The incidence of cerebrovascular disease is highest in the elderly population. However, the pathophysiological mechanisms of brain response to cerebral ischemia in old age are currently poorly understood. Ischemic changes in the commonly used young animal stroke models do not reflect the molecular changes associated with the aged brain. Neuroinflammation and oxidative stress are important pathogenic processes occurring during the acute phase of cerebral ischemia. Free radical generation is also implicated in the aging process, and the combination of these effects in elderly stroke patients could explain the higher risk of morbidity and mortality. A better understanding of stroke pathophysiology in the elderly patient would assist in the development of new therapeutic strategies for this vulnerable age group. With the increasing use of reperfusion therapies, inflammatory pathways and oxidative stress remain attractive therapeutic targets for the development of adjuvant neuroprotective agents. This paper will discuss these molecular aspects of acute stroke and senescence from a bench-to-bedside research perspective. 1. Introduction Old age is an important risk factor for stroke and is associated with increased patient morbidity and mortality [1, 2]. Many of these patients have associated comorbidities, for example, cardiovascular and respiratory disease. This is further complicated by an increased risk of cognitive and functional decline in elderly stroke patients [3, 4]. Poor functional recovery has also been demonstrated in aged-animal models [5]. The pathophysiological mechanisms of the brains response to an ischemic insult in old age are poorly understood. Most preclinical stroke studies have been performed in young animal models and therefore do not reflect the molecular changes associated with the aged brain [6, 7]. This has been one of the criticisms of preclinical stroke neuroprotection studies and implicated in the resulting failure of clinical stroke neuroprotection trials [8, 9]. Neuroprotective therapies targeting NMDA and AMPA receptors have demonstrated reduced efficacy in aged-animal stroke models [10]. The pharmacokinetic and pharmacodynamic properties of neuroprotective agents may also be different in older patients [8]. This therefore emphasizes the importance of assessing potential neuroprotective therapies in preclinical aged animal stroke models and early clinical studies of elderly patients [6]. A better understanding of stroke pathogenesis in the aged brain would assist in the development of new therapeutic strategies for treatment
References
[1]
P. J. Modrego, M. A. Pina, and F. J. Lerin, “The impact of ageing on stroke subtypes, length of stay and mortality: study in the province of Teruel, Spain,” Acta Neurologica Scandinavica, vol. 108, no. 6, pp. 435–442, 2003.
[2]
G. Saposnik, R. Cote, S. Phillips et al., “Stroke outcome in those over 80: a multicenter cohort study across Canada,” Stroke, vol. 39, no. 8, pp. 2310–2317, 2008.
[3]
M. Inzitari, C. Pozzi, L. Ferrucci et al., “Subtle neurological abnormalities as risk factors for cognitive and functional decline, cerebrovascular events, and mortality in older community-dwelling adults,” Archives of Internal Medicine, vol. 168, no. 12, pp. 1270–1276, 2008.
[4]
G. T. Stebbins, D. L. Nyenhuis, C. Wang et al., “Gray matter atrophy in patients with ischemic stroke with cognitive impairment,” Stroke, vol. 39, no. 3, pp. 785–793, 2008.
[5]
I. Badan, I. Dinca, B. Buchhold et al., “Accelerated accumulation of N- and C-terminal beta APP fragments and delayed recovery of microtubule-associated protein 1B expression following stroke in aged rats,” European Journal of Neuroscience, vol. 19, no. 8, pp. 2270–2280, 2004.
[6]
C. L. Rosen, V. A. DiNapoli, T. Nagamine, and T. Crocco, “Influence of age on stroke outcome following transient focal ischemia,” Journal of Neurosurgery, vol. 103, no. 4, pp. 687–694, 2005.
[7]
A. Popa-Wagner, S. T. Carmichael, Z. Kokaia, C. Kessler, and L. C. Walker, “The response of the aged brain to stroke: too much, too soon?” Current Neurovascular Research, vol. 4, no. 3, pp. 216–227, 2007.
[8]
G. A. Ford, “Clinical pharmacological issues in the development of acute stroke therapies,” The British Journal of Pharmacology, vol. 153, supplement 1, pp. S112–S119, 2008.
[9]
G. Z. Feuerstein, M. M. Zaleska, M. Krams et al., “Missing steps in the STAIR case: a translational medicine perspective on the development of NXY-059 for treatment of acute ischemic stroke,” Journal of Cerebral Blood Flow & Metabolism, vol. 28, no. 1, pp. 217–219, 2008.
[10]
Y. Suzuki, Y. Takagi, R. Nakamura, K. Hashimoto, and K. Umemura, “Ability of NMDA and non-NMDA receptor antagonists to inhibit cerebral ischemic damage in aged rats,” Brain Research, vol. 964, no. 1, pp. 116–120, 2003.
[11]
H. Ay, W. J. Koroshetz, M. Vangel et al., “Conversion of ischemic brain tissue into infarction increases with age,” Stroke, vol. 36, no. 12, pp. 2632–2636, 2005.
[12]
J. M. Simard, T. A. Kent, M. Chen, K. V. Tarasov, and V. Gerzanich, “Brain oedema in focal ischaemia: molecular pathophysiology and theoretical implications,” The Lancet Neurology, vol. 6, no. 3, pp. 258–268, 2007.
[13]
J. Montaner, A. Rovira, C. A. Molina et al., “Plasmatic level of neuroinflammatory markers predict the extent of diffusion-weighted image lesions in hyperacute stroke,” Journal of Cerebral Blood Flow & Metabolism, vol. 23, no. 12, pp. 1403–1407, 2003.
[14]
L. S. Rallidis, M. Vikelis, D. B. Panagiotakos et al., “Inflammatory markers and in-hospital mortality in acute ischaemic stroke,” Atherosclerosis, vol. 189, no. 1, pp. 193–197, 2006.
[15]
C. J. Smith, H. C. Emsley, C. M. Gavin et al., “Peak plasma interleukin-6 and other peripheral markers of inflammation in the first week of ischaemic stroke correlate with brain infarct volume, stroke severity and long-term outcome,” BMC Neurology, vol. 4, article 2, 2004.
[16]
J. E. Jung, G. S. Kim, H. Chen et al., “Reperfusion and neurovascular dysfunction in stroke: from basic mechanisms to potential strategies for neuroprotection,” Molecular Neurobiology, vol. 41, no. 2-3, pp. 172–179, 2010.
[17]
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.
[18]
U. Dirnagl, J. Klehmet, J. S. Braun et al., “Stroke-induced immunodepression: experimental evidence and clinical relevance,” Stroke, vol. 38, no. 2, pp. 770–773, 2007.
[19]
C. Meisel, J. M. Schwab, K. Prass, A. Meisel, and U. Dirnagl, “Central nervous system injury-induced immune deficiency syndrome,” Nature Reviews Neuroscience, vol. 6, no. 10, pp. 775–786, 2005.
[20]
M. Endres, B. Engelhardt, J. Koistinaho et al., “Improving outcome after stroke: overcoming the translational roadblock,” Cerebrovascular Diseases, vol. 25, no. 3, pp. 268–278, 2008.
[21]
J. Vi?a, C. Borrás, and J. Miquel, “Theories of ageing,” IUBMB Life, vol. 59, no. 4-5, pp. 249–254, 2007.
[22]
H. Offner, S. Subramanian, S. M. Parker, M. E. Afentoulis, A. A. Vandenbark, and P. D. Hurn, “Experimental stroke induces massive, rapid activation of the peripheral immune system,” Journal of Cerebral Blood Flow & Metabolism, vol. 26, no. 5, pp. 654–665, 2006.
[23]
F. Perini, M. Morra, M. Alecci, E. Galloni, M. Marchi, and V. Toso, “Temporal profile of serum anti-inflammatory and pro-inflammatory interleukins in acute ischemic stroke patients,” Neurological Sciences, vol. 22, no. 4, pp. 289–296, 2001.
[24]
N. Vila, J. Castillo, A. Dávalos, and A. Chamorro, “Proinflammatory cytokines and early neurological worsening in ischemic stroke,” Stroke, vol. 31, no. 10, pp. 2325–2329, 2000.
