Skip to main content

Animal models of focal brain ischemia

Abstract

Stroke is a leading cause of disability and death in many countries. Understanding the pathophysiology of ischemic injury and developing therapies is an important endeavor that requires much additional research. Animal stroke models provide an important mechanism for these activities. A large number of stroke models have been developed and are currently used in laboratories around the world. These models are overviewed as are approaches for measuring infarct size and functional outcome.

Utility of Animal Models of Ischemic Stroke

Stroke is the third leading cause of death and a major cause of disability in the United States [1, 2]. Each year, there are approximately 731,000 new strokes and half of the survivors suffer from permanent handicap [3]. Stroke costs the United States approximately $50 billion annually in direct and indirect costs [4]. Given these facts, stroke is a major public health issue requiring urgent development of effective therapies: experimental models of focal brain ischemia help in achieving this goal.

80% of human strokes are ischemic in origin [2]. Thus, experimental models of focal cerebral ischemia have been developed in an attempt to closely mimic the changes that occur during and after human ischemic stroke. These models are used to discover the mechanisms involved in the evolution of ischemic injury which, in turn, can lead to the development of novel therapeutic strategies for stroke. These same animal models can then be used to test the safety and efficacy of these treatments in vivo.

Most human ischemic strokes are caused by occlusion of the middle cerebral artery (MCA) [5] and so animal models were developed to induce ischemia in this arterial territory. These models aim to satisfy the following criteria: (1) to mimic the pathophysiological changes found in human stroke, (2) to create reproducible lesions, (3) to employ procedures that are relatively simple and noninvasive, (4) to be of low financial cost, and (5) to enable monitoring of physiologic parameters and analysis of brain tissue for outcome measures [6].

Many higher animal species fulfill the aforementioned stroke modeling criteria; however, rats are the most commonly used animals for several reasons, including: (1) their resemblance to humans in their cerebral anatomy and physiology, (2) their small size which enables easy analysis of physiology and brain tissue, (3) their low cost, (4) the remarkable genetic homogeneity within strains, and (5) greater public and institutional ethical acceptability of use relative to larger animals. Thus, the remainder of this chapter will focus primarily on rat models of ischemic stroke [7–9].

Permanent versus Transient Ischemia

Focal brain ischemia models can be categorized into two groups: permanent and transient ischemia. Permanent ischemia results in a region of severe ischemic damage (core) surrounded by a zone of less damaged tissue [10]. Reestablishment of perfusion after 3 hours does not reduce infarct size in all animal models [11].

Transient focal ischemia produces varying degrees of ischemic damage depending on the duration of ischemia. In rats, as little as 8 minutes of ischemia causes selective neural necrosis and ischemia for more than 30 minutes is always associated with infarction [12]. Importantly, after transient ischemia, brain damage results from both the ischemia and the effects of reperfusion (reperfusion injury). Compared to permanent occlusion, which mimic only a minority of human strokes where there is no recanalization, transient models better correlate with conditions such as therapy-induced thrombolysis, spontaneous thrombolysis, and transient ischemic attack. However, both permanent and transient ischemia models are needed prior to clinical drug development studies because of the heterogeneity of human stroke [13, 14].

Long-term studies are becoming increasingly important in translational stroke research; thus, the animal survival rate should be considered when designing an experimental protocol. Generally, regardless of the employed technique, models of transient ischemia offer higher survival rates relative to permanent occlusion and thus may be more suited for long-term studies. Additionally, survival is enhanced by proper surgical technique, maintaining physiological parameters within normal ranges during experimentation, and attention to animal nutrition, among other things. For studies of neuroprotection and/or thrombolysis, the effects of putative therapies on survivability must also be taken into consideration.

Rat Models of Focal Cerebral Ischemia

Many animal models of focal cerebral ischemia exist. In this chapter, special focus is paid to the intraluminal MCA occlusion and embolus models because of their relatively widespread use in the development of treatments for stroke. Importantly, these methods can also be performed on mice, allowing for transgenic studies of the pathophysiology of stroke.

Intraluminal MCA Occlusion Model

This model was originally described by Koizumi et al and has since been modified by others [11]. It is the most commonly used of rat models of stroke due to its relative simplicity and noninvasiveness. The MCA is occluded by inserting a monofilament suture into the internal carotid artery (ICA) to block blood flow to the MCA either permanently or transiently by keeping the filament in place or withdrawing it, respectively. Several manuscripts describe in detail the technical and procedural features of this model [11–16].

