Skip to main content
Erschienen in: Translational Stroke Research 2/2011

Open Access 01.06.2011 | Review

Are Underlying Assumptions of Current Animal Models of Human Stroke Correct: from STAIRs to High Hurdles?

verfasst von: Renée J. Turner, Glen C. Jickling, Frank R. Sharp

Erschienen in: Translational Stroke Research | Ausgabe 2/2011

Abstract

Animal models of acute ischemic stroke have been criticized for failing to translate to human stroke. Nevertheless, animal models are necessary to improve our understanding of stroke pathophysiology and to guide the development of new stroke therapies. The rabbit embolic clot model is one animal model that has led to an effective therapy in human acute ischemic stroke, namely tissue plasminogen activator (tPA). We propose that potential compounds that demonstrate efficacy in non-rabbit animal models of acute ischemic stroke should also be tested in the rabbit embolic blood clot model and, where appropriate, compared to tPA prior to investigation in humans. Furthermore, the use of anesthesia needs to be considered as a major confounder in animal models of acute ischemic stroke, and death should be included as an outcome measure in animal stroke studies. These steps, along with the current STAIRs recommendations, may improve the successful translation of experimental therapies to clinical stroke treatments.
Abkürzungen
ApoE
Apolipoprotein E
FDA
Food and Drug Administration
HDL
High-density lipoprotein
ICAM-1
Intercellular adhesion molecule 1
LDL
Low-density lipoprotein
NXY-059
Free radical scavenger
RSCEM
Rabbit small clot embolism model
rt-PA
Recombinant tissue plasminogen activator
SAINT I/II
Stroke Acute Ischemic Stroke NXY-059 Trial
STAIR
Stroke Therapy Academic Industry Roundtable
tPA
Tissue plasminogen activator

Introduction

The uncertainty as to why animal studies of acute cerebral ischemia fail to translate into human stroke treatments continues with NXY-059 in the SAINT II trial [1, 2]. The Stroke Therapy Academic Industry Roundtable (STAIR) recommendations [38] have identified important issues in the experimental modeling of ischemic stroke and have sought to promote the translation of animal studies to successful human stroke trials. The STAIR recommendations outline suggestions to optimally pre-clinically assess potential neuroprotective and restorative drugs for the treatment of acute ischemic stroke. Briefly, they include recommendations for drug dose, therapeutic window, choice of animal model, physiological monitoring, outcome measures, and sex differences, amongst others. However, despite such recommendations successful experimental to clinical translation has yet to be achieved. This suggests that further modifications and changes may be required to experimental paradigms in order to achieve such results. Rather than assess these criteria, or address the many excellent reviews in the literature [912], we instead seek to highlight features of stroke models that may have been underestimated, and some assumptions that have been made in the development of animal models of acute ischemic stroke and the testing of neuroprotective agents. The goal is to promote discussion and possible modification of models to ultimately improve the translation of experimental stroke studies to the clinical setting [9, 10, 1320].
The inconsistency amongst animal models, in addition to the lack of a clear progression of testing to humans has proved a major obstacle in the translation of treatments from animal models to human stroke. One experimental model which has not been adequately considered in experimental stroke research is the rabbit embolic clot model [21]. To date, this is one of the only models which has yielded/predicted a treatment approved for use in acute ischemic stroke in humans—tissue plasminogen activator (tPA) [22]. Notably, however, tPA was first shown to be efficacious in rodent stroke models [2330]. Thus, there may be some value in comparing the two models and addressing whether one is “better” than the other particularly since the rabbit model is seldom used. Even though tPA improves outcomes in both the rabbit and rodent models, this does not mean that either model actually would translate to humans. For example, just because hundreds of neuroprotectants work in rodent stroke models, this does not mean they translated to humans. If one assumed, for the sake of argument, that it is the results in the rabbit model that predicted translation to humans, the rabbit model should be used for translational studies. Factors that might account for translation in rabbits and not rodents might include: the rabbit immune system is different from the rodent; the particular blood clot model used; the clot—endothelial cell interaction is closer to humans in the rabbit model; rabbit lipid metabolism is closer to humans; the use of death as an outcome measure in the rabbit model; the absence of anesthesia in the rabbit model; and other factors discussed below. Thus, the rabbit results—and possibly not the rodent results—predicted tPA to be a clinical stroke treatment [21, 3133].
Since it is not possible at present to decide whether the rabbit or the rodent models are “best” for translational studies, we propose that it may be beneficial for compounds that have been found to be effective in non-rabbit models (rat, mouse, non-human primate, and others) to also demonstrate efficacy in the rabbit embolic blood clot model before proceeding to clinical testing. Clearly, it may be useful, more convenient and cost-effective to initially test compounds in rodent blood clot embolic models [34]. Indeed, as highlighted by the STAIR recommendations [38], testing potential therapeutics in a second species, and importantly in a species already shown to predict improved outcome in acute ischemic stroke, would improve the likelihood of successful translation. Compounds successful in such a setting should then be given a high priority for evaluation in humans. In contrast, those compounds demonstrating efficacy in only one animal model or species are less likely to be successfully translated into human stroke. We propose that compounds that prove effective in a given model also be tested in the rabbit blood clot embolic model.
This concept also extends to neuroprotection and combination therapy with tPA. Accordingly, when compounds are tested in the rabbit embolic model, they could be evaluated on their own compared to vehicle, but more importantly, where appropriate, they should be compared directly to tPA. A compound shown to improve acute ischemic stroke outcomes comparable to tPA may be more likely to translate to human stroke. This concept would apply to all compounds proposed to improve acute ischemic stroke, including new thrombolytiics and neuroprotectants. That is, if the neuroprotection provided by a “neuroprotectant” is similar to or better than that obtained with tPA, then the “neuroprotectant” is more likely to translate to human stroke. A second consideration is the combination of compounds with tPA. Several neuroprotectants when co-administered with tPA have been identified to improve outcomes in non-rabbit models of acute ischemic stroke [35]. Evaluating such agents further in conjunction with tPA in the rabbit embolic model would provide additional support for potential clinical efficacy.

