全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Neuroprotection in Stroke: Past, Present, and Future

DOI: 10.1155/2014/515716

Full-Text   Cite this paper   Add to My Lib

Abstract:

Stroke is a devastating medical condition, killing millions of people each year and causing serious injury to many more. Despite advances in treatment, there is still little that can be done to prevent stroke-related brain damage. The concept of neuroprotection is a source of considerable interest in the search for novel therapies that have the potential to preserve brain tissue and improve overall outcome. Key points of intervention have been identified in many of the processes that are the source of damage to the brain after stroke, and numerous treatment strategies designed to exploit them have been developed. In this review, potential targets of neuroprotection in stroke are discussed, as well as the various treatments that have been targeted against them. In addition, a summary of recent progress in clinical trials of neuroprotective agents in stroke is provided. 1. Introduction Stroke is one of the leading causes of death and disability worldwide. Despite decades of research, however, treatment options remain limited. In ischemic stroke, the primary focus of treatment is reperfusion. Currently, the only drug approved for the treatment of ischemic stroke is recombinant tissue plasminogen activator (rtPA, alteplase), which has a limited time window for administration and increases the risk for subsequent hemorrhage. Consequently, only a small percentage of patients receive rtPA treatment [1]. While this treatment is effective in opening up occluded cerebral vessels in some patients and can lead to improved outcomes after ischemic stroke, there are currently no approved treatments for the myriad of damaging pathological processes that persist in the brain long after the acute stage. These include the processes of inflammation, excitotoxicity, oxidative stress, apoptosis, and edema resulting from disruption of the blood-brain barrier [2]. In hemorrhagic stroke, additional processes include physical damage from the mass of accumulated blood itself, cytotoxicity of blood components, and vasospasm in subarachnoid hemorrhage [3, 4]. A considerable amount of research has been invested into the development of novel treatments capable of protecting the brain from damage following stroke, with limited success. Numerous neuroprotective treatments have been identified that show great promise in animal models of stroke. Unfortunately, nearly all have failed to provide protection in human trials. The purpose of this review is to provide an overview of targets for neuroprotection in stroke and examples of current research on potential neuroprotective treatments.

References

[1]  S. D. Reed, S. C. Cramer, D. K. Blough, K. Meyer, and J. G. Jarvik, “Treatment with tissue plasminogen activator and inpatient mortality rates for patients with ischemic stroke treated in community hospitals,” Stroke, vol. 32, no. 8, pp. 1832–1839, 2001.
[2]  M. A. Moskowitz, E. H. Lo, and C. Iadecola, “The science of stroke: mechanisms in search of treatments,” Neuron, vol. 67, no. 2, pp. 181–198, 2010.
[3]  J. Aronowski and X. Zhao, “Molecular pathophysiology of cerebral hemorrhage: secondary brain injury,” Stroke, vol. 42, no. 6, pp. 1781–1786, 2011.
[4]  D. Zemke, M. U. Farooq, A. M. Yahia, and A. Majid, “Delayed ischemia after subarachnoid hemorrhage: result of vasospasm alone or a broader vasculopathy?” Vascular Medicine, vol. 12, no. 3, pp. 243–249, 2007.
[5]  J. N. Stankowski and R. Gupta, “Therapeutic targets for neuroprotection in acute ischemic stroke: lost in translation?” Antioxidants & Redox Signaling, vol. 14, no. 10, pp. 1841–1851, 2011.
[6]  A. R. Noorian, R. G. Nogueira, and R. Gupta, “Neuroprotection in acute ischemic stroke,” Journal of Neurosurgical Sciences, vol. 55, no. 2, pp. 127–138, 2011.
[7]  D. T. Laskowitz and B. J. Kolls, “Neuroprotection in subarachnoid hemorrhage,” Stroke, vol. 41, supplement 10, pp. S79–S84, 2010.
[8]  C. P. Kellner and E. S. Connolly, “Neuroprotective strategies for intracerebral hemorrhage: trials and translation,” Stroke, vol. 41, supplement 10, pp. S99–S102, 2010.
[9]  R. Liu, H. Yuan, F. Yuan, and S.-H. Yang, “Neuroprotection targeting ischemic penumbra and beyond for the treatment of ischemic stroke,” Neurological Research, vol. 34, no. 4, pp. 331–337, 2012.
[10]  R. J. Traystman, “Animal models of focal and global cerebral ischemia,” ILAR Journal, vol. 44, no. 2, pp. 85–95, 2003.
[11]  A. Manaenko, H. Chen, J. H. Zhang, and J. Tang, “Comparison of different preclinical models of intracerebral hemorrhage,” Acta Neurochirurgica, no. 111, pp. 9–14, 2011.
[12]  E. Titova, R. P. Ostrowski, J. H. Zhang, and J. Tang, “Experimental models of subarachnoid hemorrhage for studies of cerebral vasospasm,” Neurological Research, vol. 31, no. 6, pp. 568–581, 2009.
[13]  P. R. Krafft, E. L. Bailey, T. Lekic, et al., “Etiology of stroke and choice of models,” International Journal of Stroke, vol. 7, no. 5, pp. 398–406, 2012.
[14]  R. Jin, G. Yang, and G. Li, “Inflammatory mechanisms in ischemic stroke: role of inflammatory cells,” Journal of Leukocyte Biology, vol. 87, no. 5, pp. 779–789, 2010.
[15]  S. E. Lakhan, A. Kirchgessner, and M. Hofer, “Inflammatory mechanisms in ischemic stroke: therapeutic approaches,” Journal of Translational Medicine, vol. 7, article 97, 2009.
[16]  G. J. del Zoppo, K. J. Becker, and J. M. Hallenbeck, “Inflammation after stroke: is it harmful?” Archives of Neurology, vol. 58, no. 4, pp. 669–672, 2001.
[17]  A. Jablonska and B. Lukomska, “Stroke induced brain changes: implications for stem cell transplantation,” Acta Neurobiologiae Experimentalis, vol. 71, no. 1, pp. 74–85, 2011.
[18]  J. M. Pradillo, A. Denes, A. D. Greenhalgh, et al., “Delayed administration of interleukin-1 receptor antagonist reduces ischemic brain damage and inflammation in comorbid rats,” Journal of Cerebral Blood Flow and Metabolism, vol. 32, no. 9, pp. 1810–1819, 2012.
