Thymosinβ4及β-aescin早期应用对脊髓损伤的保护作用
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
实验一、Thymosinβ4早期应用对脊髓损伤的保护作用
     【研究背景】脊髓损伤是一个引起全球广泛关注的问题,尤其是近十年来。重度脊髓损伤往往导致患者终身瘫痪。来自临床研究和动物实验的大量证据均表明,脊髓损伤的预后,不仅取决于原发性损伤的严重程度,更是与其所引起的一系列细胞和分子水平的继发性损伤过程的发展程度密切相关。这些继发性病理过程包括缺血,缺氧,免疫反应,兴奋性毒性,自由基损伤,神经细胞变性死亡,轴突脱髓鞘和waller变性等。脊髓高度血管化,以满足它对氧和葡萄糖的高需求,因此缺血往往导致严重的破坏性后果。我们以往的研究结果发现在不断扩张的脊髓损伤区的边缘存在两个缺血区,临近的缺血区大量神经元正在变性或者已经消失,稍远的缺血区大多数神经元显得相对正常并且认为可以被拯救。有报道称,改善创伤后脊髓血流能显著促进神经功能恢复。
     免疫反应被认为在脊髓损伤中发挥类似双刃剑的作用。浸润的免疫细胞可以释放神经营养因子,介导神经保护和促进神经再生。但是已有大量研究表明,伤后早期炎症反应以及脊髓损伤后激活的巨噬细胞可能参与损伤诱导的神经病变和运动功能损害。
     神经元和少突胶质细胞均对各种继发性有害因素非常敏感而最终经历变性死亡。其中,少突胶质细胞凋亡导致原发性损伤后残存的轴突进一步脱髓鞘,使那些轴突的传导能力受损,这必然显著影响病人日后神经功能的恢复。
     Tβ4是一种由43个氨基酸组成的高度保守的小分子多肽,分子量为4.9 kD,最初是从小牛胸腺组织中分离得到的。它广泛存在于几乎所有的细胞,尤其在血小板和白细胞内水平相对较高。Tβ4具有促进血管发生,抗炎,促进细胞存活和创伤修复等特性,而这些均与脊髓损伤修复密切相关。但是目前尚没有Tβ4应用于脊髓损伤的相关报道。本实验主要关注脊髓损伤后,Tβ4促进血管发生,神经元保护和抗炎三方面的作用,并观察运动功能恢复情况。
     【目的】利用我们研究所自行设计的脊髓损伤模型,观察Tβ4对脊髓损伤的治疗作用。
     【方法】所以大鼠分为3组:假手术组(n=5),Tβ4(n=15)和生理盐水组(n=15)。将大鼠麻醉后,使用改良的眼科显微镊制作大鼠脊髓夹伤模型。伤后30 min,腹腔注射Tβ4溶液,以后每3 d给药1次。至第7 d,处死所有大鼠,行组织化学检查, RECA1标记血管,NeuN标记神经元,ED1标记活化小胶质细胞和血源性巨噬细胞,GFAP标记胶质瘢痕。Western blotting检测活化小胶质细胞和血源性巨噬细胞的特异性标记物ED1和成熟少突胶质细胞的特异性标记物MBP的表达水平。所有大鼠存活7 d,在相应的时间点对大鼠进行BBB评分和足迹实验。生理盐水替代Tβ4作为对照组。
     【结果】组织化学结果显示Tβ4组大鼠脊髓空洞面积较盐水组减少32.1%。血管长度和神经元的变化主要存在于距损伤边界吻、尾侧各1 mm范围内。血管总长度,Tβ4组吻、尾侧分别是14.72±0.17 mm/mm~2和13.35±0.13 mm/mm~2,盐水组吻、尾侧分别是12.97±0.45 mm/mm~2和11.40±0.39 mm/mm~2;神经元数量,Tβ4组吻尾侧分别是335.1±11.1/mm~2和317.7±9.3/mm~2,盐水组吻、尾侧分别是284.8±15.4/mm~2和262.1±16.6 /mm~2。Tβ4和盐水两组之间密切相关。Western blotting结果显示,Tβ4组ED1表达量较盐水组下降了35.9%;Tβ4组MBP水平仅下降了20.1%,较盐水组高44.3%。各种运动功能评估方法均显示,Tβ4组大鼠运功功能较盐水组明显改善。
     【结论】在本实验,我们率先发现应用Tβ4可以显著减少SCI后损伤面积,加速运动功能恢复,很可能与其刺激血管发生,发挥神经保护和减少炎症浸润有关。Tβ4的临床安全性已经明确,因此它可以作为未来SCI治疗的选择之一。
     实验二、β-aescin早期应用对脊髓损伤的保护作用
     【研究背景】在SCI,除了原发性创伤造成的直接损伤外,在其周围还将发生不断扩张的继发性病理改变,包括缺血,自由基损伤,炎症反应,兴奋性毒性,神经元变性坏死及凋亡等。脂质过氧化(lipid peroxydation LPO)是脊髓损伤后最早出现的生化改变之一,是自由基损伤的延续,被认为是导致原始损伤区域向外恶化蔓延的重要因素。丙二醛(MDA)是膜结构中多不饱和脂肪酸过氧化的主要终产物,被认为是LPO的生物学标记。炎症反应在SCI中的作用类似双刃剑,炎症细胞早期广泛聚集并分泌大量毒性细胞因子和自由基而加重SCI,后期合成保护性细胞因子及生长因子介导神经保护和促进神经再生。
     Aescin属三萜类化合物,是从欧洲七叶树中提取的主要活性成分。Aescin含α和β两种异构体,β-aescin作为主要的活性异构体,具有抗炎、抗水肿、清除活性氧、增强静脉张力和保护内皮细胞的作用,已广泛应用于药学研究及临床治疗。
     本实验我们采用成年SD大鼠建立脊髓中度撞击伤模型。检测伤后24 h脊髓MDA的水平和髓过氧化物酶(MPO)的活性,观察伤后14 d炎症细胞的变化。观察β-aescin对它们的影响,并探讨β-aescin的SCI早期治疗作用机制。
     【目的】采用成年SD大鼠建立脊髓中度撞击伤模型,观察β-aescin对大鼠脊髓损伤后继发性损害的保护作用。
     【方法】所有大鼠分为3组:假手术组(n=10),Tβ4(n=18)和生理盐水组(n=18)。造模成功后30 min,开始经腹腔给予β-aescin(1.0 mg/kg)或生理盐水,3次/d,连续给药3 d。假手术组为正常对照。于各时间点采用BBB评分和足迹实验对大鼠双后肢运动功能进行评估;术后24 h,对损伤部位脊髓组织MDA含量及MPO活性进行检测;术后14 d行免疫组织化学检查,利用GFAP观察损伤星形胶质瘢痕及其包围的坏死空洞,利用ED1观察活性小胶质细胞和巨噬细胞。
     【结果】β-aescin组运动功能明显改善。伤后24 h,β-aescin组大鼠脊髓组织中MDA水平降低67%,MPO活性降低50%。伤后14 d,β-aescin组空洞面积减小29.3%,ED1染色阳性面积减少32.3%。
     【结论】β-aescin可以显著减轻SCI后自由基的氧化损伤作用,减少炎症细胞的活化和浸润,减小空洞面积,促进SCI后神经功能的恢复。
【Background】Spinal cord injury (SCI) is an issue that has aroused worldwide interest, particularly in recent decades. Severe SCI often leads to lifelong paralysis. Evidence from both clinical and experimental studies indicates that, the pathologic process following SCI is divided into two chronological phases: the primary mechanical injury and a wave of secondary injury triggered by the initial insult The multifaceted secondary injury that results in evolution of the pathological changes are mainly associated with ischemia, edema, immune responses, excitotoxicity, free radical injury, and apoptosis. The spinal cord is highly vascularized to meet its high demand of oxygen and glucose supply, thus ischemia may cause devastating outcomes. Our previous study found that, there were two ischemic zones at the front of the expanding secondary injury, a zone neighboring the lesion where most of the neurons are degenerating or have disappeared, and a farther zone in which most of the neurons appeared relatively normal and considered possible to be rescued. In fact, it has been reported that improving posttraumatic spinal cord blood flow can significantly facilitate functional recovery. That the immune responses considered as a double-edged sword in SCI has been well documented. Although infiltrating immune cells produce neurotrophic factors mediating neuroprotection and regeneration after SCI, it has also been shown that an early inflammatory response and activated macrophages initiated by SCI may participate in neuropathology and loss of motor function. In those secondary damaging processes after SCI, both neurons and oligodendrocytes are vulnerable to various harmful factors. Due to the loss of oligodendrocytes and subsequent axonal demyelination of surviving axons and impair their conductive capacity, it seems reasonable to expect that reducing oligodendrocyte death and improving axonal remyelination holds potential for the treatment of SCI.
     Tβ4, a highly conserved 4.9 kD polypeptide, comprises of 43 amino acids, which was first isolated from bovine thymus. It is a natural polypeptide that is ubiquitously distributed in almost all cells with relatively higher levels in platelets and white blood cells. The angiogenesis, anti-inflammation, wound repair and cell survival properties of Tβ4 are highly relevant to the SCI. We have recently formulated an idea of early neurosurgery of spinal cord contusion with great success. The patients received only surgery and rehabilitation. No medication that might improve recovery was given to avoid confusion of credit of the surgery. Therefore, search for effective drugs that may fill in the big space left for improving the outcome of operation may contribute substantially to the treatment of spinal cord contusion. The present study was focused on its angiogenic, neuronal protective and anti-inflammatory effects and the results showed that these three aspects of Tβ4 worked well in reducing the secondary injury and improve locomotion.