Acute Ischemic Stroke Models in the Rat and Mouse are not Equivalent to the Rabbit

Certainly, the rabbit model of ischemic stroke has been used to a lesser extent than rodent models. This may be in part due to the increased cost associated with rabbit experiments. Nevertheless, the predictive power of these animal models may differ considerably. Although tPA improves outcomes in both rat and mice blood clot embolic models [25, 3638], as well as in the rabbit embolic clot model [21], it does not necessarily follow that since tPA works in both rodents and rabbits that the predictive power of the rabbit model is shared by that of the rodent model. Ideally, one should show efficacy in the rodent and the rabbit clot embolic models. Thus, the rabbit becomes the confirmatory “second animal model” as suggested by the STAIRS recommendations.
Human stroke generally involves progression of underlying vascular disease, and is associated with increasing age, hypertension, diabetes, hyperlipidemia, smoking, and heart disease [39, 40]. Accordingly, models that incorporate such factors are more likely to be predictive of clinical efficacy than those that do not. None of the animal models truly replicate human stroke. They are simply models of stroke. However, choosing the most appropriate animal models for the research question is essential for the success of both experimental and clinical testing. Accordingly, there may be several explanations for the efficacy of the rabbit model and indeed several advantages of this model.

Atherosclerosis

Rodents and rabbits differ in the development of atherosclerosis. Rabbits develop atherosclerosis and share many aspects of human lipoprotein metabolism. For example, the composition of lipoproteins, production of Apoβ100-containing VLDLs by liver, cholesteryl ester transfer activity, and high absorption of dietary cholesterol are similar in humans and rabbits [41, 42]. Transgenic rodents are available which have increased plasma cholesterol and triglycerides as well as low high-density lipoprotein levels [42]. Rabbits, however, rapidly develop atherosclerosis on hypercholesterolemic diets (0.5–4%/weight) where dietary cholesterol supplementation leads to the development of fatty streaks [41]. Though rodents may also develop atherosclerosis, they are inherently much more resistant than rabbits [43]. Atherosclerosis may be induced in rodents with dietary and genetic manipulations, such as apolipoprotein E (ApoE; −/−) and low-density lipoprotein (LDL; −/−), although they develop very unstable atherosclerotic lesions [4446]. Furthermore, the cholesterol metabolism of rodents is more geared towards HDL, rather that LDL, like that in both humans and rabbits [45]. This is of clinical relevance as the expression of ApoE and LDL receptors differs in young adult versus old rats following cerebral ischemia [47]. Atherosclerosis is an important risk factor in patients with ischemia [48]. Therefore, consideration of this pathology in animal stroke models is crucial. The simple fact that rabbits develop atherosclerosis whereas normal rodents do not may make the rabbit a better model in which to test compounds for stroke due to the lipid metabolism and/or endothelial differences in rabbits versus rodents. Rabbits are commonly used for the study of atherosclerosis and for cardiovascular diseases for these very reasons [49, 50].

The Immune System

Another issue that has not been considered in animal models of stroke is whether the immune system of the rodent is appropriate for modeling that of human stroke. The immune system is critical in human stroke [5153] as exemplified by the enlimomab clinical trial [54] where anti-ICAM-1 antibody significantly worsened stroke outcome [55]. The composition and type of immune response are important factors in modeling human stroke remains to be seen [56]. The rodent immune cell composition is remarkably different from that of rabbits and humans. Specifically, rodents have a lymphocyte predominance with a 1:5 ratio of neutrophils to lymphocytes. In contrast, rabbits have a 1:1 ratio of neutrophils to lymphocytes, which is similar to the immune system in humans who have a 2:1 ratio of neutrophils to lymphocytes [57, 58]. Rodents and humans also differ significantly in the systemic immune cell gene expression response to ischemic stroke [5963]. Although, this represents only one aspect of the immune system, it suggests that the way the rodent immune system responds to cerebral ischemia may not reflect that of the human immune response to acute ischemic stroke. It is not known whether the rabbit immune response to cerebral ischemia is similar to humans or not.

Anesthesia

The majority of patients with ischemic stroke are not anesthetized. The effect of a stroke or response to stroke may differ greatly with anesthesia. Accordingly, anesthesia may markedly confound experimental stroke studies. However, there are animal models of stroke that are able to induce an ischemic stroke in conscious animals [21, 6466]. Such models are likely to model human stroke more closely than those models using anesthesia during the induction of stroke. Specifically, the rabbit blood clot embolism model [21] involves the preparation of animals under anesthesia and then later, the stroke is induced in awake, un-anesthetized animals. Moreover, numerous studies have demonstrated anesthetic agents afford a degree of protection from cerebral ischemia [6770]. Even light surgical anesthesia may substantially reduce infarct size following stroke [71]. Accordingly, the contribution of anesthesia to the experimental paradigm and potential neuroprotection requires careful consideration, and ideally the confounding effects of anesthesia must be eliminated from animal stroke models.

tPA Effectiveness in Human Stroke Provides Insight into Animal Models

Thrombolysis with tPA is effective in humans in cardioembolic, large vessel thromboembolic, and small vessel lacunar stroke [22, 72]. This finding has important implications for animal models of stroke and for testing of drugs to treat human stroke. Since cardioembolic stroke is due to embolic blood clot, then animal models of embolic autologous clot likely models this type of stroke. There are no accepted animal models that mimic large vessel thromboembolic stroke and small vessel lacunar stroke in humans [73, 74]. However, since tPA improves outcome in all three human ischemic stroke subtypes, this suggests that all three types of human stroke are caused at least in part by clots that are acted upon by tPA. For cardioembolic strokes, the clots would usually come from the heart. For large vessel strokes the clots would come from the parent vessels likely related to atherosclerosis. For lacunar strokes, clots might form because of abnormalities in the vessel wall or platelet vessel wall interactions. Thus, the blood clot embolic animal models—be they rabbit or rodent—could be viewed as reasonable mimics of human cardioembolic, large vessel and lacunar causes of human stroke. Thus, animal models in which tPA was effective might also be considered as potential models of important aspects of the pathophysiology of these three human stroke subtypes. Addressing treatments for all three causes of human stroke is essential for translation to human stroke trials because few human stroke trials to date, with the exception of cardioembolic stroke, have considered the cause of stroke when choosing subjects to treat.

Outcome Measures in Animal Models—What is the Best One?