[19]  J. S. Yoon, J.-H. Lee, and D. Tweedie, “3, 6′-dithiothalidomide improves experimental stroke outcome by suppressing neuroinflammation,” Journal of Neuroscience Research, vol. 91, no. 5, pp. 671–680, 2013.
[20]  S. Wang, H. Guo, L. Hu, et al., “Caffeic acid ester fraction from Erigeron breviscapus inhibits microglial activation and provides neuroprotection,” Chinese Journal of Integrative Medicine, vol. 18, no. 6, pp. 437–444, 2012.
[21]  T. Ewen, L. Qiuting, T. Chaogang, et al., “Neuroprotective effect of atorvastatin involves suppression of TNF-α and upregulation of IL-10 in a rat model of intracerebral hemorrhage,” Cell Biochemistry and Biophysics, vol. 66, no. 2, pp. 337–346, 2013.
[22]  R. K. Sumbria, R. J. Boado, and W. M. Pardridge, “Brain protection from stroke with intravenous TNFα decoy receptor-Trojan horse fusion protein,” Journal of Cerebral Blood Flow and Metabolism, vol. 32, no. 10, pp. 1933–1938, 2012.
[23]  P. Zhang, X. Liu, Y. Zhu, S. Chen, D. Zhou, and Y. Wang, “Honokiol inhibits the inflammatory reaction during cerebral ischemia reperfusion by suppressing NF-κB activation and cytokine production of glial cells,” Neuroscience Letters, vol. 534, pp. 123–127, 2013.
[24]  H. Luan, Z. Kan, Y. Xu, C. Lv, and W. Jiang, “Rosmarinic acid protects against experimental diabetes with cerebral ischemia: relation to inflammation response,” Journal of Neuroinflammation, vol. 10, article 28, 2013.
[25]  T. Jiang, L. Gao, J. Guo, J. Lu, Y. Wang, and Y. Zhang, “Suppressing inflammation by inhibiting the NF-κB pathway contributes to the neuroprotective effect of angiotensin-(1-7) in rats with permanent cerebral ischaemia,” British Journal of Pharmacology, vol. 167, no. 7, pp. 1520–1532, 2012.
[26]  L. Yu, C. Chen, L.-F. Wang, et al., “Neuroprotective effect of kaempferol glycosides against brain injury and neuroinflammation by inhibiting the activation of NF-κB and STAT3 in transient focal stroke,” PLoS ONE, vol. 8, no. 2, Article ID e55839, 2013.
[27]  A. Lanzillotta, G. Pignataro, C. Branca, et al., “Targeted acetylation of NF-κB/RelA and histones by epigenetic drugs reduces post-ischemic brain injury in mice with an extended therapeutic window,” Neurobiology of Disease, vol. 49, pp. 177–189, 2012.
[28]  D. Michalski, M. Heindl, J. Kacza, et al., “Spatio-temporal course of macrophage-like cell accumulation after experimental embolic stroke depending on treatment with tissue plasminogen activator and its combination with hyperbaric oxygenation,” European Journal of Histochemistry, vol. 56, no. 2, artivle e14, 2012.
[29]  J.-S. Park, J. A. Shin, J.-S. Jung et al., “Anti-inflammatory mechanism of compound K in activated microglia and its neuroprotective effect on experimental stroke in mice,” Journal of Pharmacology and Experimental Therapeutics, vol. 341, no. 1, pp. 59–67, 2012.
[30]  M. Ohnishi, A. Monda, R. Takemoto, et al., “Sesamin suppresses activation of microglia and p44/42 MAPK pathway, which confers neuroprotection in rat intracerebral hemorrhage,” Neuroscience, vol. 232, pp. 45–52, 2012.
[31]  H. Matsushita, M. Hijioka, A. Hisatsune, Y. Isohama, K. Shudo, and H. Katsuki, “Natural and synthetic retinoids afford therapeutic effects on intracerebral hemorrhage in mice,” European Journal of Pharmacology, vol. 683, no. 1–3, pp. 125–131, 2012.
[32]  F. J. Ortega, J. Jolkkonen, N. Mahy, and M. J. Rodríguez, “Glibenclamide enhances neurogenesis and improves long-term functional recovery after transient focal cerebral ischemia,” Journal of Cerebral Blood Flow and Metabolism, vol. 33, no. 3, pp. 356–364, 2013.
[33]  F. J. Ortega, J. Gimeno-Bayon, J. F. Espinosa-Parrilla et al., “ATP-dependent potassium channel blockade strengthens microglial neuroprotection after hypoxia-ischemia in rats,” Experimental Neurology, vol. 235, no. 1, pp. 282–296, 2012.
[34]  B. Wali, T. Ishrat, F. Atif, F. Hua, D. G. Stein, and I. Sayeed, “Glibenclamide administration attenuates infarct volume, hemispheric swelling, and functional impairments following permanent focal cerebral ischemia in rats,” Stroke Research and Treatment, vol. 2012, Article ID 460909, 6 pages, 2012.
[35]  E. Parada, J. Egea, I. Buendia, et al., “The microglial α7-acetylcholine nicotinic receptor is a key element in promoting neuroprotection by inducing heme oxygenase-1 via nuclear factor erythroid-2-related factor 2,” Antioxidants & Redox Signaling, vol. 19, no. 11, pp. 1135–1148, 2013.
[36]  H. Pradeep, J. B. Diya, S. Shashikumar, and G. K. Rajanikant, “Oxidative stress—assassin behind the ischemic stroke,” Folia Neuropathologica, vol. 50, no. 3, pp. 219–230, 2012.
[37]  G. H. Heeba and A. A. El-Hanafy, “Nebivolol regulates eNOS and iNOS expressions and alleviates oxidative stress in cerebral ischemia/reperfusion injury in rats,” Life Sciences, vol. 90, no. 11-12, pp. 388–395, 2012.
[38]  K. A. Radermacher, K. Wingler, F. Langhauser, et al., “Neuroprotection after stroke by targeting NOX4 as a source of oxidative stress,” Antioxidants & Redox Signaling, vol. 18, no. 12, pp. 1418–1427, 2013.
