Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Encouraging an excitable brain state: mechanisms of brain repair in stroke

Abstract

Stroke induces a plastic state in the brain. This period of enhanced plasticity leads to the sprouting of new axons, the formation of new synapses and the remapping of sensory-motor functions, and is associated with motor recovery. This is a remarkable process in the adult brain, which is normally constrained in its levels of neuronal plasticity and connectional change. Recent evidence indicates that these changes are driven by molecular systems that underlie learning and memory, such as changes in cellular excitability during memory formation. This Review examines circuit changes after stroke, the shared mechanisms between memory formation and brain repair, the changes in neuronal excitability that underlie stroke recovery, and the molecular and pharmacological interventions that follow from these findings to promote motor recovery in animal models. From these findings, a framework emerges for understanding recovery after stroke, central to which is the concept of neuronal allocation to damaged circuits. The translation of the concepts discussed here to recovery in humans is underway in clinical trials for stroke recovery drugs.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Endogenous plasticity in subacute stroke.
Fig. 2: Parallels between windows of plasticity in development and stroke.
Fig. 3: Storage of memory in coactive neuronal networks, or engrams.
Fig. 4: Recovery engram in stroke.

Similar content being viewed by others

References

  1. Benjamin, E. J. et al. Heart disease and stroke statistics — 2019 update: a report from the American Heart Association. Circulation 139, e56–e528 (2019).

    PubMed  Google Scholar 

  2. Faul, M., Xu, L., Wald, M. M. & Coronado, V. G. Traumatic brain injury in the United States: emergency department visits hospitalizations, and deaths (CDC, 2010).

  3. Song, S. S. Advanced imaging in acute ischemic stroke. Semin. Neurol. 33, 436–440 (2013).

    CAS  PubMed  Google Scholar 

  4. Fisher, M. & Albers, G. W. Advanced imaging to extend the therapeutic time window of acute ischemic stroke. Ann. Neurol. 73, 4–9 (2013).

    PubMed  Google Scholar 

  5. Sandhu, G. S. & Sunshine, J. L. Advanced neuroimaging to guide acute stroke therapy. Curr. Cardiol. Rep. 14, 741–753 (2012).

    PubMed  Google Scholar 

  6. Yoo, A. J., Pulli, B. & Gonzalez, R. G. Imaging-based treatment selection for intravenous and intra-arterial stroke therapies: a comprehensive review. Expert Rev. Cardiovasc. Ther. 9, 857–876 (2011).

    PubMed  PubMed Central  Google Scholar 

  7. Schaechter, J. D. Motor rehabilitation and brain plasticity after hemiparetic stroke. Prog. Neurobiol. 73, 61–72 (2004).

    PubMed  Google Scholar 

  8. Leng, T. & Xiong, Z.-G. Treatment for ischemic stroke: from thrombolysis to thrombectomy and remaining challenges. Brain Circ. 5, 8 (2019).

    PubMed  PubMed Central  Google Scholar 

  9. Cirillo, C. et al. Post-stroke remodeling processes in animal models and humans. J. Cereb. Blood Flow Metab. 40, 3–22 (2020).

    PubMed  Google Scholar 

  10. Carmichael, S. T. The 3 Rs of stroke biology: radial, relayed, and regenerative. Neurotherapeutics 3, 348–359 (2016).

    Google Scholar 

  11. Sharbrough, F. W., Messick, J. M. & Sundt, T. M. Correlation of continuous electroencephalograms with cerebral blood flow measurements during carotid endarterectomy. Stroke 4, 674–683 (1973).

    CAS  PubMed  Google Scholar 

  12. Heiss, W. D., Hayakawa, T. & Waltz, A. G. Cortical neuronal function during ischemia: effects of occlusion of one middle cerebral artery on single-unit activity in cats. Arch. Neurol. 33, 813–820 (1976).

    CAS  PubMed  Google Scholar 

  13. Branston, N. M., Symon, L., Crockard, H. A. & Pasztor, E. Relationship between the cortical evoked potential and local cortical blood flow following acute middle cerebral artery occlusion in the baboon. Exp. Neurol. 45, 195–208 (1974).

    CAS  PubMed  Google Scholar 

  14. Mies, G., Ishimaru, S., Xie, Y., Seo, K. & Hossmann, K. A. Ischemic thresholds of cerebral protein synthesis and energy state following middle cerebral artery occlusion in rat. J. Cereb. Blood Flow Metab. 11, 753–761 (1991).

    CAS  PubMed  Google Scholar 

  15. Taylor, R. A. & Sansing, L. H. Microglial responses after ischemic stroke and intracerebral hemorrhage. Clin. Dev. Immunol. 2013, 746068 (2013).

    PubMed  PubMed Central  Google Scholar 

  16. Anrather, J. & Iadecola, C. Inflammation and stroke: an overview. Neurotherapeutics 13, 661–670 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Carmichael, S. T. Brain excitability in stroke: the yin and yang of stroke progression. Arch. Neurol. 69, 161–167 (2012).

    PubMed  Google Scholar 

  18. Bernhardt, J. et al. Efficacy and safety of very early mobilisation within 24 h of stroke onset (AVERT): a randomised controlled trial. Lancet 386, 46–55 (2015).

    Google Scholar 

  19. Dromerick, A. W. et al. Very early constraint-induced movement during stroke rehabilitation (VECTORS): a single-center RCT. Neurology 73, 195–201 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Clarkson, A. N. et al. AMPA receptor-induced local brain-derived neurotrophic factor signaling mediates motor recovery after stroke. J. Neurosci. 31, 3766–3775 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Bernhardt, J. et al. Agreed definitions and a shared vision for new standards in stroke recovery research: The Stroke Recovery and Rehabilitation Roundtable taskforce. Neurorehabil. Neural Repair 31, 793–799 (2017).

    PubMed  Google Scholar 

  22. Corbett, D. et al. Enhancing the alignment of the preclinical and clinical stroke recovery research pipeline: consensus-based core recommendations from the Stroke Recovery and Rehabilitation Roundtable Translational Working Group. Neurorehabil. Neural Repair 31, 699–707 (2017).

    PubMed  Google Scholar 

  23. Ward, N. S., Brander, F. & Kelly, K. Intensive upper limb neurorehabilitation in chronic stroke: outcomes from the Queen Square programme. J. Neurol. Neurosurg. Psychiatry 90, 498–506 (2019).