In animal studies, a “neuroprotective” compound is typically compared to a “saline or vehicle control.” The common outcome measures are either a statistically significant decrease in infarct volume or an improvement in a given behavior [34]. However, these outcome measures in animals may not translate to improving outcomes in human stroke. Thus, alternate outcomes need to also be considered. The original rabbit clot embolic model used death as an outcome measure. This is in contrast to most other animal stroke models. In fact, most rodent studies discard animals that die from as a result of ischemic stroke [75]. Indeed, survival studies are becoming increasingly important as death is an important outcome measure in human studies and may be one of the reasons that the rabbit embolic clot model was predictive of efficacy in humans. Therefore, the inclusion of survival rates in experimental stroke studies is critical in determining the true efficacy of a potential therapeutic agent as excluding animals that have died from the study significantly skews the results so that findings are biased towards surviving animals [34, 75]. In the various trials of tPA, the drug decreased morbidity and mortality [76]. Even if one accepted standard behavioral measures short of death, the behaviors in rats, mice, rabbits and even primates that predict clinical efficacy of therapies for stroke in humans are unknown. Additionally, outcome measures are evaluated at short times following ischemia and not at the times used in clinical trials. Thus, if nothing else death provides at least one additional outcome measure. Using death as the outcome measure in the rabbit embolic clot model likely means that other behavioral assays do not have to be performed and there would be no need to find the behaviors that translate to humans. Moreover, since death was the major behavioral outcome used to demonstrate tPA efficacy in the rabbit blood clot embolic stroke model, this same behavioral endpoint may be useful when testing new neuroprotectants and/or thrombolytics in the rabbit embolic clot model and probably should be included in rodent stroke models.

Concluding Remarks

To date, there has been a lack of translation of stroke treatments from animals to humans. The rabbit blood clot model of embolic stroke, in which tPA improved stroke outcomes, may be a relevant animal model for predicting efficacy of a drug in human stroke. We suggest that blood clot embolic models should be the primary ones evaluated, and head to head comparison with tPA should be performed in the rabbit model and other models. The contribution of the immune system and atherosclerosis must be included in experimental models along with the other STAIR recommendations. Additionally, death needs to be included as an outcome measure. Ultimately, the goal of translational stroke research is to find the simplest yet most predictive animal model of ischemic stroke so that potential treatments can be successfully and rapidly moved to the clinic and improve the care of patients with stroke.

Acknowledgment

Sources of Support: National Health and Medical Research Council (RJT Australia, #519365), Canadian Institutes of Health and Research (GJ, Canada), and NIH NS054652 and NS066845 (FRS).