[39]  G. Li, H.-K. Luo, L.-F. Li, et al., “Dual effects of hydrogen sulphide on focal cerebral ischaemic injury via modulation of oxidative stress-induced apoptosis,” Clinical and Experimental Pharmacology & Physiology, vol. 39, no. 9, pp. 765–771, 2012.
[40]  J. Li, Y. Dong, H. Chen, et al., “Protective effects of hydrogen-rich saline in a rat model of permanent focal cerebral ischemia via reducing oxidative stress and inflammatory cytokines,” Brain Research, vol. 1486, pp. 103–111, 2012.
[41]  P. Y. Lam, N. Chen, P. Y. Chiu, H. Y. Leung, and K. M. Ko, “Neuroprotection against oxidative injury by a nucleic acid-based health product (Squina DNA) through enhancing mitochondrial antioxidant status and functional capacity,” Journal of Medicinal Food, vol. 15, no. 7, pp. 629–638, 2012.
[42]  H.-F. Huang, F. Guo, Y.-Z. Cao, W. Shi, and Q. Xia, “Neuroprotection by manganese superoxide dismutase (MnSOD) mimics: antioxidant effect and oxidative stress regulation in acute experimental stroke,” CNS Neuroscience & Therapeutics, vol. 18, no. 10, pp. 811–818, 2012.
[43]  A. Ahmad, M. M. Khan, S. S. Raza et al., “Ocimum sanctum attenuates oxidative damage and neurological deficits following focal cerebral ischemia/reperfusion injury in rats,” Neurological Sciences, vol. 33, no. 6, pp. 1239–1247, 2012.
[44]  Y. Zhan, C. Chen, H. Suzuki, Q. Hu, X. Zhi, and J. H. Zhang, “Hydrogen gas ameliorates oxidative stress in early brain injury after subarachnoid hemorrhage in rats,” Critical Care Medicine, vol. 40, no. 4, pp. 1291–1296, 2012.
[45]  B. J. Connell and T. M. Saleh, “Co-administration of apocynin with lipoic acid enhances neuroprotection in a rat model of ischemia/reperfusion,” Neuroscience Letters, vol. 507, no. 1, pp. 43–46, 2012.
[46]  J. Yang, H.-N. Ahn, M. Chang, P. Narasimhan, P. H. Chan, and Y. S. Song, “Complement component 3 inhibition by an antioxidant is neuroprotective after cerebral ischemia and reperfusion in mice,” Journal of Neurochemistry, vol. 124, no. 4, pp. 523–535, 2013.
[47]  X. Zeng, K. Asmaro, C. Ren et al., “Acute ethanol treatment reduces blood-brain barrier dysfunction following ischemia/reperfusion injury,” Brain Research, vol. 1437, pp. 127–133, 2012.
[48]  J. Xiang, R. Lan, Y.-P. Tang, Y.-P. Chen, and D.-F. Cai, “Apocynum venetum leaf extract attenuates disruption of the blood-brain barrier and upregulation of matrix metalloproteinase-9/-2 in a rat model of cerebral ischemia-reperfusion injury,” Neurochemical Research, vol. 37, no. 8, pp. 1820–1828, 2012.
[49]  D. Michalski, C. Hobohm, C. Weise, et al., “Interrelations between blood-brain barrier permeability and matrix metalloproteinases are differently affected by tissue plasminogen activator and hyperoxia in a rat model of embolic stroke,” Medical Gas Research, vol. 2, no. 1, article 2, 2012.
[50]  Z. Wang, Y. Xue, H. Jiao, Y. Liu, and P. Wang, “Doxycycline-mediated protective effect against focal cerebral ischemia-reperfusion injury through the modulation of tight junctions and PKCδ signaling in rats,” Journal of Molecular Neuroscience, vol. 47, no. 1, pp. 89–100, 2012.
[51]  H. Shi, B. Sheng, F. Zhang, et al., “Kruppel-like factor 2 protects against ischemic stroke by regulating endothelial blood brain barrier function,” American Journal of Physiology: Heart and Circulatory Physiology, vol. 304, no. 6, pp. H796–H805, 2013.
[52]  D. Chen, X. Wei, J. Guan, et al., “Inhibition of c-Jun N-terminal kinase prevents blood-brain barrier disruption and normalizes the expression of tight junction proteins clautin-5 and ZO-1 in a rat model of subarachnoid hemorrhage,” Acta Neurochirurgica, vol. 154, no. 8, pp. 1469–1476, 2012.
[53]  B. Huang, P. R. Krafft, Q. Ma et al., “Fibroblast growth factors preserve blood-brain barrier integrity through RhoA inhibition after intracerebral hemorrhage in mice,” Neurobiology of Disease, vol. 46, no. 1, pp. 204–214, 2012.
[54]  B. Lapergue, B. Q. Dang, J.-P. Desilles, et al., “High-density lipoprotein-based therapy reduces the hemorrhagic complications associated with tissue plasminogen activator treatment in experimental stroke,” Stroke, vol. 44, no. 3, pp. 699–707, 2013.
[55]  D. R. Pillai, N. C. Shanbhag, M. S. Dittmar, U. Bogdahn, and F. Schlachetzki, “Neurovascular protection by targeting early blood-brain barrier disruption with neurotrophic factors after ischemia-reperfusion in rats*,” Journal of Cerebral Blood Flow and Metabolism, vol. 33, no. 4, pp. 557–566, 2013.
[56]  A. Mdzinarishvili, R. Sumbria, D. Langc, and J. Klein, “Ginkgo extract EGb761 confers neuroprotection by reduction of glutamate release in ischemic brain,” Journal of Pharmacy & Pharmaceutical Sciences, vol. 15, no. 1, pp. 94–102, 2012.
[57]  S. E. Nada and Z. A. Shah, “Preconditioning with Ginkgo biloba (EGb 761) provides neuroprotection through HO1 and CRMP2,” Neurobiology of Disease, vol. 46, no. 1, pp. 180–189, 2012.
[58]  M. M. Harraz, S. M. Eacker, X. Wang, T. M. Dawson, and V. L. Dawson, “MicroRNA-223 is neuroprotective by targeting glutamate receptors,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, no. 46, pp. 2–5, 2012.