This model typically induces infarcts in the lateral caudatoputamen and frontoparietal cortex [17]. The infarct is reproducible and there is a significant ischemic penumbra early after MCA occlusion, making this model suitable for testing neuroprotective agents [18]. However, several technical factors may influence infarct size, such as: 1) physical differences in the employed monofilament suture, (2) insertion distance of the suture, and (3) accidental premature reperfusion [19–21]. It is therefore essential that standardized surgical and technical procedures be used by adequately trained personnel in order to generate reproducible lesions. There are also some complications with the intraluminal MCA occlusion model such as subarachnoid hemorrhage secondary to suture-induced arterial rupture, spontaneous hyperthermia when the duration of ischemia is longer than 2 h, and mechanical damage of endothelium which can complicate reperfusion [13, 22, 23]. The complication rates may be reduced by using silicone coated sutures [13].

The intraluminal MCAO model is suitable for neuroprotection drug experiments because it produces a substantial amount of penumbra (salvageable tissue) in the first 60-90 minutes after onset [16]. Also, the location, volume, and temporal evolution of infarction are similar to those produced by proximal electrocoagulation of the MCA [16]. The suture is easily withdrawn, enabling investigators to study the aspects of reperfusion

The suture MCA occlusion model has recently been modified to induce ischemia in a magnetic resonance imaging (MRI) unit by remotely advancing the suture occluder [16]. This in-bore occlusion method has achieved a high reproducibility rate and enables investigators to monitor in vivo ischemic changes at pre-occlusion, acute, subacute and chronic post-occlusion time points [16]. Combined with multiparametric MRI techniques, the MCA occlusion model enables anatomic, diffusion, perfusion, and functional data to be obtained longitudinally and noninvasively in the same animal, making it a powerful tool for studying the pathophysiology of brain ischemia [23, 24].

Thromboembolic Model

Though thromboembolic ischemia can be induced by a photochemical approach, the most commonly used thromboembolic model is blood clot injection, first described in the dog by Hill et al. and later applied to the rat [25–27]. This model is of great interest to researchers because of its close resemblance to human ischemic stroke and its utility in evaluating thrombolytic therapies [28]. Thrombolytic therapy with recombinant tissue plasminogen activator (rt-PA) administered intravenously within 3 h of onset of ischemic stroke in select patients is the only FDA-approved treatment for human stroke and has been shown to improve neurological outcome [29]. Recently, there has been heightened interest in studying the efficacy of combining thrombolytic and neuroprotective agents in the treatment of stroke, giving thromboembolic animal models an increasingly important role in this respect [30].

Several disadvantages were common to the early thromboembolic models, such as diffuse and inhomogenous infarction in the MCA territory from microembolization to peripheral branches [27]. Additionally, spontaneous recanalization frequently occurred, making it difficult to study thrombolytic therapies [16]. Infarct sizes were variable, contralateral strokes were common, and ischemia caused by multiple small clots did not mimic typical clinical ischemic stroke [16]. Later, it was determined that size (length and diameter) and the biological characteristics of the blood clot (fibrin-rich) are crucial to the relevance and reproducibility of this model [16].

Busch et al developed a rat clot model that surmounted the above issues in which a single fibrin-rich autologous clot was injected to produce reliable occlusion of the proximal MCA, with consistent reduction of cerebral blood flow (CBF) and histological damage in the MCA territory seen [31]. No spontaneous thrombolysis was observed and, in separate experiments, thrombolytic therapy with rt-PA or prourokinase recanalized the occluded MCA [31, 32]. Recently, Henninger et al used the embolic model in conjunction with multimodal MRI to investigate the pathophysiological mechanisms underlying the relatively rare clinical phenomenon of "spectacular shrinking deficit" in stroke patients [33].

In conclusion, the single fibrin-rich clot model induces reproducible infarcts in the MCA territory similar to those produced by the intraluminal MCA occlusion model. The clot model has the added advantages of bearing closer similarity to the mechanism underlying human ischemic stroke and better utility for studying thrombolytic therapy. Combined with modern imaging techniques, thromboembolic models have the potential to take the experimental study of stroke to new frontiers.

