001, 104(2):197-202.
    [97] Sattler KJ, Woodrum JE, Galili O, et al. Concurrent treatment with renin-angiotensin system blockers and acetylsalicylic acid reduces nuclear factor kappaB activation and C-reactive protein expression in human carotid artery plaques. Stroke JT-Stroke; a journal of cerebral circulation, 2005, 36(1):14-20.
    [98] Regensteiner JG, Hiatt WR. Current medical therapies for patients with peripheral arterial disease: a critical review. Am J Med JT-The American journal of medicine, 2002, 112(1):49-57.
    [99] Hermanowski-Vosatka A, Balkovec JM, Cheng K, et al. 11beta-HSD1 inhibition ameliorates metabolic syndrome and prevents progression of atherosclerosis in mice. J Exp Med JT- The Journal of experimental medicine, 2005, 202(4):517-27.
    [100] Beaudeux JL, Giral P, Bruckert E, et al. Matrix metalloproteinases, inflammation and atherosclerosis: therapeutic perspectives. Clin Chem Lab Med JT-Clinical chemistry and laboratory medicine: CCLM / FESCC, 2004, 42(2):121-31.
    [101] Axisa B, Loftus IM, Naylor AR, et al. Prospective, randomized, double-blind trialinvestigating the effect of doxycycline on matrix metalloproteinase expression within atherosclerotic carotid plaques. Stroke JT-Stroke; a journal of cerebral circulation, 2002, 33(12):2858-64.
    [102] Kopp CW, Holzenbein T, Steiner S, et al. Inhibition of restenosis by tissue factor pathway inhibitor: in vivo and in vitro evidence for suppressed monocyte chemoattraction and reduced gelatinolytic activity. Blood JT-Blood, 2004, 103(5):1653-61.
    [103] Steffens S, Veillard NR, Arnaud C, et al. Low dose oral cannabinoid therapy reduces progression of atherosclerosis in mice. Nature JT-Nature, 2005, 434(7034):782-6.
    [104] Chyu KY, Zhao X, Reyes OS, et al. Immunization using an Apo B-100 related epitope reduces atherosclerosis and plaque inflammation in hypercholesterolemic apo E (-/-) mice. Biochem Biophys Res Commun JT-Biochemical and biophysical research communications, 2005, 338(4):1982-9.
    [105] Takeda H, Spatz M, Ruetzler C, et al. Induction of mucosal tolerance to E-selectin prevents ischemic and hemorrhagic stroke in spontaneously hypertensive genetically stroke-prone rats. Stroke JT-Stroke; a journal of cerebral circulation, 2002, 33(9):2156-63.
    [106] Frenkel D, Huang Z, Maron R, et al. Nasal vaccination with myelin oligodendrocyte glycoprotein reduces stroke size by inducing IL-10-producing CD4+ T cells. J Immunol JT-Journal of immunology (Baltimore, Md: 1950), 2003, 171(12):6549-55.
    [107] Maron R, Sukhova G, Faria AM, et al. Mucosal administration of heat shock protein-65 decreases atherosclerosis and inflammation in aortic arch of low-density lipoprotein receptor-deficient mice. Circulation JT-Circulation, 2002, 106(13):1708-15.
    [108] Boekholdt SM, Agema WR, Peters RJ, et al. Variants of toll-like receptor 4 modify the efficacy of statin therapy and the risk of cardiovascular events. Circulation JT- Circulation, 2003, 107(19):2416-21.
    [109] Binder CJ, Horkko S, Dewan A, et al. Pneumococcal vaccination decreases atherosclerotic lesion formation: molecular mimicry between Streptococcus pneumoniae and oxidized LDL. Nat Med JT-Nature medicine, 2003, 9(6):736-43.
    [110] Momiyama Y, Hirano R, Taniguchi H, et al. Effects of interleukin-1 gene polymorphisms on the development of coronary artery disease associated withChlamydia pneumoniae infection. J Am Coll Cardiol JT-Journal of the American College of Cardiology, 2001, 38(3):712-7.