[59]  L. Li, W. Qu, L. Zhou, et al., “Activation of transient receptor potential vanilloid 4 increases NMDA-activated current in hippocampal pyramidal neurons,” Frontiers in Cellular Neuroscience, vol. 7, article 17, 2013.
[60]  D. Afshari, N. Moradian, and M. Rezaei, “Evaluation of the intravenous magnesium sulfate effect in clinical improvement of patients with acute ischemic stroke,” Clinical Neurology and Neurosurgery, vol. 115, no. 4, pp. 400–404, 2013.
[61]  K. R. Lees, K. W. Muir, I. Ford et al., “Magnesium for acute stroke (Intravenous Magnesium Efficacy in Stroke Trial): randomised controlled trial,” The Lancet, vol. 363, no. 9407, pp. 439–445, 2004.
[62]  J. L. Saver, C. Kidwell, M. Eckstein, and S. Starkman, “Prehospital neuroprotective therapy for acute stroke: results of the Field Administration of Stroke Therapy-Magnesium (FAST-MAG) pilot trial,” Stroke, vol. 35, no. 5, pp. e106–e108, 2004.
[63]  H. Li, J. Huang, W. Du, C. Jia, H. Yao, and Y. Wang, “TRPC6 inhibited NMDA receptor activities and protected neurons from ischemic excitotoxicity,” Journal of Neurochemistry, vol. 123, no. 6, pp. 1010–1018, 2012.
[64]  Y. Lin, J.-C. Zhang, J. Fu, et al., “Hyperforin attenuates brain damage induced by transient middle cerebral artery occlusion (MCAO) in rats via inhibition of TRPC6 channels degradation,” Journal of Cerebral Blood Flow and Metabolism, vol. 33, no. 2, pp. 253–262, 2013.
[65]  Y. Zhang, L. Zhou, X. Zhang, J. Bai, M. Shi, and G. Zhao, “Ginsenoside-Rd attenuates TRPM7 and ASIC1a but promotes ASIC2a expression in rats after focal cerebral ischemia,” Neurological Sciences, vol. 33, no. 5, pp. 1125–1131, 2012.
[66]  A. Majid, D. Zemke, and M. Kassab, “Pathophysiology of ischemic stroke,” in UpToDate, D. Basow, Ed., UpToDate, Waltham, Mass, USA, 2013.
[67]  T. Zgavc, D. de Geyter, A.-G. Ceulemans, et al., “Mild hypothermia reduces activated caspase-3 up to 1 week after a focal cerebral ischemia induced by endothelin-1 in rats,” Brain Research, vol. 1501, pp. 81–88, 2013.
[68]  T. Kinouchi, K. T. Kitazato, K. Shimada et al., “Activation of signal transducer and activator of transcription-3 by a peroxisome proliferator-activated receptor gamma agonist contributes to neuroprotection in the peri-infarct region after ischemia in oophorectomized rats,” Stroke, vol. 43, no. 2, pp. 478–483, 2012.
[69]  W.-Y. Lin, Y.-C. Chang, C.-J. Ho, and C.-C. Huang, “Ischemic preconditioning reduces neurovascular damage after hypoxia-ischemia via the cellular inhibitor of apoptosis 1 in neonatal brain,” Stroke, vol. 44, no. 1, pp. 162–169, 2013.
[70]  P. Fu, C. Peng, J. Y. Ding, et al., “Acute administration of ethanol reduces apoptosis following ischemic stroke in rats,” Neuroscience Research, vol. 76, no. 1-2, pp. 93–97, 2013.
[71]  Q. Zhao, C. Zhang, X. Wang, L. Chen, H. Ji, and Y. Zhang, “(S)-ZJM-289, a nitric oxide-releasing derivative of 3-n-butylphthalide, protects against ischemic neuronal injury by attenuating mitochondrial dysfunction and associated cell death,” Neurochemistry International, vol. 60, no. 2, pp. 134–144, 2012.
[72]  Z. Xie, B. Lei, Q. Huang, et al., “Neuroprotective effect of Cyclosporin A on the development of early brain injury in a subarachnoid hemorrhage model: a pilot study,” Brain Research, vol. 1472, pp. 113–123, 2012.
[73]  G. Hu, Z. Wu, F. Yang, et al., “Ginsenoside Rdblocks AIF mitochondrio-nuclear translocation and NF-κB nuclear accumulation by inhibiting poly(ADP-ribose) polymerase-1 after focal cerebral ischemia in rats,” Neurological Sciences, vol. 34, no. 12, pp. 2101–2106, 2013.
[74]  G. Wei, D.-F. Chen, X.-P. Lai, et al., “Muscone exerts neuroprotection in an experimental model of stroke via inhibition of the fas pathway,” Natural Product Communications, vol. 7, no. 8, pp. 1069–1074, 2012.
[75]  Y. Jiang, D.-W. Liu, X.-Y. Han et al., “Neuroprotective effects of anti-tumor necrosis factor-alpha antibody on apoptosis following subarachnoid hemorrhage in a rat model,” Journal of Clinical Neuroscience, vol. 19, no. 6, pp. 866–872, 2012.
[76]  B. Gabryel, A. Kost, and D. Kasprowska, “Neuronal autophagy in cerebral ischemia—a potential target for neuroprotective strategies?” Pharmacological Reports, vol. 64, no. 1, pp. 1–15, 2012.
[77]  D. Zemke, S. Azhar, and A. Majid, “The mTOR pathway as a potential target for the development of therapies against neurological disease,” Drug News and Perspectives, vol. 20, no. 8, pp. 495–499, 2007.
[78]  C.-H. Jing, L. Wang, P.-P. Liu, C. Wu, D. Ruan, and G. Chen, “Autophagy activation is associated with neuroprotection against apoptosis via a mitochondrial pathway in a rat model of subarachnoid hemorrhage,” Neuroscience, vol. 213, pp. 144–153, 2012.
[79]  M. Papadakis, G. Hadley, M. Xilouri, et al., “Tsc1 (hamartin) confers neuroprotection against ischemia by inducing autophagy,” Nature Medicine, vol. 19, no. 3, pp. 351–357, 2013.