Non-clot Embolus Models

Numerous compounds have been used to produce artificial emboli which are typically injected into the ICA, most commonly in rats [34]. Microsphere embolization is the most widely used model, with the severity of ischemic damage related to the number of emboli used [35]. The lesions require longer time to develop (24 hours on average) than in the intraluminal models, allowing for a larger therapeutic window for drug testing in microsphere models. However, the permanency of the ischemia does not simulate most clinical situations which limits the applicability of these models. Also, lesions are multifocal and have low reproducibility, though recent macrosphere models have resulted in more reproducible infarcts by increasing the diameter of the spheres and using less of them [36].

Direct Surgical MCAO

Numerous techniques have been developed to surgically approach and occlude the MCA, with the rat being the species most widely used [13]. The orbital route is the least traumatic and, compared to procedures requiring craniotomy, results in less blood loss and artifacts [37]. Electrocautarization of the MCA results in permanent occlusion, whereas clipping or ligature snares enable reperfusion [38]. Occlusion of the MCA following a transient hypotension produces a larger infarct area [39]. Tandem occlusion of the distal MCA and ipsilateral CCA results in more reproducible infarcts [40]. Recently, a three-vessel occlusion model has been shown to produce reproducible and selective neocortical infarction [41]. Common to all of these variants, the procedure is always invasive and requires extreme surgical skill which limits their utility.

Photochemically Induced Thrombosis

This model induces a cortical infarct by systemic injection of a photoactive dye in combination with irradiation by a light beam transmitted through the intact skull [42]. Oxidative damage to the endothelium caused by the altered dye leads to platelet aggregation in the irradiated area [43]. This model is used primarily in spontaneously hypertensive rats [44]. A disadvantage of this model is that vasogenic edema and blood-brain barrier breakdown occur within minutes which does not allow for the formation of penumbra--therefore, this model has been considered by many to be unsuitable for preclinical drug studies [44]. However, a new model overcomes this limitation and induces ischemia over a greatly extended time period, consistently producing a penumbra-like region [45]. This, combined with the ability to noninvasively and reproducibly induce infarct in any cortical location, are obvious advantages of the photochemical infarct model. A major disadvantage is the atypical features of the lesion (prominent vascular injury and early vasogenic edema) which are unlike human stroke [13].

Endothelin-induced MCAO

Endothelin-1 (ET-1) is a natural peptide that causes vasoconstriction and several models use this as an agent to induce MCA stroke [13]. Invasive approaches have been largely replaced by stereotactic intracerebral injection of ET-1 adjacent to the MCA, which avoid complications of surgery [46]. When ET-1 is applied to the MCA there is a significant decrease of cerebral blood flow (CBF) in its territory, resulting in an ischemic lesion pattern similar to that of direct surgical MCAO [47]. This model may be useful in restorative drug studies. Notably, after a period (~20 minutes) of severe CBF reduction, there is a slow and progressive return of blood flow to normal with the rate being dose-dependent [47]. This can be disadvantageous source of variability unless the dose is carefully standardized in the experiment.

Outcome Measures

There are several ways of measuring the severity of ischemic insult in animals: assessment of neurological status, pathological assessment, and in-vivo evaluation with magnetic resonance imaging (MRI). Most prior studies of MCAO focus on the acute phase of ischemia but since neuronal damage can occur days to weeks after insult, it is reasonable that new studies include evaluation of outcome measures both in the acute phase (1-3 days) and chronically (up to 4 weeks) [48].

Assessment of Neurological Status

Many neurological deficits are difficult to assess in animals. Motor deficits are perhaps the easiest to quantify and simple measures of motor function are available in rodent models [49]. Refined tests that assess sensorimotor function include limb placing, beam walking, sticky label test, grid walking, and rotarod [50]. A number of cognitive tests examining learning and short term memory are available for rats, including the Morris water maze [51]. Combining neurological assessment with histological measurements is becoming more critical with the heightened interest in neuroprotective drugs, the effectiveness of which may be reflected more by subtle structural and chemical changes rather than changes in gross infarct volume [52]. This statement is supported by the fact that animal data suggest a poor correlation between reductions in infarct size with neurological or behavioral deficits [13]. Also of importance is the need to extend the period of testing of neurological status for at least 1 month post-insult in animal studies, per the recommendations of the STAIR Committee [14]. However, this may be difficult to implement as survival times are 48 hours or less in many animal stroke models [53].