[25]
A. Y. Lai and K. G. Todd, “Microglia in cerebral ischemia: molecular actions and interactions,” Canadian Journal of Physiology and Pharmacology, vol. 84, no. 1, pp. 49–59, 2006.
[26]
B. H. Buck, D. S. Liebeskind, J. L. Saver et al., “Early neutrophilia is associated with volume of ischemic tissue in acute stroke,” Stroke, vol. 39, no. 2, pp. 355–360, 2008.
[27]
S. Amaro and A. Chamorro, “Translational stroke research of the combination of thrombolysis and antioxidant therapy,” Stroke, vol. 42, no. 5, pp. 1495–1499, 2011.
[28]
D. Sherman, A. Bes, J. D. Easton et al., “Use of anti-ICAM-1 therapy in ischemic stroke: results of the enlimomab acute stroke trial,” Neurology, vol. 57, no. 8, pp. 1428–1434, 2001.
[29]
M. Fisher, “New approaches to neuroprotective drug development,” Stroke, vol. 42, no. 1, pp. S24–S27, 2010.
[30]
V. Kaushal and L. C. Schlichter, “Mechanisms of microglia-mediated neurotoxicity in a new model of the stroke penumbra,” Journal of Neuroscience, vol. 28, no. 9, pp. 2221–2230, 2008.
[31]
Y. Chen, J. M. Hallenbeck, C. Ruetzler et al., “Overexpression of monocyte chemoattractant protein 1 in the brain exacerbates ischemic brain injury and is associated with recruitment of inflammatory cells,” Journal of Cerebral Blood Flow & Metabolism, vol. 23, no. 6, pp. 748–755, 2003.
[32]
X. H. Deng, G. Bertini, Y. Z. Xu, Z. Yan, and M. Bentivoglio, “Cytokine-induced activation of glial cells in the mouse brain is enhanced at an advanced age,” Neuroscience, vol. 141, no. 2, pp. 645–661, 2006.
[33]
A. Popa-Wagner, I. Badan, L. Walker, S. Groppa, N. Patrana, and C. Kessler, “Accelerated infarct development, cytogenesis and apoptosis following transient cerebral ischemia in aged rats,” Acta Neuropathologica, vol. 113, no. 3, pp. 277–293, 2007.
[34]
J. C. Lee, G. S. Cho, B. O. Choi, C. K. Hyoung, Y. S. Kim, and W. K. Kim, “Intracerebral hemorrhage-induced brain injury is aggravated in senescence-accelerated prone mice,” Stroke, vol. 37, no. 1, pp. 216–222, 2006.
[35]
J. B. Buchanan, N. L. Sparkman, J. Chen, and R. W. Johnson, “Cognitive and neuroinflammatory consequences of mild repeated stress are exacerbated in aged mice,” Psychoneuroendocrinology, vol. 33, no. 6, pp. 755–765, 2008.
[36]
M. P. Gavilán, E. Revilla, C. Pintado et al., “Molecular and cellular characterization of the age-related neuroinflammatory processes occurring in normal rat hippocampus: potential relation with the loss of somatostatin GABAergic neurons,” Journal of Neurochemistry, vol. 103, no. 3, pp. 984–996, 2007.
[37]
B. T. Baune, G. Ponath, M. Rothermundt, A. Roesler, and K. Berger, “Association between cytokines and cerebral MRI changes in the aging brain,” Journal of Geriatric Psychiatry and Neurology, vol. 22, no. 1, pp. 23–34, 2009.
[38]
S. X. Leng, Q. L. Xue, J. Tian, J. D. Walston, and L. P. Fried, “Inflammation and frailty in older women,” Journal of the American Geriatrics Society, vol. 55, no. 6, pp. 864–871, 2007.
[39]
S. Spulber, M. Oprica, T. Bartfai, B. Winblad, and M. Schultzberg, “Blunted neurogenesis and gliosis due to transgenic overexpression of human soluble IL-1ra in the mouse,” European Journal of Neuroscience, vol. 27, no. 3, pp. 549–558, 2008.
[40]
J. K. Kiecolt-Glaser, K. J. Preacher, R. C. MacCallum, C. Atkinson, W. B. Malarkey, and R. Glaser, “Chronic stress and age-related increases in the proinflammatory cytokine IL-6,” Proceedings of the National Academy of Sciences of the United States of America, vol. 100, no. 15, pp. 9090–9095, 2003.
[41]
S. M. Allan, P. J. Tyrrell, and N. J. Rothwell, “Interleukin-1 and neuronal injury,” Nature Reviews Immunology, vol. 5, no. 8, pp. 629–640, 2005.
[42]
J. R. Caso, M. A. Moro, P. Lorenzo, I. Lizasoain, and J. C. Leza, “Involvement of IL-1β in acute stress-induced worsening of cerebral ischaemia in rats,” European Neuropsychopharmacology, vol. 17, no. 9, pp. 600–607, 2007.
[43]
N. J. Mulcahy, J. Ross, N. J. Rothwell, and S. A. Loddick, “Delayed administration of interleukin-1 receptor antagonist protects against transient cerebral ischaemia in the rat,” The British Journal of Pharmacology, vol. 140, no. 3, pp. 471–476, 2003.
[44]
N. Rothwell, “Interleukin-1 and neuronal injury: mechanisms, modification, and therapeutic potential,” Brain, Behavior, and Immunity, vol. 17, no. 3, pp. 152–157, 2003.
[45]
H. C. Emsley, C. J. Smith, R. F. Georgiou et al., “A randomised phase II study of interleukin-1 receptor antagonist in acute stroke patients,” Journal of Neurology, Neurosurgery and Psychiatry, vol. 76, no. 10, pp. 1366–1372, 2005.
[46]
S. R. Clark, C. J. McMahon, I. Gueorguieva et al., “Interleukin-1 receptor antagonist penetrates human brain at experimentally therapeutic concentrations,” Journal of Cerebral Blood Flow & Metabolism, vol. 28, no. 2, pp. 387–394, 2008.
[47]
J. Galea, K. Ogungbenro, S. Hulme, et al., “Intravenous anakinra can achieve experimentally effective concentrations in the central nervous system within a therapeutic time window: results of a dose-ranging study,” Journal of Cerebral Blood Flow & Metabolism, vol. 31, no. 2, pp. 439–447, 2011.
[48]
M. Krams, K. R. Lees, W. Hacke, A. P. Grieve, J. M. Orgogozo, and G. A. Ford, “Acute stroke therapy by inhibition of neutrophils (ASTIN): an adaptive dose-response study of UK-279,276 in acute ischemic stroke,” Stroke, vol. 34, no. 11, pp. 2543–2548, 2003.
[49]
P. D. Lyden, A. Shuaib, K. R. Lees et al., “Safety and tolerability of NXY-059 for acute intracerebral hemorrhage: the CHANT Trial,” Stroke, vol. 38, no. 8, pp. 2262–2269, 2007.
[50]
A. Shuaib, K. R. Lees, P. Lyden et al., “NXY-059 for the treatment of acute ischemic stroke,” The New England Journal of Medicine, vol. 357, no. 6, pp. 562–571, 2007.
[51]
T. Nakase, S. Yoshioka, and A. Suzuki, “Free radical scavenger, edaravone, reduces the lesion size of lacunar infarction in human brain ischemic stroke,” BMC Neurology, vol. 11, p. 39, 2011.
[52]
A. Popa-Wagner, D. Pirici, E. B. Petcu et al., “Pathophysiology of the vascular wall and its relevance for cerebrovascular disorders in aged rodents,” Current Neurovascular Research, vol. 7, no. 3, pp. 251–267, 2010.
[53]
S. Amaro, D. Soy, V. Obach, A. Cervera, A. M. Planas, and A. Chamorro, “A pilot study of dual treatment with recombinant tissue plasminogen activator and uric acid in acute ischemic stroke,” Stroke, vol. 38, no. 7, pp. 2173–2175, 2007.
[54]
S. Amaro, D. Cánovas, M. Castellanos et al., “The URICO-ICTUS study, a phase 3 study of combined treatment with uric acid and rtPA administered intravenously in acute ischaemic stroke patients within the first 4·5 h of onset of symptoms,” International Journal of Stroke, vol. 5, no. 4, pp. 325–328, 2010.