    PubMed  Google Scholar 

  24. Lo, A. C. et al. Robot-assisted therapy for long-term upper-limb impairment after stroke. N. Engl. J. Med. 362, 1772–1783 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Duncan, P. W., Goldstein, L. B., Matchar, D., Divine, G. W. & Feussner, J. Measurement of motor recovery after stroke. Outcome assessment and sample size requirements. Stroke 23, 1084–1089 (1992).

    CAS  PubMed  Google Scholar 

  26. Duncan, P. W., Lai, S. M. & Keighley, J. Defining post-stroke recovery: implications for design and interpretation of drug trials. Neuropharmacology 39, 835–841 (2000).

    CAS  PubMed  Google Scholar 

  27. Wolf, S. L. et al. The EXCITE stroke trial: comparing early and delayed constraint-induced movement therapy. Stroke 41, 2309–2315 (2010).

    PubMed  PubMed Central  Google Scholar 

  28. Gladstone, D. J., Danells, C. J. & Black, S. E. The Fugl-Meyer assessment of motor recovery after stroke: a critical review of its measurement properties. Neurorehabil. Neural Repair 16, 232–240 (2002).

    PubMed  Google Scholar 

  29. McCabe, J. et al. Comparison of robotics, functional electrical stimulation, and motor learning methods for treatment of persistent upper extremity dysfunction after stroke: a randomized controlled trial. Arch. Phys. Med. Rehabil. 96, 981–990 (2015).

    PubMed  Google Scholar 

  30. Li, S. et al. An age-related sprouting transcriptome provides molecular control of axonal sprouting after stroke. Nat. Neurosci. 13, 1496–1506 (2010). This study provides evidence for a transcriptional programme unique to post-stroke periods in the peri-infarct cortex that supports axonal outgrowth in the adult and aged brain.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Li, S. et al. GDF10 is a signal for axonal sprouting and functional recovery after stroke. Nat. Neurosci. 18, 1737–1745 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Overman, J. J. et al. A role for ephrin-A5 in axonal sprouting, recovery, and activity-dependent plasticity after stroke. Proc. Natl Acad. Sci. USA 109, E2230–E2239 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Joy, M. T. et al. CCR5 is a therapeutic target for recovery after stroke and traumatic brain injury. Cell 176, 1143–1157.e13 (2019). This paper describes a gene system that is involved in learning and stroke recovery in mouse models and patients with stroke.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Luke, L. M., Allred, R. P. & Jones, T. A. Unilateral ischemic sensorimotor cortical damage induces contralesional synaptogenesis and enhances skilled reaching with the ipsilateral forelimb in adult male rats. Synapse 54, 187–199 (2004).

    CAS  PubMed  Google Scholar 

  35. Brown, C. E., Aminoltejari, K., Erb, H., Winship, I. R. & Murphy, T. H. In vivo voltage-sensitive dye imaging in adult mice reveals that somatosensory maps lost to stroke are replaced over weeks by new structural and functional circuits with prolonged modes of activation within both the peri-infarct zone and distant sites. J. Neurosci. 29, 1719–1734 (2009). This study reports reorganization of limb representations in the motor cortex following stroke.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Wahl, A. S. et al. Neuronal repair. Asynchronous therapy restores motor control by rewiring of the rat corticospinal tract after stroke. Science 344, 1250–1255 (2014).

    CAS  PubMed  Google Scholar 

  37. Brown, C. E., Wong, C. & Murphy, T. H. Rapid morphologic plasticity of peri-infarct dendritic spines after focal ischemic stroke. Stroke 39, 1286–1291 (2008).

    PubMed  Google Scholar 

  38. Mostany, R. et al. Local hemodynamics dictate long-term dendritic plasticity in peri-infarct cortex. J. Neurosci. 30, 14116–14126 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Brown, C. E., Li, P., Boyd, J. D., Delaney, K. R. & Murphy, T. H. Extensive turnover of dendritic spines and vascular remodeling in cortical tissues recovering from stroke. J. Neurosci. 27, 4101–4109 (2007). This study provides evidence for dendritic spine turnover after stroke.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Bundy, D. T. & Nudo, R. J. Preclinical studies of neuroplasticity following experimental brain injury. Stroke 50, 2626–2633 (2019).

    PubMed  PubMed Central  Google Scholar 

  41. Harrison, T. C., Silasi, G., Boyd, J. D. & Murphy, T. H. Displacement of sensory maps and disorganization of motor cortex after targeted stroke in mice. Stroke 44, 2300–2306 (2013).

    PubMed  Google Scholar 

  42. Nudo, R. J. & Milliken, G. W. Reorganization of movement representations in primary motor cortex following focal ischemic infarcts in adult squirrel monkeys. J. Neurophysiol. 75, 2144–2149 (1996).

    CAS  PubMed  Google Scholar 

  43. Jaillard, A., Martin, C. D., Garambois, K., Lebas, J. F. & Hommel, M. Vicarious function within the human primary motor cortex? A longitudinal fMRI stroke study. Brain 128, 1122–1138 (2005).

    PubMed  Google Scholar 

  44. Cramer, S. C. & Crafton, K. R. Somatotopy and movement representation sites following cortical stroke. Exp. Brain Res. 168, 25–32 (2006).

    PubMed  Google Scholar 

  45. Nishibe, M., Urban, E. T. R., Barbay, S. & Nudo, R. J. Rehabilitative training promotes rapid motor recovery but delayed motor map reorganization in a rat cortical ischemic infarct model. Neurorehabil. Neural Repair 29, 472–482 (2015).

    PubMed  Google Scholar 

  46. Zeiler, S. R. et al. Paradoxical motor recovery from a first stroke after induction of a second stroke: reopening a postischemic sensitive period. Neurorehabil. Neural Repair 30, 794–800 (2016). This study provides evidence for the existence of a sensitive period after stroke.

    PubMed  Google Scholar 

  47. Dromerick, A. Critical Periods After Stroke Study (CPASS): does a critical period exist during stroke recovery? Int. J. Stroke 11 (Suppl. 3), 227 (2016).

    Google Scholar 

  48. Dromerick, A. W. et al. Critical periods after stroke study: translating animal stroke recovery experiments into a clinical trial. Front. Hum. Neurosci. 9, 231 (2015).