     【Objective】The present study was aimed to investigate the therapeutic value for SCI of its angiogenic, neuroprotective and anti-inflammatory properties. We used a rat SCI model of bilateral compression of the spinal cord.
     【Methods】The SD rats were randomly divided into 3 groups: sham (n = 5), Tβ4 (n = 15), and normal saline (n = 15). After anesthesia, a rat SCI model of lateral compression of the spinal cord was produced using a pair of modified forceps. Tβ4 or normal saline was administrated i.p. 30 min after injury, and 2 doses on the first day of every other 3 days. Histological tests were performed to assess blood vessels, neuronal survival, and lesion size. Western blot assay was executed to detect the expression of specifical marker ED1 for activated microglia and infiltrated macrophages and specifical marker MBP for mature oligodendrocytes. All rats were allowed to surviv for 7 days. All rats underwent behavioral assessments at every time point.
     【Results】Our data demonstrated that the size of lesion area delineated by GFAP was decreased by 32.1% in the Tβ4 group compared with the saline group. The changes of total length of the blood vessels and total number of neurons within the gray matter occurred in the areas 1 mm rostral and caudal to the lesion edge. The density of total length of blood vessels in the rostral 1 mm was 14.72±0.17 mm/mm~2 in the Tβ4 treated group versus 12.97±0.45 mm/mm~2 in the saline group, in the caudal 1 mm was 13.35±0.13 mm/mm~2 versus 11.40±0.39 mm/mm~2, respectively. The number of neurons in the Tβ4 treated group in the rostral and caudal 1 mm zones were 335.1±11.1/mm~2 and 317.7±9.3 /mm~2, respectively, whereas in the saline group the rostral and caudal zones hosted far less NeuN-immunoreactive cells, 284.8±15.4/mm~2 and 262.1±16.6 /mm~2, respectively. Furthermore, there was a close correlation between the effects on blood vessel length and neuronal survival. Western blotting assay showed that, the ED1 expression in the Tβ4 group was 35.9% less than that in the saline group; the MBP level was only decreased by 20.1% in the Tβ4 group but clearly decreased by 44.3% in the saline group compared with normal control. Locomotor recovery was tested using the BBB score and footprint analysis. All of the behavioral assessments were markedly improved.
     【Conclusions】Here, for the first time, our present study shows that, systemic administration of Tβ4 after acute SCI in rats, remarkably reduces the lesion size and some pathological changes of the injured spinal cord, and accelerates locomotion recovery, most probably, by stimulating angiogenesis, increasing neuronal survival, reducing inflammatory infiltration and demyelination. Given its known safety in clinical uses and the beneficial effects on spinal cord injury, Tβ4 can well be a good candidate for treating spinal cord contusion.
     Part II: Beneficial effects ofβ-aescin on spinal cord injury in the rat
     【Background】The severity of neurological disturbance, induced by traumatic SCI, is not only associated with the degree of the mechanical insult to the tissue, but also closely correlated to progressive tissue loss beyond the primary injury site. Multiple secondary damaging factors are involved in the latter process, including ischemia, immune response, glutamate excitotoxicity, free radical generation, reactive oxygen species, lipid peroxidation (LPO), and apoptosis, to name a few. LPO is a self-perpetuating form of free radical damage that is believed to be a major factor in the spread of deterioration after SCI to regions of spared tissue outside the original zone of trauma. Malondialdehyde (MDA), the principal end product of polyunsaturated fatty acid peroxidation in cell membranes, considered as a biological marker of LPO, is a highly toxic molecule, aggravating the damage beyond membrane lipids by interacting with amino acids, proteins and DNA. The immune system likely plays a critical role in the secondary degenerative process. An intense local inflammatory response, typed by activation of resident microglia and influx of hematogenous neutrophils and macrophages, is rapidly triggered by traumatic SCI. Those cells produce free radicals including superoxide anion and nitric oxide during these processes inducing apoptosis in neurons and glia via the irreversible oxidation of proteins, lipids and nucleic acids. The LPO and inflammatory responses are therefore good targets for the exploration of pharmacological strategies to treat acute SCI. At present, the methylprednisolone having the properties of anti-peroxidation and anti-inflammation remains the only neuroprotective agent in clinic pharmacotherapy for acute SCI, but its use existing extensive controversies due to undesirable side effects. Thus we are interested in seeking for a novel therapeutic approach for acute phase SCI without significant side effects.