Pathological Assessment

In models of focal ischemia, the chief outcome has been the infarct volume, traditionally measured by quantitative histology. Among a number of histopathological methods, 2,3,5-triphenyltetrazolium chloride (TTC) and hematoxylin-eosin (H&E) staining are the two most commonly employed. TTC can be used to stain tissue much more rapidly, easily, and cheaply than H&E [54]. TTC is a colorless chemical that is reduced by mitochondrial enzymes into a compound that stains intact brain regions dark red whereas infarcted regions remain white. Studies show TTC staining to be reliable between 6 and 72 hours post-ischemia [13]. Prior to 6 hours, there may not be sufficient number of damaged mitochondria to create contrast between normal and infarcted tissue, and after 72 hours pathophysiologic inflammatory response often obscures the line of demarcation in the periphery of the damaged area [13].

Notwithstanding staining techniques, infarcts have complex shapes with sometimes indistinct margins, making it difficult to measure their volumes. Many methods have been developed to deal with these complexities, each with their own advantages and disadvantages. Perhaps one of the most important considerations to account for is the effect of vasogenic edema on infarct volume. Edema in the rat MCAO stroke model typically accounts for 20-30% of the total apparent infarct volume [55]. Separate measurement of "corrected" infarct volume (which accounts for edema) and "uncorrected" volume (which does not account for edema) is important because of the possibility that some interventions may reduce edema but not salvage brain tissue and vice-versa [13].

References

  1. Bonita R: Stroke prevention: global perspective. In Stroke Prevention. Edited by: Norris JW, Hachinksi V. New York: Oxford University Press; 2001:259–274.

    Google Scholar 

  2. Sacco RL, Wolf PA, Gorelick PB: Risk factors and their management for stroke prevention: outlook for 1999 and beyond. Neurology 1999, 53: S15-S24.

    CAS  PubMed  Google Scholar 

  3. Kaste M, Fogelholm R, Rissanen A: Economic burden of stroke and the evaluation of new therapies. Public Health 1998, 112: 103–112.

    Article  CAS  PubMed  Google Scholar 

  4. Sacco RL, Boden-Albala B: Stroke risk factors: identification and modification. In Stroke Therapy. 2nd edition. Edited by: Fisher M. Woburn, MA: Butterworth-Heinemann; 2001:1–24.

    Google Scholar 

  5. del Zoppo GJ, Poeck K, Pessin MS, Wolpert SM, Furlan AJ, Ferbert A, Alberts MJ, Zivin JA, Wechsler L, Busse O: Recombinant tissue plasminogen activator in acute thrombotic and embolic stroke. Ann Neurol 1992, 32: 78–86. 10.1002/ana.410320113

    Article  CAS  PubMed  Google Scholar 

  6. Hsu CY: Criteria for valid preclinical trials using animal stroke models. Stroke 1993, 24: 633–636.

    Article  CAS  PubMed  Google Scholar 

  7. Yamori Y, Horie R, Handa H, Sato M, Fukase M: Pathogenetic similarity of strokes in stroke-prone spontaneously hypertensive rats and humans. Stroke 1976, 7: 46–53.

    Article  CAS  PubMed  Google Scholar 

  8. Coyle P: Middle cerebral artery occlusion in the young rat. Stroke 1982, 13: 855–859.

    Article  CAS  PubMed  Google Scholar 

  9. Ginsberg MD, Busto R: Rodent models of cerebral ischemia. Stroke 1989, 20: 1627–1642.

    Article  CAS  PubMed  Google Scholar 

  10. Hunter AJ, Green AR, Cross AJ: Animal models of acute ischaemic stroke: can they predict clinically successful neuroprotective drugs? Trends Pharmacol Sci 1995, 16: 123–128. 10.1016/S0165-6147(00)88999-3

    Article  CAS  PubMed  Google Scholar 

  11. Grotta J: Stroke treatment in the human versus animal models. In Cerebrovascular Disease: Pathophysiology, Diagnosis, and Management. Edited by: Ginsberg MD, Bogousslavsky J. Blackwell Scientific, Malden, MA; 1998.

    Google Scholar 

  12. Li F, Han SS, Tatlisumak T, Liu KF, Garcia JH, Sotak CH, Fisher M: Reversal of acute apparent diffusion coefficient abnormalities and delayed neuronal death following transient focal cerebral ischemia in rats. Ann Neurol 1999, 46: 333–342. 10.1002/1531-8249(199909)46:3<333::AID-ANA9>3.0.CO;2-X

    Article  CAS  PubMed  Google Scholar 

  13. Durukan A, Tatlisumak T: Animal models of ischemic stroke. In Handbook of Clinical Neurology., (3rd series), Stroke, Part 1 Basic and epidemiological aspects. Volume 92. Edited by: Fisher M. New York. Elsevier; 2009:1–464.