    PubMed  PubMed Central  Google Scholar 

  49. Kraft, A. W., Bauer, A. Q., Culver, J. P. & Lee, J. M. Sensory deprivation after focal ischemia in mice accelerates brain remapping and improves functional recovery through Arc-dependent synaptic plasticity. Sci. Transl. Med. 10, eaag1328 (2018).

    PubMed  Google Scholar 

  50. Maravall, M., Stern, E. A. & Svoboda, K. Development of intrinsic properties and excitability of layer 2/3 pyramidal neurons during a critical period for sensory maps in rat barrel cortex. J. Neurophysiol. 92, 144–156 (2004).

    CAS  PubMed  Google Scholar 

  51. Takesian, A. E., Bogart, L. J., Lichtman, J. W. & Hensch, T. K. Inhibitory circuit gating of auditory critical-period plasticity. Nat. Neurosci. 21, 218–227 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Hensch, T. K. Critical period plasticity in local cortical circuits. Nat. Rev. Neurosci. 6, 877–888 (2005).

    CAS  PubMed  Google Scholar 

  53. Wiesel, T. N. & Hubel, D. H. Single-cell responses in striate cortex of kittens deprived of vision in one eye. J. Neurophysiol. 26, 1003–1017 (1963).

    CAS  PubMed  Google Scholar 

  54. Wang, B. S., Sarnaik, R. & Cang, J. Critical period plasticity matches binocular orientation preference in the visual cortex. Neuron 65, 246–256 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Che, A. et al. Layer I interneurons sharpen sensory maps during neonatal development. Neuron 99, 98–116.e7 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Biernaskie, J., Chernenko, G. & Corbett, D. Efficacy of rehabilitative experience declines with time after focal ischemic brain injury. J. Neurosci. 24, 1245–1254 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Carmichael, S. T. & Chesselet, M. F. Synchronous neuronal activity is a signal for axonal sprouting after cortical lesions in the adult. J. Neurosci. 22, 6062–6070 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. McLaughlin, T., Torborg, C. L., Feller, M. B. & O’Leary, D. D. M. Retinotopic map refinement requires spontaneous retinal waves during a brief critical period of development. Neuron 40, 1147–1160 (2003).

    CAS  PubMed  Google Scholar 

  59. Miyata, S., Komatsu, Y., Yoshimura, Y., Taya, C. & Kitagawa, H. Persistent cortical plasticity by upregulation of chondroitin 6-sulfation. Nat. Neurosci. 15, 414–422 (2012).

    CAS  PubMed  Google Scholar 

  60. Pizzorusso, T. et al. Reactivation of ocular dominance plasticity in the adult visual cortex. Science 298, 1248–1251 (2002).

    CAS  PubMed  Google Scholar 

  61. Gherardini, L., Gennaro, M. & Pizzorusso, T. Perilesional treatment with chondroitinase ABC and motor training promote functional recovery after stroke in rats. Cereb. Cortex 25, 202–212 (2015).

    PubMed  Google Scholar 

  62. Hill, J. J., Jin, K., Mao, X. O., Xie, L. & Greenberg, D. A. Intracerebral chondroitinase ABC and heparan sulfate proteoglycan glypican improve outcome from chronic stroke in rats. Proc. Natl Acad. Sci. USA 109, 9155–9160 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Yaeger, C. E., Ringach, D. L. & Trachtenberg, J. T. Neuromodulatory control of localized dendritic spiking in critical period cortex. Nature 567, 100–104 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Conner, J. M., Chiba, A. A. & Tuszynski, M. H. The basal forebrain cholinergic system is essential for cortical plasticity and functional recovery following brain injury. Neuron 46, 173–179 (2005).

    CAS  PubMed  Google Scholar 

  65. Clarkson, A. N., Huang, B. S., MacIsaac, S. E., Mody, I. & Carmichael, S. T. Reducing excessive GABA-mediated tonic inhibition promotes functional recovery after stroke. Nature 468, 305–309 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Orfila, J. E. et al. Delayed inhibition of tonic inhibition enhances functional recovery following experimental ischemic stroke. J. Cereb. Blood Flow Metab. 39, 1005–1014 (2019).

    PubMed  Google Scholar 

  67. Lake, E. M. R. et al. The effects of delayed reduction of tonic inhibition on ischemic lesion and sensorimotor function. J. Cereb. Blood Flow Metab. 35, 1601–1609 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Takeshi, H. et al. Enhanced phasic GABA inhibition during the repair phase of stroke: a novel therapeutic target. Brain 139, 468–480 (2016).

    Google Scholar 

  69. McGee, A. W., Yang, Y., Fischer, Q. S., Daw, N. W. & Strittmatter, S. M. Experience-driven plasticity of visual cortex limited by myelin and Nogo receptor. Science 309, 2222–2226 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Sozmen, E. G. et al. Nogo receptor blockade overcomes remyelination failure after white matter stroke and stimulates functional recovery in aged mice. Proc. Natl Acad. Sci. USA 113, E8453–E8462 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Papadopoulos, C. M. et al. Functional recovery and neuroanatomical plasticity following middle cerebral artery occlusion and IN‐1 antibody treatment in the adult rat. Ann. Neurol. 51, 433–441 (2002).

    PubMed  Google Scholar 

  72. Markus, T. M. et al. Recovery and brain reorganization after stroke in adult and aged rats. Ann. Neurol. 58, 950–953 (2005).

    PubMed  Google Scholar 

  73. Lindau, N. T. et al. Rewiring of the corticospinal tract in the adult rat after unilateral stroke and anti-Nogo-A therapy. Brain 137, 739–756 (2014).

    PubMed  Google Scholar 

  74. Lim, D. H., Ledue, J. M., Mohajerani, M. H. & Murphy, T. H. Optogenetic mapping after stroke reveals network-wide scaling of functional connections and heterogeneous recovery of the peri-infarct. J. Neurosci. 34, 16455–16466 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. McDonnell, M. N., Stinear, C. M., McDonnell, M. N. & Stinear, C. M. TMS measures of motor cortex function after stroke: a meta-analysis. Brain Stimul. 10, 721–734 (2017).

    PubMed  Google Scholar 

  76. Shohami, E., Biegon, A., Shohami, E. & Biegon, A. Novel approach to the role of NMDA receptors in traumatic brain injury. CNS Neurol. Disord. Drug Targets 13, 567–573 ((2014).

