Semin Neurol 2021; 41(02): 147-156
DOI: 10.1055/s-0041-1725136
Review Article

Learning and Stroke Recovery: Parallelism of Biological Substrates

Mary Teena Joy
1   Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California
,
1   Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California
› Institutsangaben
Funding Supported by the Dr. Miriam and Sheldon G Adelson Medical Research Foundation, NIIH grants NS102185 and NS085019.

Abstract

Stroke is a debilitating disease. Current effective therapies for stroke recovery are limited to neurorehabilitation. Most stroke recovery occurs in a limited and early time window. Many of the mechanisms of spontaneous recovery after stroke parallel mechanisms of normal learning and memory. While various efforts are in place to identify potential drug targets, an emerging approach is to understand biological correlates between learning and stroke recovery. This review assesses parallels between biological changes at the molecular, structural, and functional levels during learning and recovery after stroke, with a focus on drug and cellular targets for therapeutics.



Publikationsverlauf

Artikel online veröffentlicht:
09. März 2021

© 2021. Thieme. All rights reserved.

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

 
  • References

  • 1 Benjamin EJ, Muntner P, Alonso A. et al; American Heart Association Council on Epidemiology and Prevention Statistics Committee and Stroke Statistics Subcommittee. Heart Disease and Stroke Statistics-2019 Update: a report from the American Heart Association. Circulation 2019; 139 (10) e56-e528
  • 2 Lang CE, Lohse KR, Birkenmeier RL. Dose and timing in neurorehabilitation: prescribing motor therapy after stroke. Curr Opin Neurol 2015; 28 (06) 549-555
  • 3 Billinger SA, Arena R, Bernhardt J. et al; American Heart Association Stroke Council, Council on Cardiovascular and Stroke Nursing, Council on Lifestyle and Cardiometabolic Health, Council on Epidemiology and Prevention, Council on Clinical Cardiology. Physical activity and exercise recommendations for stroke survivors: a statement for healthcare professionals from the American Heart Association/American Stroke Association. Stroke 2014; 45 (08) 2532-2553
  • 4 Harris JE, Eng JJ. Strength training improves upper-limb function in individuals with stroke: a meta-analysis. Stroke 2010; 41 (01) 136-140
  • 5 Kwakkel G, Veerbeek JM, van Wegen EE, Wolf SL. Constraint-induced movement therapy after stroke. Lancet Neurol 2015; 14 (02) 224-234
  • 6 Duncan PW, Sullivan KJ, Behrman AL. et al; LEAPS Investigative Team. Body-weight-supported treadmill rehabilitation after stroke. N Engl J Med 2011; 364 (21) 2026-2036
  • 7 Laver KE, Lange B, George S, Deutsch JE, Saposnik G, Crotty M. Virtual reality for stroke rehabilitation. Cochrane Database Syst Rev 2017; 11: CD008349
  • 8 Maier M, Rubio Ballester B, Duff A, Duarte Oller E, Verschure PFMJ. Effect of specific over nonspecific VR-based rehabilitation on poststroke motor recovery: a systematic meta-analysis. Neurorehabil Neural Repair 2019; 33 (02) 112-129
  • 9 Takesian AE, Bogart LJ, Lichtman JW, Hensch TK. Inhibitory circuit gating of auditory critical-period plasticity. Nat Neurosci 2018; 21 (02) 218-227
  • 10 Hartshorne JK, Tenenbaum JB, Pinker S. A critical period for second language acquisition: evidence from 2/3 million English speakers. Cognition 2018; 177: 263-277
  • 11 Clarkson AN, Overman JJ, Zhong S, Mueller R, Lynch G, Carmichael ST. AMPA receptor-induced local brain-derived neurotrophic factor signaling mediates motor recovery after stroke. J Neurosci 2011; 31 (10) 3766-3775
  • 12 Dromerick AW, Edwardson MA, Edwards DF. et al. Critical periods after stroke study: translating animal stroke recovery experiments into a clinical trial. Front Hum Neurosci 2015; 9: 231