[80]  L. Gao, T. Jiang, J. Guo, et al., “Inhibition of autophagy contributes to ischemic postconditioning-induced neuroprotection against focal cerebral ischemia in rats,” PLoS ONE, vol. 7, no. 9, 2012.
[81]  D. Zemke, J. L. Smith, M. J. Reeves, and A. Majid, “Ischemia and ischemic tolerance in the brain: an overview,” NeuroToxicology, vol. 25, no. 6, pp. 895–904, 2004.
[82]  J. Lehotsky, J. Burda, V. Danielisová, M. Gottlieb, P. Kaplán, and B. Saniová, “Ischemic tolerance: the mechanisms of neuroprotective strategy,” Anatomical Record, vol. 292, no. 12, pp. 2002–2012, 2009.
[83]  U. Dirnagl, R. P. Simon, and J. M. Hallenbeck, “Ischemic tolerance and endogenous neuroprotection,” Trends in Neurosciences, vol. 26, no. 5, pp. 248–254, 2003.
[84]  T. Kirino, “Ischemic tolerance,” Journal of Cerebral Blood Flow and Metabolism, vol. 22, no. 11, pp. 1283–1296, 2002.
[85]  T. P. Obrenovitch, “Molecular physiology of preconditioning-induced brain tolerance to ischemia,” Physiological Reviews, vol. 88, no. 1, pp. 211–247, 2008.
[86]  J. M. Gidday, “Cerebral preconditioning and ischaemic tolerance,” Nature Reviews Neuroscience, vol. 7, no. 6, pp. 437–448, 2006.
[87]  C. Sommer, “Ischemic preconditioning: postischemic structural changes in the brain,” Journal of Neuropathology and Experimental Neurology, vol. 67, no. 2, pp. 85–92, 2008.
[88]  V. R. Venna, J. Li, S. E. Benashski, S. Tarabishy, and L. D. McCullough, “Preconditioning induces sustained neuroprotection by downregulation of adenosine 5′-monophosphate-activated protein kinase,” Neuroscience, vol. 201, pp. 280–287, 2012.
[89]  N. Bedirli, E. U. Bagriacik, H. Emmez, G. Yilmaz, Y. Unal, and Z. Ozkose, “Sevoflurane and isoflurane preconditioning provides neuroprotection by inhibition of apoptosis-related mRNA expression in a rat model of focal cerebral ischemia,” Journal of Neurosurgical Anesthesiology, vol. 24, no. 4, pp. 336–344, 2012.
[90]  O.-N. Bae, K. Rajanikant, J. Min, et al., “Lymphocyte cell kinase activation mediates neuroprotection during ischemic preconditioning,” The Journal of Neuroscience, vol. 32, no. 21, pp. 7278–7286, 2012.
[91]  G. Pignataro, O. Cuomo, A. Vinciguerra, et al., “NCX as a key player in the neuroprotection exerted by ischemic preconditioning and postconditioning,” in Sodium Calcium Exchange: A Growing Spectrum of Pathophysiological Implications, L. Annunziato, Ed., vol. 961 of Advances in Experimental Medicine and Biology, pp. 223–240, 2013.
[92]  G. Pignataro, F. Boscia, E. Esposito et al., “NCX1 and NCX3: two new effectors of delayed preconditioning in brain ischemia,” Neurobiology of Disease, vol. 45, no. 1, pp. 616–623, 2012.
[93]  X.-R. Liu, M. Luo, F. Yan, et al., “Ischemic postconditioning diminishes matrix metalloproteinase 9 expression and attenuates loss of the extracellular matrix proteins in rats following middle cerebral artery occlusion and reperfusion,” CNS Neuroscience & Therapeutics, vol. 18, no. 10, pp. 855–863, 2012.
[94]  M. N. Hoda, S. Siddiqui, S. Herberg, et al., “Remote ischemic perconditioning is effective alone and in combination with intravenous tissue-type plasminogen activator in murine model of embolic stroke,” Stroke, vol. 43, no. 10, pp. 2794–2799, 2012.
[95]  B. Peng, Q.-L. Guo, Z.-J. He et al., “Remote ischemic postconditioning protects the brain from global cerebral ischemia/reperfusion injury by up-regulating endothelial nitric oxide synthase through the PI3K/Akt pathway,” Brain Research, vol. 1445, pp. 92–102, 2012.
[96]  N. R. Gonzalez, R. Hamilton, A. Bilgin-Freiert, et al., “Cerebral hemodynamic and metabolic effects of remote ischemic preconditioning in patients with subarachnoid hemorrhage,” Acta Neurochirurgica, vol. 115, pp. 193–198, 2013.
[97]  K. J. Kwon, J. N. Kim, M. K. Kim, et al., “Neuroprotective effects of valproic acid against hemin toxicity: possible involvement of the down-regulation of heme oxygenase-1 by regulating ubiquitin-proteasomal pathway,” Neurochemistry International, vol. 62, no. 3, pp. 240–250, 2013.
[98]  H.-J. Chun, D. W. Kim, H.-J. Yi, et al., “Effects of statin and deferoxamine administration on neurological outcomes in a rat model of intracerebral hemorrhage,” Neurological Sciences, vol. 33, no. 2, pp. 289–296, 2012.
[99]  Y. Hong, S. Guo, S. Chen, C. Sun, J. Zhang, and X. Sun, “Beneficial effect of hydrogen-rich saline on cerebral vasospasm after experimental subarachnoid hemorrhage in rats,” Journal of Neuroscience Research, vol. 90, no. 8, pp. 1670–1680, 2012.
[100]  S. Gatti, C. Lonati, F. Acerbi et al., “Protective action of NDP-MSH in experimental subarachnoid hemorrhage,” Experimental Neurology, vol. 234, no. 1, pp. 230–238, 2012.
[101]  F. Atif, S. Yousuf, I. Sayeed, T. Ishrat, F. Hua, and D. G. Stein, “Combination treatment with progesterone and vitamin D hormone is more effective than monotherapy in ischemic stroke: the role of BDNF/TrkB/Erk1/2 signaling in neuroprotection,” Neuropharmacology, vol. 67, pp. 78–87, 2013.