    CAS  PubMed  Google Scholar 

  77. Di Lazzaro, V. et al. Motor cortex plasticity predicts recovery in acute stroke. Cereb. Cortex 20, 1523–1528 (2010).

    PubMed  Google Scholar 

  78. Winship, I. R. & Murphy, T. H. In vivo calcium imaging reveals functional rewiring of single somatosensory neurons after stroke. J. Neurosci. 28, 6592–6606 (2008). This report provides evidence for neural activity underlying limb selectivity and reorganization in the periods after stroke.

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Glykys, J. & Mody, I. Activation of GABAA receptors: views from outside the synaptic cleft. Neuron 56, 763–770 (2007).

    CAS  PubMed  Google Scholar 

  80. Atack, J. R. Preclinical and clinical pharmacology of the GABAA receptor α5 subtype-selective inverse agonist α5IA. Pharmacol. Ther. 125, 11–26 (2010).

    CAS  PubMed  Google Scholar 

  81. Kim, Y. K., Yang, E. J., Cho, K., Lim, J. Y. & Paik, N. J. Functional recovery after ischemic stroke is associated with reduced gabaergic inhibition in the cerebral cortex: a GABA PET study. Neurorehabil. Neural Repair 28, 576–583 (2014).

    PubMed  Google Scholar 

  82. Wang, Y. C. et al. Postacute delivery of GABAA α5 antagonist promotes postischemic neurological recovery and peri-infarct brain remodeling. Stroke 49, 2495–2503 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Jourdi, H. et al. Positive AMPA receptor modulation rapidly stimulates BDNF release and increases dendritic mRNA translation. J. Neurosci. 29, 8688–8697 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Clarkson, A. N., Parker, K., Nilsson, M., Walker, F. R. & Gowing, E. K. Combined ampakine and BDNF treatments enhance poststroke functional recovery in aged mice via AKT-CREB signaling. J. Cereb. Blood Flow Metab. 35, 1272–1279 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Abe, H. et al. CRMP2-binding compound, edonerpic maleate, accelerates motor function recovery from brain damage. Science 360, 50–57 (2018).

    CAS  PubMed  Google Scholar 

  86. Attneave, F. The organization of behavior: a neuropsychological theory. Am. J. Psych. 63, 633–635 (1950).

    Google Scholar 

  87. Rioult-Pedotti, M. S., Friedman, D., Hess, G. & Donoghue, J. P. Strengthening of horizontal cortical connections following skill learning. Nat. Neurosci. 1, 230–234 (1998).

    CAS  PubMed  Google Scholar 

  88. Xu, T. et al. Rapid formation and selective stabilization of synapses for enduring motor memories. Nature 462, 915–919 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Bailey, C. H., Kandel, E. R. & Harris, K. M. Structural components of synaptic plasticity and memory consolidation. Cold Spring Harb. Perspect. Biol. 7, a021758 (2015).

    PubMed  PubMed Central  Google Scholar 

  90. Cheng, M. Y. et al. Optogenetic neuronal stimulation promotes functional recovery after stroke. Proc. Natl Acad. Sci. USA 111, 12913–12918 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Tennant, K. A., Taylor, S. L., White, E. R. & Brown, C. E. Optogenetic rewiring of thalamocortical circuits to restore function in the stroke injured brain. Nat. Commun. 8, 15879 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Wahl, A. S. et al. Optogenetically stimulating intact rat corticospinal tract post-stroke restores motor control through regionalized functional circuit formation. Nat. Commun. 8, 1187 (2017). This study shows that optogenetic silencing of corticospinal neurons that have sprouted new connections after stroke impairs motor function that was regained.

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Blomstedt, P. & Hariz, M. I. Are complications less common in deep brain stimulation than in ablative procedures for movement disorders? Stereotact. Funct. Neurosurg. 84, 72–81 (2006).

    PubMed  Google Scholar 

  94. Mandat, T. S., Hurwitz, T. & Honey, C. R. Hypomania as an adverse effect of subthalamic nucleus stimulation: report of two cases. Acta Neurochir. 148, 895–897 (2006).

    CAS  PubMed  Google Scholar 

  95. Ramanathan, D. S. et al. Low-frequency cortical activity is a neuromodulatory target that tracks recovery after stroke. Nat. Med. 24, 1257–1267 (2018). This study reports that timed electrical stimulation in rodent models and in a patient with stroke can restore lost motor function.

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Biasiucci, A. et al. Brain-actuated functional electrical stimulation elicits lasting arm motor recovery after stroke. Nat. Commun. 9, 2421 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Engineer, N. D. et al. Targeted vagus nerve stimulation for rehabilitation after stroke. Front. Neurosci. 13, 280 (2019).

    PubMed  PubMed Central  Google Scholar 

  98. Hays, S. A. et al. The timing and amount of vagus nerve stimulation during rehabilitative training affect poststroke recovery of forelimb strength. Neuroreport 25, 676–682 (2014).

    PubMed  Google Scholar 

  99. Bönstrup, M. et al. Low-frequency brain oscillations track motor recovery in human stroke. Ann. Neurol. 86, 853–865 (2019).

    PubMed  Google Scholar 

  100. Guggenmos, D. J. et al. Restoration of function after brain damage using a neural prosthesis. Proc. Natl Acad. Sci. USA 110, 21177–21182 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Kandel, E. R. The molecular biology of memory: CAMP, PKA, CRE, CREB-1, CREB-2, and CPEB. Mol. Brain 5, 14 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Sano, Y. et al. CREB regulates memory allocation in the insular cortex. Curr. Biol. 24, 2833–2837 (2014). This study details neuronal allocation as a result of CREB function.

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Caracciolo, L. et al. CREB controls cortical circuit plasticity and functional recovery after stroke. Nat. Commun. 9, 2250 (2018). This study shows that overexpression of CREB improves motor function, presumably through neuronal allocation.

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Birjandi, S. Z. et al. Phosphodiesterase 10A inhibition leads to brain region-specific recovery based on stroke type. Transl. Stroke Res. https://doi.org/10.1007/s12975-020-00819-8 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  105. Zhou, M. et al. CCR5 is a suppressor for cortical plasticity and hippocampal learning and memory. eLife 5, e20985 (2016).

    PubMed  PubMed Central  Google Scholar 

  106. Arber, S. Motor circuits in action: specification, connectivity, and function. Neuron 74, 975–989 (2012).

    CAS  PubMed  Google Scholar 

  107. Ohashi, H., Gribble, P. L. & Ostry, D. J. Somatosensory cortical excitability changes precede those in motor cortex during human motor learning. J. Neurophysiol. 122, 1397–1405 (2019).