[55]
H. M. den Hertog, H. B. van der Worp, H. M. A. van Gemert et al., “The Paracetamol (Acetaminophen) In Stroke (PAIS) trial: a multicentre, randomised, placebo-controlled, phase III trial,” The Lancet Neurology, vol. 8, no. 5, pp. 434–440, 2009.
[56]
Y. Lampl, M. Boaz, R. Gilad et al., “Minocycline treatment in acute stroke: an open-label, evaluator-blinded study,” Neurology, vol. 69, no. 14, pp. 1404–1410, 2007.
[57]
S. C. Fagan, J. L. Waller, F. T. Nichols et al., “Minocycline to improve neurologic outcome in stroke (MINOS): a dose-finding study,” Stroke, vol. 41, no. 10, pp. 2283–2287, 2010.
[58]
D. Orion, Y. Schwammenthal, T. Reshef et al., “Interleukin-6 and soluble intercellular adhesion molecule-1 in acute brain ischaemia,” European Journal of Neurology, vol. 15, no. 4, pp. 323–328, 2008.
[59]
D. Acalovschi, T. Wiest, M. Hartmann et al., “Multiple levels of regulation of the interleukin-6 system in stroke,” Stroke, vol. 34, no. 8, pp. 1864–1869, 2003.
[60]
O. Herrmann, V. Tarabin, S. Suzuki et al., “Regulation of body temperature and neuroprotection by endogenous interleukin-6 in cerebral ischemia,” Journal of Cerebral Blood Flow & Metabolism, vol. 23, no. 4, pp. 406–415, 2003.
[61]
S. Sotgiu, B. Zanda, B. Marchetti et al., “Inflammatory biomarkers in blood of patients with acute brain ischemia,” European Journal of Neurology, vol. 13, no. 5, pp. 505–513, 2006.
[62]
M. Fornage, Y. A. Chiang, E. S. O'Meara et al., “Biomarkers of inflammation and MRI-defined small vessel disease of the brain: the cardiovascular health study,” Stroke, vol. 39, no. 7, pp. 1952–1959, 2008.
[63]
M. Rodríguez-Yá?ez and J. Castillo, “Role of inflammatory markers in brain ischemia,” Current Opinion in Neurology, vol. 21, no. 3, pp. 353–357, 2008.
[64]
M. J. McKenzie, S. Yu, R. F. Macko, J. C. McLenithan, and C. E. Hafer-Macko, “Human genome comparison of paretic and nonparetic vastus lateralis muscle in patients with hemiparetic stroke,” Journal of Rehabilitation Research and Development, vol. 45, no. 2, pp. 273–282, 2008.
[65]
G. Zuliani, G. Guerra, M. Ranzini et al., “High interleukin-6 plasma levels are associated with functional impairment in older patients with vascular dementia,” International Journal of Geriatric Psychiatry, vol. 22, no. 4, pp. 305–311, 2007.
[66]
F. C. Barone, B. Arvin, R. F. White et al., “Tumor necrosis factor-α: a mediator of focal ischemic brain injury,” Stroke, vol. 28, no. 6, pp. 1233–1244, 1997.
[67]
T. Liu, R. K. Clark, P. C. McDonnell, R. F. White, F. C. Barone, and G. Z. Feuerstein, “Tumor necrosis factor-α expression in ischemic neurons,” Stroke, vol. 25, no. 7, pp. 1481–1488, 1994.
[68]
M. K. McCoy and M. G. Tansey, “TNF signaling inhibition in the CNS: implications for normal brain function and neurodegenerative disease,” Journal of Neuroinflammation, vol. 5, article 45, 2008.
[69]
K. L. Lambertsen, B. H. Clausen, A. A. Babcock et al., “Microglia protect neurons against ischemia by synthesis of tumor necrosis factor,” Journal of Neuroscience, vol. 29, no. 5, pp. 1319–1330, 2009.
[70]
L. C. Pettigrew, M. S. Kindy, S. Scheff et al., “Focal cerebral ischemia in the TNFalpha-transgenic rat,” Journal of Neuroinflammation, vol. 5, article 47, 2008.
[71]
S. D. Lavine, F. M. Hofman, and B. V. Zlokovic, “Circulating antibody against tumor necrosis factor-alpha protects rat brain from reperfusion injury,” Journal of Cerebral Blood Flow & Metabolism, vol. 18, no. 1, pp. 52–58, 1998.
[72]
F. Lovering and Y. Zhang, “Therapeutic potential of TACE inhibitors in stroke,” Current Drug Targets, vol. 4, no. 2, pp. 161–168, 2005.
[73]
H. L. Rosenzweig, M. Minami, N. S. Lessov, et al., “Endotoxin preconditioning protects against the cytotoxic effects of TNFalpha after stroke: a novel role for TNFalpha in LPS-ischemic tolerance,” Journal of Cerebral Blood Flow & Metabolism, vol. 27, no. 10, pp. 1663–1674, 2007.
[74]
N. A. Brabers and H. S. Nottet, “Role of the pro-inflammatory cytokines TNF-α and IL-1β in HIV-associated dementia,” European Journal of Clinical Investigation, vol. 36, no. 7, pp. 447–458, 2006.
[75]
J. Y. Zou and F. T. Crews, “TNFα potentiates glutamate neurotoxicity by inhibiting glutamate uptake in organotypic brain slice cultures: neuroprotection by NFκB inhibition,” Brain Research, vol. 1034, no. 1-2, pp. 11–24, 2005.
[76]
J. M. Gidday, “Cerebral preconditioning and ischaemic tolerance,” Nature Reviews Neuroscience, vol. 7, no. 6, pp. 437–448, 2006.
[77]
T. Kirino, “Ischemic tolerance,” Journal of Cerebral Blood Flow & Metabolism, vol. 22, no. 11, pp. 1283–1296, 2002.
[78]
M. P. Stenzel-Poore, S. L. Stevens, and R. P. Simon, “Genomics of preconditioning,” Stroke, vol. 35, no. 11, supplement 1, pp. 2683–2686, 2004.
[79]
M. Orio, A. Kunz, T. Kawano, J. Anrather, P. Zhou, and C. Iadecola, “Lipopolysaccharide induces early tolerance to excitotoxicity via nitric oxide and cGMP,” Stroke, vol. 38, no. 10, pp. 2812–2817, 2007.
[80]
K. Tasaki, C. A. Ruetzler, T. Ohtsuki, D. Martin, H. Nawashiro, and J. M. Hallenbeck, “Lipopolysaccharide pre-treatment induces resistance against subsequent focal cerebral ischemic damage in spontaneously hypertensive rats,” Brain Research, vol. 748, no. 1-2, pp. 267–270, 1997.
[81]
E. B. Petcu, T. Kocher, A. Kuhr et al., “Mild systemic inflammation has a neuroprotective effect after stroke in rats,” Current Neurovascular Research, vol. 5, no. 4, pp. 214–223, 2008.
[82]
A. Kunz, L. Park, T. Abe et al., “Neurovascular protection by ischemic tolerance: role of nitric oxide and reactive oxygen species,” Journal of Neuroscience, vol. 27, no. 27, pp. 7083–7093, 2007.
[83]
H. L. Rosenzweig, N. S. Lessov, D. C. Henshall, M. Minami, R. P. Simon, and M. P. Stenzel-Poore, “Endotoxin preconditioning prevents cellular inflammatory response during ischemic neuroprotection in mice,” Stroke, vol. 35, no. 11, pp. 2576–2581, 2004.
[84]
D. Della Morte, P. Abete, F. Gallucci et al., “Transient ischemic attack before nonlacunar ischemic stroke in the elderly,” Journal of Stroke and Cerebrovascular Diseases, vol. 17, no. 5, pp. 257–262, 2008.
[85]
C. Iadecola and M. Alexander, “Cerebral ischemia and inflammation,” Current Opinion in Neurology, vol. 14, no. 1, pp. 89–94, 2001.