     Aescin, the major active principle from aesculus hippocastanum, was subjected to a natural mixture of triterpene saponins. It has two isomers ofαandβ. Among which,β-aescin shown numerous pharmacological properties of anti-edema, anti-inflammation, anti-oxidation, increasing venous tone, and protecting vascular endothelial cells.β-aescin appears to be the active component of the mixture and is the molecular form present in major available pharmaceutical products. It has been widely applied in haemorrhoids, chronic venous insuf?ciency, peripheral vascular diseases, and postoperative, postburn or posttraumatic edema in clinical practices with very few adverse effects. The aim of this study was to investigate the possible effects ofβ-aescin on LPO and cellular in?ammation after traumatic SCI when the agent was administered intraperitoneally. We also determined whetherβ-aescin could improve functional deficits.
     【objective】To investigate the protective effects ofβ-aescin on acute traumatic spinal cord injury (SCI) in rats
     【Methods】We examined this hypothesis in a rat model of moderate spinal cord contusion. All rats were assigned into 3 groups: 1. Sham-operated control group (n = 10): only received a laminectomy with no additional treatment; 2.β-aescin group (n = 18): SCI plus 1 mlβ-aescin.i.p; 3. Saline control group (n = 18): SCI plus 1 ml saline.i.p. 30 min after injury, the spinal cord-injured rats were treated withβ-aescin (1.0 mg/kg bw i.p.) or normal saline, and received additional dose t.i.d for 3 consecutive days. Locomotor deficits induced by SCI were assessed using the BBB score and footprint analysis. Biochemical assays were executed to detect the level of malondialdehyde (MDA) and the activity of myeloperoxydase (MPO). Histological tests were performed to analyze the lesion size, and activated microglia and recruited macrophages.
     【Results】Our data indicated that all of the behavioral assessments were markedly improved. In addition,β-aescin treatment significantly reduced MDA level by 67% and MPO activity by 50% at 24 hr after injury, decreased lesion cavitation by 29.3% and ED1 positive area by 32.3% at 14 days after injury.
     【Conclision】The present study showed thatβ-aescin treatment strikingly reduced lipid peroxydation (LPO), suppressed microglia activation and hematogenous leukocyte infiltration, and facilitated reparation after SCI.
引文
[1] Carlson GD, Gorden C. Current developments in spinal cord injury research. Spine J 2002; 2(2):116-28.
    [2] Rossignol S, Schwab M, Schwartz M, Fehlings MG. Spinal cord injury: time to move. J Neurosci 2007; 27(44):11782-92.
    [3] Hamann, K., Shi, R. (2009) Acrolein scavenging: a potential novel mechanism of attenuating oxidative stress following spinal cord injury. J Neurochem, 111,1348-56.
    [4] Montopoli M, Froldi G, Comelli MC, Prosdocimi M, Caparrotta L. Aescin protection of human vascular endothelial cells exposed to cobalt chloride mimicked hypoxia and inflammatory stimuli. Planta Med 2007; 73(3):285-8.
    [5] Sosne, G., Szliter, E.A., Barrett, R., Kernacki, K.A., Kleinman, H.& Hazlett, L.D. (2002) Thymosin beta 4 promotes corneal wound healing and decreases inflammation in vivo following alkali injury. Exp Eye Res, 74,293-9.
    [6] Bock-Marquette, I., Saxena, A., White, M.D., Dimaio, J.M.& Srivastava, D. (2004) Thymosin beta4 activates integrin-linked kinase and promotes cardiac cell migration, survival and cardiac repair. Nature, 432,466-72.
    [7] Bao F, Liu D. Hydroxyl radicals generated in the rat spinal cord at the level produced by impact injury induce cell death by necrosis and apoptosis: protection by a metalloporphyrin. Neuroscience 2004; 126(2):285-95.
    [8] Smart, N., Rossdeutsch, A.& Riley, P.R. (2007) Thymosin beta4 and angiogenesis: modes of action and therapeutic potential. Angiogenesis, 10,229-41.
    [9] Sosne, G., Qiu, P., Christopherson, P.L.& Wheater, M.K. (2007) Thymosin beta 4 suppression of corneal NFkappaB: a potential anti-inflammatory pathway. Exp Eye Res, 84,663-9.
    [10] Sirtori CR. Aescin: pharmacology, pharmacokinetics and therapeutic profile. Pharmacol Res, 2001,44(3):183-93.
    [11] Tator, C.H., Fehlings, M.G. (1991) Review of the secondary injury theory of acute spinal cord trauma with emphasis on vascular mechanisms. J Neurosurg, 75,15-26.
    [12] Mautes, A.E., Weinzierl, M.R., Donovan, F.& Noble, L.J. (2000) Vascular events after spinal cord injury: contribution to secondary pathogenesis. Phys Ther, 80,673-87.
    [13] Beattie MS, Hermann GE, Rogers RC, Bresnahan JC. Cell death in models of spinal cord injury. Prog Brain Res 2002; 137:37-47.
    [14] Profyris C, Cheema SS, Zang D, et al. Degenerative and regenerative mechanisms governing spinal cord injury. Neurobiol Dis, 2004,15(3):415-36.
    [15] Ferguson AR, Christensen RN, Gensel JC, et al. Cell death after spinal cord injury is exacerbated by rapid TNF alpha-induced trafficking of GluR2-lacking AMPARs to the plasma membrane. J Neurosci 2008; 28(44):11391-400.
    [16] McDonald JW, Sadowsky C. Spinal-cord injury. Lancet 2002; 359(9304):417-25.
    [17] Tator CH. Update on the pathophysiology and pathology of acute spinalcord injury. Brain Pathol 1995; 5(4):407-13.
    [18] Blight A. Mechanical factors in experimental spinal cord injury. J Am Paraplegia Soc 1988; 11(2):26-34.
    [19] Young W. Spinal cord contusion models. Prog Brain Res 2002; 137:231-55.
