Cellular and molecular mechanisms of action of transcranial direct current stimulation: evidence from in vitro and in vivo models

Simon J Pelletier, Francesca Cicchetti, Simon J Pelletier, Francesca Cicchetti

Abstract

Transcranial direct current stimulation is a noninvasive technique that has been experimentally tested for a number of psychiatric and neurological conditions. Preliminary observations suggest that this approach can indeed influence a number of cellular and molecular pathways that may be disease relevant. However, the mechanisms of action underlying its beneficial effects are largely unknown and need to be better understood to allow this therapy to be used optimally. In this review, we summarize the physiological responses observed in vitro and in vivo, with a particular emphasis on cellular and molecular cascades associated with inflammation, angiogenesis, neurogenesis, and neuroplasticity recruited by direct current stimulation, a topic that has been largely neglected in the literature. A better understanding of the neural responses to transcranial direct current stimulation is critical if this therapy is to be used in large-scale clinical trials with a view of being routinely offered to patients suffering from various conditions affecting the central nervous system.

Keywords: inflammation; long-term potentiation; neurogenesis.

© The Author 2015. Published by Oxford University Press on behalf of CINP.

Figures

Figure 1.
Figure 1.
Putative molecular mechanisms of action of anodal transcranial direct current stimulation (tDCS). Schematic illustrating the effects of anodal tDCS on the synapses of pyramidal neurons in the primary motor cortex. Note that cathodal tDCS is not represented, as it largely generates the opposite effects of anodal tDCS, except for the mechanisms involving brain-derived neurotrophic factor (BDNF), which is described below. Anodal tDCS hyperpolarizes the membrane of the axon terminal facing the anode (Bikson et al., 2004). Despite the hyperpolarization, there is greater neurotransmitter release, which is caused by an increase in intracellular Ca2+ in response to anodal tDCS, whereas a decrease of Ca2+ leads to lower neurotransmitter release (Perret et al., 1999; Stagg et al., 2009a). Tropomyosin-receptor kinase (Trk) receptors may also be attracted to the synapse in anodal tDCS (if the presynaptic synapse faces the skull, as illustrated here) (McCaig et al., 2000; Viard et al., 2004). The activation of Trk receptors suggests a role for BDNF in anodal tDCS, which further increases the probability of synaptic vesicle docking and neurotransmitter release (Pozzo-Miller et al., 1999). Direct current electric fields (DCEFs) also directly affect the postsynaptic neuron by depolarizing (basal dendrites and soma, represented in the figure) or hyperpolarizing (apical dendrites) the membrane in anodal tDCS, or the opposite with cathodal tDCS (Kabakov et al., 2012), which further facilitates/inhibits α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR)- and N-methyl-d-aspartate receptor (NMDAR)-mediated ionic changes. Overall, with long-term potentiation (LTP) responses, there is an upregulation of neurotransmitter release that facilitates the opening of AMPARs and indirectly that of NMDARs (Derkach et al., 2007). The opposite is true for long-term depression (LTD). The Ca2+ influx has been demonstrated to increase AMPAR phosphorylation and their incorporation into the membrane (Opazo et al., 2010). Ca2+ further increases the release of neurotrophic factors into the synaptic cleft and its absence decreases it (Neal and Guilarte, 2010). Once activated, postsynaptic Trk receptor induces later phase LTP (L-LTP) and favors the opening of NMDARs, which also promotes L-LTP, whereas the opposite is involved in cathodal tDCS, promoting later phase LTD (L-LTD) (Minichiello, 2009). Both L-LTP and L-LTD are dependent on modifications of gene expression (Frey et al., 1996; Smolen, 2007). PSD, postsynaptic domain; Cav; voltage-gated calcium channel.
Figure 2.
Figure 2.
Cell types and their responses to transcranial direct current stimulation (tDCS).
Figure 3.
Figure 3.