  • 13 Humm JL, Kozlowski DA, James DC, Gotts JE, Schallert T. Use-dependent exacerbation of brain damage occurs during an early post-lesion vulnerable period. Brain Res 1998; 783 (02) 286-292
  • 14 Caracciolo L, Marosi M, Mazzitelli J. et al. CREB controls cortical circuit plasticity and functional recovery after stroke. Nat Commun 2018; 9 (01) 2250
  • 15 Joy MT, Ben Assayag E, Shabashov-Stone D. et al. CCR5 is a therapeutic target for recovery after stroke and traumatic brain injury. Cell 2019; 176 (05) 1143-1157.e13
  • 16 Brown TG, Sherrington CS. On the instability of a cortical point. Proc R Soc Lond 1912; 85: 250-277
  • 17 Globus A, Rosenzweig MR, Bennett EL, Diamond MC. Effects of differential experience on dendritic spine counts in rat cerebral cortex. J Comp Physiol Psychol 1973; 82 (02) 175-181
  • 18 Greenough WT, Larson JR, Withers GS. Effects of unilateral and bilateral training in a reaching task on dendritic branching of neurons in the rat motor-sensory forelimb cortex. Behav Neural Biol 1985; 44 (02) 301-314
  • 19 Fu M, Yu X, Lu J, Zuo Y. Repetitive motor learning induces coordinated formation of clustered dendritic spines in vivo. Nature 2012; 483 (7387): 92-95
  • 20 Xu T, Yu X, Perlik AJ. et al. Rapid formation and selective stabilization of synapses for enduring motor memories. Nature 2009; 462 (7275): 915-919
  • 21 Yang G, Pan F, Gan WB. Stably maintained dendritic spines are associated with lifelong memories. Nature 2009; 462 (7275): 920-924
  • 22 Hayashi-Takagi A, Yagishita S, Nakamura M. et al. Labelling and optical erasure of synaptic memory traces in the motor cortex. Nature 2015; 525 (7569): 333-338
  • 23 Clark TA, Fu M, Dunn AK, Zuo Y, Jones TA. Preferential stabilization of newly formed dendritic spines in motor cortex during manual skill learning predicts performance gains, but not memory endurance. Neurobiol Learn Mem 2018; 152: 50-60
  • 24 Brown CE, Li P, Boyd JD, Delaney KR, Murphy TH. Extensive turnover of dendritic spines and vascular remodeling in cortical tissues recovering from stroke. J Neurosci 2007; 27 (15) 4101-4109
  • 25 Mostany R, Chowdhury TG, Johnston DG, Portonovo SA, Carmichael ST, Portera-Cailliau C. Local hemodynamics dictate long-term dendritic plasticity in peri-infarct cortex. J Neurosci 2010; 30 (42) 14116-14126
  • 26 Takatsuru Y, Fukumoto D, Yoshitomo M, Nemoto T, Tsukada H, Nabekura J. Neuronal circuit remodeling in the contralateral cortical hemisphere during functional recovery from cerebral infarction. J Neurosci 2009; 29 (32) 10081-10086
  • 27 Clark TA, Sullender C, Jacob D, Zuo Y, Dunn AK, Jones TA. Rehabilitative training interacts with ischemia-instigated spine dynamics to promote a lasting population of new synapses in peri-infarct motor cortex. J Neurosci 2019; 39 (43) 8471-8483
  • 28 Wang L, Conner JM, Nagahara AH, Tuszynski MH. Rehabilitation drives enhancement of neuronal structure in functionally relevant neuronal subsets. Proc Natl Acad Sci U S A 2016; 113 (10) 2750-2755
  • 29 Cheng MY, Wang EH, Woodson WJ. et al. Optogenetic neuronal stimulation promotes functional recovery after stroke. Proc Natl Acad Sci U S A 2014; 111 (35) 12913-12918
  • 30 Tennant KA, Taylor SL, White ER, Brown CE. Optogenetic rewiring of thalamocortical circuits to restore function in the stroke injured brain. Nat Commun 2017; 8: 15879
  • 31 Carmichael ST. Brain excitability in stroke: the yin and yang of stroke progression. Arch Neurol 2012; 69 (02) 161-167
  • 32 Li S, Carmichael ST. Growth-associated gene and protein expression in the region of axonal sprouting in the aged brain after stroke. Neurobiol Dis 2006; 23 (02) 362-373
  • 33 Jones TA. Multiple synapse formation in the motor cortex opposite unilateral sensorimotor cortex lesions in adult rats. J Comp Neurol 1999; 414 (01) 57-66