    Google Scholar 

  14. Fisher M: Recommendations for advancing development of acute stroke therapies: Stroke Therapy Academic Industry Roundtable 3. Stroke 2003, 34: 1539–1546. 10.1161/01.STR.0000072983.64326.53

    Article  CAS  PubMed  Google Scholar 

  15. Li F, Han S, Tatlisumak T, Carano RA, Irie K, Sotak CH, Fisher M: A new method to improve in-bore middle cerebral artery occlusion in rats: demonstration with diffusion- and perfusion-weighted imaging. Stroke 1998, 29: 1715–1720.

    Article  CAS  PubMed  Google Scholar 

  16. Li F, Tatlisumak T: Focal brain ischemia models in rodents. In Handbook of Experimental Neurology: Methods and Techniques in Animal Research. Edited by: Tatlisumak, Fisher M. Cambridge: Cambridge University Press; 2006:311–328.

    Chapter  Google Scholar 

  17. Meng X, Fisher M, Shen Q, Sotak CH, Duong TQ: Characterizing the diffusion/perfusion mismatch in experimental focal cerebral ischemia. Ann Neurol 2004, 55: 207–212. 10.1002/ana.10803

    Article  PubMed Central  PubMed  Google Scholar 

  18. Kuge Y, Minematsu K, Yamaguchi T, Miyake Y: Nylon monofilament for intraluminal middle cerebral occlusion in rats. Stroke 1995, 26: 1655–1658.

    Article  CAS  PubMed  Google Scholar 

  19. Zarow GJ, Karibe H, States BA, Graham SH, Weinstein PR: Endovascular suture occlusion of the middle cerebral artery in rats: effect of suture insertion distance on cerebral blood flow, infarct distribution and infarct volume. Neurol Res 1997, 19: 409–416.

    CAS  PubMed  Google Scholar 

  20. Schmid-Elsaesser R, Zausinger S, Hungerhuber E, Baethmann A, Reulen HJ: A critical reevaluation of the intraluminal thread model of focal cerebral ischemia: evidence of inadvertent premature reperfusion and subarachnoid hemorrhage in rats by laser-Doppler flowmetry. Stroke 1998, 29: 2162–2170.

    Article  CAS  PubMed  Google Scholar 

  21. Zhao Q, Memezawa H, Smith ML, Siesjo BK: Hyperthermia complicates middle cerebral artery occlusion induced by an intraluminal filament. Brain Res 1994, 649: 253–259. 10.1016/0006-8993(94)91071-5

    Article  CAS  PubMed  Google Scholar 

  22. Li F, Omae T, Fisher M: Spontaneous hyperthermia and its mechanism in the intraluminal suture middle cerebral artery occlusion model of rats. Stroke 1999, 30: 2464–2471.

    Article  CAS  PubMed  Google Scholar 

  23. Sicard KM, Henninger N, Fisher M, Duong TQ, Ferris CF: Differential recovery of multimodal MRI and behavior after transient focal cerebral ischemia in rats. J Cereb Blood Flow Metab 2006, 11: 1451–1462. 10.1038/sj.jcbfm.9600299

    Article  Google Scholar 

  24. Sicard KM, Henninger N, Fisher M, Duong TQ, Ferris CF: Long-term changes of functional MRI-based brain function, behavioral status, and histopathology after transient focal cerebral ischemia in rats. Stroke 2006, 10: 2593–2600. 10.1161/01.STR.0000239667.15532.c1

    Article  Google Scholar 

  25. Hill NC, Millikan CH, Wakim KG, Sayre GP: Studies in cerebrovascular disease. VII. Experimental production of cerebral infarction by intracarotid injection of homologous blood clot: preliminary report. Mayo Clin Proc 1955, 30: 625–633.

    CAS  Google Scholar 

  26. Futrell N, Watson BD, Dietrich WD, Prado R, Millikan C, Ginsberg MD: A new model of embolic stroke produced by photochemical injury to the carotid artery in the rat. Ann Neurol 1988, 23: 251–257. 10.1002/ana.410230307

    Article  CAS  PubMed  Google Scholar 

  27. Kudo M, Aoyama A, Ichimori S, Fukunaga N: An animal model of cerebral infarction: homologous blood clot emboli in rats. Stroke 1982, 13: 505–508.