    [20] Maxwell WL. Histopathological changes at central nodes of Ranvier after stretch-injury. Microsc Res Tech 1996; 34(6):522-35.
    [21] Hall ED, Springer JE. Neuroprotection and acute spinal cord injury: a reappraisal. NeuroRx 2004; 1(1):80-100.
    [22] Springer JE, Rao RR, Lim HR, et al. The functional and neuroprotective actions of Neu2000, a dual-acting pharmacological agent, in the treatment of acute spinal cord injury. J Neurotrauma 2010; 27(1):139-49.
    [23] Guha A, Tator CH. Acute cardiovascular effects of experimental spinal cord injury. J Trauma 1988; 28(4):481-90.
    [24] Holtz A, Nystrom B, Gerdin B. Relation between spinal cord blood flow and functional recovery after blocking weight-induced spinal cord injury in rats. Neurosurgery 1990; 26(6):952-7.
    [25] Faden AI, Jacobs TP, Holaday JW. Opiate antagonist improves neurologic recovery after spinal injury. Science 1981; 211(4481):493-4.
    [26] Faden AI, Jacobs TP, Mougey E, Holaday JW. Endorphins in experimental spinal injury: therapeutic effect of naloxone. Ann Neurol 1981; 10(4):326-32.
    [27] Faden AI, Jacobs TP. Opiate antagonist WIN44,441-3 stereospecifically improves neurologic recovery after ischemic spinal injury. Neurology 1985; 35(9):1311-5.
    [28] Anthes DL, Theriault E, Tator CH. Ultrastructural evidence for arteriolarvasospasm after spinal cord trauma. Neurosurgery 1996; 39(4):804-14.
    [29] Koyanagi I, Tator CH, Theriault E. Silicone rubber microangiography of acute spinal cord injury in the rat. Neurosurgery 1993; 32(2):260-8; discussion 268.
    [30] Moriya T, Hassan AZ, Young W, Chesler M. Dynamics of extracellular calcium activity following contusion of the rat spinal cord. J Neurotrauma 1994; 11(3):255-63.
    [31] Young W. Role of calcium in central nervous system injuries. J Neurotrauma 1992; 9 Suppl 1:S9-25.
    [32] Braughler JM, Duncan LA, Goodman T. Calcium enhances in vitro free radical-induced damage to brain synaptosomes, mitochondria, and cultured spinal cord neurons. J Neurochem 1985; 45(4):1288-93.
    [33] Beck KD, Nguyen HX, Galvan MD, Salazar DL, Woodruff TM, Anderson AJ. Quantitative analysis of cellular inflammation after traumatic spinal cord injury: evidence for a multiphasic inflammatory response in the acute to chronic environment. Brain 2010; 133(Pt 2):433-47.
    [34] Popovich PG, Wei P, Stokes BT. Cellular inflammatory response after spinal cord injury in Sprague-Dawley and Lewis rats. J Comp Neurol 1997; 377(3):443-64.
    [35] Bethea JR, Dietrich WD. Targeting the host inflammatory response in traumatic spinal cord injury. Curr Opin Neurol 2002; 15(3):355-60.
    [36] Bethea, J.R. (2000) Spinal cord injury-induced inflammation: a dual-edged sword. Prog Brain Res, 128,33-42.
    [37] Popovich PG, Stuckman S, Gienapp IE, Whitacre CC. Alterations in immune cell phenotype and function after experimental spinal cordinjury. J Neurotrauma 2001; 18(9):957-66.
    [38] Chan CC. Inflammation: beneficial or detrimental after spinal cord injury. Recent Pat CNS Drug Discov 2008; 3(3):189-99.
    [39] Ankeny DP, Popovich PG. Mechanisms and implications of adaptive immune responses after traumatic spinal cord injury. Neuroscience 2009; 158(3):1112-21.
    [40] Popovich PG, Guan Z, Wei P, et al. Depletion of hematogenous macrophages promotes partial hindlimb recovery and neuroanatomical repair after experimental spinal cord injury. Exp Neurol, 1999,158(2):351-65.
    [41] Gonzalez R, Glaser J, Liu MT, et al. Reducing inflammation decreases secondary degeneration and functional deficit after spinal cord injury. Exp Neurol, 2003,184(1):456-63.
    [42] Hauben E, Nevo U, Yoles E, et al. Autoimmune T cells as potential neuroprotective therapy for spinal cord injury. Lancet 2000; 355(9200):286-7.
    [43] Hohlfeld R, Kerschensteiner M, Stadelmann C, Lassmann H, Wekerle H. The neuroprotective effect of inflammation: implications for the therapy of multiple sclerosis. J Neuroimmunol 2000; 107(2):161-6.
    [44] Schwartz M. Macrophages and microglia in central nervous system injury: are they helpful or harmful. J Cereb Blood Flow Metab 2003; 23(4):385-94.
    [45] Crutcher KA, Gendelman HE, Kipnis J, et al. Debate: "is increasing neuroinflammation beneficial for neural repair?". J Neuroimmune Pharmacol 2006; 1(3):195-211.
    [46] Yin Y, Henzl MT, Lorber B, et al. Oncomodulin is a macrophage-derivedsignal for axon regeneration in retinal ganglion cells. Nat Neurosci 2006; 9(6):843-52.
    [47] Hendrix S, Nitsch R. The role of T helper cells in neuroprotection and regeneration. J Neuroimmunol 2007; 184(1-2):100-12.
    [48] Donnelly DJ, Popovich PG. Inflammation and its role in neuroprotection, axonal regeneration and functional recovery after spinal cord injury. Exp Neurol, 2008,209(2):378-88.
    [49] Dusart I, Schwab ME. Secondary cell death and the inflammatory reaction after dorsal hemisection of the rat spinal cord. Eur J Neurosci 1994; 6(5):712-24.
    [50] Schwartz M, Butovsky O, Bruck W, Hanisch UK. Microglial phenotype: is the commitment reversible. Trends Neurosci 2006; 29(2):68-74.
    [51] Hashimoto M, Nitta A, Fukumitsu H, Nomoto H, Shen L, Furukawa S. Inflammation-induced GDNF improves locomotor function after spinal cord injury. Neuroreport 2005; 16(2):99-102.
    [52] Schwartz M. Sell Memorial Lecture. Helping the body to cure itself: immune modulation by therapeutic vaccination for spinal cord injury. J Spinal Cord Med 2003; 26 Suppl 1:S6-10.
    [53] McTigue DM, Popovich PG, Jakeman LB, Stokes BT. Strategies for spinal cord injury repair. Prog Brain Res 2000; 128:3-8.
    [54] Popovich PG. Immunological regulation of neuronal degeneration and regeneration in the injured spinal cord. Prog Brain Res 2000; 128:43-58.