    [111] Dinarello CA. Interleukin-1. Cytokine Growth Factor Rev JT-Cytokine & growth factor reviews, 1997, 8(4):253-65.
    [112] Wang X, Feuerstein GZ, Gu JL, et al. Interleukin-1 beta induces expression of adhesion molecules in human vascular smooth muscle cells and enhances adhesion of leukocytes to smooth muscle cells. Atherosclerosis JT-Atherosclerosis, 1995, 115(1):89-98.
    [113]赵庆斌,刘艳,祝家庆,等.急性冠脉综合征患者血清白介素1, 6, 10的变化.第四军医大学学报, 2003(15):1400-1401.
    [114] Oyama J, Shimokawa H, Morita S, et al. Elevated interleukin-1beta in pericardial fluid of patients with ischemic heart disease. Coron Artery Dis JT-Coronary artery disease, 2001, 12(7):567-71.
    [115] Porreca E, Di Febbo C, Barbacane RC, et al. Effect of interleukin-1 receptor antagonist on vascular smooth muscle cell proliferation. Atherosclerosis JT-Atherosclerosis, 1993, 99(1):71-8.
    [116] Nicklin MJ, Hughes DE, Barton JL, et al. Arterial inflammation in mice lacking the interleukin 1 receptor antagonist gene. J Exp Med JT-The Journal of experimental medicine, 2000, 191(2):303-12.
    [117] Francis SE, Camp NJ, Burton AJ, et al. Interleukin 1 receptor antagonist gene polymorphism and restenosis after coronary angioplasty. Heart JT-Heart (British Cardiac Society), 2001, 86(3):336-40.
    [118] Leonard WJ, Lin JX. Cytokine receptor signaling pathways. J Allergy Clin Immunol JT- The Journal of allergy and clinical immunology, 2000, 105(5):877-88.
    [119] Noguchi M, Yi H, Rosenblatt HM, et al. Interleukin-2 receptor gamma chain mutation results in X-linked severe combined immunodeficiency in humans, Cell 73:147-157. J Immunol JT- Journal of immunology (Baltimore, Md: 1950), 2008, 181(9):5817-27.
    [120] Cao X, Shores EW, Hu-Li J, et al. Defective lymphoid development in mice lacking expression of the common cytokine receptor gamma chain. Immunity JT-Immunity, 1995, 2(3):223-38.
    [121] Frostegard J, Ulfgren AK, Nyberg P, et al. Cytokine expression in advanced human atherosclerotic plaques: dominance of pro-inflammatory (Th1) and macrophage- stimulating cytokines. Atherosclerosis JT-Atherosclerosis, 1999, 145(1):33-43.
    [122] Mazzone A, De Servi S, Vezzoli M, et al. Plasma levels of interleukin 2, 6, 10 and phenotypic characterization of circulating T lymphocytes in ischemic heart disease. Atherosclerosis JT-Atherosclerosis, 1999, 145(2):369-74.
    [123] Mizia-Stec K, Mandecki T, Zahorska-Markiewicz B, et al. Selected cytokines and soluble forms of cytokine receptors in coronary artery disease. Eur J Intern Med JT- European journal of internal medicine, 2002, 13(2):115-122.
    [124] Huber SA, Sakkinen P, Conze D, et al. Interleukin-6 exacerbates early atherosclerosis in mice. Arterioscler Thromb Vasc Biol JT-Arteriosclerosis, thrombosis, and vascular biology, 1999, 19(10):2364-7.
    [125] George J, Shoenfeld Y, Gilburd B, et al. Requisite role for interleukin-4 in the acceleration of fatty streaks induced by heat shock protein 65 or Mycobacterium tuberculosis. Circ Res JT-Circulation research, 2000, 86(12):1203-10.
    [126] King VL, Szilvassy SJ, Daugherty A. Interleukin-4 deficiency decreases atherosclerotic lesion formation in a site-specific manner in female LDL receptor-/- mice. Arterioscler Thromb Vasc Biol JT-Arteriosclerosis, thrombosis, and vascular biology, 2002, 22(3): 456-61.