[86]
S. Doré, T. Otsuka, T. Mito et al., “Neuronal overexpression of cyclooxygenase-2 increases cerebral infarction,” Annals of Neurology, vol. 54, no. 2, pp. 155–162, 2003.
[87]
F. R. Sharp, A. Lu, Y. Tang, and D. E. Millhorn, “Multiple molecular penumbras after focal cerebral ischemia,” Journal of Cerebral Blood Flow & Metabolism, vol. 20, no. 7, pp. 1011–1032, 2000.
[88]
K. Sriram and J. P. O'Callaghan, “Divergent roles for tumor necrosis factor-α in the brain,” Journal of Neuroimmune Pharmacology, vol. 2, no. 2, pp. 140–153, 2007.
[89]
K. Strle, J. H. Zhou, W. H. Shen et al., “Interleukin-10 in the brain,” Critical Reviews in Immunology, vol. 21, no. 5, pp. 427–449, 2001.
[90]
E. van Exel, J. Gussekloo, A. J. M. De Craen, A. Bootsma-van Der Wiel, M. Fr?lich, and R. G. J. Westendorp, “Inflammation and stroke: the Leiden 85-Plus Study,” Stroke, vol. 33, no. 4, pp. 1135–1138, 2002.
[91]
M. Makwana, L. L. Jones, D. Cuthill et al., “Endogenous transforming growth factor β1 suppresses inflammation and promotes survival in adult CNS,” Journal of Neuroscience, vol. 27, no. 42, pp. 11201–11213, 2007.
[92]
E. Lehrmann, R. Kiefer, T. Christensen et al., “Microglia and macrophages are major sources of locally produced transforming growth factor-β1 after transient middle cerebral artery occlusion in rats,” GLIA, vol. 24, no. 4, pp. 437–448, 1998.
[93]
L. Pang, W. Ye, X. M. Che, B. J. Roessler, A. L. Betz, and G. Y. Yang, “Reduction of inflammatory response in the mouse brain with adenoviral-mediated transforming growth factor-β1 expression,” Stroke, vol. 32, no. 2, pp. 544–552, 2001.
[94]
K. M. Dhandapani and D. W. Brann, “Transforming growth factor-β: a neuroprotective factor in cerebral ischemia,” Cell Biochemistry and Biophysics, vol. 39, no. 1, pp. 13–22, 2003.
[95]
A. Popa-Wagner, K. St?cker, A. T. Balseanu et al., “Effects of granulocyte-colony stimulating factor after stroke in aged rats,” Stroke, vol. 41, no. 5, pp. 1027–1031, 2010.
[96]
Y. Sugiyama, Y. Yagita, N. Oyama, et al., “Granulocyte colony-stimulating factor enhances arteriogenesis and ameliorates cerebral damage in a mouse model of ischemic stroke,” Stroke, vol. 42, no. 3, pp. 770–775, 2011.
[97]
A. Floel, T. Warnecke, T. Duning, et al., “Granulocyte-colony stimulating factor (G-CSF) in stroke patients with concomitant vascular disease-a randomized controlled trial,” PLoS One, vol. 6, no. 5, Article ID e19767, 2011.
[98]
W. R. Sch?bitz, R. Laage, G. Vogt et al., “AXIS: a trial of intravenous granulocyte colony-stimulating factor in acute ischemic stroke,” Stroke, vol. 41, no. 11, pp. 2545–2551, 2010.
[99]
M. Endres, U. Laufs, J. K. Liao, and M. A. Moskowitz, “Targeting eNOS for stroke protection,” Trends in Neurosciences, vol. 27, no. 5, pp. 283–289, 2004.
[100]
I. Badan, B. Buchhold, A. Hamm et al., “Accelerated glial reactivity to stroke in aged rats correlates with reduced functional recovery,” Journal of Cerebral Blood Flow & Metabolism, vol. 23, no. 7, pp. 845–854, 2003.
[101]
K. P. Doyle, E. Cekanaviciute, L. E. Mamer, and M. S. Buckwalter, “TGFβ signaling in the brain increases with aging and signals to astrocytes and innate immune cells in the weeks after stroke,” Journal of Neuroinflammation, vol. 7, article 62, 2010.
[102]
M. Minami and M. Satoh, “Chemokines and their receptors in the brain: pathophysiological roles in ischemic brain injury,” Life Sciences, vol. 74, no. 2-3, pp. 321–327, 2003.
[103]
S. Yamagami, M. Tamura, M. Hayashi et al., “Differential production of MCP-1 and cytokine-induced neutrophil chemoattractant in the ischemic brain after transient focal ischemia in rats,” Journal of Leukocyte Biology, vol. 65, no. 6, pp. 744–749, 1999.
[104]
B. W. McColl, N. J. Rothwell, and S. M. Allan, “Systemic inflammatory stimulus potentiates the acute phase and CXC chemokine responses to experimental stroke and exacerbates brain damage via interleukin-1- and neutrophil-dependent mechanisms,” Journal of Neuroscience, vol. 27, no. 16, pp. 4403–4412, 2007.
[105]
S. J. Campbell, V. H. Perry, F. J. Pitossi et al., “Central nervous system injury triggers hepatic CC and CXC chemokine expression that is associated with leukocyte mobilization and recruitment to both the central nervous system and the liver,” The American Journal of Pathology, vol. 166, no. 5, pp. 1487–1497, 2005.
[106]
N. Kumagai, Y. Chiba, M. Hosono et al., “Involvement of pro-inflammatory cytokines and microglia in an age-associated neurodegeneration model, the SAMP10 mouse,” Brain Research, vol. 1185, no. 1, pp. 75–85, 2007.
[107]
V. A. DiNapoli, J. D. Huber, K. Houser, X. Li, and C. L. Rosen, “Early disruptions of the blood-brain barrier may contribute to exacerbated neuronal damage and prolonged functional recovery following stroke in aged rats,” Neurobiology of Aging, vol. 29, no. 5, pp. 753–764, 2008.
[108]
S. M. Stamatovic, P. Shakui, R. F. Keep et al., “Monocyte chemoattractant protein-1 regulation of blood-brain barrier permeability,” Journal of Cerebral Blood Flow & Metabolism, vol. 25, no. 5, pp. 593–606, 2005.
[109]
N. Kostulas, P. Kivis?kk, Y. Huang, D. Matusevicius, V. Kostulas, and H. Link, “Ischemic stroke is associated with a systemic increase of blood mononuclear cells expressing interleukin-8 mRNA,” Stroke, vol. 29, no. 2, pp. 462–466, 1998.
[110]
T. Matsumoto, K. Ikeda, N. Mukaida et al., “Prevention of cerebral edema and infarct in cerebral reperfusion injury by an antibody to interleukin-8,” Laboratory Investigation, vol. 77, no. 2, pp. 119–125, 1997.
[111]
J. Dawson, W. Miltz, A. K. Mir, and C. Wiessner, “Targeting monocyte chemoattractant protein-1 signalling in disease,” Expert Opinion on Therapeutic Targets, vol. 7, no. 1, pp. 35–48, 2003.
[112]
J. S. Beech, J. Reckless, D. E. Mosedale, D. J. Grainger, S. C. Williams, and D. K. Menon, “Neuroprotection in ischemia-reperfusion injury: an antiinflammatory approach using a novel broad-spectrum chemokine inhibitor,” Journal of Cerebral Blood Flow & Metabolism, vol. 21, no. 6, pp. 683–689, 2001.
[113]
N. Mitsios, M. Saka, J. Krupinski et al., “A microarray study of gene and protein regulation in human and rat brain following middle cerebral artery occlusion,” BMC Neuroscience, vol. 8, article 93, 2007.
[114]
N. Kawahara, Y. Wang, A. Mukasa, et al., “Genome-wide gene expression analysis for induced ischemic tolerance and delayed neuronal death following transient global ischemia in rats,” Journal of Cerebral Blood Flow & Metabolism, vol. 24, no. 2, pp. 212–223, 2004.
[115]
M. Schwaninger, I. Inta, and O. Herrmann, “NF-κB signalling in cerebral ischaemia,” Biochemical Society Transactions, vol. 34, no. 6, pp. 1291–1294, 2006.