Putative cellular mechanisms of action of anodal transcranial direct current stimulation (tDCS). Anodal tDCS has been demonstrated to increase the amplitude and reduce the timing of glutamatergic neuronal firing (the opposite effect is observed for cathodal stimulation) (Cambiaghi et al., 2010; Hunter et al., 2013). In contrast, the firing of interneurons is decreased in both anodal and cathodal tDCS in healthy human subjects, as suggested by decreased γ-aminobutyric acid (GABA) release (Stagg et al., 2009a). Glutamate is also reduced by cathodal tDCS in healthy human subjects, whereas dopamine has been reported to be increased in normal rats with such therapy (Stagg et al., 2009a; Tanaka et al., 2013). Increases in growth associated protein-43 (GAP-43), a protein synthesized during axonal growth, and microtubule-associated protein-2 (MAP-2), involved in dendritic remodeling along with an increase in dendritic density, has been reported in stimulated brain structures in rats with ischemic lesions, which further suggests that anodal tDCS may have neuroprotective as well as neurorestorative properties (Yoon et al., 2012). There are, however, no data on their modulation by cathodal tDCS. In ischemic mice, cathodal tDCS also decreases ionized calcium-binding apater molecule-1 (Iba1+), CD45+, and caspase-3+ cell numbers, whereas the opposite is seen with anodal tDCS (Peruzzotti-Jametti et al., 2013). In a rat model of chronic stress-induced pain, tumor necrosis factor-α (TNF-α) is also downregulated by anodal tDCS, but an increase in the number of Iba1+ cells is observed in both anodal and cathodal tDCS at high intensity in normal rats (Spezia Adachi et al., 2012). Angiogenesis and increases in vascular endothelial growth factor (VEGF) levels have been reported in peripheral tissues exposed to DCEF (Bai et al., 2011). Finally, anodal tDCS in ischemic mice may also exacerbate cortical hemorrhage and provoke the disruption of the blood-brain barrier (BBB; Peruzzotti-Jametti et al., 2013). Taken together, all of this suggests that tDCS affects a number of physiological processes in both the central and peripheral nervous systems that may be relevant to its effects in disease states. AP, action potential; BDNF, brain-derived neurotrophic factor; DA, dopamine; Glu, glutamate.

References

    1. Alexander JK, Fuss B, Colello RJ. (2006). Electric field-induced astrocyte alignment directs neurite outgrowth. Neuron Glia Biol 2:93–103.
    1. Anastassiou CA, Perin R, Markram H, Koch C. (2011). Ephaptic coupling of cortical neurons. Nat Neurosci 14:217–223.
    1. Andrade C. (2013). Once- to twice-daily, 3-year domiciliary maintenance transcranial direct current stimulation for severe, disabling, clozapine-refractory continuous auditory hallucinations in schizophrenia. J ECT 29:239–242.
    1. Ariza CA, Fleury AT, Tormos CJ, Petruk V, Chawla S, Oh J, Sakaguchi DS, Mallapragada SK. (2010). The influence of electric fields on hippocampal neural progenitor cells. Stem Cell Rev 6:585–600.
    1. Arlotti M, Rahman A, Minhas P, Bikson M. (2012). Axon terminal polarization induced by weak uniform DC electric fields: a modeling study. Conf Proc IEEE Eng Med Biol Soc 2012:4575–4578.
    1. Auvichayapat N, Rotenberg A, Gersner R, Ngodklang S, Tiamkao S, Tassaneeyakul W, Auvichayapat P. (2013). Transcranial direct current stimulation for treatment of refractory childhood focal epilepsy. Brain Stimul 6:696–700.
    1. Au-Yeung SS, Wang J, Chen Y, Chua E. (2014). Transcranial direct current stimulation to primary motor area improves hand dexterity and selective attention in chronic stroke. Am J Phys Med Rehabil 10.1097/PHM.0000000000000127.
    1. Babona-Pilipos R, Droujinine IA, Popovic MR, Morshead CM. (2011). Adult subependymal neural precursors, but not differentiated cells, undergo rapid cathodal migration in the presence of direct current electric fields. PLoS One 6:e23808.
    1. Bai H, Forrester JV, Zhao M. (2011). DC electric stimulation upregulates angiogenic factors in endothelial cells through activation of VEGF receptors. Cytokine 55:110–115.
    1. Batsikadze G, Moliadze V, Paulus W, Kuo MF, Nitsche MA. (2013). Partially non-linear stimulation intensity-dependent effects of direct current stimulation on motor cortex excitability in humans. J Physiol 591:1987–2000.