    [127] Cornicelli JA, Butteiger D, Rateri DL, et al. Interleukin-4 augments acetylated LDL-induced cholesterol esterification in macrophages. J Lipid Res JT-Journal of lipid research, 2000, 41(3):376-83.
    [128] Dumoutier L, Louahed J, Renauld JC. Cloning and characterization of IL-10-related T cell-derived inducible factor (IL-TIF), a novel cytokine structurally related to IL-10 and inducible by IL-9. J Immunol JT-Journal of immunology (Baltimore, Md: 1950), 2000, 164(4):1814-9.
    [129] Musso T, Calosso L, Zucca M, et al. Human monocytes constitutively express membrane-bound, biologically active, and interferon-gamma-upregulated interleukin-15. Blood JT-Blood, 1999, 93(10):3531-9.
    [130] Houtkamp MA, van Der Wal AC, de Boer OJ, et al. Interleukin-15 expression in atherosclerotic plaques: an alternative pathway for T-cell activation in atherosclerosis?. Arterioscler Thromb Vasc Biol JT-Arteriosclerosis, thrombosis, and vascular biology, 2001, 21(7):1208-13.
    [131] Bonecchi R, Facchetti F, Dusi S, et al. Induction of functional IL-8 receptors by IL-4 and IL-13 in human monocytes. J Immunol JT-Journal of immunology (Baltimore, Md: 1950), 2000, 164(7):3862-9.
    [132] Oppmann B, Lesley R, Blom B, et al. Novel p19 protein engages IL-12p40 to form a cytokine, IL-23, with biological activities similar as well as distinct from IL-12. Immunity JT-Immunity, 2000, 13(5):715-25.
    [133] Parham C, Chirica M, Timans J, et al. A receptor for the heterodimeric cytokine IL-23 is composed of IL-12Rbeta1 and a novel cytokine receptor subunit, IL-23R. J Immunol JT -Journal of immunology (Baltimore, Md: 1950), 2002, 168(11):5699-708.
    [134] Pflanz S, Timans JC, Cheung J, et al. IL-27, a heterodimeric cytokine composed of EBI3 and p28 protein, induces proliferation of naive CD4(+) T cells. Immunity JT- Immunity, 2002, 16(6):779-90.
    [135] Stenvinkel P, Heimburger O, Jogestrand T. Elevated interleukin-6 predicts progressive carotid artery atherosclerosis in dialysis patients: association with Chlamydia pneumoniae seropositivity. Am J Kidney Dis JT-American journal of kidney diseases: the official journal of the National Kidney Foundation, 2002, 39(2):274-82.
    [136] Balbay Y, Tikiz H, Baptiste RJ, et al. Circulating interleukin-1 beta, interleukin-6, tumor necrosis factor-alpha, and soluble ICAM-1 in patients with chronic stable angina and myocardial infarction. Angiology JT-Angiology, 2001, 52(2):109-14.
    [137] Georges JL, Loukaci V, Poirier O, et al. Interleukin-6 gene polymorphisms and susceptibility to myocardial infarction: the ECTIM study. Etude Cas-Temoin de l'Infarctus du Myocarde. J Mol Med JT-Journal of molecular medicine (Berlin, Germany), 2001, 79(5-6):300-5.
    [138] Bozza M, Bliss JL, Dorner AJ, et al. Interleukin-11 modulates Th1/Th2 cytokine production from activated CD4+ T cells. J Interferon Cytokine Res JT-Journal ofinterferon & cytokine research: the official journal of the International Society for Interferon and Cytokine Research, 2001, 21(1):21-30.
    [139]姜德谦,邓蔚,林礼茂.急性冠状动脉综合征患者血清IL-10及IL-12水平测定.临床心血管病杂志, 2001, 17(5):201-203.
    [140] Brizzi MF, Formato L, Dentelli P, et al. Interleukin-3 stimulates migration and proliferation of vascular smooth muscle cells: a potential role in atherogenesis. Circulation JT-Circulation, 2001, 103(4):549-54.