Open Access

This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.
Open AccessThis is an open access article distributed under the terms of the Creative Commons Attribution Noncommercial License (https://​creativecommons.​org/​licenses/​by-nc/​2.​0), which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Literatur
1.
Zurück zum Zitat Diener HC, Lees KR, Lyden P, Grotta J, Davalos A, Davis SM, et al. NXY-059 for the treatment of acute stroke: pooled analysis of the SAINT I and II Trials. Stroke. 2008;39:1751–8.PubMedCrossRef Diener HC, Lees KR, Lyden P, Grotta J, Davalos A, Davis SM, et al. NXY-059 for the treatment of acute stroke: pooled analysis of the SAINT I and II Trials. Stroke. 2008;39:1751–8.PubMedCrossRef
2.
Zurück zum Zitat Shuaib A, Lees KR, Lyden P, Grotta J, Davalos A, Davis SM, et al. NXY-059 for the treatment of acute ischemic stroke. N Engl J Med. 2007;357:562–71.PubMedCrossRef Shuaib A, Lees KR, Lyden P, Grotta J, Davalos A, Davis SM, et al. NXY-059 for the treatment of acute ischemic stroke. N Engl J Med. 2007;357:562–71.PubMedCrossRef
3.
Zurück zum Zitat Fisher M. Recommendations for advancing development of acute stroke therapies: Stroke Therapy Academic Industry Roundtable 3. Stroke. 2003;34:1539–46.PubMedCrossRef Fisher M. Recommendations for advancing development of acute stroke therapies: Stroke Therapy Academic Industry Roundtable 3. Stroke. 2003;34:1539–46.PubMedCrossRef
4.
Zurück zum Zitat Fisher M, Albers GW, Donnan GA, Furlan AJ, Grotta JC, Kidwell CS, et al. Enhancing the development and approval of acute stroke therapies: Stroke Therapy Academic Industry Roundtable. Stroke. 2005;36:1808–13.PubMedCrossRef Fisher M, Albers GW, Donnan GA, Furlan AJ, Grotta JC, Kidwell CS, et al. Enhancing the development and approval of acute stroke therapies: Stroke Therapy Academic Industry Roundtable. Stroke. 2005;36:1808–13.PubMedCrossRef
5.
Zurück zum Zitat Fisher M, Feuerstein G, Howells DW, Hurn PD, Kent TA, Savitz SI, et al. Update of the stroke therapy academic industry roundtable preclinical recommendations. Stroke. 2009;40:2244–50.PubMedCrossRef Fisher M, Feuerstein G, Howells DW, Hurn PD, Kent TA, Savitz SI, et al. Update of the stroke therapy academic industry roundtable preclinical recommendations. Stroke. 2009;40:2244–50.PubMedCrossRef
6.
Zurück zum Zitat Fisher M, Hanley DF, Howard G, Jauch EC, Warach S. Recommendations from the STAIR V meeting on acute stroke trials, technology and outcomes. Stroke. 2007;38:245–8.PubMedCrossRef Fisher M, Hanley DF, Howard G, Jauch EC, Warach S. Recommendations from the STAIR V meeting on acute stroke trials, technology and outcomes. Stroke. 2007;38:245–8.PubMedCrossRef
7.
Zurück zum Zitat Saver JL, Albers GW, Dunn B, Johnston KC, Fisher M. Stroke Therapy Academic Industry Roundtable (STAIR) recommendations for extended window acute stroke therapy trials. Stroke. 2009;40:2594–600.PubMedCrossRef Saver JL, Albers GW, Dunn B, Johnston KC, Fisher M. Stroke Therapy Academic Industry Roundtable (STAIR) recommendations for extended window acute stroke therapy trials. Stroke. 2009;40:2594–600.PubMedCrossRef
8.
Zurück zum Zitat STAIR. Recommendations for clinical trial evaluation of acute stroke therapies. Stroke. 2001;32:1598–606. STAIR. Recommendations for clinical trial evaluation of acute stroke therapies. Stroke. 2001;32:1598–606.
9.
Zurück zum Zitat Dirnagl U. Bench to bedside: the quest for quality in experimental stroke research. J Cereb Blood Flow Metab. 2006;26:1465–78.PubMedCrossRef Dirnagl U. Bench to bedside: the quest for quality in experimental stroke research. J Cereb Blood Flow Metab. 2006;26:1465–78.PubMedCrossRef
10.
Zurück zum Zitat Endres M, Engelhardt B, Koistinaho J, Lindvall O, Meairs S, Mohr JP, et al. Improving outcome after stroke: overcoming the translational roadblock. Cerebrovasc Dis. 2008;25:268–78.PubMedCrossRef Endres M, Engelhardt B, Koistinaho J, Lindvall O, Meairs S, Mohr JP, et al. Improving outcome after stroke: overcoming the translational roadblock. Cerebrovasc Dis. 2008;25:268–78.PubMedCrossRef
11.
Zurück zum Zitat Ginsberg MD, Busto R. Rodent models of cerebral ischemia. Stroke. 1989;20:1627–42.PubMed Ginsberg MD, Busto R. Rodent models of cerebral ischemia. Stroke. 1989;20:1627–42.PubMed
12.
Zurück zum Zitat Lang JT, McCullough LD. Pathways to ischemic neuronal cell death: are sex differences relevant? J Transl Med. 2008;6:33.PubMedCrossRef Lang JT, McCullough LD. Pathways to ischemic neuronal cell death: are sex differences relevant? J Transl Med. 2008;6:33.PubMedCrossRef
13.
Zurück zum Zitat Herson PS, Koerner IP, Hurn PD. Sex, sex steroids, and brain injury. Semin Reprod Med. 2009;27:229–39.PubMedCrossRef Herson PS, Koerner IP, Hurn PD. Sex, sex steroids, and brain injury. Semin Reprod Med. 2009;27:229–39.PubMedCrossRef
14.
Zurück zum Zitat Howells DW, Porritt MJ, Rewell SS, O'Collins V, Sena ES, van der Worp HB, et al. Different strokes for different folks: the rich diversity of animal models of focal cerebral ischemia. J Cereb Blood Flow Metab. 2010;30:1412–31.PubMedCrossRef Howells DW, Porritt MJ, Rewell SS, O'Collins V, Sena ES, van der Worp HB, et al. Different strokes for different folks: the rich diversity of animal models of focal cerebral ischemia. J Cereb Blood Flow Metab. 2010;30:1412–31.PubMedCrossRef
15.
Zurück zum Zitat Liu S, Zhen G, Meloni BP, Campbell K, Winn HR. Rodent Stroke Model Guidelines for Preclinical Stroke Trials (1st Edition). J Exp Stroke Transl Med. 2009;2:2–27.PubMed Liu S, Zhen G, Meloni BP, Campbell K, Winn HR. Rodent Stroke Model Guidelines for Preclinical Stroke Trials (1st Edition). J Exp Stroke Transl Med. 2009;2:2–27.PubMed
16.