    1. Bedlack RS, Jr., Wei M, Loew LM. (1992). Localized membrane depolarizations and localized calcium influx during electric field-guided neurite growth. Neuron 9:393–403.
    1. Ben Taib NO, Manto M. (2009). Trains of transcranial direct current stimulation antagonize motor cortex hypoexcitability induced by acute hemicerebellectomy. J Neurosurg 111:796–806.
    1. Benninger DH, Lomarev M, Lopez G, Wassermann EM, Li X, Considine E, Hallett M. (2010). Transcranial direct current stimulation for the treatment of Parkinson’s disease. J Neurol Neurosurg Psychiatry 81:1105–1111.
    1. Bikson M, Inoue M, Akiyama H, Deans JK, Fox JE, Miyakawa H, Jefferys JG. (2004). Effects of uniform extracellular DC electric fields on excitability in rat hippocampal slices in vitro. J Physiol 557:175–190.
    1. Bikson M, Reato D, Rahman A. (2012). Cellular and network effects of transcranial direct current stimulation: insights from animal models and brain slices. In: Frontiers in neuroscience, pp 55–91 Boca Raton, FL: CRC Press.
    1. Bolzoni F, Pettersson LG, Jankowska E. (2013a) Evidence for long–lasting subcortical facilitation by transcranial direct current stimulation in the cat. J Physiol 591:3381–3399.
    1. Bolzoni F, Baczyk M, Jankowska E. (2013b) Subcortical effects of transcranial direct current stimulation (tDCS) in the rat. J Physiol 15;591:4027–4042.
    1. Borgens RB, Blight AR, McGinnis ME. (1987). Behavioral recovery induced by applied electric fields after spinal cord hemisection in guinea pig. Science 238:366–369.
    1. Borgens RB, Blight AR, McGinnis ME. (1990). Functional recovery after spinal cord hemisection in guinea pigs: the effects of applied electric fields. J Comp Neurol 296:634–653.
    1. Borgens RB, Toombs JP, Blight AR, McGinnis ME, Bauer MS, Widmer WR, Cook JR., Jr (1993). Effects of applied electric fields on clinical cases of complete paraplegia in dogs. Restor Neurol Neurosci 5:305–322.
    1. Borgens RB, Shi R, Mohr TJ, Jaeger CB. (1994). Mammalian cortical astrocytes align themselves in a physiological voltage gradient. Exp Neurol 128:41–49.
    1. Brunoni AR, Nitsche MA, Bolognini N, Bikson M, Wagner T, Merabet L, Edwards DJ, Valero-Cabre A, Rotenberg A, Pascual-Leone A, Ferrucci R, Priori A, Boggio PS, Fregni F. (2012). Clinical research with transcranial direct current stimulation (tDCS): challenges and future directions. Brain Stimul 5:175–195.
    1. Cambiaghi M, Velikova S, Gonzalez-Rosa JJ, Cursi M, Comi G, Leocani L. (2010). Brain transcranial direct current stimulation modulates motor excitability in mice. Eur J Neurosci 31:704–709.
    1. Cambiaghi M, Teneud L, Velikova S, Gonzalez-Rosa JJ, Cursi M, Comi G, Leocani L. (2011). Flash visual evoked potentials in mice can be modulated by transcranial direct current stimulation. Neuroscience 185:161–165.
    1. Cooper MS, Keller RE. (1984). Perpendicular orientation and directional migration of amphibian neural crest cells in dc electrical fields. Proc Natl Acad Sci U S A 81:160–164.
    1. Cork RJ, McGinnis ME, Tsai J, Robinson KR. (1994). The growth of PC12 neurites is biased towards the anode of an applied electrical field. J Neurobiol 25:1509–1516.
    1. Cormie P, Robinson KR. (2007). Embryonic zebrafish neuronal growth is not affected by an applied electric field in vitro. Neurosci Lett 411:128–132.
    1. Dell’Osso B, Zanoni S, Ferrucci R, Vergari M, Castellano F, D’Urso N, Dobrea C, Benatti B, Arici C, Priori A, Altamura AC. (2012). Transcranial direct current stimulation for the outpatient treatment of poor-responder depressed patients. Eur Psychiatry 27:513–517.