[102]  Y. Du, X. Zhang, H. Ji, H. Liu, S. Li, and L. Li, “Probucol and atorvastatin in combination protect rat brains in MCAO model: upregulating Peroxiredoxin2, Foxo3a and Nrf2 expression,” Neuroscience Letters, vol. 509, no. 2, pp. 110–115, 2012.
[103]  X. Guo, X. Bu, J. Jiang, P. Cheng, and Z. Yan, “Enhanced neuroprotective effects of co-administration of G-CSF with simvastatin on intracerebral hemorrhage in rats,” Turkish Neurosurgery, vol. 22, no. 6, pp. 732–739, 2012.
[104]  B. Kallmünzer, S. Schwab, and R. Kollmar, “Mild hypothermia of 34°C reduces side effects of rt-PA treatment after thromboembolic stroke in rats,” Experimental & Translational Stroke Medicine, vol. 4, no. 1, article 3, 2012.
[105]  Y. Wang, Z. Zhang, N. Chow, et al., “An activated protein C analog with reduced anticoagulant activity extends the therapeutic window of tissue plasminogen activator for ischemic stroke in rodents,” Stroke, vol. 43, no. 9, pp. 2444–2449, 2012.
[106]  F. Campos, T. Qin, J. Castillo, et al., “Fingolimod reduces hemorrhagic transformation associated with delayed tissue plasminogen activator treatment in a mouse thromboembolic model,” Stroke, vol. 44, no. 2, pp. 505–511, 2013.
[107]  D. Zemke and A. Majid, “The potential of minocycline for neuroprotection in human neurologic disease,” Clinical Neuropharmacology, vol. 27, no. 6, pp. 293–298, 2004.
[108]  M. V. P. Srivastava, A. Bhasin, R. Bhatia et al., “Efficacy of minocycline in acute ischemic stroke: a single-blinded, placebo-controlled trial,” Neurology India, vol. 60, no. 1, pp. 23–28, 2012.
[109]  X. Fan, E. H. Lo, and X. Wang, “Effects of minocycline plus tissue plasminogen activator combination therapy after focal embolic stroke in type 1 diabetic rats,” Stroke, vol. 44, no. 3, pp. 745–752, 2013.
[110]  X. Jin, J. Liu, K. J. Liu, G. A. Rosenberg, Y. Yang, and W. Liu, “Normobaric hyperoxia combined with minocycline provides greater neuroprotection than either alone in transient focal cerebral ischemia,” Experimental Neurology, vol. 240, pp. 9–16, 2013.
[111]  E. C. S. Franco, M. M. Cardoso, A. Gouvêia, A. Pereira, and W. Gomes-Leal, “Modulation of microglial activation enhances neuroprotection and functional recovery derived from bone marrow mononuclear cell transplantation after cortical ischemia,” Neuroscience Research, vol. 73, no. 2, pp. 122–132, 2012.
[112]  M. M. Cardoso, E. C. S. Franco, C. C. de Souza, M. C. Da Silva, A. Gouveia, and W. Gomes-Leal, “Minocycline treatment and bone marrow mononuclear cell transplantation after endothelin-1 induced striatal ischemia,” Inflammation, vol. 36, no. 1, pp. 197–205, 2013.
[113]  H. Sakata, K. Niizuma, H. Yoshioka et al., “Minocycline-preconditioned neural stem cells enhance neuroprotection after ischemic stroke in rats,” Journal of Neuroscience, vol. 32, no. 10, pp. 3462–3473, 2012.
[114]  F. Bellia, G. Vecchio, S. Cuzzocrea, V. Calabrese, and E. Rizzarelli, “Neuroprotective features of carnosine in oxidative driven diseases,” Molecular Aspects of Medicine, vol. 32, no. 4–6, pp. 258–266, 2011.
[115]  A. A. Boldyrev, S. L. Stvolinsky, T. N. Fedorova, and Z. A. Suslina, “Carnosine as a natural antioxidant and geroprotector: from molecular mechanisms to clinical trials,” Rejuvenation Research, vol. 13, no. 2-3, pp. 156–158, 2010.
[116]  O.-N. Bae, K. Serfozo, S.-H. Baek, et al., “Safety and efficacy evaluation of carnosine, an endogenous neuroprotective agent for ischemic stroke,” Stroke, vol. 44, no. 1, pp. 205–212, 2013.
[117]  Y. Shen, P. He, Y.-Y. Fan et al., “Carnosine protects against permanent cerebral ischemia in histidine decarboxylase knockout mice by reducing glutamate excitotoxicity,” Free Radical Biology & Medicine, vol. 48, no. 5, pp. 727–735, 2010.
[118]  G. K. Rajanikant, D. Zemke, M.-C. Senut et al., “Carnosine is neuroprotective against permanent focal cerebral ischemia in mice,” Stroke, vol. 38, no. 11, pp. 3023–3031, 2007.
[119]  J. Min, M.-C. Senut, K. Rajanikant et al., “Differential neuroprotective effects of carnosine, anserine, and N-acetyl carnosine against permanent focal ischemia,” Journal of Neuroscience Research, vol. 86, no. 13, pp. 2984–2991, 2008.
[120]  R. G. Krishnamurthy, M.-C. Senut, D. Zemke et al., “Asiatic acid, a pentacyclic triterpene from Centella asiatica, is neuroprotective in a mouse model of focal cerebral ischemia,” Journal of Neuroscience Research, vol. 87, no. 11, pp. 2541–2550, 2009.
[121]  K. Y. Lee, O.-N. Bae, K. Serfozo et al., “Asiatic acid attenuates infarct volume, mitochondrial dysfunction, and matrix metalloproteinase-9 induction after focal cerebral ischemia,” Stroke, vol. 43, pp. 1632–1638, 2012.
[122]  M. Xu, Y. Xiong, J. Liu, J. Qian, L. Zhu, and J. Gao, “Asiatic acid, a pentacyclic triterpene in Centella asiatica, attenuates glutamate-induced cognitive deficits in mice and apoptosis in SH-SY5Y cells,” Acta Pharmacologica Sinica, vol. 33, no. 5, pp. 578–587, 2012.