  • 34 Brown CE, Aminoltejari K, Erb H, Winship IR, Murphy TH. 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 2009; 29 (06) 1719-1734
  • 35 Tonegawa S, Morrissey MD, Kitamura T. The role of engram cells in the systems consolidation of memory. Nat Rev Neurosci 2018; 19 (08) 485-498
  • 36 Chittajallu R, Isaac JT. Emergence of cortical inhibition by coordinated sensory-driven plasticity at distinct synaptic loci. Nat Neurosci 2010; 13 (10) 1240-1248
  • 37 Diering GH, Huganir RL. The AMPA receptor code of synaptic plasticity. Neuron 2018; 100 (02) 314-329
  • 38 Yoon JH, Grandelis A, Maddock RJ. Dorsolateral prefrontal cortex GABA concentration in humans predicts working memory load processing capacity. J Neurosci 2016; 36 (46) 11788-11794
  • 39 Collinson N, Kuenzi FM, Jarolimek W. et al. Enhanced learning and memory and altered GABAergic synaptic transmission in mice lacking the alpha 5 subunit of the GABAA receptor. J Neurosci 2002; 22 (13) 5572-5580
  • 40 Clarkson AN, Huang BS, Macisaac SE, Mody I, Carmichael ST. Reducing excessive GABA-mediated tonic inhibition promotes functional recovery after stroke. Nature 2010; 468 (7321): 305-309
  • 41 Feng G, Laskowski MB, Feldheim DA. et al. Roles for ephrins in positionally selective synaptogenesis between motor neurons and muscle fibers. Neuron 2000; 25 (02) 295-306
  • 42 Giger RJ, Hollis II ER, Tuszynski MH. Guidance molecules in axon regeneration. Cold Spring Harb Perspect Biol 2010; 2 (07) a001867
  • 43 Shen K, Cowan CW. Guidance molecules in synapse formation and plasticity. Cold Spring Harb Perspect Biol 2010; 2 (04) a001842
  • 44 Filosa A, Paixão S, Honsek SD. et al. Neuron-glia communication via EphA4/ephrin-A3 modulates LTP through glial glutamate transport. Nat Neurosci 2009; 12 (10) 1285-1292
  • 45 Overman JJ, Clarkson AN, Wanner IB. et al. A role for ephrin-A5 in axonal sprouting, recovery, and activity-dependent plasticity after stroke. Proc Natl Acad Sci U S A 2012; 109 (33) E2230-E2239
  • 46 Overman JJ, Carmichael ST. Plasticity in the injured brain: more than molecules matter. Neuroscientist 2014; 20 (01) 15-28
  • 47 de Boer A, Storm A, Rué L, Poppe L, Robberecht W, Lemmens R. Heterozygous deletion of ephrinA5 does not improve functional recovery after experimental stroke. Stroke 2019; 50 (04) e101
  • 48 El-Brolosy MA, Stainier DYR. Genetic compensation: a phenomenon in search of mechanisms. PLoS Genet 2017; 13 (07) e1006780
  • 49 Tsokas P, Hsieh C, Yao Y. et al. Compensation for PKMζ in long-term potentiation and spatial long-term memory in mutant mice. eLife 2016; 5: e14846
  • 50 Kempf A, Montani L, Petrinovic MM. 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 2013; 38 (11) 3567-3579
  • 51 Cramer SC. Issues important to the design of stroke recovery trials. Lancet Neurol 2020; 19 (03) 197-198
  • 52 Dobkin BH, Carmichael ST. The specific requirements of neural repair trials for stroke. Neurorehabil Neural Repair 2016; 30 (05) 470-478
  • 53 Hicks A, Schallert T, Jolkkonen J. Cell-based therapies and functional outcome in experimental stroke. Cell Stem Cell 2009; 5 (02) 139-140
  • 54 Piao J, Major T, Auyeung G. et al. Human embryonic stem cell-derived oligodendrocyte progenitors remyelinate the brain and rescue behavioral deficits following radiation. Cell Stem Cell 2015; 16 (02) 198-210
  • 55 Gladstone DJ, Danells CJ, Black SE. The Fugl-Meyer assessment of motor recovery after stroke: a critical review of its measurement properties. Neurorehabil Neural Repair 2002; 16 (03) 232-240
  • 56 Lang CE, Edwards DF, Birkenmeier RL, Dromerick AW. Estimating minimal clinically important differences of upper-extremity measures early after stroke. Arch Phys Med Rehabil 2008; 89 (09) 1693-1700