    [55] Yang L, Blumbergs PC, Jones NR, Manavis J, Sarvestani GT, Ghabriel MN. Early expression and cellular localization of proinflammatory cytokines interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha in human traumatic spinal cord injury. Spine (Phila Pa 1976)2004; 29(9):966-71.
    [56] Carlson SL, Parrish ME, Springer JE, Doty K, Dossett L. Acute inflammatory response in spinal cord following impact injury. Exp Neurol 1998; 151(1):77-88.
    [57] Town T, Nikolic V, Tan J. The microglial "activation" continuum: from innate to adaptive responses. J Neuroinflammation 2005; 2:24.
    [58] Jones LL, Tuszynski MH. Spinal cord injury elicits expression of keratan sulfate proteoglycans by macrophages, reactive microglia, and oligodendrocyte progenitors. J Neurosci 2002; 22(11):4611-24.
    [59] Trivedi A, Olivas AD, Noble-Haeusslein LJ. Inflammation and Spinal Cord Injury: Infiltrating Leukocytes as Determinants of Injury and Repair Processes. Clin Neurosci Res 2006; 6(5):283-292.
    [60] Taoka Y, Okajima K. Spinal cord injury in the rat. Prog Neurobiol 1998; 56(3):341-58.
    [61] Noble LJ, Donovan F, Igarashi T, Goussev S, Werb Z. Matrix metalloproteinases limit functional recovery after spinal cord injury by modulation of early vascular events. J Neurosci 2002; 22(17):7526-35.
    [62] Bao F, Chen Y, Dekaban GA, Weaver LC. Early anti-inflammatory treatment reduces lipid peroxidation and protein nitration after spinal cord injury in rats. J Neurochem 2004; 88(6):1335-44.
    [63] Hamada Y, Ikata T, Katoh S, et al. Involvement of an intercellular adhesion molecule 1-dependent pathway in the pathogenesis of secondary changes after spinal cord injury in rats. J Neurochem 1996; 66(4):1525-31.
    [64] Kigerl KA, McGaughy VM, Popovich PG. Comparative analysis of lesion development and intraspinal inflammation in four strains of micefollowing spinal contusion injury. J Comp Neurol 2006; 494(4):578-94.
    [65] McDonald, J.W., Belegu, V. (2006) Demyelination and remyelination after spinal cord injury. J Neurotrauma, 23,345-59.
    [66] Blight AR. Delayed demyelination and macrophage invasion: a candidate for secondary cell damage in spinal cord injury. Cent Nerv Syst Trauma 1985; 2(4):299-315.
    [67] Boyles JK, Zoellner CD, Anderson LJ, et al. A role for apolipoprotein E, apolipoprotein A-I, and low density lipoprotein receptors in cholesterol transport during regeneration and remyelination of the rat sciatic nerve. J Clin Invest 1989; 83(3):1015-31.
    [68] Blight AR. Macrophages and inflammatory damage in spinal cord injury. J Neurotrauma 1992; 9 Suppl 1:S83-91.
    [69] Blight AR. Effects of silica on the outcome from experimental spinal cord injury: implication of macrophages in secondary tissue damage. Neuroscience 1994; 60(1):263-73.
    [70] Popovich PG, Reinhard JF Jr, Flanagan EM, Stokes BT. Elevation of the neurotoxin quinolinic acid occurs following spinal cord trauma. Brain Res 1994; 633(1-2):348-52.
    [71] Minc-Golomb D, Yadid G, Tsarfaty I, Resau JH, Schwartz JP. In vivo expression of inducible nitric oxide synthase in cerebellar neurons. J Neurochem 1996; 66(4):1504-9.
    [72] Soares HD, Hicks RR, Smith D, McIntosh TK. Inflammatory leukocytic recruitment and diffuse neuronal degeneration are separate pathological processes resulting from traumatic brain injury. J Neurosci 1995; 15(12):8223-33.
    [73] Holmin S, Soderlund J, Biberfeld P, Mathiesen T. Intracerebralinflammation after human brain contusion. Neurosurgery 1998; 42(2):291-8; discussion 298-9.
    [74] Fleming JC, Norenberg MD, Ramsay DA, et al. The cellular inflammatory response in human spinal cords after injury. Brain 2006; 129(Pt 12):3249-69.
    [75] Nagamoto-Combs K, McNeal DW, Morecraft RJ, Combs CK. Prolonged microgliosis in the rhesus monkey central nervous system after traumatic brain injury. J Neurotrauma 2007; 24(11):1719-42.
    [76] Kigerl KA, Gensel JC, Ankeny DP, Alexander JK, Donnelly DJ, Popovich PG. Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J Neurosci 2009; 29(43):13435-44.
    [77] Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity 2010; 32(5):593-604.
    [78] Popovich PG, Stuckman S, Gienapp IE, Whitacre CC. Alterations in immune cell phenotype and function after experimental spinal cord injury. J Neurotrauma 2001; 18(9):957-66.
    [79] Schnell L, Schneider R, Berman MA, Perry VH, Schwab ME. Lymphocyte recruitment following spinal cord injury in mice is altered by prior viral exposure. Eur J Neurosci 1997; 9(5):1000-7.
    [80] Lyons JA, San M, Happ MP, Cross AH. B cells are critical to induction of experimental allergic encephalomyelitis by protein but not by a short encephalitogenic peptide. Eur J Immunol 1999; 29(11):3432-9.
    [81] Raine CS, Cannella B, Hauser SL, Genain CP. Demyelination in primate autoimmune encephalomyelitis and acute multiple sclerosis lesions: a case for antigen-specific antibody mediation. Ann Neurol 1999;46(2):144-60.
    [82] Chavarria A, Alcocer-Varela J. Is damage in central nervous system due to inflammation. Autoimmun Rev 2004; 3(4):251-60.
    [83] Upender MB, Naegele JR. Activation of microglia during developmentally regulated cell death in the cerebral cortex. Dev Neurosci 1999; 21(6):491-505.
    [84] Yasojima K, Schwab C, McGeer EG, McGeer PL. Up-regulated production and activation of the complement system in Alzheimer's disease brain. Am J Pathol 1999; 154(3):927-36.
    [85] Chouchakova N, Skokowa J, Baumann U, et al. Fc gamma RIII-mediated production of TNF-alpha induces immune complex alveolitis independently of CXC chemokine generation. J Immunol 2001; 166(8):5193-200.
    [86] Abdul-Majid KB, Stefferl A, Bourquin C, et al. Fc receptors are critical for autoimmune inflammatory damage to the central nervous system in experimental autoimmune encephalomyelitis. Scand J Immunol 2002; 55(1):70-81.