    1. Derkach VA, Oh MC, Guire ES, Soderling TR. (2007). Regulatory mechanisms of AMPA receptors in synaptic plasticity. Nat Rev Neurosci 8:101–113.
    1. Dockery CA, Liebetanz D, Birbaumer N, Malinowska M, Wesierska MJ. (2011). Cumulative benefits of frontal transcranial direct current stimulation on visuospatial working memory training and skill learning in rats. Neurobiol Learn Mem 96:452–460.
    1. Erskine L, Stewart R, McCaig CD. (1995). Electric field-directed growth and branching of cultured frog nerves: effects of aminoglycosides and polycations. J Neurobiol 26:523–536.
    1. Fecteau S, Agosta S, Hone-Blanchet A, Fregni F, Boggio P, Ciraulo D, Pascual-Leone A. (2014). Modulation of smoking and decision-making behaviors with transcranial direct current stimulation in tobacco smokers: a preliminary study. Drug Alcohol Depend 140:78–84.
    1. Feng JF, Liu J, Zhang XZ, Zhang L, Jiang JY, Nolta J, Zhao M. (2012). Guided migration of neural stem cells derived from human embryonic stem cells by an electric field. Stem Cells 30:349–355.
    1. Ferrucci R, Vergari M, Cogiamanian F, Bocci T, Ciocca M, Tomasini E, De Riz M, Scarpini E, Priori A. (2014). Transcranial direct current stimulation (tDCS) for fatigue in multiple sclerosis. NeuroRehabilitation 34:121–127.
    1. Franke K, Gruler H. (1990). Galvanotaxis of human granulocytes: electric field jump studies. Eur Biophys J 18:335–346.
    1. Fregni F, Pascual-Leone A. (2007). Technology insight: noninvasive brain stimulation in neurology-perspectives on the therapeutic potential of rTMS and tDCS. Nat Clin Pract Neurol 3:383–393.
    1. Frey U, Frey S, Schollmeier F, Krug M. (1996). Influence of actinomycin D, a RNA synthesis inhibitor, on long-term potentiation in rat hippocampal neurons in vivo and in vitro. J Physiol 490 (Pt 3):703–711.
    1. Fritsch B, Reis J, Martinowich K, Schambra HM, Ji Y, Cohen LG, Lu B. (2010). Direct current stimulation promotes BDNF-dependent synaptic plasticity: potential implications for motor learning. Neuron 66:198–204.
    1. Gersner R, Kravetz E, Feil J, Pell G, Zangen A. (2011). Long-term effects of repetitive transcranial magnetic stimulation on markers for neuroplasticity: differential outcomes in anesthetized and awake animals. J Neurosci 31:7521–7526.
    1. Huang R, Peng L, Hertz L. (1997). Effects of a low-voltage static electric field on energy metabolism in astrocytes. Bioelectromagnetics 18:77–80.
    1. Hubbard JI, Willis WD. (1962). Hyperpolarization of mammalian motor nerve terminals. J Physiol 163:115–137.
    1. Hunter MA, Coffman BA, Trumbo MC, Clark VP. (2013). Tracking the neuroplastic changes associated with transcranial direct current stimulation: a push for multimodal imaging. Front Hum Neurosci 7:495.
    1. Im CH, Park JH, Shim M, Chang WH, Kim YH. (2012). Evaluation of local electric fields generated by transcranial direct current stimulation with an extracephalic reference electrode based on realistic 3D body modeling. Phys Med Biol 57:2137–2150.
    1. Ingvar S. (1920). Reaction of cells to the galvanic current in tissue cultures. Exp Biol Med 17:198–199.
    1. Jennings J, Chen D, Feldman D. (2008). Transcriptional response of dermal fibroblasts in direct current electric fields. Bioelectromagnetics 29:394–405.
    1. Jiang T, Xu RX, Zhang AW, Di W, Xiao ZJ, Miao JY, Luo N, Fang YN. (2012). Effects of transcranial direct current stimulation on hemichannel pannexin-1 and neural plasticity in rat model of cerebral infarction. Neuroscience 226:421–426.