    PubMed  PubMed Central  Google Scholar 

  108. Biane, J. S., Scanziani, M., Tuszynski, M. H. & Conner, J. M. Motor cortex maturation is associated with reductions in recurrent connectivity among functional subpopulations and increases in intrinsic excitability. J. Neurosci. 35, 4719–4728 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Costa, R. M., Cohen, D. & Nicolelis, M. A. L. Differential corticostriatal plasticity during fast and slow motor skill learning in mice. Curr. Biol. 14, 1124–1134 (2004).

    CAS  PubMed  Google Scholar 

  110. Peters, A. J., Chen, S. X. & Komiyama, T. Emergence of reproducible spatiotemporal activity during motor learning. Nature 510, 263–267 (2014). This study details searching, selection and refinement of the motor engram during stages of motor learning.

    CAS  PubMed  Google Scholar 

  111. Li, Q. et al. Refinement of learned skilled movement representation in motor cortex deep output layer. Nat. Commun. 8, 15834 (2017).

    PubMed  PubMed Central  Google Scholar 

  112. Masamizu, Y. et al. Two distinct layer-specific dynamics of cortical ensembles during learning of a motor task. Nat. Neurosci. 17, 987–994 (2014).

    CAS  PubMed  Google Scholar 

  113. Makino, H., Hwang, E. J., Hedrick, N. G. & Komiyama, T. Circuit mechanisms of sensorimotor learning. Neuron 92, 705–721 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Costa, R. M., Ganguly, K., Costa, R. M. & Carmena, J. M. Emergence of coordinated neural dynamics underlies neuroprosthetic learning and skillful control. Neuron 93, 955–970.e5 (2017).

    PubMed  Google Scholar 

  115. Kern, M., Bert, S., Glanz, O., Schulze-Bonhage, A. & Ball, T. Human motor cortex relies on sparse and action-specific activation during laughing, smiling and speech production. Commun. Biol. 2, 118 (2019).

    PubMed  PubMed Central  Google Scholar 

  116. Chalk, M., Marre, O. & Tkačik, G. Toward a unified theory of efficient, predictive, and sparse coding. Proc. Natl Acad. Sci. USA 115, 186–191 (2018).

    CAS  PubMed  Google Scholar 

  117. Tang, E. et al. Effective learning is accompanied by high-dimensional and efficient representations of neural activity. Nat. Neurosci. 22, 1000–1009 (2019).

    CAS  PubMed  Google Scholar 

  118. Simmons, P. J. & De Ruyter Van Steveninck, R. R. Sparse but specific temporal coding by spikes in an insect sensory-motor ocellar pathway. J. Exp. Biol. 213, 2629–2639 (2010).

    PubMed  Google Scholar 

  119. Clemens, J., Kutzki, O., Ronacher, B., Schreiber, S. & Wohlgemuth, S. Efficient transformation of an auditory population code in a small sensory system. Proc. Natl Acad. Sci. USA 108, 13812–13817 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Wiestler, T. & Diedrichsen, J. Skill learning strengthens cortical representations of motor sequences. eLife 2, e00801 (2013).

    PubMed  PubMed Central  Google Scholar 

  121. Vahdat, S., Darainy, M., Milner, T. E. & Ostry, D. J. Functionally specific changes in resting-state sensorimotor networks after motor learning. J. Neurosci. 31, 16907–16915 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Makino, H. et al. Transformation of cortex-wide emergent properties during motor learning. Neuron 94, 880–890.e8 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Lewis, J. E. & Maler, L. Synaptic dynamics on different time scales in a parallel fiber feedback pathway of the weakly electric fish. J. Neurophysiol. 91, 1064–1070 (2004).

    PubMed  Google Scholar 

  124. Mooney, R. Neurobiology of song learning. Curr. Opin. Neurobiol. 19, 654–660 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Pidoux, L., Leblanc, P., Levenes, C. & Leblois, A. A subcortical circuit linking the cerebellum to the basal ganglia engaged in vocal learning. eLife 7, e32167 (2018).

    PubMed  PubMed Central  Google Scholar 

  126. Ghilardi, M. F. et al. Patterns of regional brain activation associated with different forms of motor learning. Brain Res. 871, 127–145 (2000).

    CAS  PubMed  Google Scholar 

  127. Seidler, R. D. et al. Neural correlates of encoding and expression in implicit sequence learning. Exp. Brain Res. 165, 114–124 (2005).

    CAS  PubMed  Google Scholar 

  128. Ward, N. S., Brown, M. M., Thompson, A. J. & Frackowiak, R. S. J. Neural correlates of outcome after stroke: a cross-sectional fMRI study. Brain 126, 1430–1448 (2003).

    CAS  PubMed  Google Scholar 

  129. Ward, N. S., Brown, M. M., Thompson, A. J. & Frackowiak, R. S. J. The influence of time after stroke on brain activations during a motor task. Ann. Neurol. 55, 829–834 (2004).

    PubMed  PubMed Central  Google Scholar 

  130. Carey, L. M. et al. Evolution of brain activation with good and poor motor recovery after stroke. Neurorehabil. Neural Repair 20, 24–41 (2006).

    PubMed  Google Scholar 

  131. Askim, T., Indredavik, B., Vangberg, T. & Håberg, A. Motor network changes associated with successful motor skill relearning after acute ischemic stroke: a longitudinal functional magnetic resonance imaging study. Neurorehabil. Neural Repair 23, 295–304 (2009).

    PubMed  Google Scholar 

  132. Rehme, A. K., Eickhoff, S. B., Wang, L. E., Fink, G. R. & Grefkes, C. Dynamic causal modeling of cortical activity from the acute to the chronic stage after stroke. Neuroimage 55, 1147–1158 (2011).

    PubMed  Google Scholar 

  133. Kawai, R. et al. Motor cortex is required for learning but not for executing a motor skill. Neuron 86, 800–812 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Whishaw, I. Q. Loss of the innate cortical engram for action patterns used in skilled reaching and the development of behavioral compensation following motor cortex lesions in the rat. Neuropharmacology 39, 788–805 (2000). This report provides a description of compensated skilled behaviour following stroke to the rodent cortex.