Zurück zum Zitat Lo EH. Experimental models, neurovascular mechanisms and translational issues in stroke research. Br J Pharmacol. 2008;153 Suppl 1:S396–405.PubMed Lo EH. Experimental models, neurovascular mechanisms and translational issues in stroke research. Br J Pharmacol. 2008;153 Suppl 1:S396–405.PubMed
17.
Zurück zum Zitat Macleod MR, Fisher M, O'Collins V, Sena ES, Dirnagl U, Bath PM, et al. Reprint: good laboratory practice: preventing introduction of bias at the bench. J Cereb Blood Flow Metab. 2009;29:221–3.PubMedCrossRef Macleod MR, Fisher M, O'Collins V, Sena ES, Dirnagl U, Bath PM, et al. Reprint: good laboratory practice: preventing introduction of bias at the bench. J Cereb Blood Flow Metab. 2009;29:221–3.PubMedCrossRef
18.
Zurück zum Zitat Savitz SI. A critical appraisal of the NXY-059 neuroprotection studies for acute stroke: a need for more rigorous testing of neuroprotective agents in animal models of stroke. Exp Neurol. 2007;205:20–5.PubMedCrossRef Savitz SI. A critical appraisal of the NXY-059 neuroprotection studies for acute stroke: a need for more rigorous testing of neuroprotective agents in animal models of stroke. Exp Neurol. 2007;205:20–5.PubMedCrossRef
19.
Zurück zum Zitat Sena ES, van der Worp HB, Bath PM, Howells DW, Macleod MR. Publication bias in reports of animal stroke studies leads to major overstatement of efficacy. PLoS Biol. 2010;8:e1000344.PubMedCrossRef Sena ES, van der Worp HB, Bath PM, Howells DW, Macleod MR. Publication bias in reports of animal stroke studies leads to major overstatement of efficacy. PLoS Biol. 2010;8:e1000344.PubMedCrossRef
21.
Zurück zum Zitat Zivin JA, Fisher M, DeGirolami U, Hemenway CC, Stashak JA. Tissue plasminogen activator reduces neurological damage after cerebral embolism. Science. 1985;230:1289–92.PubMedCrossRef Zivin JA, Fisher M, DeGirolami U, Hemenway CC, Stashak JA. Tissue plasminogen activator reduces neurological damage after cerebral embolism. Science. 1985;230:1289–92.PubMedCrossRef
22.
Zurück zum Zitat NINDS. Tissue plasminogen activator for acute ischemic stroke. The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group. N Engl J Med. 1995;333:1581–7.CrossRef NINDS. Tissue plasminogen activator for acute ischemic stroke. The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group. N Engl J Med. 1995;333:1581–7.CrossRef
23.
Zurück zum Zitat Busch E, Kruger K, Allegrini PR, Kerskens CM, Gyngell ML, Hoehn-Berlage M, et al. Reperfusion after thrombolytic therapy of embolic stroke in the rat: magnetic resonance and biochemical imaging. J Cereb Blood Flow Metab. 1998;18:407–18.PubMedCrossRef Busch E, Kruger K, Allegrini PR, Kerskens CM, Gyngell ML, Hoehn-Berlage M, et al. Reperfusion after thrombolytic therapy of embolic stroke in the rat: magnetic resonance and biochemical imaging. J Cereb Blood Flow Metab. 1998;18:407–18.PubMedCrossRef
24.
Zurück zum Zitat Jiang Q, Zhang RL, Zhang ZG, Ewing JR, Divine GW, Chopp M. Diffusion-, T2-, and perfusion-weighted nuclear magnetic resonance imaging of middle cerebral artery embolic stroke and recombinant tissue plasminogen activator intervention in the rat. J Cereb Blood Flow Metab. 1998;18:758–67.PubMedCrossRef Jiang Q, Zhang RL, Zhang ZG, Ewing JR, Divine GW, Chopp M. Diffusion-, T2-, and perfusion-weighted nuclear magnetic resonance imaging of middle cerebral artery embolic stroke and recombinant tissue plasminogen activator intervention in the rat. J Cereb Blood Flow Metab. 1998;18:758–67.PubMedCrossRef
25.
Zurück zum Zitat Papadopoulos SM, Chandler WF, Salamat MS, Topol EJ, Sackellares JC. Recombinant human tissue-type plasminogen activator therapy in acute thromboembolic stroke. J Neurosurg. 1987;67:394–8.PubMedCrossRef Papadopoulos SM, Chandler WF, Salamat MS, Topol EJ, Sackellares JC. Recombinant human tissue-type plasminogen activator therapy in acute thromboembolic stroke. J Neurosurg. 1987;67:394–8.PubMedCrossRef
26.
Zurück zum Zitat Asahi M, Asahi K, Wang X, Lo EH. Reduction of tissue plasminogen activator-induced hemorrhage and brain injury by free radical spin trapping after embolic focal cerebral ischemia in rats. J Cereb Blood Flow Metab. 2000;20:452–7.PubMedCrossRef Asahi M, Asahi K, Wang X, Lo EH. Reduction of tissue plasminogen activator-induced hemorrhage and brain injury by free radical spin trapping after embolic focal cerebral ischemia in rats. J Cereb Blood Flow Metab. 2000;20:452–7.PubMedCrossRef
27.
Zurück zum Zitat Kano T, Harada T, Katayama Y. Attenuation of extravasation of tissue plasminogen activator by the free radical scavenger, edaravone: evaluation in a rat thromboembolic stroke model. Neurol Res. 2005;27:499–502.PubMedCrossRef Kano T, Harada T, Katayama Y. Attenuation of extravasation of tissue plasminogen activator by the free radical scavenger, edaravone: evaluation in a rat thromboembolic stroke model. Neurol Res. 2005;27:499–502.PubMedCrossRef
28.
Zurück zum Zitat Kano T, Katayama Y, Tejima E, Lo EH. Hemorrhagic transformation after fibrinolytic therapy with tissue plasminogen activator in a rat thromboembolic model of stroke. Brain Res. 2000;854:245–8.PubMedCrossRef Kano T, Katayama Y, Tejima E, Lo EH. Hemorrhagic transformation after fibrinolytic therapy with tissue plasminogen activator in a rat thromboembolic model of stroke. Brain Res. 2000;854:245–8.PubMedCrossRef
29.
Zurück zum Zitat Overgaard K, Sereghy T, Pedersen H, Boysen G. Effect of delayed thrombolysis with rt-PA in a rat embolic stroke model. J Cereb Blood Flow Metab. 1994;14:472–7.PubMedCrossRef Overgaard K, Sereghy T, Pedersen H, Boysen G. Effect of delayed thrombolysis with rt-PA in a rat embolic stroke model. J Cereb Blood Flow Metab. 1994;14:472–7.PubMedCrossRef
30.