    [141] Fickenscher H, Hor S, Kupers H, et al. The interleukin-10 family of cytokines. Trends Immunol JT-Trends in immunology, 2002, 23(2):89-96.
    [142] Lang R, Patel D, Morris JJ, et al. Shaping gene expression in activated and resting primary macrophages by IL-10. J Immunol JT-Journal of immunology (Baltimore, Md: 1950), 2002, 169(5):2253-63.
    [143] Smith DA, Irving SD, Sheldon J, et al. Serum levels of the antiinflammatory cytokine interleukin-10 are decreased in patients with unstable angina. Circulation JT- Circulation, 2001, 104(7):746-9.
    [144] Cruikshank WW, Kornfeld H, Center DM. Interleukin-16. J Leukoc Biol JT-Journal of leukocyte biology, 2000, 67(6):757-66.
    [145] Wang N, Tabas I, Winchester R, et al. Interleukin 8 is induced by cholesterol loading of macrophages and expressed by macrophage foam cells in human atheroma. J Biol Chem JT-The Journal of biological chemistry, 1996, 271(15):8837-42.
    [146] Boisvert WA, Santiago R, Curtiss LK, et al. A leukocyte homologue of the IL-8 receptor CXCR-2 mediates the accumulation of macrophages in atherosclerotic lesions of LDL receptor-deficient mice. J Clin Invest JT-The Journal of clinical investigation, 1998, 101(2):353-63.
    [147] Lee J, Ho WH, Maruoka M, et al. IL-17E, a novel proinflammatory ligand for the IL-17 receptor homolog IL-17Rh1. J Biol Chem JT-The Journal of biological chemistry, 2001, 276(2):1660-4.
    [148] Starnes T, Robertson MJ, Sledge G, et al. Cutting edge: IL-17F, a novel cytokine selectively expressed in activated T cells and monocytes, regulates angiogenesis andendothelial cell cytokine production. J Immunol JT-Journal of immunology (Baltimore, Md: 1950), 2001, 167(8):4137-40.
    [149]李明才,柳晓金,苏绍波.白介素-33与炎症.生命的化学, 2008(02): 137-139.
    [150] Baekkevold ES, Roussigne M, Yamanaka T, et al. Molecular characterization of NF-HEV, a nuclear factor preferentially expressed in human high endothelial venules. Am J Pathol JT-The American journal of pathology, 2003, 163(1):69-79.
    [151] Carriere V, Roussel L, Ortega N, et al. IL-33, the IL-1-like cytokine ligand for ST2 receptor, is a chromatin-associated nuclear factor in vivo. Proc Natl Acad Sci USA JT- Proceedings of the National Academy of Sciences of the United States of America, 2007, 104(1):282-7.
    [152] Moussion C, Ortega N, Girard JP. The IL-1-like cytokine IL-33 is constitutively expressed in the nucleus of endothelial cells and epithelial cells in vivo: a novel 'alarmin'?. PLoS One JT- PloS one, 2008, 3(10):e3331.
    [153] Dinarello CA. Biologic basis for interleukin-1 in disease. Blood JT-Blood, 1996, 87(6): 2095-147.
    [154] Schmitz J, Owyang A, Oldham E, et al. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity JT-Immunity, 2005, 23(5):479-90.
    [155] Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature JT-Nature, 2002, 418(6894):191-5.
    [156] Bonaldi T, Talamo F, Scaffidi P, et al. Monocytic cells hyperacetylate chromatin protein HMGB1 to redirect it towards secretion. EMBO J JT-The EMBO journal, 2003, 22(20): 5551-60.
    [157] Bianchi ME. DAMPs, PAMPs and alarmins: all we need to know about danger. J Leukoc Biol JT-Journal of leukocyte biology, 2007, 81(1):1-5.
    [158] Allakhverdi Z, Smith DE, Comeau MR, et al. Cutting edge: The ST2 ligand IL-33 potently activates and drives maturation of human mast cells. J Immunol JT-Journal of immunology (Baltimore, Md: 1950), 2007, 179(4):2051-4.