  • 57 Kim DY, Quinlan EB, Gramer R, Cramer SC. BDNF Val66Met polymorphism is related to motor system function after stroke. Phys Ther 2016; 96 (04) 533-539
  • 58 Kraft AW, Bauer AQ, Culver JP, Lee JM. Sensory deprivation after focal ischemia in mice accelerates brain remapping and improves functional recovery through Arc-dependent synaptic plasticity. Sci Transl Med 2018; 10 (426) 10
  • 59 Lindau NT, Bänninger BJ, Gullo M. et al. Rewiring of the corticospinal tract in the adult rat after unilateral stroke and anti-Nogo-A therapy. Brain 2014; 137 (Pt 3): 739-756
  • 60 Kandel ER. The molecular biology of memory: cAMP, PKA, CRE, CREB-1, CREB-2, and CPEB. Mol Brain 2012; 5: 14
  • 61 Silva AJ, Kogan JH, Frankland PW, Kida S. CREB and memory. Annu Rev Neurosci 1998; 21: 127-148
  • 62 Zhou Y, Won J, Karlsson MG. et al. CREB regulates excitability and the allocation of memory to subsets of neurons in the amygdala. Nat Neurosci 2009; 12 (11) 1438-1443
  • 63 Yiu AP, Mercaldo V, Yan C. et al. Neurons are recruited to a memory trace based on relative neuronal excitability immediately before training. Neuron 2014; 83 (03) 722-735
  • 64 Middei S, Spalloni A, Longone P. et al. CREB selectively controls learning-induced structural remodeling of neurons. Learn Mem 2012; 19 (08) 330-336
  • 65 Gao Y, Deng K, Hou J. et al. Activated CREB is sufficient to overcome inhibitors in myelin and promote spinal axon regeneration in vivo. Neuron 2004; 44 (04) 609-621
  • 66 Deng K, He H, Qiu J, Lorber B, Bryson JB, Filbin MT. Increased synthesis of spermidine as a result of upregulation of arginase I promotes axonal regeneration in culture and in vivo. J Neurosci 2009; 29 (30) 9545-9552
  • 67 Roth BL. DREADDs for neuroscientists. Neuron 2016; 89 (04) 683-694
  • 68 Graham V, Khudyakov J, Ellis P, Pevny L. SOX2 functions to maintain neural progenitor identity. Neuron 2003; 39 (05) 749-765
  • 69 Dayer AG, Jenny B, Sauvain MO. et al. Expression of FGF-2 in neural progenitor cells enhances their potential for cellular brain repair in the rodent cortex. Brain 2007; 130 (Pt 11): 2962-2976
  • 70 Yoshimura S, Teramoto T, Whalen MJ. et al. FGF-2 regulates neurogenesis and degeneration in the dentate gyrus after traumatic brain injury in mice. J Clin Invest 2003; 112 (08) 1202-1210
  • 71 Lee TT, Green BA, Dietrich WD, Yezierski RP. Neuroprotective effects of basic fibroblast growth factor following spinal cord contusion injury in the rat. J Neurotrauma 1999; 16 (05) 347-356
  • 72 Rabchevsky AG, Fugaccia I, Fletcher-Turner A, Blades DA, Mattson MP, Scheff SW. Basic fibroblast growth factor (bFGF) enhances tissue sparing and functional recovery following moderate spinal cord injury. J Neurotrauma 1999; 16 (09) 817-830
  • 73 Liang H, Zhao H, Gleichman A. et al. Region-specific and activity-dependent regulation of SVZ neurogenesis and recovery after stroke. Proc Natl Acad Sci U S A 2019; 116 (27) 13621-13630
  • 74 Martin-Blondel G, Brassat D, Bauer J, Lassmann H, Liblau RS. CCR5 blockade for neuroinflammatory diseases--beyond control of HIV. Nat Rev Neurol 2016; 12 (02) 95-105
  • 75 Zhou M, Greenhill S, Huang S. et al. CCR5 is a suppressor for cortical plasticity and hippocampal learning and memory. eLife 2016; 5: 5
  • 76 Guyon A. CXCL12 chemokine and its receptors as major players in the interactions between immune and nervous systems. Front Cell Neurosci 2014; 8: 65
  • 77 Lorenzen E, Ceraudo E, Berchiche YA. et al. G protein subtype-specific signaling bias in a series of CCR5 chemokine analogs. Sci Signal 2018; 11 (552) 11
  • 78 Hannila SS, Filbin MT. The role of cyclic AMP signaling in promoting axonal regeneration after spinal cord injury. Exp Neurol 2008; 209 (02) 321-332
  • 79 Galasso JM, Harrison JK, Silverstein FS. Excitotoxic brain injury stimulates expression of the chemokine receptor CCR5 in neonatal rats. Am J Pathol 1998; 153 (05) 1631-1640