Zurück zum Zitat Zhang L, Zhang ZG, Zhang RL, Lu M, Adams J, Elliott PJ, et al. Postischemic (6-Hour) treatment with recombinant human tissue plasminogen activator and proteasome inhibitor PS-519 reduces infarction in a rat model of embolic focal cerebral ischemia. Stroke. 2001;32:2926–31.PubMedCrossRef Zhang L, Zhang ZG, Zhang RL, Lu M, Adams J, Elliott PJ, et al. Postischemic (6-Hour) treatment with recombinant human tissue plasminogen activator and proteasome inhibitor PS-519 reduces infarction in a rat model of embolic focal cerebral ischemia. Stroke. 2001;32:2926–31.PubMedCrossRef
31.
Zurück zum Zitat Chehrazi BB, Seibert JA, Kissel P, Hein L, Brock JM. Evaluation of recombinant tissue plasminogen activator in embolic stroke. Neurosurgery. 1989;24:355–60.PubMedCrossRef Chehrazi BB, Seibert JA, Kissel P, Hein L, Brock JM. Evaluation of recombinant tissue plasminogen activator in embolic stroke. Neurosurgery. 1989;24:355–60.PubMedCrossRef
32.
Zurück zum Zitat Phillips DA, Davis MA, Fisher M. Selective embolization and clot dissolution with tPA in the internal carotid artery circulation of the rabbit. AJNR Am J Neuroradiol. 1988;9:899–902.PubMed Phillips DA, Davis MA, Fisher M. Selective embolization and clot dissolution with tPA in the internal carotid artery circulation of the rabbit. AJNR Am J Neuroradiol. 1988;9:899–902.PubMed
33.
Zurück zum Zitat Phillips DA, Fisher M, Smith TW, Davis MA. The safety and angiographic efficacy of tissue plasminogen activator in a cerebral embolization model. Ann Neurol. 1988;23:391–4.PubMedCrossRef Phillips DA, Fisher M, Smith TW, Davis MA. The safety and angiographic efficacy of tissue plasminogen activator in a cerebral embolization model. Ann Neurol. 1988;23:391–4.PubMedCrossRef
34.
35.
Zurück zum Zitat Savitz SI, Fisher M. Future of neuroprotection for acute stroke: in the aftermath of the SAINT trials. Ann Neurol. 2007;61:396–402.PubMedCrossRef Savitz SI, Fisher M. Future of neuroprotection for acute stroke: in the aftermath of the SAINT trials. Ann Neurol. 2007;61:396–402.PubMedCrossRef
36.
Zurück zum Zitat Kudo M, Aoyama A, Ichimori S, Fukunaga N. An animal model of cerebral infarction. Homologous blood clot emboli in rats. Stroke. 1982;13:505–8.PubMed Kudo M, Aoyama A, Ichimori S, Fukunaga N. An animal model of cerebral infarction. Homologous blood clot emboli in rats. Stroke. 1982;13:505–8.PubMed
37.
Zurück zum Zitat Meng W, Wang X, Asahi M, Kano T, Asahi K, Ackerman RH, et al. Effects of tissue type plasminogen activator in embolic versus mechanical models of focal cerebral ischemia in rats. J Cereb Blood Flow Metab. 1999;19:1316–21.PubMedCrossRef Meng W, Wang X, Asahi M, Kano T, Asahi K, Ackerman RH, et al. Effects of tissue type plasminogen activator in embolic versus mechanical models of focal cerebral ischemia in rats. J Cereb Blood Flow Metab. 1999;19:1316–21.PubMedCrossRef
38.
Zurück zum Zitat Zhang RL, Zhang L, Jiang Q, Zhang ZG, Goussev A, Chopp M. Postischemic intracarotid treatment with TNK-tPA reduces infarct volume and improves neurological deficits in embolic stroke in the unanesthetized rat. Brain Res. 2000;878:64–71.PubMedCrossRef Zhang RL, Zhang L, Jiang Q, Zhang ZG, Goussev A, Chopp M. Postischemic intracarotid treatment with TNK-tPA reduces infarct volume and improves neurological deficits in embolic stroke in the unanesthetized rat. Brain Res. 2000;878:64–71.PubMedCrossRef
39.
Zurück zum Zitat Elkind MS, Sacco RL. Stroke risk factors and stroke prevention. Semin Neurol. 1998;18:429–40.PubMedCrossRef Elkind MS, Sacco RL. Stroke risk factors and stroke prevention. Semin Neurol. 1998;18:429–40.PubMedCrossRef
40.
Zurück zum Zitat Sacco RL. Newer risk factors for stroke. Neurology. 2001;57:S31–4.PubMed Sacco RL. Newer risk factors for stroke. Neurology. 2001;57:S31–4.PubMed
41.
Zurück zum Zitat Dornas WC, Oliveira TT, Augusto LE, Nagem TJ. Experimental atherosclerosis in rabbits. Arq Bras Cardiol. 2010;95:272–8.PubMedCrossRef Dornas WC, Oliveira TT, Augusto LE, Nagem TJ. Experimental atherosclerosis in rabbits. Arq Bras Cardiol. 2010;95:272–8.PubMedCrossRef
42.
Zurück zum Zitat Moghadasian MH, Frohlich JJ, McManus BM. Advances in experimental dyslipidemia and atherosclerosis. Lab Invest. 2001;81:1173–83.PubMedCrossRef Moghadasian MH, Frohlich JJ, McManus BM. Advances in experimental dyslipidemia and atherosclerosis. Lab Invest. 2001;81:1173–83.PubMedCrossRef
43.
Zurück zum Zitat Yanni AE. The laboratory rabbit: an animal model of atherosclerosis research. Lab Anim. 2004;38:246–56.PubMedCrossRef Yanni AE. The laboratory rabbit: an animal model of atherosclerosis research. Lab Anim. 2004;38:246–56.PubMedCrossRef
44.
Zurück zum Zitat Daugherty A, Lu H, Howatt DA, Rateri DL. Modes of defining atherosclerosis in mouse models: relative merits and evolving standards. Methods Mol Biol. 2009;573:1–15.PubMedCrossRef Daugherty A, Lu H, Howatt DA, Rateri DL. Modes of defining atherosclerosis in mouse models: relative merits and evolving standards. Methods Mol Biol. 2009;573:1–15.PubMedCrossRef
45.
Zurück zum Zitat Russell JC, Proctor SD. Small animal models of cardiovascular disease: tools for the study of the roles of metabolic syndrome, dyslipidemia, and atherosclerosis. Cardiovasc Pathol. 2006;15:318–30.PubMedCrossRef Russell JC, Proctor SD. Small animal models of cardiovascular disease: tools for the study of the roles of metabolic syndrome, dyslipidemia, and atherosclerosis. Cardiovasc Pathol. 2006;15:318–30.PubMedCrossRef
46.
Zurück zum Zitat Verbeuren TJ. Experimental models of thrombosis and atherosclerosis. Therapie. 2006;61:379–87.PubMedCrossRef Verbeuren TJ. Experimental models of thrombosis and atherosclerosis. Therapie. 2006;61:379–87.PubMedCrossRef
47.
Zurück zum Zitat Hayashi T, Kamada H, Jin G, Deguchi K, Nagotani S, Sehara Y, et al. Different expression of low density lipoprotein receptor and ApoE between young adult and old rat brains after ischemia. Neurol Res. 2006;28:822–5.PubMedCrossRef Hayashi T, Kamada H, Jin G, Deguchi K, Nagotani S, Sehara Y, et al. Different expression of low density lipoprotein receptor and ApoE between young adult and old rat brains after ischemia. Neurol Res. 2006;28:822–5.PubMedCrossRef