    [159] Galli SJ, Nakae S, Tsai M. Mast cells in the development of adaptive immune responses.Nat Immunol JT-Nature immunology, 2005, 6(2):135-42.
    [160] Allakhverdi Z, Comeau MR, Jessup HK, et al. Thymic stromal lymphopoietin is released by human epithelial cells in response to microbes, trauma, or inflammation and potently activates mast cells. J Exp Med JT-The Journal of experimental medicine, 2007, 204(2):253-8.
    [161] Soumelis V, Reche PA, Kanzler H, et al. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nat Immunol JT-Nature immunology, 2002, 3(7):673-80.
    [162] Bajenoff M, Egen JG, Koo LY, et al. Stromal cell networks regulate lymphocyte entry, migration, and territoriality in lymph nodes. Immunity JT-Immunity, 2006, 25(6):989-1001.
    [163] Link A, Vogt TK, Favre S, et al. Fibroblastic reticular cells in lymph nodes regulate the homeostasis of naive T cells. Nat Immunol JT-Nature immunology, 2007,8(11):1255-65.
    [164] Luther SA, Tang HL, Hyman PL, et al. Coexpression of the chemokines ELC and SLC by T zone stromal cells and deletion of the ELC gene in the plt/plt mouse. Proc Natl Acad Sci USA JT-Proceedings of the National Academy of Sciences of the United States of America, 2000, 97(23):12694-9.
    [165] Hinz B, Phan SH, Thannickal VJ, et al. The myofibroblast: one function, multiple origins. Am J Pathol JT-The American journal of pathology, 2007, 170(6):1807-16.
    [166] Tomasek JJ, Gabbiani G, Hinz B, et al. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat Rev Mol Cell Biol JT-Nature reviews. Molecular cell biology, 2002, 3(5):349-63.
    [167] Sanada S, Hakuno D, Higgins LJ, et al. IL-33 and ST2 comprise a critical biomechanically induced and cardioprotective signaling system. J Clin Invest JT-The Journal of clinical investigation, 2007, 117(6):1538-49.
    [168] Onda H, Kasuya H, Takakura K, et al. Identification of genes differentially expressed in canine vasospastic cerebral arteries after subarachnoid hemorrhage. J Cereb Blood Flow Metab JT-Journal of cerebral blood flow and metabolism: official journal of the International Society of Cerebral Blood Flow and Metabolism, 1999, 19(11):1279-88.
    [169] Weinberg EO, Shimpo M, De Keulenaer GW, et al. Expression and regulation of ST2,an interleukin-1 receptor family member, in cardiomyocytes and myocardial infarction. Circulation JT-Circulation, 2002, 106(23):2961-6.
    [170] Dunne A, O'Neill LA. The interleukin-1 receptor/Toll-like receptor superfamily: signal transduction during inflammation and host defense. Sci STKE JT-Science's STKE: signal transduction knowledge environment, 2003, 2003(171):re3.
    [171] Matsuzawa A, Saegusa K, Noguchi T, et al. ROS-dependent activation of the TRAF6- ASK1-p38 pathway is selectively required for TLR4-mediated innate immunity. Nat Immunol JT-Nature immunology, 2005, 6(6):587-92.
    [172] Ninomiya-Tsuji J, Kishimoto K, Hiyama A, et al. The kinase TAK1 can activate the NIK-I kappaB as well as the MAP kinase cascade in the IL-1 signalling pathway. Nature JT-Nature, 1999, 398(6724):252-6.
    [173] Zhang D, Gaussin V, Taffet GE, et al. TAK1 is activated in the myocardium after pressure overload and is sufficient to provoke heart failure in transgenic mice. Nat Med JT-Nature medicine, 2000, 6(5):556-63.
    [174] Iwahana H, Yanagisawa K, Ito-Kosaka A, et al. Different promoter usage and multiple transcription initiation sites of the interleukin-1 receptor-related human ST2 gene in UT-7 and TM12 cells. Eur J Biochem JT-European journal of biochemistry / FEBS, 1999, 264(2):397-406.