    [87] Schwab C, McGeer PL. Complement activated C4d immunoreactive oligodendrocytes delineate small cortical plaques in multiple sclerosis. Exp Neurol 2002; 174(1):81-8.
    [88] Rodriguez M, Lennon VA. Immunoglobulins promote remyelination in the central nervous system. Ann Neurol 1990; 27(1):12-7.
    [89] Schnell L, Schwab ME. Axonal regeneration in the rat spinal cord produced by an antibody against myelin-associated neurite growth inhibitors. Nature 1990; 343(6255):269-72.
    [90] Huang DW, McKerracher L, Braun PE, David S. A therapeutic vaccineapproach to stimulate axon regeneration in the adult mammalian spinal cord. Neuron 1999; 24(3):639-47.
    [91] Mizrachi Y, Ohry A, Aviel A, Rozin R, Brooks ME, Schwartz M. Systemic humoral factors participating in the course of spinal cord injury. Paraplegia 1983; 21(5):287-93.
    [92] Hayes KC, Hull TC, Delaney GA, et al. Elevated serum titers of proinflammatory cytokines and CNS autoantibodies in patients with chronic spinal cord injury. J Neurotrauma 2002; 19(6):753-61.
    [93] Hayes KC, Hull TC, Delaney GA, et al. Elevated serum titers of proinflammatory cytokines and CNS autoantibodies in patients with chronic spinal cord injury. J Neurotrauma 2002; 19(6):753-61.
    [94] Ankeny DP, Popovich PG. B cells and autoantibodies: complex roles in CNS injury. Trends Immunol 2010; 31(9):332-8.
    [95] Ankeny DP, Lucin KM, Sanders VM, McGaughy VM, Popovich PG. Spinal cord injury triggers systemic autoimmunity: evidence for chronic B lymphocyte activation and lupus-like autoantibody synthesis. J Neurochem 2006; 99(4):1073-87.
    [96] Ankeny DP, Guan Z, Popovich PG. B cells produce pathogenic antibodies and impair recovery after spinal cord injury in mice. J Clin Invest 2009; 119(10):2990-9.
    [97] Sroga JM, Jones TB, Kigerl KA, McGaughy VM, Popovich PG. Rats and mice exhibit distinct inflammatory reactions after spinal cord injury. J Comp Neurol 2003; 462(2):223-40.
    [98] Popovich PG, Stokes BT, Whitacre CC. Concept of autoimmunity following spinal cord injury: possible roles for T lymphocytes in the traumatized central nervous system. J Neurosci Res 1996; 45(4):349-63.
    [99] Velardo MJ, Burger C, Williams PR, et al. Patterns of gene expression reveal a temporally orchestrated wound healing response in the injured spinal cord. J Neurosci 2004; 24(39):8562-76.
    [100] Hauben E, Schwartz M. Therapeutic vaccination for spinal cord injury: helping the body to cure itself. Trends Pharmacol Sci 2003; 24(1):7-12.
    [101] Jones TB, Hart RP, Popovich PG. Molecular control of physiological and pathological T-cell recruitment after mouse spinal cord injury. J Neurosci 2005; 25(28):6576-83.
    [102] Crutcher KA, Gendelman HE, Kipnis J, et al. Debate: "is increasing neuroinflammation beneficial for neural repair?". J Neuroimmune Pharmacol 2006; 1(3):195-211.
    [103] Fee D, Crumbaugh A, Jacques T, et al. Activated/effector CD4+ T cells exacerbate acute damage in the central nervous system following traumatic injury. J Neuroimmunol 2003; 136(1-2):54-66.
    [104] Michaelis EK. Molecular biology of glutamate receptors in the central nervous system and their role in excitotoxicity, oxidative stress and aging. Prog Neurobiol 1998; 54(4):369-415.
    [105] Traynelis SF, Wollmuth LP, McBain CJ, et al. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev 2010; 62(3):405-96.
    [106] Dingledine R, Borges K, Bowie D, Traynelis SF. The glutamate receptor ion channels. Pharmacol Rev 1999; 51(1):7-61.
    [107] Gottlieb M, Matute C. Expression of ionotropic glutamate receptor subunits in glial cells of the hippocampal CA1 area following transient forebrain ischemia. J Cereb Blood Flow Metab 1997; 17(3):290-300.
    [108] Noda M, Nakanishi H, Nabekura J, Akaike N. AMPA-kainate subtypesof glutamate receptor in rat cerebral microglia. J Neurosci 2000; 20(1):251-8.
    [109] Steinhauser C, Gallo V. News on glutamate receptors in glial cells. Trends Neurosci 1996; 19(8):339-45.
    [110] Panter SS, Yum SW, Faden AI. Alteration in extracellular amino acids after traumatic spinal cord injury. Ann Neurol 1990; 27(1):96-9.
    [111] Doble A. The role of excitotoxicity in neurodegenerative disease: implications for therapy. Pharmacol Ther 1999; 81(3):163-221.
    [112] Puchalski RB, Louis JC, Brose N, et al. Selective RNA editing and subunit assembly of native glutamate receptors. Neuron 1994; 13(1):131-47.
    [113] Burnashev N. Calcium permeability of glutamate-gated channels in the central nervous system. Curr Opin Neurobiol 1996; 6(3):311-7.
    [114] Matute C, Alberdi E, Domercq M, Perez-Cerda F, Perez-Samartin A, Sanchez-Gomez MV. The link between excitotoxic oligodendroglial death and demyelinating diseases. Trends Neurosci 2001; 24(4):224-30.
    [115] Didier M, Bursztajn S, Adamec E, et al. DNA strand breaks induced by sustained glutamate excitotoxicity in primary neuronal cultures. J Neurosci 1996; 16(7):2238-50.
    [116] Larm JA, Cheung NS, Beart PM. Apoptosis induced via AMPA-selective glutamate receptors in cultured murine cortical neurons. J Neurochem 1997; 69(2):617-22.
    [117] Qin ZH, Wang Y, Chase TN. Stimulation of N-methyl-D-aspartate receptors induces apoptosis in rat brain. Brain Res 1996; 725(2):166-76.
    [118] Portera-Cailliau C, Price DL, Martin LJ. Non-NMDA and NMDA receptor-mediated excitotoxic neuronal deaths in adult brain aremorphologically distinct: further evidence for an apoptosis-necrosis continuum. J Comp Neurol 1997; 378(1):88-104.
    [119] Morioka M, Hamada J, Ushio Y, Miyamoto E. Potential role of calcineurin for brain ischemia and traumatic injury. Prog Neurobiol 1999; 58(1):1-30.