48.
Zurück zum Zitat Mostaza JM, Martin-Jadraque R, Vicente I, San Martin MA, Lahoz C. Patients at high risk of cerebrovascular disease: the REACH study. Cerebrovasc Dis. 2009;27 Suppl 1:77–81.PubMedCrossRef Mostaza JM, Martin-Jadraque R, Vicente I, San Martin MA, Lahoz C. Patients at high risk of cerebrovascular disease: the REACH study. Cerebrovasc Dis. 2009;27 Suppl 1:77–81.PubMedCrossRef
49.
Zurück zum Zitat Qureshi AI, Taylor RA. Research priorities for intracranial atherosclerotic diseases. J Neuroimaging. 2009;19 Suppl 1:39S–42S.PubMedCrossRef Qureshi AI, Taylor RA. Research priorities for intracranial atherosclerotic diseases. J Neuroimaging. 2009;19 Suppl 1:39S–42S.PubMedCrossRef
50.
Zurück zum Zitat Zhang GY, Chen ZQ, Ling F, Li YJ, Wang Y, Gu BX. Establishment of a novel hemodynamic cerebral ischemia model of atherosclerotic rabbit. Neurol India. 2010;58:191–4.PubMedCrossRef Zhang GY, Chen ZQ, Ling F, Li YJ, Wang Y, Gu BX. Establishment of a novel hemodynamic cerebral ischemia model of atherosclerotic rabbit. Neurol India. 2010;58:191–4.PubMedCrossRef
51.
Zurück zum Zitat Jordan J, Segura T, Brea D, Galindo MF, Castillo J. Inflammation as therapeutic objective in stroke. Curr Pharm Des. 2008;14:3549–64.PubMedCrossRef Jordan J, Segura T, Brea D, Galindo MF, Castillo J. Inflammation as therapeutic objective in stroke. Curr Pharm Des. 2008;14:3549–64.PubMedCrossRef
52.
Zurück zum Zitat Tanasescu R, Nicolau A, Ticmeanu M, Luca D, Caraiola S, Cojocaru IM, et al. An immunological approach to cerebral ischemia (I). Immune cells and adhesion molecules. Rom J Intern Med. 2008;46:3–8.PubMed Tanasescu R, Nicolau A, Ticmeanu M, Luca D, Caraiola S, Cojocaru IM, et al. An immunological approach to cerebral ischemia (I). Immune cells and adhesion molecules. Rom J Intern Med. 2008;46:3–8.PubMed
53.
Zurück zum Zitat Tuttolomondo A, Di Sciacca R, Di Raimondo D, Renda C, Pinto A, Licata G. Inflammation as a therapeutic target in acute ischemic stroke treatment. Curr Top Med Chem. 2009;9:1240–60.PubMedCrossRef Tuttolomondo A, Di Sciacca R, Di Raimondo D, Renda C, Pinto A, Licata G. Inflammation as a therapeutic target in acute ischemic stroke treatment. Curr Top Med Chem. 2009;9:1240–60.PubMedCrossRef
54.
Zurück zum Zitat Enlimomab. Use of anti-ICAM-1 therapy in ischemic stroke: results of the Enlimomab Acute Stroke Trial. Neurology. 2001;57:1428–34. Enlimomab. Use of anti-ICAM-1 therapy in ischemic stroke: results of the Enlimomab Acute Stroke Trial. Neurology. 2001;57:1428–34.
55.
Zurück zum Zitat Furuya K, Takeda H, Azhar S, McCarron RM, Chen Y, Ruetzler CA, et al. Examination of several potential mechanisms for the negative outcome in a clinical stroke trial of enlimomab, a murine anti-human intercellular adhesion molecule-1 antibody: a bedside-to-bench study. Stroke. 2001;32:2665–74.PubMedCrossRef Furuya K, Takeda H, Azhar S, McCarron RM, Chen Y, Ruetzler CA, et al. Examination of several potential mechanisms for the negative outcome in a clinical stroke trial of enlimomab, a murine anti-human intercellular adhesion molecule-1 antibody: a bedside-to-bench study. Stroke. 2001;32:2665–74.PubMedCrossRef
56.
Zurück zum Zitat Shah IM, Macrae IM, Di Napoli M. Neuroinflammation and neuroprotective strategies in acute ischaemic stroke—from bench to bedside. Curr Mol Med. 2009;9:336–54.PubMedCrossRef Shah IM, Macrae IM, Di Napoli M. Neuroinflammation and neuroprotective strategies in acute ischaemic stroke—from bench to bedside. Curr Mol Med. 2009;9:336–54.PubMedCrossRef
57.
Zurück zum Zitat Fauci AS, Braunwald E, Kasper DL, Hauser SL, Longo DL, Jameson JL, et al. Laboratory Values of Clinical Importance. 2009. Fauci AS, Braunwald E, Kasper DL, Hauser SL, Longo DL, Jameson JL, et al. Laboratory Values of Clinical Importance. 2009.
58.
Zurück zum Zitat Fox JG, Anderson LC, Leow FM, Quimby FW. Laboratory Animal Medicine. Academic Press; 2002. Fox JG, Anderson LC, Leow FM, Quimby FW. Laboratory Animal Medicine. Academic Press; 2002.
59.
Zurück zum Zitat Jickling GC, Xu H, Stamova B, Ander BP, Zhan X, Tian Y, et al. Signatures of cardioembolic and large-vessel ischemic stroke. Ann Neurol. 2010;68:681–92.PubMedCrossRef Jickling GC, Xu H, Stamova B, Ander BP, Zhan X, Tian Y, et al. Signatures of cardioembolic and large-vessel ischemic stroke. Ann Neurol. 2010;68:681–92.PubMedCrossRef
60.
Zurück zum Zitat Moore DF, Li H, Jeffries N, Wright V, Cooper Jr RA, Elkahloun A, et al. Using peripheral blood mononuclear cells to determine a gene expression profile of acute ischemic stroke: a pilot investigation. Circulation. 2005;111:212–21.PubMedCrossRef Moore DF, Li H, Jeffries N, Wright V, Cooper Jr RA, Elkahloun A, et al. Using peripheral blood mononuclear cells to determine a gene expression profile of acute ischemic stroke: a pilot investigation. Circulation. 2005;111:212–21.PubMedCrossRef
61.
Zurück zum Zitat Stamova B, Xu H, Jickling G, Bushnell C, Tian Y, Ander BP, et al. Gene expression profiling of blood for the prediction of ischemic stroke. Stroke. 2010;41:2171–7.PubMedCrossRef Stamova B, Xu H, Jickling G, Bushnell C, Tian Y, Ander BP, et al. Gene expression profiling of blood for the prediction of ischemic stroke. Stroke. 2010;41:2171–7.PubMedCrossRef
62.
Zurück zum Zitat Tang Y, Lu A, Aronow BJ, Sharp FR. Blood genomic responses differ after stroke, seizures, hypoglycemia, and hypoxia: blood genomic fingerprints of disease. Ann Neurol. 2001;50:699–707.PubMedCrossRef Tang Y, Lu A, Aronow BJ, Sharp FR. Blood genomic responses differ after stroke, seizures, hypoglycemia, and hypoxia: blood genomic fingerprints of disease. Ann Neurol. 2001;50:699–707.PubMedCrossRef