    [175] Ali S, Huber M, Kollewe C, et al. IL-1 receptor accessory protein is essential for IL-33-induced activation of T lymphocytes and mast cells. Proc Natl Acad Sci USA JT- Proceedings of the National Academy of Sciences of the United States of America, 2007, 104(47):18660-5.
    [176] Brunner M, Krenn C, Roth G, et al. Increased levels of soluble ST2 protein and IgG1 production in patients with sepsis and trauma. Intensive Care Med JT-Intensive care medicine, 2004, 30(7):1468-73.
    [177] Shimpo M, Morrow DA, Weinberg EO, et al. Serum levels of the interleukin-1 receptor family member ST2 predict mortality and clinical outcome in acute myocardial infarction. Circulation JT-Circulation, 2004, 109(18):2186-90.
    [178] Weinberg EO, Shimpo M, Hurwitz S, et al. Identification of serum soluble ST2 receptoras a novel heart failure biomarker. Circulation JT-Circulation, 2003, 107(5):721-6.
    [179] Tajima S, Oshikawa K, Tominaga S, et al. The increase in serum soluble ST2 protein upon acute exacerbation of idiopathic pulmonary fibrosis. Chest JT-Chest, 2003, 124(4): 1206-14.
    [180] Oshikawa K, Kuroiwa K, Tago K, et al. Elevated soluble ST2 protein levels in sera of patients with asthma with an acute exacerbation. Am J Respir Crit Care Med JT- American journal of respiratory and critical care medicine, 2001, 164(2):277-81.
    [181] Kuroiwa K, Arai T, Okazaki H, et al. Identification of human ST2 protein in the sera of patients with autoimmune diseases. Biochem Biophys Res Commun JT- Biochemical and biophysical research communications, 2001, 284(5):1104-8.
    [182] May LT, Ndubuisi MI, Patel K, et al. Interleukin-6 chaperones in blood. Ann N Y Acad Sci JT-Annals of the New York Academy of Sciences, 1995, 762:120-8.
    [183] Economides AN, Carpenter LR, Rudge JS, et al. Cytokine traps: multi-component, high- affinity blockers of cytokine action. Nat Med JT-Nature medicine, 2003, 9(1):47-52.
    [184] Sadoshima J, Izumo S. The cellular and molecular response of cardiac myocytes to mechanical stress. Annu Rev Physiol JT-Annual review of physiology, 1997, 59:551-71.
    [185] Grossman W, Jones D, McLaurin LP. Wall stress and patterns of hypertrophy in the human left ventricle. J Clin Invest JT-The Journal of clinical investigation, 1975, 56(1): 56-64.
    [186] Baudino TA, Carver W, Giles W, et al. Cardiac fibroblasts: friend or foe?. Am J Physiol Heart Circ Physiol JT-American journal of physiology. Heart and circulatory physiology, 2006, 291(3):H1015-26.
    [187] Ha T, Hua F, Li Y, et al. Blockade of MyD88 attenuates cardiac hypertrophy and decreases cardiac myocyte apoptosis in pressure overload-induced cardiac hypertrophy in vivo. Am J Physiol Heart Circ Physiol JT-American journal of physiology. Heart and circulatory physiology, 2006, 290(3):H985-94.
    [188] Sarkar S, Vellaichamy E, Young D, et al. Influence of cytokines and growth factors in ANG II-mediated collagen upregulation by fibroblasts in rats: role of myocytes. Am J Physiol Heart Circ Physiol JT-American journal of physiology. Heart and circulatoryphysiology, 2004, 287(1):H107-17.
    [189] Freund C, Schmidt-Ullrich R, Baurand A, et al. Requirement of nuclear factor-kappaB in angiotensin II- and isoproterenol-induced cardiac hypertrophy in vivo. Circulation JT- Circulation, 2005, 111(18):2319-25.
    [190] Hoffmann A, Baltimore D. Circuitry of nuclear factor kappaB signaling. Immunol Rev JT- Immunological reviews, 2006, 210:171-86.