[123]  R. Tabassum, K. Vaibhav, P. Shrivastava, et al., “Centella asiatica attenuates the neurobehavioral, neurochemical and histological changes in transient focal middle cerebral artery occlusion rats,” Neurological Sciences, vol. 34, no. 6, pp. 925–933, 2013.
[124]  R. Kollmar and S. Schwab, “Hypothermia and ischemic stroke,” Current Treatment Options in Neurology, vol. 14, no. 2, pp. 188–196, 2012.
[125]  S. S. Song and P. D. Lyden, “Overview of therapeutic hypothermia,” Current Treatment Options in Neurology, vol. 14, no. 6, pp. 541–548, 2012.
[126]  H. A. Choi, N. Badjatia, and S. A. Mayer, “Hypothermia for acute brain injury—mechanisms and practical aspects,” Nature Reviews Neurology, vol. 8, no. 4, pp. 214–222, 2012.
[127]  S. Nagel, M. Papadakis, K. Pfleger, C. Grond-Ginsbach, A. M. Buchan, and S. Wagner, “Microarray analysis of the global gene expression profile following hypothermia and transient focal cerebral ischemia,” Neuroscience, vol. 208, pp. 109–122, 2012.
[128]  K.-E. Choi, C. L. Hall, J.-M. Sun, et al., “A novel stroke therapy of pharmacologically induced hypothermia after focal cerebral ischemia in mice,” FASEB Journal, vol. 26, no. 7, pp. 2799–2810, 2012.
[129]  S. E. Lakhan and F. Pamplona, “Application of mild therapeutic hypothermia on stroke: a systematic review and meta-analysis,” Stroke Research and Treatment, vol. 2012, Article ID 295906, 12 pages, 2012.
[130]  R. Kollmar, B. Gebhardt, and S. Schwab, “EuroHYP-1 trial: EU-funded therapy study on the effectiveness of mild therapeutic hypothermia for acute ischemic stroke,” Der Nervenarzt, vol. 83, no. 10, pp. 1252–1259, 2012.
[131]  S. Lorrio, V. Gómez-Rangel, P. Negredo, et al., “Novel multitarget ligand ITH33/IQM9.21 provides neuroprotection in in vitro and in vivo models related to brain ischemia,” Neuropharmacology, vol. 67, pp. 403–411, 2013.
[132]  F. Guo, W.-L. Jin, L.-Y. Li, et al., “M9, a novel region of Amino-Nogo-A, attenuates cerebral ischemic injury by inhibiting NADPH oxidase-derived superoxide production in mice,” CNS Neuroscience & Therapeutics, vol. 19, no. 5, pp. 319–328, 2013.
[133]  S.-W. Kim, Y. Jin, J.-H. Shin et al., “Glycyrrhizic acid affords robust neuroprotection in the postischemic brain via anti-inflammatory effect by inhibiting HMGB1 phosphorylation and secretion,” Neurobiology of Disease, vol. 46, no. 1, pp. 147–156, 2012.
[134]  J. Y. Kim, H. Y. Jeong, H. K. Lee et al., “Neuroprotection of the leaf and stem of Vitis amurensis and their active compounds against ischemic brain damage in rats and excitotoxicity in cultured neurons,” Phytomedicine, vol. 19, no. 2, pp. 150–159, 2012.
[135]  C. Ju, S. Hwang, G.-S. Cho, et al., “Differential anti-ischemic efficacy and therapeutic time window of trans- and cis-hinokiresinols: stereo-specific antioxidant and anti-inflammatory activities,” Neuropharmacology, vol. 67, pp. 465–475, 2013.
[136]  X. Zhang, X. Zhang, C. Wang et al., “Neuroprotection of early and short-time applying berberine in the acute phase of cerebral ischemia: up-regulated pAkt, pGSK and pCREB, down-regulated NF-κB expression, ameliorated BBB permeability,” Brain Research, vol. 1459, pp. 61–70, 2012.
[137]  M. Khan, T. S. Dhammu, H. Sakakima, et al., “The inhibitory effect of S-nitrosoglutathione on blood-brain barrier disruption and peroxynitrite formation in a rat model of experimental stroke,” Journal of Neurochemistry, vol. 123, supplement s2, pp. 86–97, 2012.
[138]  P. M. Gharibani, J. Modi, C. Pan, et al., “The mechanism of taurine protection against endoplasmic reticulum stress in an animal stroke model of cerebral artery occlusion and stroke-related conditions in primary neuronal cell culture,” Advances in Experimental Medicine and Biology, vol. 776, pp. 241–258, 2013.
[139]  C. Cheyuo, A. Jacob, R. Wu et al., “Recombinant human MFG-E8 attenuates cerebral ischemic injury: its role in anti-inflammation and anti-apoptosis,” Neuropharmacology, vol. 62, no. 2, pp. 890–900, 2012.
[140]  Y. Sun, P. Yu, G. Zhang et al., “Therapeutic effects of tetramethylpyrazine nitrone in rat ischemic stroke models,” Journal of Neuroscience Research, vol. 90, no. 8, pp. 1662–1669, 2012.
[141]  A. Nehlig, “The neuroprotective effects of cocoa flavanol and its influence on cognitive performance,” British Journal of Clinical Pharmacology, vol. 75, no. 3, pp. 716–727, 2013.
[142]  T.-L. Yen, C.-K. Hsu, W.-J. Lu et al., “Neuroprotective effects of xanthohumol, a prenylated flavonoid from hops (humulus lupulus), in ischemic stroke of rats,” Journal of Agricultural and Food Chemistry, vol. 60, no. 8, pp. 1937–1944, 2012.
[143]  S. S. Raza, M. M. Khan, A. Ahmad, et al., “Neuroprotective effect of naringenin is mediated through suppression of NF-κB signaling pathway in experimental stroke,” Neuroscience, vol. 230, pp. 157–171, 2013.
[144]  S. Li, C. Wu, L. Zhu, et al., “By improving regional cortical blood flow, attenuating mitochondrial dysfunction and sequential apoptosis galangin acts as a potential neuroprotective agent after acute ischemic stroke,” Molecules, vol. 17, no. 11, pp. 13403–13423, 2012.