63.
Zurück zum Zitat Tang Y, Xu H, Du X, Lit L, Walker W, Lu A, et al. Gene expression in blood changes rapidly in neutrophils and monocytes after ischemic stroke in humans: a microarray study. J Cereb Blood Flow Metab. 2006;26:1089–102.PubMedCrossRef Tang Y, Xu H, Du X, Lit L, Walker W, Lu A, et al. Gene expression in blood changes rapidly in neutrophils and monocytes after ischemic stroke in humans: a microarray study. J Cereb Blood Flow Metab. 2006;26:1089–102.PubMedCrossRef
64.
Zurück zum Zitat Callaway JK, Knight MJ, Watkins DJ, Beart PM, Jarrott B. Delayed treatment with AM-36, a novel neuroprotective agent, reduces neuronal damage after endothelin-1-induced middle cerebral artery occlusion in conscious rats. Stroke. 1999;30:2704–12. discussion 2712.PubMed Callaway JK, Knight MJ, Watkins DJ, Beart PM, Jarrott B. Delayed treatment with AM-36, a novel neuroprotective agent, reduces neuronal damage after endothelin-1-induced middle cerebral artery occlusion in conscious rats. Stroke. 1999;30:2704–12. discussion 2712.PubMed
65.
Zurück zum Zitat Huang SS, Tsai SK, Chih CL, Chiang LY, Hsieh HM, Teng CM, et al. Neuroprotective effect of hexasulfobutylated C60 on rats subjected to focal cerebral ischemia. Free Radic Biol Med. 2001;30:643–9.PubMedCrossRef Huang SS, Tsai SK, Chih CL, Chiang LY, Hsieh HM, Teng CM, et al. Neuroprotective effect of hexasulfobutylated C60 on rats subjected to focal cerebral ischemia. Free Radic Biol Med. 2001;30:643–9.PubMedCrossRef
66.
Zurück zum Zitat Sharkey J, Ritchie IM, Kelly PA. Perivascular microapplication of endothelin-1: a new model of focal cerebral ischaemia in the rat. J Cereb Blood Flow Metab. 1993;13:865–71.PubMedCrossRef Sharkey J, Ritchie IM, Kelly PA. Perivascular microapplication of endothelin-1: a new model of focal cerebral ischaemia in the rat. J Cereb Blood Flow Metab. 1993;13:865–71.PubMedCrossRef
67.
68.
Zurück zum Zitat Kirsch JR, Traystman RJ, Hurn PD. Anesthetics and cerebroprotection: experimental aspects. Int Anesthesiol Clin. 1996;34:73–93.PubMedCrossRef Kirsch JR, Traystman RJ, Hurn PD. Anesthetics and cerebroprotection: experimental aspects. Int Anesthesiol Clin. 1996;34:73–93.PubMedCrossRef
69.
Zurück zum Zitat Koerner IP, Brambrink AM. Brain protection by anesthetic agents. Curr Opin Anaesthesiol. 2006;19:481–6.PubMedCrossRef Koerner IP, Brambrink AM. Brain protection by anesthetic agents. Curr Opin Anaesthesiol. 2006;19:481–6.PubMedCrossRef
70.
Zurück zum Zitat Wang L, Jing W, Hang YN. Glutamate-induced c-Jun expression in neuronal PC12 cells: the effects of ketamine and propofol. J Neurosurg Anesthesiol. 2008;20:124–30.PubMedCrossRef Wang L, Jing W, Hang YN. Glutamate-induced c-Jun expression in neuronal PC12 cells: the effects of ketamine and propofol. J Neurosurg Anesthesiol. 2008;20:124–30.PubMedCrossRef
71.
Zurück zum Zitat Gelb AW, Bayona NA, Wilson JX, Cechetto DF. Propofol anesthesia compared to awake reduces infarct size in rats. Anesthesiology. 2002;96:1183–90.PubMedCrossRef Gelb AW, Bayona NA, Wilson JX, Cechetto DF. Propofol anesthesia compared to awake reduces infarct size in rats. Anesthesiology. 2002;96:1183–90.PubMedCrossRef
72.
Zurück zum Zitat Hsia AW, Sachdev HS, Tomlinson J, Hamilton SA, Tong DC. Efficacy of IV tissue plasminogen activator in acute stroke: does stroke subtype really matter? Neurology. 2003;61:71–5.PubMed Hsia AW, Sachdev HS, Tomlinson J, Hamilton SA, Tong DC. Efficacy of IV tissue plasminogen activator in acute stroke: does stroke subtype really matter? Neurology. 2003;61:71–5.PubMed
73.
Zurück zum Zitat Bailey EL, McCulloch J, Sudlow C, Wardlaw JM. Potential animal models of lacunar stroke: a systematic review. Stroke. 2009;40:e451–8.PubMedCrossRef Bailey EL, McCulloch J, Sudlow C, Wardlaw JM. Potential animal models of lacunar stroke: a systematic review. Stroke. 2009;40:e451–8.PubMedCrossRef
74.
Zurück zum Zitat Hainsworth AH, Markus HS. Do in vivo experimental models reflect human cerebral small vessel disease? A systematic review. J Cereb Blood Flow Metab. 2008;28:1877–91.PubMedCrossRef Hainsworth AH, Markus HS. Do in vivo experimental models reflect human cerebral small vessel disease? A systematic review. J Cereb Blood Flow Metab. 2008;28:1877–91.PubMedCrossRef
75.
Zurück zum Zitat Lapchak P. Translational stroke research using a rabbit embolic stroke model: a correlative analysis hypothesis for novel therapy development. Transl Stroke Res. 2010;1:96–107.PubMedCrossRef Lapchak P. Translational stroke research using a rabbit embolic stroke model: a correlative analysis hypothesis for novel therapy development. Transl Stroke Res. 2010;1:96–107.PubMedCrossRef
76.
Zurück zum Zitat Wardlaw JM, Murray V, Berge E, Del Zoppo GJ. Thrombolysis for acute ischaemic stroke. Cochrane Database Syst Rev 2009: CD000213. Wardlaw JM, Murray V, Berge E, Del Zoppo GJ. Thrombolysis for acute ischaemic stroke. Cochrane Database Syst Rev 2009: CD000213.
Metadaten
Titel
Are Underlying Assumptions of Current Animal Models of Human Stroke Correct: from STAIRs to High Hurdles?
verfasst von
Renée J. Turner
Glen C. Jickling
Frank R. Sharp
Publikationsdatum
01.06.2011
Verlag
Springer-Verlag
Erschienen in
Translational Stroke Research / Ausgabe 2/2011
Print ISSN: 1868-4483
Elektronische ISSN: 1868-601X
DOI
https://doi.org/10.1007/s12975-011-0067-3

Weitere Artikel der Ausgabe 2/2011

Translational Stroke Research 2/2011 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.

Demenzkranke durch Antipsychotika vielfach gefährdet

23.04.2024 Demenz Nachrichten

Wenn Demenzkranke aufgrund von Symptomen wie Agitation oder Aggressivität mit Antipsychotika behandelt werden, sind damit offenbar noch mehr Risiken verbunden als bislang angenommen.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.