[116]
J. R. Patel and G. J. Brewer, “Age-related differences in NFkappaB translocation and Bcl-2/Bax ratio caused by TNFalpha and Abeta42 promote survival in middle-age neurons and death in old neurons,” Experimental Neurology, vol. 213, no. 1, pp. 93–100, 2008.
[117]
W. Zhang, I. Potrovita, V. Tarabin, et al., “Neuronal activation of NF-kappaB contributes to cell death in cerebral ischemia,” Journal of Cerebral Blood Flow & Metabolism, vol. 25, no. 1, pp. 30–40, 2005.
[118]
D. Stephenson, T. Yin, E. B. Smalstig, et al., “Transcription factor nuclear factor-kappa B is activated in neurons after focal cerebral ischemia,” Journal of Cerebral Blood Flow & Metabolism, vol. 20, no. 3, pp. 592–603, 2000.
[119]
J. L. Luo, H. Kamata, and M. Karin, “The anti-death machinery in IKK/NF-κB signaling,” Journal of Clinical Immunology, vol. 25, no. 6, pp. 541–550, 2005.
[120]
N. D. Perkins, “The Rel/NF-κB family: friend and foe,” Trends in Biochemical Sciences, vol. 25, no. 9, pp. 434–440, 2000.
[121]
J. Li, Z. Lu, W. L. Li, P. Y. Shan, and L. Wei, “Cell death and proliferation in NF-κB p50 knockout mouse after cerebral ischemia,” Brain Research, vol. 1230, pp. 281–289, 2008.
[122]
A. J. Williams, J. R. Dave, and F. C. Tortella, “Neuroprotection with the proteasome inhibitor MLN519 in focal ischemic brain injury: relation to nuclear factor κB (NF-κB), inflammatory gene expression, and leukocyte infiltration,” Neurochemistry International, vol. 49, no. 2, pp. 106–112, 2006.
[123]
I. M. Shah and M. Di Napoli, “The ubiquitin-proteasome system and proteasome inhibitors in central nervous system diseases,” Cardiovascular & Hematological Disorders Drug Targets, vol. 7, no. 4, pp. 250–273, 2007.
[124]
B. Florian, R. Vintilescu, A. T. Balseanu et al., “Long-term hypothermia reduces infarct volume in aged rats after focal ischemia,” Neuroscience Letters, vol. 438, no. 2, pp. 180–185, 2008.
[125]
M. R. Macleod, J. Petersson, B. Norrving et al., “Hypothermia for Stroke: call to action 2010,” International Journal of Stroke, vol. 5, no. 6, pp. 489–492, 2010.
[126]
P. Ge, Y. Luo, C. L. Liu, and B. Hu, “Protein aggregation and proteasome dysfunction after brain ischemia,” Stroke, vol. 38, no. 12, pp. 3230–3236, 2007.
[127]
R. Marfella, C. Di Filippo, M. T. Laieta et al., “Effects of ubiquitin-proteasome system deregulation on the vascular senescence and atherosclerosis process in elderly patients,” Journals of Gerontology—Series A, vol. 63, no. 2, pp. 200–203, 2008.
[128]
S. Lavu, O. Boss, P. J. Elliott, and P. D. Lambert, “Sirtuins—novel therapeutic targets to treat age-associated diseases,” Nature Reviews Drug Discovery, vol. 7, no. 10, pp. 841–853, 2008.
[129]
A. Salminen and K. Kaarniranta, “NF-κB signaling in the aging process,” Journal of Clinical Immunology, vol. 29, no. 4, pp. 397–405, 2009.
[130]
L. Zhang, S. Huang, Y. Chen, Z. Wang, E. Li, and Y. Xu, “Icariin inhibits hydrogen peroxide-mediated cytotoxicity by up-regulating sirtuin type 1-dependent catalase and peroxiredoxin,” Basic & Clinical Pharmacology & Toxicology, vol. 107, no. 5, pp. 899–905, 2010.
[131]
J. A. Shin, H. Lee, Y. K. Lim, Y. Koh, J. H. Choi, and E. M. Park, “Therapeutic effects of resveratrol during acute periods following experimental ischemic stroke,” Journal of Neuroimmunology, vol. 227, no. 1-2, pp. 93–100, 2010.
[132]
A. P. Raval, H. W. Lin, K. R. Dave et al., “Resveratrol and ischemic preconditioning in the brain,” Current Medicinal Chemistry, vol. 15, no. 15, pp. 1545–1551, 2008.
[133]
K. R. Patel, E. Scott, V. A. Brown, A. J. Gescher, W. P. Steward, and K. Brown, “Clinical trials of resveratrol,” Annals of the New York Academy of Sciences, vol. 1215, pp. 161–169, 2011.
[134]
G. Saretzki, “Telomerase, mitochondria and oxidative stress,” Experimental Gerontology, vol. 44, no. 8, pp. 485–492, 2009.
[135]
P. Willeit, J. Willeit, A. Brandstatter, et al., “Cellular aging reflected by leukocyte telomere length predicts advanced atherosclerosis and cardiovascular disease risk,” Arteriosclerosis, Thrombosis, and Vascular Biology, vol. 30, no. 8, pp. 1649–1656, 2010.
[136]
B. Zhang, L. Chen, K. R. Swartz et al., “Deficiency of telomerase activity aggravates the blood-brain barrier disruption and neuroinflammatory responses in a model of experimental stroke,” Journal of Neuroscience Research, vol. 88, no. 13, pp. 2859–2868, 2010.
[137]
J. A. Palacios, D. Herranz, M. L. De Bonis, S. Velasco, M. Serrano, and M. A. Blasco, “SIRT1 contributes to telomere maintenance and augments global homologous recombination,” Journal of Cell Biology, vol. 191, no. 7, pp. 1299–1313, 2010.
[138]
Z. Zheng, J. Y. Kim, H. Ma, J. E. Lee, and M. A. Yenari, “Anti-inflammatory effects of the 70 kDa heat shock protein in experimental stroke,” Journal of Cerebral Blood Flow & Metabolism, vol. 28, no. 1, pp. 53–63, 2008.
[139]
S. H. Lee, M. Kim, B. W. Yoon et al., “Targeted hsp70.1 disruption increases infarction volume after focal cerebral ischemia in mice,” Stroke, vol. 32, no. 12, pp. 2905–2912, 2001.
[140]
D. I. Sinn, K. Chu, S. T. Lee et al., “Pharmacological induction of heat shock protein exerts neuroprotective effects in experimental intracerebral hemorrhage,” Brain Research, vol. 1135, no. 1, pp. 167–176, 2007.
[141]
R. Singh, S. Kolvraa, and S. I. Rattan, “Genetics of human longevity with emphasis on the relevance of HSP70 as candidate genes,” Frontiers in Bioscience, vol. 12, pp. 4504–4513, 2007.
[142]
A. J. L. Macario and E. C. De Macario, “Chaperonopathies by defect, excess, or mistake,” Annals of the New York Academy of Sciences, vol. 1113, pp. 178–191, 2007.
[143]
B. Kalmar and L. Greensmith, “Induction of heat shock proteins for protection against oxidative stress,” Advanced Drug Delivery Reviews, vol. 61, no. 4, pp. 310–318, 2009.
[144]
C. J. M. Frijns and L. J. Kappell, “Inflammatory cell adhesion molecules in ischemic cerebrovascular disease,” Stroke, vol. 33, no. 8, pp. 2115–2122, 2002.
[145]
A. M. Simundic, V. Basic, E. Topic et al., “Soluble adhesion molecules in acute ischemic stroke,” Clinical and Investigative Medicine, vol. 27, no. 2, pp. 86–92, 2004.
[146]
E. S. Connolly, C. J. Winfree, C. J. Prestigiacomo et al., “Exacerbation of cerebral injury in mice that express the P-selectin gene: identification of P-selectin blockade as a new target for the treatment of stroke,” Circulation Research, vol. 81, no. 3, pp. 304–310, 1997.
[147]
A. V. Goussev, Z. Zhang, D. C. Anderson, and M. Chopp, “P-selectin antibody reduces hemorrhage and infarct volume resulting from MCA occlusion in the rat,” Journal of the Neurological Sciences, vol. 161, no. 1, pp. 16–22, 1998.