    [120] Wang HG, Pathan N, Ethell IM, et al. Ca2+-induced apoptosis through calcineurin dephosphorylation of BAD. Science 1999; 284(5412):339-43.
    [121] Morioka M, Hamada J, Ushio Y, Miyamoto E. Potential role of calcineurin for brain ischemia and traumatic injury. Prog Neurobiol 1999; 58(1):1-30.
    [122] Ankarcrona M, Dypbukt JM, Orrenius S, Nicotera P. Calcineurin and mitochondrial function in glutamate-induced neuronal cell death. FEBS Lett 1996; 394(3):321-4.
    [123] Nagata S. Apoptosis by death factor. Cell 1997; 88(3):355-65.
    [124] Arends MJ, Wyllie AH. Apoptosis: mechanisms and roles in pathology. Int Rev Exp Pathol 1991; 32:223-54.
    [125] Majno G, Joris I. Apoptosis, oncosis, and necrosis. An overview of cell death. Am J Pathol 1995; 146(1):3-15.
    [126] Cohen GM. Caspases: the executioners of apoptosis. Biochem J 1997; 326 ( Pt 1):1-16.
    [127] Grutter MG. Caspases: key players in programmed cell death. Curr Opin Struct Biol 2000; 10(6):649-55.
    [128] Emery E, Aldana P, Bunge MB, et al. Apoptosis after traumatic human spinal cord injury. J Neurosurg 1998; 89(6):911-20.
    [129] Kwon, B.K., Tetzlaff, W., Grauer, J.N., Beiner, J.& Vaccaro, A.R. (2004)Pathophysiology and pharmacologic treatment of acute spinal cord injury. Spine J, 4,451-64.
    [130] Kuida K, Haydar TF, Kuan CY, et al. Reduced apoptosis and cytochrome c-mediated caspase activation in mice lacking caspase 9. Cell 1998; 94(3):325-37.
    [131] Hakem R, Hakem A, Duncan GS, et al. Differential requirement for caspase 9 in apoptotic pathways in vivo. Cell 1998; 94(3):339-52.
    [132] Slee EA, Harte MT, Kluck RM, et al. Ordering the cytochrome c-initiated caspase cascade: hierarchical activation of caspases-2, -3, -6, -7, -8, and -10 in a caspase-9-dependent manner. J Cell Biol 1999; 144(2):281-92.
    [133] Zou H, Henzel WJ, Liu X, Lutschg A, Wang X. Apaf-1, a human protein homologous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3. Cell 1997; 90(3):405-13.
    [134] Daugas E, Susin SA, Zamzami N, et al. Mitochondrio-nuclear translocation of AIF in apoptosis and necrosis. FASEB J 2000; 14(5):729-39.
    [135] Plesnila N, Zhu C, Culmsee C, Groger M, Moskowitz MA, Blomgren K. Nuclear translocation of apoptosis-inducing factor after focal cerebral ischemia. J Cereb Blood Flow Metab 2004; 24(4):458-66.
    [136] Susin SA, Lorenzo HK, Zamzami N, et al. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 1999; 397(6718):441-6.
    [137] Joza N, Susin SA, Daugas E, et al. Essential role of the mitochondrial apoptosis-inducing factor in programmed cell death. Nature 2001; 410(6828):549-54.
    [138] Beattie MS, Hermann GE, Rogers RC, Bresnahan JC. Cell death inmodels of spinal cord injury. Prog Brain Res 2002; 137:37-47.
    [139] Beattie MS, Farooqui AA, Bresnahan JC. Review of current evidence for apoptosis after spinal cord injury. J Neurotrauma 2000; 17(10):915-25.
    [140] Crowe MJ, Bresnahan JC, Shuman SL, Masters JN, Beattie MS. Apoptosis and delayed degeneration after spinal cord injury in rats and monkeys. Nat Med 1997; 3(1):73-6.
    [141] Pfeiffer, S.E., Warrington, A.E.& Bansal, R. (1993) The oligodendrocyte and its many cellular processes. Trends Cell Biol, 3,191-7.
    [142] Wu, B., Ren, X. (2009) Promoting axonal myelination for improving neurological recovery in spinal cord injury. J Neurotrauma, 26,1847-56.
    [143] Basso DM. Neuroanatomical substrates of functional recovery after experimental spinal cord injury: implications of basic science research for human spinal cord injury. Phys Ther 2000; 80(8):808-17.
    [144] Basso DM, Beattie MS, Bresnahan JC. Graded histological and locomotor outcomes after spinal cord contusion using the NYU weight-drop device versus transection. Exp Neurol 1996; 139(2):244-56.
    [145] Bunge RP, Puckett WR, Becerra JL, Marcillo A, Quencer RM. Observations on the pathology of human spinal cord injury. A review and classification of 22 new cases with details from a case of chronic cord compression with extensive focal demyelination. Adv Neurol 1993; 59:75-89.
    [146] Betz AL. Identification of hypoxanthine transport and xanthine oxidase activity in brain capillaries. J Neurochem 1985; 44(2):574-9.
    [147] Bredt DS, Snyder SH. Isolation of nitric oxide synthetase, a calmodulin-requiring enzyme. Proc Natl Acad Sci U S A 1990; 87(2):682-5.
    [148] Forstermann U, Schmidt HH, Pollock JS, et al. Isoforms of nitric oxide synthase. Characterization and purification from different cell types. Biochem Pharmacol 1991; 42(10):1849-57.
    [149] Wu W, Liuzzi FJ, Schinco FP, et al. Neuronal nitric oxide synthase is induced in spinal neurons by traumatic injury. Neuroscience 1994; 61(4):719-26.
    [150] Paterniti I, Genovese T, Crisafulli C, et al. Treatment with green tea extract attenuates secondary inflammatory response in an experimental model of spinal cord trauma. Naunyn Schmiedebergs Arch Pharmacol, 2009,380(2):179-92.
    [151] Jiang H, Ren Y, Zhao J, Feng J. Parkin protects human dopaminergic neuroblastoma cells against dopamine-induced apoptosis. Hum Mol Genet 2004; 13(16):1745-54.
    [152] Choi J, Forster MJ, McDonald SR, Weintraub ST, Carroll CA, Gracy RW. Proteomic identification of specific oxidized proteins in ApoE-knockout mice: relevance to Alzheimer's disease. Free Radic Biol Med 2004; 36(9):1155-62.
    [153] Yune TY, Chang MJ, Kim SJ, et al. Increased production of tumor necrosis factor-alpha induces apoptosis after traumatic spinal cord injury in rats. J Neurotrauma 2003; 20(2):207-19.
    [154] Szabo C. Multiple pathways of peroxynitrite cytotoxicity. Toxicol Lett 2003; 140-141:105-12.