  • 80 Watkins TA, Wang B, Huntwork-Rodriguez S. et al. DLK initiates a transcriptional program that couples apoptotic and regenerative responses to axonal injury. Proc Natl Acad Sci U S A 2013; 110 (10) 4039-4044
  • 81 Biane JS, Takashima Y, Scanziani M, Conner JM, Tuszynski MH. Thalamocortical projections onto behaviorally relevant neurons exhibit plasticity during adult motor learning. Neuron 2016; 89 (06) 1173-1179
  • 82 Boele HJ, Koekkoek SK, De Zeeuw CI, Ruigrok TJ. Axonal sprouting and formation of terminals in the adult cerebellum during associative motor learning. J Neurosci 2013; 33 (45) 17897-17907
  • 83 Hihara S, Notoya T, Tanaka M. et al. Extension of corticocortical afferents into the anterior bank of the intraparietal sulcus by tool-use training in adult monkeys. Neuropsychologia 2006; 44 (13) 2636-2646
  • 84 Zatorre RJ, Fields RD, Johansen-Berg H. Plasticity in gray and white: neuroimaging changes in brain structure during learning. Nat Neurosci 2012; 15 (04) 528-536
  • 85 Dayan E, Cohen LG. Neuroplasticity subserving motor skill learning. Neuron 2011; 72 (03) 443-454
  • 86 Carmichael ST, Wei L, Rovainen CM, Woolsey TA. New patterns of intracortical projections after focal cortical stroke. Neurobiol Dis 2001; 8 (05) 910-922
  • 87 Li S, Nie EH, Yin Y. et al. GDF10 is a signal for axonal sprouting and functional recovery after stroke. Nat Neurosci 2015; 18 (12) 1737-1745
  • 88 Morecraft RJ, Ge J, Stilwell-Morecraft KS. et al. Frontal and frontoparietal injury differentially affect the ipsilateral corticospinal projection from the nonlesioned hemisphere in monkey (Macaca mulatta). J Comp Neurol 2016; 524 (02) 380-407
  • 89 Wahl AS, Omlor W, Rubio JC. et al. Neuronal repair. Asynchronous therapy restores motor control by rewiring of the rat corticospinal tract after stroke. Science 2014; 344 (6189): 1250-1255
  • 90 Hamadjida A, Wyss AF, Mir A, Schwab ME, Belhaj-Saif A, Rouiller EM. Influence of anti-Nogo-A antibody treatment on the reorganization of callosal connectivity of the premotor cortical areas following unilateral lesion of primary motor cortex (M1) in adult macaque monkeys. Exp Brain Res 2012; 223 (03) 321-340
  • 91 Zai L, Ferrari C, Subbaiah S. et al. Inosine alters gene expression and axonal projections in neurons contralateral to a cortical infarct and improves skilled use of the impaired limb. J Neurosci 2009; 29 (25) 8187-8197
  • 92 Tennant KA, Adkins DL, Donlan NA. et al. The organization of the forelimb representation of the C57BL/6 mouse motor cortex as defined by intracortical microstimulation and cytoarchitecture. Cereb Cortex 2011; 21 (04) 865-876
  • 93 Tennant KA, Kerr AL, Adkins DL. et al. Age-dependent reorganization of peri-infarct “premotor” cortex with task-specific rehabilitative training in mice. Neurorehabil Neural Repair 2015; 29 (02) 193-202
  • 94 Harrison TC, Silasi G, Boyd JD, Murphy TH. Displacement of sensory maps and disorganization of motor cortex after targeted stroke in mice. Stroke 2013; 44 (08) 2300-2306
  • 95 Sawaki L, Butler AJ, Leng X. et al. Constraint-induced movement therapy results in increased motor map area in subjects 3 to 9 months after stroke. Neurorehabil Neural Repair 2008; 22 (05) 505-513
  • 96 Kleim JA, Barbay S, Nudo RJ. Functional reorganization of the rat motor cortex following motor skill learning. J Neurophysiol 1998; 80 (06) 3321-3325
  • 97 Nudo RJ, Milliken GW, Jenkins WM, Merzenich MM. Use-dependent alterations of movement representations in primary motor cortex of adult squirrel monkeys. J Neurosci 1996; 16 (02) 785-807
  • 98 Reed A, Riley J, Carraway R. et al. Cortical map plasticity improves learning but is not necessary for improved performance. Neuron 2011; 70 (01) 121-131