    CAS  PubMed  Google Scholar 

  135. Biane, J. S., Takashima, Y., Scanziani, M., Conner, J. M. & Tuszynski, M. H. Reorganization of recurrent layer 5 corticospinal networks following adult motor training. J. Neurosci. 39, 4684–4693 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Biane, J. S., Takashima, Y., Scanziani, M., Conner, J. M. & Tuszynski, M. H. Thalamocortical projections onto behaviorally relevant neurons exhibit plasticity during adult motor learning. Neuron 89, 1173–1179 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Wang, L., Conner, J. M., Rickert, J. & Tuszynski, M. H. Structural plasticity within highly specific neuronal populations identifies a unique parcellation of motor learning in the adult brain. Proc. Natl Acad. Sci. USA 108, 2545–2550 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Wang, L., Conner, J. M., Nagahara, A. H. & Tuszynski, M. H. Rehabilitation drives enhancement of neuronal structure in functionally relevant neuronal subsets. Proc. Natl Acad. Sci. USA 113, 2750–2755 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Tonegawa, S., Liu, X., Ramirez, S. & Redondo, R. Memory engram cells have come of age. Neuron 87, 918–931 (2015).

    CAS  PubMed  Google Scholar 

  140. Josselyn, S. A., Köhler, S. & Frankland, P. W. Finding the engram. Nat. Rev. Neurosci. 16, 521–534 (2015).

    CAS  PubMed  Google Scholar 

  141. Josselyn, S. A. & Tonegawa, S. Memory engrams: recalling the past and imagining the future. Science 367, eaaw4325 (2020). This paper is a thorough review of memory engrams during normal learning.

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Zhou, Y. et al. CREB regulates excitability and the allocation of memory to subsets of neurons in the amygdala. Nat. Neurosci. 12, 1438–1443 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Gouty-Colomer, L. et al. Arc expression identifies the lateral amygdala fear memory trace. Mol. Psychiatry 21, 364–375 (2016).

    CAS  PubMed  Google Scholar 

  144. Han, J. H. et al. Neuronal competition and selection during memory formation. Science 316, 457–460 (2007).

    CAS  PubMed  Google Scholar 

  145. Yiu, A. P. et al. Neurons are recruited to a memory trace based on relative neuronal excitability immediately before training. Neuron 83, 722–735 (2014).

    CAS  PubMed  Google Scholar 

  146. Cowansage, K. K. et al. Direct reactivation of a coherent neocortical memory of context. Neuron 84, 432–441 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Goshen, I. et al. Dynamics of retrieval strategies for remote memories. Cell 147, 678–689 (2011).

    CAS  PubMed  Google Scholar 

  148. Frank, A. C. et al. Hotspots of dendritic spine turnover facilitate clustered spine addition and learning and memory. Nat. Commun. 9, 422 (2018).

    PubMed  PubMed Central  Google Scholar 

  149. Roy, D. S., Muralidhar, S., Smith, L. M. & Tonegawa, S. Silent memory engrams as the basis for retrograde amnesia. Proc. Natl Acad. Sci. USA 114, E9972–E9979 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Hayashi-Takagi, A. et al. Labelling and optical erasure of synaptic memory traces in the motor cortex. Nature 525, 333–338 (2015). This study shows that dendritic spine plasticity is an anatomical manifestation of a motor memory, the inhibition of which impairs motor memory and performance.

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Han, J. H. et al. Selective erasure of a fear memory. Science 323, 1492–1496 (2009).

    CAS  PubMed  Google Scholar 

  152. Kheirbek, M. A. et al. Differential control of learning and anxiety along the dorsoventral axis of the dentate gyrus. Neuron 77, 955–968 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Matos, M. R. et al. Memory strength gates the involvement of a CREB-dependent cortical fear engram in remote memory. Nat. Commun. 10, 2315 (2019).

    PubMed  PubMed Central  Google Scholar 

  154. Park, S. et al. Neuronal allocation to a hippocampal engram. Neuropsychopharmacology 41, 2987–2993 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Liu, X. et al. Optogenetic stimulation of a hippocampal engram activates fear memory recall. Nature 484, 381–385 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Ramirez, S. et al. Activating positive memory engrams suppresses depression-like behaviour. Nature 522, 335–339 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Tanaka, Y. H. et al. Thalamocortical axonal activity in motor cortex exhibits layer-specific dynamics during motor learning. Neuron 100, 244–258.e12 (2018).

    CAS  PubMed  Google Scholar 

  158. Sauerbrei, B. A. et al. Cortical pattern generation during dexterous movement is input-driven. Nature 577, 386–391 (2020).

    CAS  PubMed  Google Scholar 

  159. Guo, J. Z. et al. Cortex commands the performance of skilled movement. eLife 4, e10774 (2015).

    PubMed  PubMed Central  Google Scholar 

  160. Peters, A. J., Lee, J., Hedrick, N. G., O’neil, K. & Komiyama, T. Reorganization of corticospinal output during motor learning. Nat. Neurosci. 20, 1133–1141 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Omlor, W. et al. Context-dependent limb movement encoding in neuronal populations of motor cortex. Nat. Commun. 10, 4812 (2019).

    PubMed  PubMed Central  Google Scholar 

  162. Park, A. et al. A time-dependent role for the transcription factor CREB in neuronal allocation to an engram underlying a fear memory revealed using a novel in vivo optogenetic tool to modulate CREB function. Neuropsychopharmacology 45, 916–924 (2019).

    PubMed  PubMed Central  Google Scholar 

  163. Tanaka, K. Z. et al. Cortical representations are reinstated by the hippocampus during memory retrieval. Neuron 84, 347–354 (2014).

    CAS  PubMed  Google Scholar 

  164. Jones, T. A. Motor compensation and its effects on neural reorganization after stroke. Nat. Rev. Neurosci. 18, 267–280 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Kwakkel, G., Kollen, B. & Lindeman, E. Understanding the pattern of functional recovery after stroke: facts and theories. Restor. Neurol. Neurosci. 22, 281–299 (2004).

    PubMed  Google Scholar 

  166. Levin, M. F., Michaelsen, S. M., Cirstea, C. M. & Roby-Brami, A. Use of the trunk for reaching targets placed within and beyond the reach in adult hemiparesis. Exp. Brain Res. 143, 171–180 (2002).

    PubMed  Google Scholar 

  167. Shaikh, T., Goussev, V., Feldman, A. G. & Levin, M. F. Arm–trunk coordination for beyond-the-reach movements in adults with stroke. Neurorehabil. Neural Repair 28, 355–366 (2014).