[148]
H. Suzuki, T. Hayashi, S. J. Tojo et al., “Anti-P-selectin antibody attenuates rat brain ischemic injury,” Neuroscience Letters, vol. 265, no. 3, pp. 163–166, 1999.
[149]
A. Kalinowska and J. Losy, “PECAM-1, a key player in neuroinflammation,” European Journal of Neurology, vol. 13, no. 12, pp. 1284–1290, 2006.
[150]
V. Richter, F. Rassoul, K. Purschwitz, B. Hentschel, W. Reuter, and T. Kuntze, “Circulating vascular cell adhesion molecules VCAM-1, ICAM-1, and E-selectin in dependence on aging,” Gerontology, vol. 49, no. 5, pp. 293–300, 2003.
[151]
R. L. Zhang, M. Chopp, Y. Li et al., “Anti-ICAM-1 antibody reduces ischemic cell damage after transient middle cerebral artery occlusion in the rat,” Neurology, vol. 44, no. 9, pp. 1747–1751, 1994.
[152]
M. E. Sughrue, A. Mehra, E. S. Connolly Jr., and A. L. D'Ambrosio, “Anti-adhesion molecule strategies as potential neuroprotective agents in cerebral ischemia: a critical review of the literature,” Inflammation Research, vol. 53, no. 10, pp. 497–508, 2004.
[153]
S. A. Mousa, “Cell adhesion molecules: potential therapeutic & diagnostic implications,” Molecular Biotechnology, vol. 38, no. 1, pp. 33–40, 2008.
[154]
C. J. Price, D. K. Menon, A. M. Peters et al., “Cerebral neutrophil recruitment, histology, and outcome in acute ischemic stroke: an imaging-based study,” Stroke, vol. 35, no. 7, pp. 1659–1664, 2004.
[155]
V. A. DiNapoli, S. A. Benkovic, X. Li et al., “Age exaggerates proinflammatory cytokine signaling and truncates signal transducers and activators of transcription 3 signaling following ischemic stroke in the rat,” Neuroscience, vol. 170, no. 2, pp. 633–644, 2010.
[156]
R. D. Engberink, E. L. Blezer, E. I. Hoff, et al., “MRI of monocyte infiltration in an animal model of neuroinflammation using SPIO-labeled monocytes or free USPIO,” Journal of Cerebral Blood Flow & Metabolism, vol. 28, no. 4, pp. 841–851, 2008.
[157]
A. Denes, R. Vidyasagar, J. Feng, et al., “Proliferating resident microglia after focal cerebral ischaemia in mice,” Journal of Cerebral Blood Flow & Metabolism, vol. 27, no. 12, pp. 1941–1953, 2007.
[158]
A. Denes, P. Thornton, N. J. Rothwell, and S. M. Allan, “Inflammation and brain injury: acute cerebral ischaemia, peripheral and central inflammation,” Brain, Behavior, and Immunity, vol. 24, no. 5, pp. 708–723, 2010.
[159]
A. Thiel, B. A. Radlinska, C. Paquette et al., “The temporal dynamics of poststroke neuroinflammation: a longitudinal diffusion tensor imaging-guided PET study with 11C-PK11195 in acute subcortical stroke,” Journal of Nuclear Medicine, vol. 51, no. 9, pp. 1404–1412, 2010.
[160]
C. T. Ekdahl, Z. Kokaia, and O. Lindvall, “Brain inflammation and adult neurogenesis: the dual role of microglia,” Neuroscience, vol. 158, no. 3, pp. 1021–1029, 2009.
[161]
M. Lalancette-Hébert, G. Gowing, A. Simard, C. W. Yuan, and J. Kriz, “Selective ablation of proliferating microglial cells exacerbates ischemic injury in the brain,” Journal of Neuroscience, vol. 27, no. 10, pp. 2596–2605, 2007.
[162]
J. R. Caso, J. M. Pradillo, O. Hurtado, J. C. Leza, M. A. Moro, and I. Lizasoain, “Toll-like receptor 4 is involved in subacute stress-induced neuroinflammation and in the worsening of experimental stroke,” Stroke, vol. 39, no. 4, pp. 1314–1320, 2008.
[163]
D. van Duin and A. C. Shaw, “Toll-like receptors in older adults,” Journal of the American Geriatrics Society, vol. 55, no. 9, pp. 1438–1444, 2007.
[164]
R. Hartl, L. Schurer, G. W. Schmid-Schonbein, and G. J. del Zoppo, “Experimental antileukocyte interventions in cerebral ischemia,” Journal of Cerebral Blood Flow & Metabolism, vol. 16, no. 6, pp. 1108–1119, 1996.
[165]
S. Jander, M. Schroeter, and A. Saleh, “Imaging inflammation in acute brain ischemia,” Stroke, vol. 38, no. 2, pp. 642–645, 2007.
[166]
P. H. Chan, “Reactive oxygen radicals in signaling and damage in the ischemic brain,” Journal of Cerebral Blood Flow & Metabolism, vol. 21, no. 1, pp. 2–14, 2001.
[167]
K. Xu, M. A. Puchowicz, X. Sun, and J. C. Lamanna, “Mitochondrial dysfunction in aging rat brain following transient global ischemia,” Advances in Experimental Medicine and Biology, vol. 614, pp. 379–386, 2008.
[168]
A Canuelo, E. Siles, R. Martinez-Romero, M. A. Peinado, and E. Martinez-Lara, “The nitric oxide system response to hypoxia/reoxygenation in the aged cerebral cortex,” Experimental Gerontology, vol. 42, no. 12, pp. 1137–1145, 2007.
[169]
Z. Huang, P. L. Huang, J. Ma, et al., “Enlarged infarcts in endothelial nitric oxide synthase knockout mice are attenuated by nitro-L-arginine,” Journal of Cerebral Blood Flow & Metabolism, vol. 16, no. 5, pp. 981–987, 1996.
[170]
H. Hara, P. L. Huang, N. Panahian, M. C. Fishman, and M. A. Moskowitz, “Reduced brain edema and infarction volume in mice lacking the neuronal isoform of nitric oxide synthase after transient MCA occlusion,” Journal of Cerebral Blood Flow & Metabolism, vol. 16, no. 4, pp. 605–611, 1996.
[171]
M. J. Eliasson, Z. Huang, R. J. Ferrante et al., “Neuronal nitric oxide synthase activation and peroxynitrite formation in ischemic stroke linked to neural damage,” Journal of Neuroscience, vol. 19, no. 14, pp. 5910–5918, 1999.
[172]
E. Martinez-Lara, A. R. Canuelo, E. Siles, et al., “Constitutive nitric oxide synthases are responsible for the nitric oxide production in the ischemic aged cerebral cortex,” Brain Research, vol. 1054, no. 1, pp. 88–94, 2005.
[173]
P. A. Lapchak and D. M. Araujo, “Advances in ischemic stroke treatment: neuroprotective and combination therapies,” Expert Opinion on Emerging Drugs, vol. 12, no. 1, pp. 97–112, 2007.
[174]
K. Qu, C. P. Chen, B. Halliwell, P. K. Moore, and P. T. H. Wong, “Hydrogen sulfide is a mediator of cerebral ischemic damage,” Stroke, vol. 37, no. 3, pp. 889–893, 2006.
[175]
P. T. Wong, K. Qu, G. N. Chimon, et al., “High plasma cyst(e)ine level may indicate poor clinical outcome in patients with acute stroke: possible involvement of hydrogen sulfide,” Journal of Neuropathology & Experimental Neurology, vol. 65, no. 2, pp. 109–115, 2006.
[176]
L. F. Hu, P. T. H. Wong, P. K. Moore, and J. S. Bian, “Hydrogen sulfide attenuates lipopolysaccharide-induced inflammation by inhibition of p38 mitogen-activated protein kinase in microglia,” Journal of Neurochemistry, vol. 100, no. 4, pp. 1121–1128, 2007.
[177]
E. Lowicka and J. Beltowski, “Hydrogen sulfide (H2S)—the third gas of interest for pharmacologists,” Pharmacological Reports, vol. 59, no. 1, pp. 4–24, 2007.