[145]  M. Gelderblom, F. Leypoldt, J. Lewerenz et al., “The flavonoid fisetin attenuates postischemic immune cell infiltration, activation and infarct size after transient cerebral middle artery occlusion in mice,” Journal of Cerebral Blood Flow and Metabolism, vol. 32, no. 5, pp. 835–843, 2012.
[146]  Q.-B. Zhou, Q. Jia, Y. Zhang, L.-Y. Li, Z.-F. Chi, and P. Liu, “Effects of baicalin on protease-activated receptor-1 expression and brain injury in a rat model of intracerebral hemorrhage,” The Chinese Journal of Physiology, vol. 55, no. 3, pp. 202–209, 2012.
[147]  L. S. A. Capettini, S. Q. Savergnini, R. F. Da Silva, et al., “Update on the role of cannabinoid receptors after ischemic stroke,” Mediators of Inflammation, vol. 2012, Article ID 824093, 8 pages, 2012.
[148]  P. Pacher and K. Mackie, “Interplay of cannabinoid 2 (CB2) receptors with nitric oxide synthases, oxidative and nitrative stress, and cell death during remote neurodegeneration,” Journal of Molecular Medicine, vol. 90, no. 4, pp. 347–351, 2012.
[149]  J. G. Zarruk, D. Fernández-López, I. García-Yébenes et al., “Cannabinoid type 2 receptor activation downregulates stroke-induced classic and alternative brain macrophage/microglial activation concomitant to neuroprotection,” Stroke, vol. 43, no. 1, pp. 211–219, 2012.
[150]  J. Sun, Y. Fang, H. Ren, et al., “WIN55, 212-2 protects oligodendrocyte precursor cells in stroke penumbra following permanent focal cerebral ischemia in rats,” Acta Pharmacologica Sinica, vol. 34, no. 1, pp. 119–128, 2013.
[151]  D. Fernández-López, J. Faustino, N. Derugin et al., “Reduced infarct size and accumulation of microglia in rats treated with WIN 55,212-2 after neonatal stroke,” Neuroscience, vol. 207, pp. 307–315, 2012.
[152]  N. Suzuki, M. Suzuki, K. Hamajo, K. Murakami, T. Tsukamoto, and M. Shimojo, “Contribution of hypothermia and CB1 receptor activation to protective effects of TAK-937, a cannabinoid receptor agonist, in rat transient MCAO model,” PLoS ONE, vol. 7, no. 7, Article ID e40889, 2012.
[153]  N. Suzuki, M. Suzuki, K. Murakami, K. Hamajo, T. Tsukamoto, and M. Shimojo, “Cerebroprotective effects of TAK-937, a cannabinoid receptor agonist, on ischemic brain damage in middle cerebral artery occluded rats and non-human primates,” Brain Research, vol. 1430, pp. 93–100, 2012.
[154]  S.-Y. Xu and S.-Y. Pan, “The failure of animal models of neuroprotection in acute ischemic stroke to translate to clinical efficacy,” Medical Science Monitor Basic Research, vol. 19, pp. 37–45, 2013.
[155]  B. A. Sutherland, J. Minnerup, J. S. Balami, F. Arba, A. M. Buchan, and C. Kleinschnitz, “Neuroprotection for ischaemic stroke translation from the bench to the bedside,” International Journal of Stroke, vol. 7, no. 5, pp. 407–418, 2012.
[156]  A. Dávalos, J. Alvarez-Sabín, J. Castillo, et al., “Citicoline in the treatment of acute ischaemic stroke: an international, randomised, multicentre, placebo-controlled study (ICTUS trial),” The Lancet, vol. 380, no. 9839, pp. 349–357, 2012.
[157]  W.-D. Heiss, M. Brainin, N. M. Bornstein, J. Tuomilehto, and Z. Hong, “Cerebrolysin in patients with acute ischemic stroke in Asia: results of a double-blind, placebo-controlled randomized trial,” Stroke, vol. 43, no. 3, pp. 630–636, 2012.
[158]  X. Liu, L. Wang, A. Wen et al., “Ginsenoside-Rd improves outcome of acute ischaemic stroke—a randomized, double-blind, placebo-controlled, multicenter trial,” European Journal of Neurology, vol. 19, no. 6, pp. 855–863, 2012.
[159]  T. J. England, M. Abaei, D. P. Auer et al., “Granulocyte-colony stimulating factor for mobilizing bone marrow stem cells in subacute stroke: the stem cell trial of recovery enhancement after stroke 2 randomized controlled trial,” Stroke, vol. 43, no. 2, pp. 405–411, 2012.
[160]  M. D. Hill, R. H. Martin, D. Mikulis, et al., “Safety and efficacy of NA-1 in patients with iatrogenic stroke after endovascular aneurysm repair (ENACT): a phase 2, randomised, double-blind, placebo-controlled trial,” The Lancet Neurology, vol. 11, no. 11, pp. 942–950, 2012.
[161]  S. M. D. Mees, A. Algra, W. P. Vandertop, et al., “Magnesium for aneurysmal subarachnoid haemorrhage (MASH-2): a randomised placebo-controlled trial,” The Lancet, vol. 380, no. 9836, pp. 44–49, 2012.
[162]  J. I. Suarez, R. H. Martin, E. Calvillo et al., “The albumin in subarachnoid hemorrhage (ALISAH) multicenter pilot clinical trial: safety and neurologic outcomes,” Stroke, vol. 43, no. 3, pp. 683–690, 2012.
[163]  M. Fisher, “Recommendations for standards regarding preclinical neuroprotective and restorative drug development,” Stroke, vol. 30, no. 12, pp. 2752–2758, 1999.
[164]  G. W. Albers, J. Bogousslavsky, M. A. Bozik et al., “Recommendations for clinical trial evaluation of acute stroke therapies,” Stroke, vol. 32, no. 7, pp. 1598–1606, 2001.
[165]  U. Dirnagl and M. Fisher, “International, multicenter randomized preclinical trials in translational stroke research: it's time to act,” Journal of Cerebral Blood Flow and Metabolism, vol. 32, no. 6, pp. 933–935, 2012.
[166]  U. Dirnagl, A. Hakim, M. Macleod, et al., “A concerted appeal for international cooperation in preclinical stroke research,” Stroke, vol. 44, no. 6, pp. 1754–1760, 2013.

Full-Text

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133