    1. Kabakov AY, Muller PA, Pascual-Leone A, Jensen FE, Rotenberg A. (2012). Contribution of axonal orientation to pathway-dependent modulation of excitatory transmission by direct current stimulation in isolated rat hippocampus. J Neurophysiol 107:1881–1889.
    1. Kamida T, Kong S, Eshima N, Abe T, Fujiki M, Kobayashi H. (2011). Transcranial direct current stimulation decreases convulsions and spatial memory deficits following pilocarpine-induced status epilepticus in immature rats. Behav Brain Res 217:99–103.
    1. Kampa BM, Clements J, Jonas P, Stuart GJ. (2004). Kinetics of Mg2+ unblock of NMDA receptors: implications for spike-timing dependent synaptic plasticity. J Physiol 556:337–345.
    1. Kim SJ, Kim BK, Ko YJ, Bang MS, Kim MH, Han TR. (2010). Functional and histologic changes after repeated transcranial direct current stimulation in rat stroke model. J Korean Med Sci 25:1499–1505.
    1. Koppes AN, Seggio AM, Thompson DM. (2011). Neurite outgrowth is significantly increased by the simultaneous presentation of Schwann cells and moderate exogenous electric fields. J Neural Eng 8:046023.
    1. Lamy JC, Boakye M. (2013). BDNF Val66Met polymorphism alters spinal DC stimulation-induced plasticity in humans. J Neurophysiol 110:109–116.
    1. Lang N, Siebner HR, Ward NS, Lee L, Nitsche MA, Paulus W, Rothwell JC, Lemon RN, Frackowiak RS. (2005). How does transcranial DC stimulation of the primary motor cortex alter regional neuronal activity in the human brain? Eur J Neurosci 22:495–504.
    1. Laste G, Caumo W, Adachi LN, Rozisky JR, de Macedo IC, Filho PR, Partata WA, Fregni F, Torres IL. (2012). After-effects of consecutive sessions of transcranial direct current stimulation (tDCS) in a rat model of chronic inflammation. Exp Brain Res 221:75–83.
    1. Li L, El-Hayek YH, Liu B, Chen Y, Gomez E, Wu X, Ning K, Li L, Chang N, Zhang L, Wang Z, Hu X, Wan Q. (2008). Direct-current electrical field guides neuronal stem/progenitor cell migration. Stem Cells 26:2193–2200.
    1. Li J, Nandagopal S, Wu D, Romanuik SF, Paul K, Thomson DJ, Lin F. (2011a) Activated T lymphocytes migrate toward the cathode of DC electric fields in microfluidic devices. Lab Chip 11:1298–1304.
    1. Li Y, Tian X, Qian L, Yu X, Jiang W. (2011b) Anodal transcranial direct current stimulation relieves the unilateral bias of a rat model of Parkinson’s disease. Conf Proc IEEE Eng Med Biol Soc 2011:765–768.
    1. Liebetanz D, Nitsche MA, Tergau F, Paulus W. (2002). Pharmacological approach to the mechanisms of transcranial DC-stimulation-induced after-effects of human motor cortex excitability. Brain 125:2238–2247.
    1. Liebetanz D, Fregni F, Monte-Silva KK, Oliveira MB, Amancio-dos-Santos A, Nitsche MA, Guedes RC. (2006a) After-effects of transcranial direct current stimulation (tDCS) on cortical spreading depression. Neurosci Lett 398:85–90.
    1. Liebetanz D, Klinker F, Hering D, Koch R, Nitsche MA, Potschka H, Loscher W, Paulus W, Tergau F. (2006b) Anticonvulsant effects of transcranial direct-current stimulation (tDCS) in the rat cortical ramp model of focal epilepsy. Epilepsia 47:1216–1224.
    1. Liebetanz D, Koch R, Mayenfels S, Konig F, Paulus W, Nitsche MA. (2009). Safety limits of cathodal transcranial direct current stimulation in rats. Clin Neurophysiol 120:1161–1167.
    1. Lin SL, Li Y, Woolley AT, Lee ML, Tolley HD, Warnick KF. (2008). Programmed elution and peak profiles in electric field gradient focusing. Electrophoresis 29:1058–1066.