    [155] Lucas JH, Wheeler DG, Guan Z, et al. Effect of glutathione augmentation on lipid peroxidation after spinal cord injury. J Neurotrauma, 2002,19(6):763-75.
    [156] Halliwell B, Chirico S. Lipid peroxidation: its mechanism, measurement,and significance. Am J Clin Nutr, 1993,57(5 Suppl):715S-724S; discussion 724S-725S.
    [157] Esterbauer H, Schaur RJ, Zollner H. Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radic Biol Med, 1991,11(1):81-128.
    [158] Barut S, Canbolat A, Bilge T, et al. Lipid peroxidation in experimental spinal cord injury: time-level relationship. Neurosurg Rev, 1993,16(1):53-9.
    [159] Basso DM, Beattie MS, Bresnahan JC. A sensitive and reliable locomotor rating scale for open field testing in rats. J Neurotrauma 1995; 12(1):1-21.
    [160] Brown CJ, Mackinnon SE, Evans PJ, et al. Self-evaluation of walking-track measurement using a Sciatic Function Index. Microsurgery 1989; 10(3):226-35.
    [161] Graeber, M.B., Streit, W.J., Kiefer, R., Schoen, S.W.& Kreutzberg, G.W. (1990) New expression of myelomonocytic antigens by microglia and perivascular cells following lethal motor neuron injury. J Neuroimmunol, 27,121-32.
    [162] Sinescu, C., Popa, F., Grigorean, V.T., Onose, G., Sandu, A.M., Popescu, M., Burnei, G., Strambu, V.& Popa, C. (2010) Molecular basis of vascular events following spinal cord injury. J Med Life, 3,254-61.
    [163] Koyanagi, I., Tator, C.H.& Theriault, E. (1993) Silicone rubber microangiography of acute spinal cord injury in the rat. Neurosurgery, 32,260-8; discussion 268.
    [164] Knoblach, S.M., Faden, A.I. (1998) Interleukin-10 improves outcome and alters proinflammatory cytokine expression after experimentaland promoting gastrointestinal transit to attenuate formation of postoperative adhesions. World J Surg 2005; 29(12):1614-20; discussion 1621-2.
    [172] Wang T, Fu F, Zhang L, Han B, Zhu M, Zhang X. Effects of escin on acute inflammation and the immune system in mice. Pharmacol Rep 2009; 61(4):697-704.
    [173] Pittler MH, Ernst E. Horse chestnut seed extract for chronic venous insufficiency. Cochrane Database Syst Rev 2004; (2):CD003230.
    [174] Longiave D, Omini C, Nicosia S, Berti F. The mode of action of aescin on isolated veins: relationship with PGF2 alpha. Pharmacol Res Commun 1978; 10(2):145-52.
    [175] Kucukkurt I, Ince S, Keles H, et al. Beneficial effects of Aesculus hippocastanum L. seed extract on the body's own antioxidant defense system on subacute administration. J Ethnopharmacol 2010; 129(1):18-22.
    [176] Iwasa K, Ikata T, Fukuzawa K. Protective effect of vitamin E on spinal cord injury by compression and concurrent lipid peroxidation. Free Radic Biol Med, 1989,6(6):599-606.
    [177] Taoka Y, Okajima K, Uchiba M, et al. Activated protein C reduces the severity of compression-induced spinal cord injury in rats by inhibiting activation of leukocytes. J Neurosci, 1998,18(4):1393-8.
    [178] Bao F, Chen Y, Dekaban GA, et al. Early anti-inflammatory treatment reduces lipid peroxidation and protein nitration after spinal cord injury in rats. J Neurochem, 2004,88(6):1335-44.
    [179] Liu C, Shi Z, Fan L, et al. Resveratrol improves neuron protection and functional recovery in rat model of spinal cord injury. Brain Res, 2010.and promoting gastrointestinal transit to attenuate formation of postoperative adhesions. World J Surg 2005; 29(12):1614-20; discussion 1621-2.
    [172] Wang T, Fu F, Zhang L, Han B, Zhu M, Zhang X. Effects of escin on acute inflammation and the immune system in mice. Pharmacol Rep 2009; 61(4):697-704.
    [173] Pittler MH, Ernst E. Horse chestnut seed extract for chronic venous insufficiency. Cochrane Database Syst Rev 2004; (2):CD003230.
    [174] Longiave D, Omini C, Nicosia S, Berti F. The mode of action of aescin on isolated veins: relationship with PGF2 alpha. Pharmacol Res Commun 1978; 10(2):145-52.
    [175] Kucukkurt I, Ince S, Keles H, et al. Beneficial effects of Aesculus hippocastanum L. seed extract on the body's own antioxidant defense system on subacute administration. J Ethnopharmacol 2010; 129(1):18-22.
    [176] Iwasa K, Ikata T, Fukuzawa K. Protective effect of vitamin E on spinal cord injury by compression and concurrent lipid peroxidation. Free Radic Biol Med, 1989,6(6):599-606.
    [177] Taoka Y, Okajima K, Uchiba M, et al. Activated protein C reduces the severity of compression-induced spinal cord injury in rats by inhibiting activation of leukocytes. J Neurosci, 1998,18(4):1393-8.
    [178] Bao F, Chen Y, Dekaban GA, et al. Early anti-inflammatory treatment reduces lipid peroxidation and protein nitration after spinal cord injury in rats. J Neurochem, 2004,88(6):1335-44.
    [179] Liu C, Shi Z, Fan L, et al. Resveratrol improves neuron protection and functional recovery in rat model of spinal cord injury. Brain Res, 2010.
    [180] Hakan T, Toklu HZ, Biber N, et al. Meloxicam exerts neuroprotection on spinal cord trauma in rats. Int J Neurosci, 2011,121(3):142-8.
    [181] Facino RM, Carini M, Stefani R, Aldini G, Saibene L. Anti-elastase and anti-hyaluronidase activities of saponins and sapogenins from Hedera helix, Aesculus hippocastanum, and Ruscus aculeatus: factors contributing to their efficacy in the treatment of venous insufficiency. Arch Pharm (Weinheim) 1995; 328(10):720-4.
    [182] Sirtori CR. Aescin: pharmacology, pharmacokinetics and therapeutic profile. Pharmacol Res 2001; 44(3):183-93.
    [183] Li X, Chen GP, Li L, Wang KJ, Zhang BQ, Hu SJ. Dual effects of sodium aescinate on vascular tension in rat thoracic aorta. Microvasc Res 2010; 79(1):63-9.
    [184] Bielanski TE, Piotrowski ZH. Horse-chestnut seed extract for chronic venous insufficiency. J Fam Pract 1999; 48(3):171-2.

© 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