    Article  CAS  PubMed  Google Scholar 

  28. Albers GW: Antithrombotic agents in cerebral ischemia. Am J Cardiol 1995, 75: 348–388. 10.1016/0002-9149(95)80008-G

    Article  Google Scholar 

  29. National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group T: Tissue plasminogen activator for acute ischemic stroke. New Engl J Med 1995, 333: 1581–1587. 10.1056/NEJM199512143332401

    Article  Google Scholar 

  30. Savitz SI, Fisher M: Future of neuroprotection for acute stroke: in the aftermath of the SAINT trials. Ann Neurol 2007, 5: 396–402. 10.1002/ana.21127

    Article  Google Scholar 

  31. Busch E, Kruger K, Hossmann KA: Improved model of thromboembolic stroke and rt-PA induced reperfusion in the rat. Brain Res 1997, 778: 16–24. 10.1016/S0006-8993(97)01008-1

    Article  CAS  PubMed  Google Scholar 

  32. Takano K, Carano RA, Tatlisumak T, Meiler M, Sotak CH, Kleinert HD, Fisher M: Efficacy of intra-arterial and intravenous prourokinase in an embolic stroke model evaluated by diffusion-perfusion magnetic resonance imaging. Neurology 1998, 50: 870–875.

    Article  CAS  PubMed  Google Scholar 

  33. Henninger N, Sicard KM, Fisher M: Spectacular shrinking deficit: insights from multimodal magnetic resonance imaging after embolic middle cerebral artery occlusion in Sprague-Dawley rats. J Cereb Blood Flow Metab 2007, 10: 1756–1763. 10.1038/sj.jcbfm.9600477

    Article  Google Scholar 

  34. Molnar L, Hegedus K, Fekete I: A new model for inducing transient cerebral ischemia and subsequent reperfusion in rabbits without craniectomy. Stroke 1988, 19: 1262–1266.

    Article  CAS  PubMed  Google Scholar 

  35. Zivin JA, DeGirolami U, Kochhar A, Lyden PD, Mazzarella V, Hemenway CC, Henry ME: A model for quantitative evaluation of embolic stroke therapy. Brain Res 1987, 435: 305–309. 10.1016/0006-8993(87)91613-1

    Article  CAS  PubMed  Google Scholar 

  36. Gerriets T, Li F, Silva MD, Meng X, Brevard M, Sotak CH, Fisher M: The macrosphere model: evaluation of a new stroke model for permanent middle cerebral artery occlusion in rats. J Neurosci Methods 2003, 122: 210–211. 10.1016/S0165-0270(02)00322-9

    Article  Google Scholar 

  37. Pulsinelli W, Jacewicz M: Animal models of brain ischemia. In Stroke: Pathophysiology, Diagnosis, and Management. 2nd edition. Edited by: Barnett HJ, Mohr JP, Stein BM, et al. Churchill Livingstone, New York; 1992.

    Google Scholar 

  38. Shigeno T, Teasdale GM, McCulloch J, Graham DI: Recirculation model following MCA occlusion in rats. Cerebral blood flow, cerebrovascular permeability, and brain edema. J Neurosurg 1985, 63: 272–277. 10.3171/jns.1985.63.2.0272

    Article  CAS  PubMed  Google Scholar 

  39. Osborne KA, Shigeno T, Balarsky AM, Ford I, McCulloch J, Teasdale GM, Graham DI: Quantitative assessment of early brain damage in a rat model of focal cerebral ischaemia. J Neurol Neurosurg Psychiatry 1987, 50: 402–410. 10.1136/jnnp.50.4.402

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  40. Brint S, Jacewicz M, Kiessling M, Tanabe J, Pulsinelli W: Focal brain ischemia in the rat: methods for reproducible neocortical infarction using tandem occlusion of the distal middle cerebral and ipsilateral common carotid arteries. J Cereb Blood Flow Metab 1988, 8: 474–485.