    1. Long H, Yang G, Wang Z. (2011). Galvanotactic migration of EA.Hy926 endothelial cells in a novel designed electric field bioreactor. Cell Biochem Biophys 61:481–491.
    1. Marquez-Ruiz J, Leal-Campanario R, Sanchez-Campusano R, Molaee-Ardekani B, Wendling F, Miranda PC, Ruffini G, Gruart A, Delgado-Garcia JM. (2012). Transcranial direct-current stimulation modulates synaptic mechanisms involved in associative learning in behaving rabbits. Proc Natl Acad Sci U S A 109:6710–6715.
    1. McCaig CD, Sangster L, Stewart R. (2000). Neurotrophins enhance electric field-directed growth cone guidance and directed nerve branching. Dev Dyn 217:299–308.
    1. McCaig CD, Rajnicek AM, Song B, Zhao M. (2005). Controlling cell behavior electrically: current views and future potential. Physiol Rev 85:943–978.
    1. McLaughlin S, Poo MM. (1981). The role of electro-osmosis in the electric-field-induced movement of charged macromolecules on the surfaces of cells. Biophys J 34:85–93.
    1. Meng X, Arocena M, Penninger J, Gage FH, Zhao M, Song B. (2011). PI3K mediated electrotaxis of embryonic and adult neural progenitor cells in the presence of growth factors. Exp Neurol 227:210–217.
    1. Minichiello L. (2009). TrkB signalling pathways in LTP and learning. Nat Rev Neurosci 10:850–860.
    1. Monte-Silva K, Kuo MF, Hessenthaler S, Fresnoza S, Liebetanz D, Paulus W, Nitsche MA. (2013). Induction of late LTP-like plasticity in the human motor cortex by repeated non-invasive brain stimulation. Brain Stimul 6:424–432.
    1. Mycielska ME, Djamgoz MB. (2004). Cellular mechanisms of direct-current electric field effects: galvanotaxis and metastatic disease. J Cell Sci 117:1631–1639.
    1. Neal AP, Guilarte TR. (2010). Molecular neurobiology of lead (Pb(2+)): effects on synaptic function. Mol Neurobiol 42:151–160.
    1. Nitsche MA, Paulus W. (2001). Sustained excitability elevations induced by transcranial DC motor cortex stimulation in humans. Neurology 57:1899–1901.
    1. Nitsche MA, Lampe C, Antal A, Liebetanz D, Lang N, Tergau F, Paulus W. (2006). Dopaminergic modulation of long-lasting direct current-induced cortical excitability changes in the human motor cortex. Eur J Neurosci 23:1651–1657.
    1. Nitsche MA, Cohen LG, Wassermann EM, Priori A, Lang N, Antal A, Paulus W, Hummel F, Boggio PS, Fregni F, Pascual-Leone A. (2008). Transcranial direct current stimulation: State of the art 2008. Brain Stimul 1:206–223.
    1. Nitsche MA, Kuo MF, Karrasch R, Wachter B, Liebetanz D, Paulus W. (2009). Serotonin affects transcranial direct current-induced neuroplasticity in humans. Biol Psychiatry 66:503–508.
    1. Nitsche MA, Muller-Dahlhaus F, Paulus W, Ziemann U. (2012). The pharmacology of neuroplasticity induced by non-invasive brain stimulation: building models for the clinical use of CNS active drugs. J Physiol. 590:4641–4662
    1. Opazo P, Labrecque S, Tigaret CM, Frouin A, Wiseman PW, De Koninck P, Choquet D. (2010). CaMKII triggers the diffusional trapping of surface AMPARs through phosphorylation of stargazin. Neuron 67:239–252.
    1. Orida N, Feldman JD. (1982). Directional protrusive pseudopodial activity and motility in macrophages induced by extracellular electric fields. Cell Motil 2:243–255.
    1. Palmer AM, Messerli MA, Robinson KR. (2000). Neuronal galvanotropism is independent of external Ca(2+) entry or internal Ca(2+) gradients. J Neurobiol 45:30–38.
    1. Pan L, Borgens RB. (2010). Perpendicular organization of sympathetic neurons within a required physiological voltage. Exp Neurol 222:161–164.