    [191] Hingtgen SD, Tian X, Yang J, et al. Nox2-containing NADPH oxidase and Akt activation play a key role in angiotensin II-induced cardiomyocyte hypertrophy. Physiol Genomics JT- Physiological genomics, 2006, 26(3):180-91.
    [192] Nishida M, Tanabe S, Maruyama Y, et al. G alpha 12/13- and reactive oxygen species- dependent activation of c-Jun NH2-terminal kinase and p38 mitogen-activated protein kinase by angiotensin receptor stimulation in rat neonatal cardiomyocytes. J Biol Chem JT-The Journal of biological chemistry, 2005, 280(18):18434-41.
    [193] Li Q, Harraz MM, Zhou W, et al. Nox2 and Rac1 regulate H2O2-dependent recruitment of TRAF6 to endosomal interleukin-1 receptor complexes. Mol Cell Biol JT- Molecular and cellular biology, 2006, 26(1):140-54.
    [194] Ha T, Li Y, Hua F, et al. Reduced cardiac hypertrophy in toll-like receptor 4-deficient mice following pressure overload. Cardiovasc Res JT-Cardiovascular research, 2005, 68(2):224-34.
    [195] Hansson GK, Libby P. The immune response in atherosclerosis: a double-edged sword. Nat Rev Immunol JT- Nature reviews. Immunology, 2006, 6(7):508-19.
    [196] de Boer OJ, van der Wal AC, Verhagen CE, et al. Cytokine secretion profiles of cloned T cells from human aortic atherosclerotic plaques. J Pathol JT- The Journal of pathology, 1999, 188(2):174-9.
    [197] Buono C, Binder CJ, Stavrakis G, et al. T-bet deficiency reduces atherosclerosis and alters plaque antigen-specific immune responses. Proc Natl Acad Sci USA JT- Proceedings of the National Academy of Sciences of the United States of America, 2005, 102(5):1596-601.
    [198] Bobryshev YV, Lord RS. Expression of heat shock protein-70 by dendritic cells in thearterial intima and its potential significance in atherogenesis. J Vasc Surg JT-Journal of vascular surgery: official publication, the Society for Vascular Surgery [and] International Society for Cardiovascular Surgery, North American Chapter, 2002, 35(2):368-75.
    [199]贾真,顾复生,向农.硝苯地平控释片改善稳定性心绞痛患者一氧化氮对淋巴细胞Ca2+i的调节作用.中华心血管病杂志, 2001(05):27-29.
    [200]胡雪松,孙宁玲.硝苯地平控释片改善血管内皮功能的探讨.中国循环杂志, 2002(03): 50-52.
    [201] Kawamura T, Yoshida K, Sugawara A, et al. Regulation of skeletal muscle peroxisome proliferator-activated receptor gamma expression by exercise and angiotensin- converting enzyme inhibition in fructose-fed hypertensive rats. Hypertens Res JT- Hypertension research: official journal of the Japanese Society of Hypertension, 2004, 27(1):61-70.
    [202] Gorla-Bajszczak A, Siegrist-Kaiser C, Boss O, et al. Expression of peroxisome proliferator-activated receptors in lean and obese Zucker rats. Eur J Endocrinol JT- European journal of endocrinology / European Federation of Endocrine Societies, 2000, 142(1):71-8.
    [203] Tunstall RJ, Mehan KA, Wadley GD, et al. Exercise training increases lipid metabolism gene expression in human skeletal muscle. Am J Physiol Endocrinol Metab JT - American journal of physiology. Endocrinology and metabolism, 2002, 283(1):E66-72.
    [204] Diep QN, El Mabrouk M, Cohn JS, et al. Structure, endothelial function, cell growth, and inflammation in blood vessels of angiotensin II-infused rats: role of peroxisome proliferator-activated receptor-gamma. Circulation JT- Circulation, 2002, 105(19):2296-302.
    [205] Sun MW, Qian FL, Wang J, et al. Low-intensity voluntary running lowers blood pressure with simultaneous improvement in endothelium-dependent vasodilatation and insulin sensitivity in aged spontaneously hypertensive rats. Hypertens Res JT- Hypertension research: official journal of the Japanese Society of Hypertension, 2008, 31(3):543-52.