[178]
H. Graeme, “B vitamins in patients with recent transient ischaemic attack or stroke in the VITAmins TO Prevent Stroke (VITATOPS) trial: a randomised, double-blind, parallel, placebo-controlled trial,” The Lancet Neurology, vol. 9, no. 9, pp. 855–865, 2010.
[179]
H. D. Sesso, J. E. Buring, W. G. Christen et al., “Vitamins E and C in the prevention of cardiovascular disease in men: the physicians' health study II randomized controlled trial,” Journal of the American Medical Association, vol. 300, no. 18, pp. 2123–2133, 2008.
[180]
H. Hemila and J. Kaprio, “Vitamin E may affect the life expectancy of men, depending on dietary vitamin C intake and smoking,” Age Ageing, vol. 40, no. 2, pp. 215–220, 2011.
[181]
A. R. Green, “Pharmacological approaches to acute ischaemic stroke: reperfusion certainly, neuroprotection possibly,” The British Journal of Pharmacology, vol. 153, supplement 1, pp. S325–S338, 2008.
[182]
P. A. Lapchak, “A critical assessment of edaravone acute ischemic stroke efficacy trials: Is edaravone an effective neuroprotective therapy?” Expert Opinion on Pharmacotherapy, vol. 11, no. 10, pp. 1753–1763, 2010.
[183]
K. Yagi, K. T. Kitazato, M. Uno et al., “Edaravone, a free radical scavenger, inhibits MMP-9-related brain hemorrhage in rats treated with tissue plasminogen activator,” Stroke, vol. 40, no. 2, pp. 626–631, 2009.
[184]
B. J. Lee, Y. Egi, K. van Leyen, E. H. Lo, and K. Arai, “Edaravone, a free radical scavenger, protects components of the neurovascular unit against oxidative stress in vitro,” Brain Research, vol. 1307, pp. 22–27, 2010.
[185]
S. Amaro, A. M. Planas, and A. Chamorro, “Uric acid administration in patients with acute stroke: a novel approach to neuroprotection,” Expert Review of Neurotherapeutics, vol. 8, no. 2, pp. 259–270, 2008.
[186]
F. P. Gonzalez-Ibarra, J Varon, and E. G. Lopez-Meza, “Therapeutic hypothermia: critical review of the molecular mechanisms of action,” Frontiers in Neurology, vol. 2, p. 4, 2011.
[187]
X. Ji, Y. Luo, F. Ling, et al., “Mild hypothermia diminishes oxidative DNA damage and pro-death signaling events after cerebral ischemia: a mechanism for neuroprotection,” Frontiers in Bioscience, vol. 12, pp. 1737–1747, 2007.
[188]
B. Florian, R. Vintilescu, A. T. Balseanu et al., “Long-term hypothermia reduces infarct volume in aged rats after focal ischemia,” Neuroscience Letters, vol. 438, no. 2, pp. 180–185, 2008.
[189]
W. D. Dietrich, C. M. Atkins, and H. M. Bramlett, “Protection in animal models of brain and spinal cord injury with mild to moderate hypothermia,” Journal of Neurotrauma, vol. 26, no. 3, pp. 301–312, 2009.
[190]
P. D. Lyden, D. Krieger, M. Yenari, and W. D. Dietrich, “Therapeutic hypothermia for acute stroke,” International Journal of Stroke, vol. 1, no. 1, pp. 9–19, 2006.
[191]
X. N. Tang, L. Liu, and M. A. Yenari, “Combination therapy with hypothermia for treatment of cerebral ischemia,” Journal of Neurotrauma, vol. 26, no. 3, pp. 325–331, 2009.
[192]
H. Ohta, Y. Terao, Y. Shintani, and Y. Kiyota, “Therapeutic time window of post-ischemic mild hypothermia and the gene expression associated with the neuroprotection in rat focal cerebral ischemia,” Neuroscience Research, vol. 57, no. 3, pp. 424–433, 2007.
[193]
J. H. Heo, J. Lucero, T. Abumiya, J. A. Koziol, B. R. Copeland, and G. J. del Zoppo, “Matrix metalloproteinases increase very early during experimental focal cerebral ischemia,” Journal of Cerebral Blood Flow & Metabolism, vol. 19, no. 6, pp. 624–633, 1999.
[194]
J. Montaner, J. Alvarez-Sabín, C. Molina et al., “Matrix metalloproteinase expression after human cardioembolic stroke: temporal profile and relation to neurological impairment,” Stroke, vol. 32, no. 8, pp. 1759–1766, 2001.
[195]
M. Asahi, K. Asahi, J. C. Jung, G. J. del Zoppo, M. E. Fini, and E. H. Lo, “Role for matrix metalloproteinase 9 after focal cerebral ischemia: effects of gene knockout and enzyme inhibition with BB-94,” Journal of Cerebral Blood Flow & Metabolism, vol. 20, no. 12, pp. 1681–1689, 2000.
[196]
P. Svedin, H. Hagberg, K. S?vman, C. Zhu, and C. Mallard, “Matrix metalloproteinase-9 gene knock-out protects the immature brain after cerebral hypoxia-ischemia,” Journal of Neuroscience, vol. 27, no. 7, pp. 1511–1518, 2007.
[197]
M. Castellanos, R. Leira, J. Serena et al., “Plasma metalloproteinase-9 concentration predicts hemorrhagic transformation in acute ischemic stroke,” Stroke, vol. 34, no. 1, pp. 40–46, 2003.
[198]
J. Montaner, C. A. Molina, J. Monasterio et al., “Matrix metalloproteinase-9 pretreatment level predicts intracranial hemorrhagic complications after thrombolysis in human stroke,” Circulation, vol. 107, no. 4, pp. 598–603, 2003.
[199]
R. R. Sood, S. Taheri, E. Candelario-Jalil, E. Y. Estrada, and G. A. Rosenberg, “Early beneficial effect of matrix metalloproteinase inhibition on blood-brain barrier permeability as measured by magnetic resonance imaging countered by impaired long-term recovery after stroke in rat brain,” Journal of Cerebral Blood Flow & Metabolism, vol. 28, no. 2, pp. 431–438, 2008.
[200]
M. Asahi, K. Asahi, J. C. Jung, G. J. del Zoppo, M. E. Fini, and E. H. Lo, “Role for matrix metalloproteinase 9 after focal cerebral ischemia: effects of gene knockout and enzyme inhibition with BB-94,” Journal of Cerebral Blood Flow & Metabolism, vol. 20, no. 12, pp. 1681–1689, 2000.
[201]
X. F. Jiang, S. Namura, and I. Nagata, “Matrix metalloproteinase inhibitor KB-R7785 attenuates brain damage resulting from permanent focal cerebral ischemia in mice,” Neuroscience Letters, vol. 305, no. 1, pp. 41–44, 2001.
[202]
Z. Gu, J. Cui, S. Brown et al., “A highly specific inhibitor of matrix metalloproteinase-9 rescues laminin from proteolysis and neurons from apoptosis in transient focal cerebral ischemia,” Journal of Neuroscience, vol. 25, no. 27, pp. 6401–6408, 2005.
[203]
M. Asahi, T. Sumii, M. E. Fini, S. Itohara, and E. H. Lo, “Matrix metalloproteinase 2 gene knockout has no effect on acute brain injury after focal ischemia,” NeuroReport, vol. 12, no. 13, pp. 3003–3007, 2001.
[204]
O. B. Dimitrijevic, S. M. Stamatovic, R. F. Keep, and A. V. Andjelkovic, “Effects of the chemokine CCL2 on blood-brain barrier permeability during ischemia-reperfusion injury,” Journal of Cerebral Blood Flow & Metabolism, vol. 26, no. 6, pp. 797–810, 2006.
[205]
S. Wagner, M. Tagaya, J. A. Koziol, V. Quaranta, and G. J. Del Zoppo, “Rapid disruption of an astrocyte interaction with the extracellular matrix mediated by integrin α6β4 during focal cerebral ischemia/reperfusion,” Stroke, vol. 28, no. 4, pp. 858–865, 1997.