    1. Patel N, Poo MM. (1982). Orientation of neurite growth by extracellular electric fields. J Neurosci 2:483–496.
    1. Pedron S, Monnin J, Haffen E, Sechter D, Van Waes V. (2014). Repeated transcranial direct current stimulation prevents abnormal behaviors associated with abstinence from chronic nicotine consumption. Neuropsychopharmacology 39:981–988.
    1. Pelletier SJ, Lagace M, St-Amour I, Arsenault D, Cisbani G, Chabrat A, Fecteau S, Levesque M, Cicchetti F (2014). The morphological and molecular changes of brain cells exposed to direct current electric field stimulation. Int J Neuropsychopharmacol in press.
    1. Perret S, Cantereau A, Audin J, Dufy B, Georgescauld D. (1999). Interplay between Ca2+ release and Ca2+ influx underlies localized hyperpolarization-induced [Ca2+]i waves in prostatic cells. Cell Calcium 25:297–311.
    1. Peruzzotti-Jametti L, Cambiaghi M, Bacigaluppi M, Gallizioli M, Gaude E, Mari S, Sandrone S, Cursi M, Teneud L, Comi G, Musco G, Martino G, Leocani L. (2013). Safety and efficacy of transcranial direct current stimulation in acute experimental ischemic stroke. Stroke 44:3166–3174.
    1. Pozzo-Miller LD, Gottschalk W, Zhang L, McDermott K, Du J, Gopalakrishnan R, Oho C, Sheng ZH, Lu B. (1999). Impairments in high-frequency transmission, synaptic vesicle docking, and synaptic protein distribution in the hippocampus of BDNF knockout mice. J Neurosci 19:4972–4983.
    1. Purpura DP, McMurtry JG. (1965). Intracellular activities and evoked potential changes during polarization of motor cortex. J Neurophysiol 28:166–185.
    1. Radman T, Ramos RL, Brumberg JC, Bikson M. (2009). Role of cortical cell type and morphology in subthreshold and suprathreshold uniform electric field stimulation in vitro. Brain Stimul 2:215–228, 228 e211–213.
    1. Rahman A, Reato D, Arlotti M, Gasca F, Datta A, Parra LC, Bikson M. (2013). Cellular effects of acute direct current stimulation: somatic and synaptic terminal effects. J Physiol 591:2563–2578.
    1. Rajnicek AM, Foubister LE, McCaig CD. (2006). Temporally and spatially coordinated roles for Rho, Rac, Cdc42 and their effectors in growth cone guidance by a physiological electric field. J Cell Sci 119:1723–1735.
    1. Ranieri F, Podda MV, Riccardi E, Frisullo G, Dileone M, Profice P, Pilato F, Di Lazzaro V, Grassi C. (2012). Modulation of LTP at rat hippocampal CA3-CA1 synapses by direct current stimulation. J Neurophysiol 107:1868–1880.
    1. Rueger MA, Keuters MH, Walberer M, Braun R, Klein R, Sparing R, Fink GR, Graf R, Schroeter M. (2012). Multi-session transcranial direct current stimulation (tDCS) elicits inflammatory and regenerative processes in the rat brain. PLoS One 7:e43776.
    1. Schweid L, Rushmore RJ, Valero-Cabre A. (2008). Cathodal transcranial direct current stimulation on posterior parietal cortex disrupts visuo-spatial processing in the contralateral visual field. Exp Brain Res 186:409–417.
    1. Smolen P. (2007). A model of late long-term potentiation simulates aspects of memory maintenance. PLoS One 2:e445.
    1. Sohn MK, Jee SJ, Kim YW. (2013). Effect of transcranial direct current stimulation on postural stability and lower extremity strength in hemiplegic stroke patients. Ann Rehabil Med 37:759–765.
    1. Song B, Gu Y, Pu J, Reid B, Zhao Z, Zhao M. (2007). Application of direct current electric fields to cells and tissues in vitro and modulation of wound electric field in vivo. Nat Protoc 2:1479–1489.