    [206]刘颖,赵瑛,刘会敏.罗格列酮对胰岛素抵抗大鼠肾皮质内皮素1及内皮型一氧化氮合酶表达的影响.中华肾脏病杂志, 2005(03):157-160.
    [207]张卫国,杨桂姣,孟华等.力竭游泳运动大鼠下丘脑室旁核NF-κB的表达.山西医科大学学报, 2009(05):385-388, 477.
    [208]孟华,杨桂姣,张卫国,等.力竭运动对大鼠杏仁核NF-κB表达的影响.山西医科大学学报, 2009(04): 289-291, 383.
    [209] Hollander J, Fiebig R, Gore M, et al. Superoxide dismutase gene expression is activated by a single bout of exercise in rat skeletal muscle. Pflugers Arch JT-Pflugers Archiv: European journal of physiology, 2001, 442(3):426-34.
    [210]吴潇男,田振军,张志琪.运动训练对大鼠主动脉应力和NF-κB及c-fos表达的影响.中国应用生理学杂志, 2009(01): 97-101.
    [211] Pockley AG, Calderwood SK, Multhoff G. The atheroprotective properties of Hsp70: a role for Hsp70-endothelial interactions?. Cell Stress Chaperones JT-Cell stress & chaperones, 2009, 14(6):545-53.
    [212] Jornot L, Mirault ME, Junod AF. Differential expression of hsp70 stress proteins in human endothelial cells exposed to heat shock and hydrogen peroxide. Am J Respir Cell Mol Biol JT-American journal of respiratory cell and molecular biology, 1991, 5(3): 265-75.
    [213] Madden LA, Sandstrom ME, Lovell RJ, et al. Inducible heat shock protein 70 and its role in preconditioning and exercise. Amino Acids JT-Amino acids, 2008, 34(4):511-6.
    [214]胡亚哲,陈艳,扈诗兴,等.运动训练诱导大鼠心肌细胞热休克蛋白70表达.中国运动医学杂志, 2009(05): 561-563.
    [215] Sandstrom ME, Siegler JC, Lovell RJ, et al. The effect of 15 consecutive days of heat-exercise acclimation on heat shock protein 70. Cell Stress Chaperones JT-Cell stress & chaperones, 2008, 13(2):169-75.
    [216] Melling CW, Thorp DB, Noble EG. Regulation of myocardial heat shock protein 70 gene expression following exercise. J Mol Cell Cardiol JT-Journal of molecular and cellular cardiology, 2004, 37(4):847-55.
    [217] Paroo Z, Haist JV, Karmazyn M, et al. Exercise improves postischemic cardiac function in males but not females: consequences of a novel sex-specific heat shock protein 70 response. Circ Res JT-Circulation research, 2002, 90(8):911-7.
    [218] Morton JP, Holloway K, Woods P, et al. Exercise training-induced gender-specific heatshock protein adaptations in human skeletal muscle. Muscle Nerve JT-Muscle & nerve, 2009, 39(2):230-3.
    [219] Brint EK, Xu D, Liu H, et al. ST2 is an inhibitor of interleukin 1 receptor and Toll-like receptor 4 signaling and maintains endotoxin tolerance. Nat Immunol. 2004,5(4): 373-9.
    [220] Timmers L, Sluijter JP, van KJK, et al. Toll-like receptor 4 mediates maladaptive left ventricular remodeling and impairs cardiac function after myocardial infarction. Circ Res, 2008, 102(2): 257-64.
    [221] Miller AM, Xu D, Asquith DL, et al. IL-33 reduces the development of atherosclerosis. J Exp Med JT-The Journal of experimental medicine, 2008, 205(2):339-46.
    [222]范天全,于海奕,郭艳红.急性冠状动脉综合征患者血浆白细胞介素33浓度变化及其与预后的关系.临床荟萃, 2009, 24(11):924-927. ???

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700