[206]
L. T. Grinberg and D. R. Thal, “Vascular pathology in the aged human brain,” Acta Neuropathologica, vol. 119, no. 3, pp. 277–290, 2010.
[207]
J. M. Wardlaw, F. Doubal, P. Armitage et al., “Lacunar stroke is associated with diffuse blood-brain barrier dysfunction,” Annals of Neurology, vol. 65, no. 2, pp. 194–202, 2009.
[208]
V. Vasilevko, G. F. Passos, D. Quiring et al., “Aging and cerebrovascular dysfunction: contribution of hypertension, cerebral amyloid angiopathy, and immunotherapy,” Annals of the New York Academy of Sciences, vol. 1207, pp. 58–70, 2010.
[209]
T. N. Nagaraja, K. A. Keenan, J. D. Fenstermacher, and R. A. Knight, “Acute leakage patterns of fluorescent plasma flow markers after transient focal cerebral ischemia suggest large openings in blood-brain barrier,” Microcirculation, vol. 15, no. 1, pp. 1–14, 2008.
[210]
N. Zeevi, J. Chhabra, I. E. Silverman, N. S. Lee, and L. D. McCullough, “Acute stroke management in the elderly,” Cerebrovascular Diseases, vol. 23, no. 4, pp. 304–308, 2007.
[211]
N. Zeevi, J. Pachter, L. D. McCullough, L. Wolfson, and G. A. Kuchel, “The blood-brain barrier: geriatric relevance of a critical brain-body interface,” Journal of the American Geriatrics Society, vol. 58, no. 9, pp. 1749–1757, 2010.
[212]
D. Israeli, D. Tanne, D. Daniels et al., “The application of MRI for depiction of subtle blood brain barrier disruption in stroke,” International Journal of Biological Sciences, vol. 6, no. 7, pp. 845–852, 2010.
[213]
H. C. Emsley, C. J. Smith, C. M. Gavin et al., “An early and sustained peripheral inflammatory response in acute ischaemic stroke: relationships with infection and atherosclerosis,” Journal of Neuroimmunology, vol. 139, no. 1-2, pp. 93–101, 2003.
[214]
Y. Yoshimoto, Y. Tanaka, and K. Hoya, “Acute systemic inflammatory response syndrome in subarachnoid hemorrhage,” Stroke, vol. 32, no. 9, pp. 1989–1993, 2001.
[215]
H. C. Emsley, C. J. Smith, R. F. Georgiou et al., “Correlation of systemic inflammatory response with infarct volume in acute ischemic stroke patients,” Stroke, vol. 36, no. 2, pp. 228–229, 2005.
[216]
P. E. Marchiori, A. M. M. Lino, M. T. A. Hirata, N. B. Carvalho, M. W. I. Brotto, and M. Scaff, “Occurrence of nervous system involvement in SIRS,” Journal of the Neurological Sciences, vol. 250, no. 1-2, pp. 147–152, 2006.
[217]
A. A. Wong, J. P. Davis, P. J. Schluter, R. D. Henderson, J. D. O'Sullivan, and S. J. Read, “The time course and determinants of temperature within the first 48 h after ischaemic stroke,” Cerebrovascular Diseases, vol. 24, no. 1, pp. 104–110, 2007.
[218]
R. Leira, M. Rodriguez-Yanez, M. Castellanos, et al., “Hyperthermia is a surrogate marker of inflammation-mediated cause of brain damage in acute ischaemic stroke,” Journal of Internal Medicine, vol. 260, no. 4, pp. 343–349, 2006.
[219]
R. Noor, C. X. Wang, and A. Shuaib, “Effects of hyperthermia on infarct volume in focal embolic model of cerebral ischemia in rats,” Neuroscience Letters, vol. 349, no. 2, pp. 130–132, 2003.
[220]
T. M. Hemmen, R. Raman, K. Z. Guluma et al., “Intravenous thrombolysis plus hypothermia for acute treatment of ischemic stroke (ICTuS-L): final results,” Stroke, vol. 41, no. 10, pp. 2265–2270, 2010.
[221]
H. C. Emsley and S. J. Hopkins, “Acute ischaemic stroke and infection: recent and emerging concepts,” The Lancet Neurology, vol. 7, no. 4, pp. 341–353, 2008.
[222]
A. Z. Sheng, Q. Shen, D. Cordato, Y. Y. Zhang, and D. K. Yin Chan, “Delirium within three days of stroke in a cohort of elderly patients,” Journal of the American Geriatrics Society, vol. 54, no. 8, pp. 1192–1198, 2006.
[223]
S. N. Whitehead, G. Cheng, V. C. Hachinski, and D. F. Cechetto, “Progressive increase in infarct size, neuroinflammation, and cognitive deficits in the presence of high levels of amyloid,” Stroke, vol. 38, no. 12, pp. 3245–3250, 2007.
[224]
L. S. Rothenburg, N. Herrmann, W. Swardfager et al., “The relationship between inflammatory markers and post stroke cognitive impairment,” Journal of Geriatric Psychiatry and Neurology, vol. 23, no. 3, pp. 199–205, 2010.
[225]
B. Buchhold, L. Mogoanta, Y. Suofu et al., “Environmental enrichment improves functional and neuropathological indices following stroke in young and aged rats,” Restorative Neurology and Neuroscience, vol. 25, no. 5-6, pp. 467–484, 2007.
[226]
R. A. Lobo, “Menopause and stroke and the effects of hormonal therapy,” Climacteric, vol. 10, no. 2, pp. 27–31, 2007.
[227]
M. Liu, S. Dziennis, P. D. Hurn, and N. J. Alkayed, “Mechanisms of gender-linked ischemic brain injury,” Restorative Neurology and Neuroscience, vol. 27, no. 3, pp. 163–179, 2009.
[228]
S. Basaria, A. D. Coviello, T. G. Travison et al., “Adverse events associated with testosterone administration,” The New England Journal of Medicine, vol. 363, no. 2, pp. 109–122, 2010.
[229]
T. Singh and A. B. Newman, “Inflammatory markers in population studies of aging,” Ageing Research Reviews, vol. 10, no. 3, pp. 319–329, 2010.
[230]
C. L. Wassel, E. Barrett-Connor, and G. A. Laughlin, “Association of circulating C-reactive protein and interleukin-6 with longevity into the 80s and 90s: the Rancho Bernardo Study,” Journal of Cerebral Blood Flow & Metabolism, vol. 95, no. 10, pp. 4748–4755, 2010.
[231]
M. Di Napoli, M. Schwaninger, R. Cappelli et al., “Evaluation of C-reactive protein measurement for assessing the risk and prognosis in ischemic stroke: a statement for health care professionals from the CRP Pooling Project members,” Stroke, vol. 36, no. 6, pp. 1316–1329, 2005.
[232]
L. S. Rallidis, M. Vikelis, D. B. Panagiotakos, G. K. Liakos, E. Krania, and D. T. Kremastinos, “Usefulness of inflammatory and haemostatic markers to predict short-term risk for death in middle-aged ischaemic stroke patients,” Acta Neurologica Scandinavica, vol. 117, no. 6, pp. 415–420, 2008.
[233]
K. Winbeck, H. Poppert, T. Etgen, B. Conrad, and D. Sander, “Prognostic relevance of early serial C-reactive protein measurements after first ischemic stroke,” Stroke, vol. 33, no. 10, pp. 2459–2464, 2002.
[234]
N. S. Rost, P. A. Wolf, C. S. Kase et al., “Plasma concentration of C-reactive protein and risk of ischemic stroke and transient ischemic attack: the Framingham study,” Stroke, vol. 32, no. 11, pp. 2575–2579, 2001.
[235]
P. M. Ridker, “The time for cardiovascular inflammation reduction trials has arrived: how low to go for hsCRP?” Arteriosclerosis, Thrombosis, and Vascular Biology, vol. 28, no. 7, pp. 1222–1224, 2008.
[236]
Y. Shintani, Y. Terao, and H. Ohta, “Molecular mechanisms underlying hypothermia-induced neuroprotection,” Stroke Research and Treatment, vol. 2011, Article ID 809874, 9 pages, 2011.