    1. Spezia Adachi LN, Caumo W, Laste G, Fernandes Medeiros L, Ripoll Rozisky J, de Souza A, Fregni F, Torres IL. (2012). Reversal of chronic stress-induced pain by transcranial direct current stimulation (tDCS) in an animal model. Brain Res 1489:17–26.
    1. Stagg CJ, Nitsche MA. (2011). Physiological basis of transcranial direct current stimulation. Neuroscientist 17:37–53.
    1. Stagg CJ, O’Shea J, Kincses ZT, Woolrich M, Matthews PM, Johansen-Berg H. (2009a) Modulation of movement-associated cortical activation by transcranial direct current stimulation. Eur J Neurosci 30:1412–1423.
    1. Stagg CJ, Best JG, Stephenson MC, O’Shea J, Wylezinska M, Kincses ZT, Morris PG, Matthews PM, Johansen-Berg H. (2009b) Polarity-sensitive modulation of cortical neurotransmitters by transcranial stimulation. J Neurosci 29:5202–5206.
    1. Tahtis V, Kaski D, Seemungal BM. (2014). The effect of single session bi-cephalic transcranial direct current stimulation on gait performance in sub-acute stroke: a pilot study. Restor Neurol Neurosci, 10.3233/RNN-140393.
    1. Takano Y, Yokawa T, Masuda A, Niimi J, Tanaka S, Hironaka N. (2011). A rat model for measuring the effectiveness of transcranial direct current stimulation using fMRI. Neurosci Lett 491:40–43.
    1. Tanaka T, Takano Y, Tanaka S, Hironaka N, Kobayashi K, Hanakawa T, Watanabe K, Honda M. (2013). Transcranial direct-current stimulation increases extracellular dopamine levels in the rat striatum. Front Syst Neurosci 7:6.
    1. Viard P, Butcher AJ, Halet G, Davies A, Nurnberg B, Heblich F, Dolphin AC. (2004). PI3K promotes voltage-dependent calcium channel trafficking to the plasma membrane. Nat Neurosci 7:939–946.
    1. Wachter D, Wrede A, Schulz-Schaeffer W, Taghizadeh-Waghefi A, Nitsche MA, Kutschenko A, Rohde V, Liebetanz D. (2011). Transcranial direct current stimulation induces polarity-specific changes of cortical blood perfusion in the rat. Exp Neurol 227:322–327.
    1. Wood MD, Willits RK. (2009). Applied electric field enhances DRG neurite growth: influence of stimulation media, surface coating and growth supplements. J Neural Eng 6:046003.
    1. Yoon KJ, Oh BM, Kim DY. (2012). Functional improvement and neuroplastic effects of anodal transcranial direct current stimulation (tDCS) delivered 1 day vs. 1 week after cerebral ischemia in rats. Brain Res 1452:61–72.
    1. Zhang P, Liu ZT, He GX, Liu JP, Feng J. (2011). Low-voltage direct-current stimulation is safe and promotes angiogenesis in rabbits with myocardial infarction. Cell Biochem Biophys 59:19–27.
    1. Zhao M. (2009). Electrical fields in wound healing: an overriding signal that directs cell migration. Semin Cell Dev Biol 20:674–682.
    1. Zhao M, Bai H, Wang E, Forrester JV, McCaig CD. (2004). Electrical stimulation directly induces pre-angiogenic responses in vascular endothelial cells by signaling through VEGF receptors. J Cell Sci 117:397–405.
    1. Zhao M, Song B, Pu J, Wada T, Reid B, Tai G, Wang F, Guo A, Walczysko P, Gu Y, Sasaki T, Suzuki A, Forrester JV, Bourne HR, Devreotes PN, McCaig CD, Penninger JM. (2006). Electrical signals control wound healing through phosphatidylinositol-3-OH kinase-gamma and PTEN. Nature 442:457–460.
    1. Zhao Z, Qin L, Reid B, Pu J, Hara T, Zhao M. (2012). Directing migration of endothelial progenitor cells with applied DC electric fields. Stem Cell Res 8:38–48.
    1. Zobeiri M, van Luijtelaar G. (2013). Noninvasive transcranial direct current stimulation in a genetic absence model. Epilepsy Behav 26:42–50.

Source: PubMed

3
Abonner