    PubMed  Google Scholar 

  168. Michaelsen, S. M., Jacobs, S., Roby-Brami, A. & Levin, M. F. Compensation for distal impairments of grasping in adults with hemiparesis. Exp. Brain Res. 157, 162–173 (2004).

    PubMed  Google Scholar 

  169. Nowak, D. A. et al. Dexterity is impaired at both hands following unilateral subcortical middle cerebral artery stroke. Eur. J. Neurosci. 25, 3173–3184 (2007).

    PubMed  Google Scholar 

  170. Van Kordelaar, J., Van Wegen, E. E. H., Nijland, R. H. M., Daffertshofer, A. & Kwakkel, G. Understanding adaptive motor control of the paretic upper limb early poststroke: the EXPLICIT-stroke program. Neurorehabil. Neural Repair 27, 854–863 (2013).

    PubMed  Google Scholar 

  171. Subramanian, S. K., Yamanaka, J., Chilingaryan, G. & Levin, M. F. Validity of movement pattern kinematics as measures of arm motor impairment poststroke. Stroke 41, 2303–2308 (2010).

    PubMed  Google Scholar 

  172. Harris-Love, M. L., Morton, S. M., Perez, M. A. & Cohen, L. G. Mechanisms of short-term training-induced reaching improvement in severely hemiparetic stroke patients: a TMS study. Neurorehabil. Neural Repair 25, 398–411 (2011).

    PubMed  PubMed Central  Google Scholar 

  173. Grefkes, C. & Ward, N. S. Cortical reorganization after stroke: how much and how functional? Neuroscientist 20, 56–70 (2014).

    PubMed  Google Scholar 

  174. Sargin, D. et al. CREB regulates spine density of lateral amygdala neurons: implications for memory allocation. Front. Behav. Neurosci. 7, 209 (2013).

    PubMed  PubMed Central  Google Scholar 

  175. Fu, M., Yu, X., Lu, J. & Zuo, Y. Repetitive motor learning induces coordinated formation of clustered dendritic spines in vivo. Nature 483, 92–96 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Roy, D. S. et al. Memory retrieval by activating engram cells in mouse models of early Alzheimer’s disease. Nature 531, 508–512 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Clark, T. A. et al. Rehabilitative training interacts with ischemia-instigated spine dynamics to promote a lasting population of new synapses in peri-infarct motor cortex. J. Neurosci. 39, 8471–8483 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Cramer, S. C. Repairing the human brain after stroke: I. Mechanisms of spontaneous recovery. Ann. Neurol. 63, 272–287 (2008).

    PubMed  Google Scholar 

  179. Goel, P. & Dickman, D. Distinct homeostatic modulations stabilize reduced postsynaptic receptivity in response to presynaptic DLK signaling. Nat. Commun. 9, 1856 (2018).

    PubMed  PubMed Central  Google Scholar 

  180. Rashid, A. J. et al. Competition between engrams influences fear memory formation and recall. Science 353, 383–387 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  181. Sun, X. et al. Functionally distinct neuronal ensembles within the memory engram. Cell 181, 410–423.e17 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Zeiler, S. R. et al. Medial premotor cortex shows a reduction in inhibitory markers and mediates recovery in a mouse model of focal stroke. Stroke 44, 483–489 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Chollet, F. et al. Fluoxetine for motor recovery after acute ischaemic stroke (FLAME): a randomised placebo-controlled trial. Lancet Neurol. 10, 123–130 (2011).

    CAS  PubMed  Google Scholar 

  184. MacArthur, R. D. & Novak, R. M. Maraviroc: the first of a new class of antiretroviral agents. Clin. Infect. Dis. 47, 236–241 (2008).

    CAS  PubMed  Google Scholar 

  185. Samson, M. et al. Resistance to HIV-1 infection in caucasian individuals bearing mutant alleles of the CCR-5 chemokine receptor gene. Nature 382, 722–725 (1996).

    CAS  PubMed  Google Scholar 

  186. Kuritzkes, D., Kar, S. & Kirkpatrick, P. Maraviroc. Nat. Rev. Drug Discov. 7, 15–16 (2008).

    CAS  Google Scholar 

  187. US National Library of Medicine. CinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03172026 (2019).

  188. US National Library of Medicine. CinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02928393 (2019).

  189. US National Library of Medicine. CinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02877615 (2020).

  190. Prickaerts, J., Heckman, P. R. A. & Blokland, A. Investigational phosphodiesterase inhibitors in phase I and phase II clinical trials for Alzheimer’s disease. Expert Opin. Investig. Drugs 26, 1033–1048 (2017).

    CAS  PubMed  Google Scholar 

  191. US National Library of Medicine. CinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02013310 (2015).

  192. Yurgelun-Todd, D. A., Renshaw, P. F., Goldsmith, P., Uz, T. & Macek, T. A. A randomized, placebo-controlled, phase 1 study to evaluate the effects of TAK-063 on ketamine-induced changes in fMRI BOLD signal in healthy subjects. Psychopharmacology 237, 317–328 (2020).

    CAS  PubMed  Google Scholar 

  193. Ng, K. L. et al. Fluoxetine maintains a state of heightened responsiveness to motor training early after stroke in a mouse model. Stroke 46, 2951–2960 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. FOCUS Trial Collaboration. Effects of fluoxetine on functional outcomes after acute stroke (FOCUS): a pragmatic, double-blind, randomised, controlled trial. Lancet 393, 265–274 (2019).

    Google Scholar 

  195. Dobkin, B. H. & Carmichael, S. T. The specific requirements of neural repair trials for stroke. Neurorehabil. Neural Repair 30, 470–478 (2016). This report presents discussions on the design of neural repair trials in stroke research.

    PubMed  Google Scholar 

  196. Savitz, S. I. & Fisher, M. Future of neuroprotection for acute stroke: in the aftermath of the SAINT trials. Ann. Neurol. 61, 396–402 (2007).

    CAS  PubMed  Google Scholar 

  197. O’Collins, V. E. et al. 1,026 experimental treatments in acute stroke. Ann. Neurol. 59, 467–477 (2006).

    PubMed  Google Scholar 

  198. de Boer, A. et al. Heterozygous deletion of ephrinA5 does not improve functional recovery after experimental stroke. Stroke 50, e101 (2019).