    Article  CAS  PubMed  Google Scholar 

  41. Yanamoto H, Nagata I, Niitsu Y, Xue JH, Zhang Z, Kikuchi H: Evaluation of MCAO stroke models in normotensive rats: standardized neocortical infarction by the 3VO technique. Exp Neurol 2003, 182: 261–274. 10.1016/S0014-4886(03)00116-X

    Article  PubMed  Google Scholar 

  42. Watson BD, Dietrich WD, Busto R, Wachtel MS, Ginsberg MD: Induction of reproducible brain infarction by photochemically initiated thrombosis. Ann Neurol 1985, 17: 497–504. 10.1002/ana.410170513

    Article  CAS  PubMed  Google Scholar 

  43. Dietrich WD, Watson BD, Busto R, Ginsberg MD, Bethea JR: Photochemically induced cerebral infarction. I. Early microvascular alterations. Acta Neuropathol (Berl) 1987, 72: 315–325. 10.1007/BF00687262

    Article  CAS  Google Scholar 

  44. Cai H, Yao H, Ibayashi S, et al.: Photothrombotic middle cerebral artery occlusion in spontaneously hypertensive rats: influence of substrain, gender, and distal middle cerebral artery patterns on infarct size. Stroke 1998, 29: 1982–1986.

    Article  CAS  PubMed  Google Scholar 

  45. Hilger T, Blunk JA, Hoehn M, Mies G, Wester P: Characterization of a novel chronic photothrombotic ring stroke model in rats by magnetic resonance imaging, biochemical imaging, and histology. J Cereb Blood Flow Metab 2004, 24: 789–797. 10.1097/01.WCB.0000123905.17746.DB

    Article  PubMed  Google Scholar 

  46. Sharkey J, Butcher SP: Characterisation of an experimental model of stroke produced by intracerebral microinjection of endothelin-1 adjacent to the rat middle cerebral artery. J Neurosci Methods 1995, 60: 125–131. 10.1016/0165-0270(95)00003-D

    Article  CAS  PubMed  Google Scholar 

  47. Macrae IM, Robinson MJ, Graham DI, Reid JL, McCulloch J: Endothelin- 1-induced reductions in cerebral blood flow: dose dependency, time course, and neuropathological consequences. J Cereb Blood Flow Metab 1993, 13: 276–284.

    Article  CAS  PubMed  Google Scholar 

  48. Persson L, Hårdemark HG, Bolander HG, Hillered L, Olsson Y: Neurologic and neuropathologic outcome after middle cerebral artery occlusion in rats. Stroke 1989, 20: 641–645.

    Article  CAS  PubMed  Google Scholar 

  49. Bederson JB, Pitts LH, Tsuji M, Nishimura MC, Davis RL, Bartkowski H: Rat middle cerebral artery occlusion: evaluation of the model and development of a neurologic examination. Stroke 1986, 17: 472–476.

    Article  CAS  PubMed  Google Scholar 

  50. Hunter AJ, Hatcher J, Virley D, Nelson P, Irving E, Hadingham SJ, Parsons AA: Functional assessments in mice and rats after focal stroke. Neuropharmacology 2000, 39: 806–816. 10.1016/S0028-3908(99)00262-2

    Article  CAS  PubMed  Google Scholar 

  51. D'Hooge R, DE Deyn PP: Applications of the Morris water maze in the study of learning and memory. Brain Res Brain Res Rev 2001, 36: 60–90. 10.1016/S0165-0173(01)00067-4

    Article  PubMed  Google Scholar 

  52. Roof RL, Schielke GP, Ren X, Hall ED: A comparison of long-term functional outcome after 2 middle cerebral artery occlusion models in rats. Stroke 2001, 32: 2648–2657. 10.1161/hs1101.097397

    Article  CAS  PubMed  Google Scholar 

  53. DeBow SB, Clark DL, Maclellan CL, Colbourne F: Incomplete assessment of experimental cytoprotectants in rodent ischemia studies. Can J Neurol Sci 2003, 30: 368–374.

    Article  PubMed  Google Scholar 

  54. Bederson JB, Pitts LH, Germano SM, Nishimura MC, Davis RL, Bartkowski HM: Evaluation of 2,3,5-triphenyltetrazolium chloride as a stain for detection and quantification of experimental cerebral infarction in rats. Stroke 1986, 17: 1304–1308.

    Article  CAS  PubMed  Google Scholar 

  55. Swanson RA, Sharp FR: Infarct measurement methodology. J Cereb Blood Flow Metab 1994, 14: 697–698.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Marc Fisher.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

Both KMS and MF performed background research and wrote the manuscript. All authors read and approved the final manuscript.

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Sicard, K.M., Fisher, M. Animal models of focal brain ischemia. Exp & Trans Stroke Med 1, 7 (2009). https://doi.org/10.1186/2040-7378-1-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/2040-7378-1-7

Keywords