    PubMed  Google Scholar 

  199. Kempf, A. et al. Upregulation of axon guidance molecules in the adult central nervous system of Nogo-A knockout mice restricts neuronal growth and regeneration. Eur. J. Neurosci. 38, 3567–3579 (2013).

    PubMed  Google Scholar 

  200. Lee, J. K., Kim, J. E., Sivula, M. & Strittmatter, S. M. Nogo receptor antagonism promotes stroke recovery by enhancing axonal plasticity. J. Neurosci. 24, 6209–6217 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  201. Kilic, E. et al. Role of Nogo-A in neuronal survival in the reperfused ischemic brain. J. Cereb. Blood Flow Metab. 30, 969–984 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  202. Dyer, D. P. et al. Chemokine receptor redundancy and specificity are context dependent. Immunity 50, 378–389.e5 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Sorce, S. et al. Increased brain damage after ischaemic stroke in mice lacking the chemokine receptor CCR5. Br. J. Pharmacol. 160, 311–321 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  204. Victoria, E. C. G. et al. Knockdown of C-C chemokine receptor 5 (CCR5) is protective against cerebral ischemia and reperfusion injury. Curr. Neurovasc. Res. 14, 125–131 (2017).

    PubMed  Google Scholar 

  205. Giorgio, C. et al. Inhibition of Eph/ephrin interaction with the small molecule UniPR500 improves glucose tolerance in healthy and insulin-resistant mice. Pharmacol. Res. 141, 319–330 (2019).

    CAS  PubMed  Google Scholar 

  206. Broderick, J. P., Adeoye, O. & Elm, J. Evolution of the modified Rankin Scale and its use in future stroke trials. Stroke 48, 2007–2012 (2017).

    PubMed  PubMed Central  Google Scholar 

  207. Quinn, T. J., Langhorne, P. & Stott, D. J. Barthel Index for stroke trials: development, properties, and application. Stroke 42, 1146–1151 (2011).

    PubMed  Google Scholar 

  208. Steinberg, G. K. et al. Clinical outcomes of transplanted modified bone marrow-derived mesenchymal stem cells in stroke: a phase 1/2a study. Stroke 47, 1817–1824 (2016).

    PubMed  PubMed Central  Google Scholar 

  209. Wu, W. X. et al. Effect of early and intensive rehabilitation after ischemic stroke on functional recovery of the lower limbs: a pilot, randomized trial. J. Stroke Cerebrovasc. Dis. 29, 104649 (2020).

    PubMed  Google Scholar 

  210. Mayo, N. E., Wood-Dauphinee, S., Côté, R., Durcan, L. & Carlton, J. Activity, participation, and quality of life 6 months poststroke. Arch. Phys. Med. Rehabil. 83, 1035–1042 (2002).

    PubMed  Google Scholar 

  211. Stewart, J. C. & Cramer, S. C. Patient-reported measures provide unique insights into motor function after stroke. Stroke 44, 1111–1116 (2013).

    PubMed  PubMed Central  Google Scholar 

  212. Chabriat, H. et al. RESTORE BRAIN study investigators. Safety and efficacy of GABAA α5 antagonist S44819 in patients with ischaemic stroke: a multicentre, double-blind, randomised, placebo-controlled trial. Lancet Neurol. 19, 226–233 (2020).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank the Dr. Miriam and Sheldon G. Adelson Medical Research Foundation, the American Heart Association and the National Institutes of Health (NIH; grant number NS085019) for financial support.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to S. Thomas Carmichael.

Ethics declarations

Competing interests

S.T.C. has received research grant funding from Takeda Phamaceuticals. M.T.J. declares no competing interests.

Additional information

Peer review information

Nature Reviews Neuroscience thanks D. Hermann, S, Josselyn and T. Murphy for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Stroke

A disease caused by the blockade or rupture of a blood vessel to the brain. Loss of blood flow causes death of brain tissue, resulting in long-term neurological impairments.

Plasticity

Structural or functional changes (or both) within neurons that affect the connectivity of neurons with each other in a network serving a function. These changes are usually in response to a change in neuronal input or firing patterns induced by this input, such as during learning or after injury.

Neuronal circuits

Neurons from either the same brain region or different regions that are connected to each other via synapses to form networks that subserve different brain functions.

Neuronal excitability

Changes in the electrical properties of a neuron in response to a stimulus.

Neuronal allocation

The property by which neurons are selected to integrate into a circuit that stores information on a particular stimulus.

Motor recovery

The return of a motor function where the initial motor patterns prior to injury or a stroke are regained.

Peri-infarct

Brain tissue that borders the infarct.

Infarct

The site of tissue loss in the brain from a stroke.

Dendritic spines

Sub-micrometre protrusions on dendrites of a neuron that receive synaptic input as postsynaptic elements in the synaptic connection.

Axonal sprouting

The growth and extension of a new axon and its connections. This may occur from an injured or damaged axon, or through collateralization or extension from an existing axon. The ability to grow new axons is nearly absent in the adult brain except after injury.

Critical period

A time window during development or after a stroke marked by significant changes in neuronal growth and connectivity shaped by sensory, motor and cognitive experiences.

Engram

A collection of neurons that fire together in response to a stimulus with higher connectivity with each other and store the memory for that stimulus.

Compensation

The return of a motor function using new movement patterns that allow complete or partial execution of a task, often at the expense of higher energy demands and lower movement quality.

Map reorganization

Maps are gross anatomical and functional readouts from the activity of a population of neurons that underlie or respond to movement or sensation, and they can reorganize to take a different spatial location or expand in original territory during learning or in response to injury.

Clinical translation

The transfer of an experimental paradigm identified in an animal or laboratory-based model of disease to a clinical setting in which patients are treated based on these preclinical experimental results.

Neural repair therapy

A therapeutic strategy that targets genes or signalling molecules underlying plasticity such that stroke recovery can be induced through increased connectivity between relevant brain regions to execute lost function. Neural repair therapies do not minimize cell death that occurs in the initial phases of stroke, but rather utilize and enhance inherent plasticity present in the later stages of stroke by manipulating specific genes, allowing for more effective therapies and greater treatment accessibility to patients who do not qualify for clot removal therapies.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Joy, M.T., Carmichael, S.T. Encouraging an excitable brain state: mechanisms of brain repair in stroke. Nat Rev Neurosci 22, 38–53 (2021). https://doi.org/10.1038/s41583-020-00396-7

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41583-020-00396-7

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing