Next Article in Journal
Understanding the Mechanosensitivity of the Median Nerve in Pre-Surgical Carpal Tunnel Syndrome Patients: A Correlational Study
Next Article in Special Issue
Non-Dominant Hemisphere Excitability Is Unaffected during and after Transcranial Direct Current Stimulation of the Dominant Hemisphere
Previous Article in Journal
The Role of Aging in Intracerebral Hemorrhage
Previous Article in Special Issue
Does Multisession Cathodal Transcranial Direct Current Stimulation of the Left Dorsolateral Prefrontal Cortex Prime the Effects of Cognitive Behavioral Therapy on Fear of Pain, Fear of Movement, and Disability in Patients with Nonspecific Low Back Pain? A Randomized Clinical Trial Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Transcranial Direct Current Stimulation to Ameliorate Post-Stroke Cognitive Impairment

by
Kelly L. Sloane
1,2 and
Roy H. Hamilton
1,2,*
1
Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
2
Department of Physical Medicine and Rehabilitation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
*
Author to whom correspondence should be addressed.
Brain Sci. 2024, 14(6), 614; https://doi.org/10.3390/brainsci14060614
Submission received: 22 April 2024 / Revised: 4 June 2024 / Accepted: 12 June 2024 / Published: 19 June 2024

Abstract

:
Post-stroke cognitive impairment is a common and disabling condition with few effective therapeutic options. After stroke, neural reorganization and other neuroplastic processes occur in response to ischemic injury, which can result in clinical improvement through spontaneous recovery. Neuromodulation through transcranial direct current stimulation (tDCS) is a promising intervention to augment underlying neuroplasticity in order to improve cognitive function. This form of neuromodulation leverages mechanisms of neuroplasticity post-stroke to optimize neural reorganization and improve function. In this review, we summarize the current state of cognitive neurorehabilitation post-stroke, the practical features of tDCS, its uses in stroke-related cognitive impairment across cognitive domains, and special considerations for the use of tDCS in the post-stroke patient population.

1. Introduction

With a prevalence of almost 800,000 annually in the United States, stroke is the leading cause of adult disability [1]. Estimates of the prevalence of cognitive dysfunction after stroke vary considerably depending on the type of cognitive testing and the timing after stroke, but estimates range from 30% to up to 90% [2,3]. Older adults and those with vascular risk factors are at a 3.6-fold increase in the risk of poststroke dementia [4]. Every 1-year increase in baseline age results in 17% higher odds of cognitive impairment at 1 year post-stroke [5]. Mild or subtle cognitive dysfunction can sometimes be missed early in the post-stroke period. Presence of post-stroke cognitive impairment, no matter the degree of impairment, is associated with poorer functional outcomes as well as increased caregiver burden, healthcare costs, and mortality [6,7,8]. Even among patients with good overall functional outcomes, cognitive impairment persists in over 70% of cases after 3 months [9]. Attention and memory are among the most common stroke-related deficits [3,10] with a negative impact on daily functioning and responsiveness to rehabilitation [11,12,13,14]. Despite how common and disabling post-stroke cognitive impairment can be, there are few effective therapies. In this review, we describe a promising intervention, transcranial direct current stimulation (tDCS), to treat post-stroke cognitive impairment by leveraging neuroplastic processes.
The gold standard for treatment of post-stroke cognitive impairment remains one-on-one or group cognitive therapy sessions with a licensed rehabilitation specialist. Rehabilitation strategies for specific cognitive functions, such as training of memory, have been used with the aid of external memory aides, visual scanning training, and problem-solving techniques [15]. The success of cognitive and speech therapies varies depending on insurance status, socio-economic factors, and accessibility, and time-intensive and prolonged courses of therapy are often not offered beyond the immediate post-acute recovery period [16]. Pharmacological therapies like donepezil and other anticholinesterase inhibitors, which are commonly used in the treatment of Alzheimer’s disease, have shown limited benefit in post-stroke cognitive impairment [17]. Physical activity, particularly aerobic exercise, has been associated with a reduced risk of dementia in older adults [18] and similarly in individuals with stroke-related cognitive impairment; the therapeutic effects of exercise have been identified, but the durability of these effects are not yet known [19,20]. Similarly, cognitive rehabilitation techniques have demonstrated improvement in cognitive function in the short-term post-treatment period, but the long-term effects are not known [21,22]. Other treatments, like mindfulness-based stress reduction, have also been tested in small pilot studies with short-term benefits [21].
Many treatment strategies are derived from approaches to other neurological conditions affecting cognition, like neurodegeneration or cognitive aging. However, they do not leverage post-stroke neuroplastic processes occurring in the immediate post-stroke period. Forms of non-invasive neuromodulation have the potential to modulate the spontaneous recovery already occurring in neural networks. As such, non-invasive brain stimulation has emerged as a promising intervention to treat cognitive impairment in stroke survivors [23]. One such approach, tDCS, is a promising, low-cost, accessible technique that has been demonstrated to be an effective tool for improving cognitive dysfunction in healthy adults as well as those with a variety of brain pathologies [24,25,26,27]. However, for the stroke patient population, the majority of neuromodulation studies have focused on motor rather than cognitive impairments.
In this review, we will provide an overview of the application of tDCS as an intervention for augmenting cognitive recovery after stroke. We will first discuss the current state of cognitive rehabilitation in post-stroke recovery. Next, we will describe the mechanisms of action of tDCS and the utility of tDCS to augment cognitive performance in non-stroke patient populations. We will then review the existing studies of tDCS to improve cognitive performance in post-stroke cognitive impairment and discuss future applications.

2. Current State of Cognitive Neurorehabilitation in Post-Stroke Recovery

2.1. Cognitive Profile

Cognition is an umbrella term for specific higher-order functions and can be divided into at least six neurocognitive domains: language, learning and memory, social cognition, complex attention, executive function, and perceptual-motor function [28]. Cognitive dysfunction and dementia frequently occur following an acute stroke, and they are an important cause of stroke-related morbidity [29]. Approximately 66% of patients show cognitive impairment in the acute phase [30], and 50% of stroke survivors fulfil criteria for mild cognitive impairment after 18 months [31,32]. In addition to impairments in global cognition, deficits in attention and executive functions are common [33]. Despite the prevalence of post-stroke cognitive impairment, most research in stroke recovery focuses on non-cognitive impairments. In fact, a recent review found that only 4.6% of peer-reviewed research publications involving stroke survivors included a measurement of cognition [34].
Estimates of the incidence and prevalence of cognitive dysfunction after stroke vary. Some studies suggest that vascular cognitive impairments affect up to 90% of people after stroke [3,9]. Other studies give lower estimates at 10–35%, likely because of differences in assessments or definitions of dementia in hospital and population-based studies [35]. In addition to age and ischemic small vessel white matter disease, which are known mediators of post-stroke cognitive impairment [36], neuroimaging findings associated with post-stroke cognitive impairment include: microbleeds, brain atrophy, medial temporal atrophy, and lesions in strategic brain locations (e.g., right corticospinal tract, left arcuate fasciculus, left middle frontal gyrus, and left postero-inferior cerebellum) [37,38].
Cognitive deficits are associated with worse outcomes. Individuals who are cognitively impaired 3 months after stroke have a 53% increased risk of death at 5, 10, and 15 years after stroke compared to those without cognitive impairment [6]. Among patients with motor deficits in the arm after stroke, those with attention and executive function difficulties demonstrated worse recovery than those with intact cognition [39]. Further, cognitive dysfunction after stroke, as defined by the Montreal Cognitive Assessment (MoCA) of less than 26, was associated with increased functional impairment with a modified Rankin score (mRS) greater than 2 at 6, 12, and 36 months after stroke [7]. Even among patients who show “good” functional outcomes, as defined by mRS as less than 2, more than half have persistent cognitive dysfunction, most commonly affecting memory, visuospatial, and executive functioning [3,40]. In addition to causing new post-stroke cognitive impairment, stroke can also accelerate pre-existing cognitive decline due to underlying neurodegenerative disease [2].

2.2. Impact of Cognitive Impairment on Other Modalities of Recovery

There is a growing body of evidence that post-stroke cognitive impairment is associated with worse recovery and less responsiveness to therapies for language and motor impairments [40,41,42] and functional dependence [43]. Learning in any modality post-stroke engages cognitive processes [44]. Motor performance may be affected by cognition function, especially involving tasks with increasing cognitive demands [39,45]. In a study of subacute stroke patients at 3 months after stroke, impairments in motor function were associated with impairments in memory and executive function [46]. Worse cognitive function post-stroke was associated with lesser responsiveness to a gait intervention [47] and completion of complex motor tasks [48]. The full extent of how cognitive status modulates response to rehabilitation interventions may not be fully understood, as many studies of post-stroke motor impairment limit participation based on cognitive status (i.e., cognitive impairment is an exclusion for participation) [39].

2.3. Interventions to Improve Post-Stroke Cognitive Impairment

Cognitive rehabilitation provided by speech-language pathologists or occupational therapists is the current gold standard of care for cognitive recovery. High-intensity rehabilitative training can alleviate impairments in cognitive functions [21,49], with greater than 50 min per day of cognitive therapy associated with improved cognitive outcomes, compared to lesser duration of therapy [50]. However, in real-world practice, patients participate in much less therapy than required for successful recovery [51,52]. Clinical factors, such as stroke severity and discharge destination, and non-clinical factors, such as insurance status, socio-economic factors, and accessibility, can limit opportunities for rehabilitative training [53,54,55]. In addition, even when available, the therapies themselves require substantial effort, and these motivational demands are often perceived as overwhelming and exhausting [56].
Other interventions to improve cognition after stroke have been studied with limited success. Randomized controlled trials of pharmacological interventions, such as donepezil [57], escitalopram [58], and amantadine [59], have revealed small but significant improvements in cognitive performance. Virtual reality [60], computer training [61], and aerobic exercise [62] have also shown promise.

3. tDCS Mechanism, Considerations and Other Uses

3.1. Mechanism of Action

tDCS is a non-invasive form of brain stimulation that can target specific brain regions integral to the cognitive network. tDCS uses constant, low direct current through external electrodes, which penetrate the skin, skull, and meninges to modify neural transmembrane electrical potentials and, therefore, the excitability and firing rate of neurons [63]. In normal neuronal function, the neuronal resting membrane potential is influenced by the movement of ions across the cell membrane. Depolarization refers to the increased positive membrane potential, and conversely, hyperpolarization refers to the negative membrane potential. To generate an action potential, the opening of the voltage-gated sodium channels leads to the influx of positive sodium ions into the cell such that the membrane potential becomes more positive, and depolarization occurs. In tDCS, a low intensity electrical current can alter the resting membrane potential in the neurons near the electrode [64]. Neurons in proximity to the anode exhibit increased depolarization of their transmembrane potential, and neurons underlying a cathode appear to exhibit hyperpolarization of their transmembrane potential [65,66]. The cortical effects of anodal and cathodal tDCS are often conceived of as being excitatory and inhibitory, respectively; however, the relationship between stimulation polarity and elicited cortical effects can be more complicated, depending on parameters such as stimulation intensity and duration [67]. Alterations in transmembrane potentials appear to last up to an hour, cause a local effect, and are N-methyl-D-aspartate (NMDA) receptor dependent [68]. Magnetic resonance imaging (MRI) studies have demonstrated increased perfusion and functional connectivity during stimulation [69]. The administration of tDCS has also been found to be very safe, with adverse effects that include fatigue, mild headache, nausea, and itching in the area of stimulation [70].

3.2. Practical Features of tDCS

The practical features of tDCS make it an especially useful tool for neurorehabilitation. Devices are small and portable, primarily comprising a battery-powered device, two electrode wires (anode and cathode), and a head strap. This device delivers a unidirectional, constant current, with the anode and cathode placed in different locations on the scalp, depending on the functional outcome desired. Once a stimulation target is selected, the corresponding location can be found using 10–20 electroencephalogram (EEG) system measurements. While this approach is less precise anatomically, it does not require neuronavigation technologies or physiologic localization using transcranial magnetic stimulation [71]. Several options exist for the positioning of electrodes on the head in the target location. Some device models employ adjustable rubber straps to accommodate each individual person’s head size, while others use pre-fabricated headgear, which can be used for a range of head sizes and has electrode sites at pre-determined montage locations. The components of a conventional tDCS device are illustrated in Figure 1. Many tDCS devices can be employed with relatively little training, and self-administration of tDCS or delivery by a caregiver is being explored with several devices.
Guidelines for technology and protocol implementation have been developed to direct the remote use of tDCS in clinical research [72]. Remote-use devices, which are designed be to compact and user friendly, can be pre-programmed with stimulation parameters such that the participant cannot alter the stimulation dose, duration, or frequency [72]. These parameters can be controlled through hardware or software limitations that prevent indiscriminate device use. With respect to hardware-based limitations, a device is pre-programmed to provide a set number of sessions (e.g., 5 sessions of 20 min each) with defined intensity (e.g., 2 milliamps anodal stimulation) within a set interval (e.g., 1 session per 24 h interval). Regarding software-based limitations, a device is programmed to provide stimulation at a specific intensity, duration, and session frequency and can only be activated with a unique code. Effective and safe remote tDCS administration requires not only these safeguards on device administration but also operationalization of remotely supervised tDCS, including comprehensive protocols for training participants in the use of tDCS and ongoing monitoring for compliance, treatment-associated adverse effects, and the user’s capability to self-administer tDCS. Capacity for remote use invites a broader and more accessible intervention, but it also risks inconsistencies in use when it is self-administered by the participant rather than the research staff. These challenges may be mitigated by requiring hands-on pre-intervention training for research participants to learn and practice with the technology and real-time monitoring of treatment fidelity through videoconferencing [72,73,74,75,76]. A single center reported findings from its studies involving 6779 participants who underwent remotely supervised tDCS and found no serious adverse events, few study discontinuations due to tolerability (0.04%), and there was high treatment fidelity (92–98%) [75]. Remote tDCS has been used in studies of a wide variety of neuropsychiatric conditions including multiple sclerosis [72], Parkinson’s disease [74], major depressive disorder [76], depression, [77], and stroke [78], to name a few.
Similar to conventional tDCS, wherein stimulation is performed through a single pair of large (often 5 × 7 cm or 5 × 5 cm) electrodes, high-definition tDCS (HD-tDCS) has been developed to deliver a focused current to the target site. HD-tDCS employs multiple smaller electrodes to focus the current in one or more regions of the brain and has substantially higher spatial resolution than conventional tDCS. For example, a common HD-tDCS montage is the 4 × 1 ring, in which a center electrode is placed over the target cortical region surrounded by four return electrodes [79].

3.3. Models of Neuroplasticity to Guide Stimulation Targets

The anatomic targets of tDCS are typically at the level of the cortex, as it is much more difficult to reliably direct current to subcortical or brainstem sites. Additionally, stimulation targets are typically brain regions that are hypothesized directly related to the impairment being targeted. There are two central models that have historically guided how brain regions are selected as targets for stimulation (Figure 2). The first model—the interhemispheric imbalance model—is based on the notion that functional impairments are mediated, at least in part, by a mismatch in brain activity between cerebral hemispheres induced by unilateral injury of the brain. This model is predicated on the notion that many of the connections between the two cerebral hemispheres are inhibitory in nature and that intact cerebral hemispheres, therefore, normally exert an inhibitory influence on each other. According to this account, a unilateral injury to the brain results in (1) decreased cortical excitability on the injured (i.e., ipsilesional) hemisphere, (2) decreased inhibitory projections to the intact (i.e., contralesional) hemisphere, and (3) further inhibition of the ipsilesional hemisphere mediated by increased contralesional interhemispheric inhibition [80]. This implies that two intervention strategies for improving the imbalance between ipsilesional and contralesional hemispheres would be to either increase the excitability of the ipsilesional hemisphere or decrease the activity of the contralesional hemisphere. This version of the interhemispheric imbalance model has motivated a number of studies, employing tDCS and other forms of noninvasive neuromodulation in motor recovery [81,82], neglect [83], and aphasia [83,84,85,86,87].
In the second model—the bimodal balance recovery model—the residual structural brain reserve influences physiology and functional output [88]. For individuals with low structural reserve (i.e., greater stroke-induced structural brain injury) especially, increased cortical excitability of the contralesional side will provide compensatory function and activity in residual brain networks [88]. These models have been developed from the perspective of motor recovery, where motor output is largely attributed to the corticospinal tract, but there is not consensus on an optimal region for stimulation in cognitive rehabilitation. Functional connectivity studies have demonstrated that cognitive operations are organized along interconnected networks rather than represented in discrete processing modules [89]. In certain forms of cognitive impairment like aphasia or neglect, where discrete hubs of activity have been identified, the site of stimulation is predominantly the left inferior frontal gyrus [86] and the right posterior parietal cortex [83], respectively. However, for domain-general cognitive functions, any hub within the network of interest could serve as a potential stimulation site. The dorsolateral prefrontal cortex (DLPFC) has emerged as a popular site of stimulation. The DLPFC is a crucial hub for multiple cognitive networks (dorsal attention network, salience network, frontoparietal network) that support processing and response involved in executive function, working memory, motor planning, and attention [90,91,92] and is a common target for stimulation because of its role in these cognitive domains [93].

3.4. Task-Dependency of tDCS and Approaches to Therapy

The effect of tDCS depends on the selection of the site of stimulation and the concurrent training task. The task-dependency relates to the fact that tDCS is believed to induce subthreshold changes in the neuronal resting membrane thresholds [94,95]. However, neuroplastic changes associated with tDCS are thought to be induced via Hebbian principles of neuronal activation (i.e., “neurons that fire together wire together”) and are dependent on the presence of recurrent, organized firing of relevant neuronal pathways [96,97,98]. By this account, the mechanistic goal of tDCS is to modulate existing patterns of neuronal firing in order to further strengthen or weaken those firing patterns. For this reason, it has been argued that stimulation must be paired with training tasks that activate the brain regions or networks that are being stimulated [99]. For example, the ipsilesional primary motor cortex would be an appropriate target for the amelioration of stroke-induced hemiparesis, and constraint-induced motor therapy of that hemiparetic arm would be an appropriate training task because it is designed to engage the ipsilesional motor cortex through forced use of the paretic limb.

3.5. Brain State-Dependency for tDCS and Priming

The brain state of the individual prior to stimulation may also play an important role in response to subsequent tDCS and its effects [100]. Thus, priming involves optimizing the pre-stimulation brain state to achieve greater plasticity-related changes during the stimulation period [101]. Priming has been evaluated through several methods, including physical exercise, cognitive exercises, or stimulation itself. Aerobic exercise has been shown to improve cognitive outcomes in older adults with cognitive impairment, [102] and aerobic exercise is also associated with increased cortical excitability and decreased intracortical inhibition, conditions favorable for plasticity [103]. Thus, aerobic exercise’s synergistic effects may prime the brain for focal modulation [104], which is currently being studied with an ongoing clinical trial [105]. Emotional stress may also be a priming method for enhancing the effects of tDCS. Acute stress activates stress-related neural circuits, including the DLPFC, through the release of cortisol and catecholamines to coactivate the targeted network [106,107] and, therefore, alter the neural system’s responsiveness to stimulation [106]. Studies of healthy adults have indicated that stress priming is associated with better performance on working memory tasks, particularly those involving emotional content [108]. In addition, tDCS itself can be used to prime the brain resting state to improve responsiveness to tDCS. Facilitatory tDCS protocols have been developed with subthreshold, low-intensity cathodal stimulation subthreshold, low-intensity priming sessions using cathodal tDCS to elicit low-level prior neuronal activity, enhancing the likelihood of further long-term potentiation (LTP)-like effects without significantly impacting overall excitability [109,110,111].

3.6. Limitations of tDCS

There are several key limitations of tDCS in its applications for cognitive enhancement. First, the low current of tDCS leads to a subthreshold alteration of the resting membrane potential and supports neuron depolarization but does not directly alter action potential and depolarization [112]. Therefore, the effect of tDCS is less robust than other neuromodulatory modalities like transcranial magnetic stimulation (TMS). Second, tDCS has limited spatial accuracy due to its non-invasive administration over the scalp in approximate locations. Computational models of current flow in conventional tDCS show diffuse spread over cortical regions [113]. Techniques like high-definition tDCS can constrict current to specific regions through multiple smaller electrodes to improve spatial resolution. Third, although tDCS has been studied in a number of neurologic and psychiatric conditions, the heterogeneity of study designs have made it difficult to determine the optimal stimulation parameters and treatment protocols, especially for post-stroke cognitive impairment [114].

3.7. Comparison of tDCS to Other Neuromodulatory Techniques Used in Post-Stroke Cognitive Impairment

Several other neuromodulatory techniques exist that can also be considered for intervention in post-stroke cognitive impairment. Similar to tDCS, transcranial magnetic stimulation (TMS) involves passing an electric current through conductive wires of an insulated coil to induce a local magnetic field, which then stimulates a secondary electric current in the brain underlying the coil. Repetitive TMS (rTMS), a therapeutic form of TMS, uses high frequency (at or above approximately 5 pulses per second) stimulation, which can induce the promotion of synaptogenesis and long-term potentiation [115]. Because of the mechanism of action of TMS, induction of seizures is a possible severe acute adverse effect. Though relatively rare, seizures have been described in single-pulse, pattern, and repetitive pulse uses of TMS [116,117,118]. In practice, researchers are advised to avoid individuals with risk factors for TMS-provoked seizures, which include mediations or medical conditions known to lower seizure threshold and history of epilepsy [119]. While tDCS can be administered remotely, which increases accessibility, rTMS is hindered by its size and expense. The use of the device is limited to trained practitioners rather than individuals for administration. Similar to tDCS, TMS lacks good spatial resolution, which can diffuse the cortical effects of the stimulation [120,121]. The effect of rTMS has been investigated in a number of post-stroke cognitive impairment cases, most commonly motor impairment and language. In the assessment of global cognitive function, as measured by the Mini-Mental Status Examination (MMSE) or the Montreal Cognitive Assessment (MoCA), active rTMS targeting the left DLPFC was associated with improved performance compared to sham [122], with evidence for positive effects in post-stroke language and motor impairments as well [123].
Another form of neuromodulation, transcranial focused ultrasound (TUS), delivers highly focused acoustic energy at low frequency [124]. TUS has the advantage of greater spatial resolution than other forms of non-invasive brain stimulation [125,126]. Transcranial focused ultrasound can open the blood–brain barrier [127] and when applied in short bursts, can modulate cortical neuron excitability [128]. The study of transcranial ultrasound stimulation has largely focused on its applications of clot dissolution and delivery of therapeutic drugs to specific brain regions [129], but it is a promising intervention to ameliorate post-stroke cognitive impairments needing further research.

4. tDCS Therapy in Stroke Patient Populations

The effect of tDCS on cognition has been studied in several non-stroke patient populations, including those with normal cognition, neurodegenerative disease, and traumatic brain injury [130,131]. In healthy adults, tDCS can improve performance in the learned tasks [132] and result in enhanced functional connectivity of working memory networks on fMRI evaluation [133]. In those with mild cognitive impairment or Alzheimer’s disease (AD), tDCS seems to have an enhanced effect in participants with mild cognitive impairment or AD compared to healthy controls [134]. In demyelinating disorders, like multiple sclerosis, neuromodulation has also shown benefits in cognitive performance and fatigue when paired with cognitive training for multiple sclerosis patients [135,136]. Additionally, when evaluated in participants with cognitive and behavioral changes secondary to traumatic brain injury, tDCS with stimulation to the DLPFC resulted in improvements in overall cognition as well as psychosocial wellbeing [137,138].

4.1. tDCS for Cognitive Performance in Stroke: Language

Methods of tDCS use to improve cognitive performance have varied in the stroke patient population based on the domain targeted. Aphasia, an impairment in language production and comprehension, can be caused by injury to the language-dominant hemisphere of the brain, most commonly the left. Studies of tDCS to improve language production in persons with chronic left hemisphere stroke and non-fluent aphasia have revealed positive effects associated with stimulation [139,140,141,142]. Though functional MRI data suggest that recovery is associated with restoration of the left hemispheric residual language network [143], other studies suggest that language recovery relies on an intact right hemisphere [144,145]. Therefore, there is no consensus on the optimal target for tDCS for aphasia-related impairments. The majority of clinical trials have focused on excitatory stimulation to the left hemispheric sites of language function, like the left inferior frontal gyrus or the left posterior perisylvian region, to equilibrate the interhemispheric inhibition proposed in the previously described model. To date, the largest randomized, double-blind, sham-controlled trial in this area included 74 participants with chronic non-fluent post-stroke aphasia with the stimulation target determined by pre-treatment functional MRI, in which the anode was directed over the area of greatest activation in the left hemisphere during spoken naming over the course of 15 language therapy sessions. Active tDCS resulted in a greater change in correct picture naming and in the correct naming relative to sham, with the benefit maintained for at least 3 weeks [146]. Another smaller study investigated the long-term outcome of tDCS to the left frontal (perilesional) region while participating in simultaneous naming training for 10 sessions, resulting in improvements in naming that persisted until 16 weeks post-intervention [147]. Anodal stimulation over the left inferior frontal gyrus, a site that mediates language control corresponding to Broca’s area, combined with repetition tasks, has been used in over 19 studies of post-stroke aphasia and resulted in improved accuracy in speech production and naming [148,149], speech production [150], and reading ability [151,152].
Though the left hemisphere is the most common stimulation side, other regions like the right hemisphere and the cerebellum have also been investigated for treatment of post-stroke aphasia. The effect of stimulation of the opposite hemisphere, the contralesional right hemisphere, builds on the bimodal balance model, in which cathodal (inhibitory) stimulation targets the source of compensatory activity in the contralesional hemisphere. In a randomized controlled crossover study, 10 individuals with naming impairments due to post-stroke aphasia underwent 5 days of 2 mA cathodal stimulation over the right Broca’s homologue area or sham stimulation for 20 min with simultaneous daily sessions of conventional word-retrieval training [152]. Cathodal stimulation resulted in improved naming on the Boston Naming Test post-intervention (after the final session), but durability of this result beyond the immediate period was note assessed. A meta-analysis of cathodal tDCS to the right hemisphere for post-stroke aphasia found three studies, including Kang et al. above, and pooled analysis showed no statistically significant difference between cathodal stimulation and sham stimulation [153]. Another potential site of stimulation is the right cerebellum because of its role in word retrieval and generation, somatic processing, and language learning [154]. The cerebellum, while involved in language pathways, remains intact in individuals with isolated left hemisphere lesions, therefore representing an intact gateway to the affected neural networks [155]. Hillis et al. have investigated anodal stimulation to the right posterolateral cerebellum in combination with computerized aphasia treatment in a double-blinded, within-subject crossover randomized controlled trial, in which participants undergo sham stimulation along with either anodal or cathodal (2 mA) stimulation for 15 sessions of 20 min each [156]. Active stimulation resulted in greater improvements in naming accuracy compared to sham immediately post-intervention and at 2 months post-intervention, with the cathodal stimulation showing larger (though not statistically significant) gains than anodal stimulation. The mechanism of this polarity-independent effect is not clear but may be the result of the complexity of gyral folding in the cerebellar cortex such that the structure orientation relative to the electric field direction may differ even within small regions of the cerebellum [157].

4.2. tDCS for Cognitive Performance in Stroke: Attention

In addition to aphasia, the largest body of work in post-stroke cognitive impairment and tDCS focuses on a common form of inattention, neglect. Neglect, the inability to perceive, report, and orient to sensory events towards one side of space, is typically caused by injury to the non-dominant (right) hemisphere in the parietal cortex of the brain, resulting in impairments affecting the left side. Though damage to several areas of this hemisphere can produce neglect, cortical regions of the right inferior parietal lobe or temporo-parietal junction have classically been implicated in neglect [158]. Recovery studies involving tDCS to ameliorate neglect have used the right posterior parietal cortex (PPC) as the primary target for anodal stimulation, likely because of its role in visuospatial attention, sustained attention, and spatial perception [159]. A recent systematic review found 11 studies (152 participants with stroke-related impairments) with the tDCS intervention ranging from 1 to 20 sessions and intensity ranging from 1 to 2 mA [83]. Out of the 11 studies, 9 applied anodal stimulation to the ipsilesional right PPC, with 5 of those applying cathodal stimulation to the contralesional PPC. Two studies stimulated the right primary motor cortex. All studies reported improvements in measures of neglect, including line bisection and cancellation tests in the active stimulation groups compared to sham stimulation; however, the strength and generalizability of these findings are unclear given the heterogenous study designs.
Several studies in neglect-related impairments have involved dual stimulation, in which the anodal is placed on the ispilesional PPC and the cathode is placed on the contralesional hemisphere to disrupt the inhibitory imbalance that occurs during spontaneous recovery [160,161]. A double-blind random-order cross-over experiment investigated dual tDCS in chronic stroke participants, with spatial neglect evaluated in three randomly arranged tDCS sessions of 1 mA for 20 min (anodal tDCS over the right PPC and cathodal tDCS over the left PPC, anodal tDCS over the right PPC and cathode over the left supraorbital area, and sham mode). Significant changes in line bisection test and star cancelation test performance were noted in both stimulation conditions compared to sham, with dual tDCS having a stronger effect than single tDCS [162]. A similar study tested tDCS in three similar modes (anodal tDCS over the right PPC, cathodal tDCS over the left PPC, or sham tDCS) at 2 mA for 30 min over 15 sessions, but dual stimulation was not applied. Though each group experienced improvements in the line bisection test, the star cancellation test, and the motor-free visual perception test, there was no statistically significant difference among groups [160]. A double-blinded, pilot randomized clinical trial enrolled participants with subacute stroke (within 6 months of enrollment) of 1 mA for 20 min for a total of 15 sessions in three treatment conditions: anodal tDCS over the right PPC, cathodal tDCS over the left PPC, or sham tDCS. Post-tDCS session, participants received 1 h of physical therapy involving visual scanning and task-specific activities. Anodal tDCS was associated with better performance on tests of neglect than sham, but there were no statistically significant differences between anodal and cathodal stimulation [161].
The primary motor cortex is another stimulation site that has been investigated in neuromodulation of neglect. Because the primary motor cortex mediates early consolidation of motor memories and movement dynamics [163], anodal stimulation of this region may facilitate the realignment of hand-eye coordination leftward to consolidate learning from during prism adaptation, a form of behavioral therapy [164].

4.3. tDCS for Cognitive Performance in Stroke: Other Domains of Cognition

4.3.1. Global Cognition

Unlike the consistency of stimulation targets and outcome measures in neuromodulatory studies of neglect and aphasia, studies of neuromodulation to target other domains of cognition are varied. Because cognitive function is conveyed through diffuse, interconnected, bilateral networks, there are no clear stimulation targets; however, several have been tested [165,166]. A recent study of tDCS included 26 patients with chronic stroke and post-stroke cognitive impairment who underwent 20 sessions of either tDCS plus computerized cognitive training or sham tDCS plus computerized cognitive training. Participants were trained to self-administer (or a caregiver administered) tDCS for 5 sessions per week for 4 weeks, with each session lasting 30 min. Participants undergoing active stimulation of the DLPFC showed significant improvements in the Korean version of the MoCA (p = 0.004 for real group versus p = 0.132 for sham group), more notable in individuals with lower pre-intervention MoCA scores and those with left hemispheric lesions [78].
A recent meta-analysis found 11 randomized controlled trials published in English and Chinese journals, all occurring within the last 10 years [23]. Overall, treatment with anodal tDCS was more likely to result in improved cognitive performance (p < 0.001) at the end of follow-up. Improvement appears to be more pronounced when stimulation was administered < 40 days from incident stroke.

4.3.2. Memory and Verbal Learning

Neuroimaging studies have identified regions of the prefrontal and parietal cortices as key regions for verbal learning and working memory [167]. One trial of 10 participants with chronic stroke stimulated the left dorsolateral prefrontal cortex for 2 sessions; and compared to sham stimulation, the tDCS group showed significant changes in working memory recognition accuracy and response time [168]. A similar study included 11 patients with chronic stroke who underwent stimulation 5 times per week during their inpatient rehabilitation hospitalization while participating in computer-assisted cognitive exercises. Anodal stimulation was to the bilateral prefrontal cortex and the cathode to the non-dominant arm for 30 min at an intensity of 2 mA; the intervention group demonstrating significant improvements in auditory and visual continuous performance tests compared to sham [169]. Another study involved anodal or sham tDCS to the left temporoparietal area to enhance audiovisual memory in 12 stroke patients. There was marked improvement in the number of correctly recalled words across 5 trials on Rey Auditory Verbal Learning in the tDCS group compared to sham [165]. In Yun et al. [166], the anterior temporal lobe, a key hub for semantic memory [170], was the focus of stimulation. Forty-five participants with a history of stroke were randomized to anodal stimulation of the left anterior temporal lobe, the right anterior temporal lobe, or sham stimulation with intervention delivered for 30 min for 15 sessions. There was greater improvement seen in the left anodal stimulation group compared to the two other groups across multiple cognitive assessments but only a statistically significant difference in the verbal learning test-delayed recall (p < 0.05) [131].

4.3.3. Executive Function

Executive function, a group of cognitive processes related to self-regulation, planning, and reasoning for goal-directed behavior [22], is thought to be regulated by the frontal (particularly DLPFC) [171] and the temporal lobes [172]. As with working-memory-related impairments, tDCS for executive-function-related impairments have included several studies targeting the left DLPFC. A meta-analysis of 27 studies that targeted the DLPFC found substantial heterogeneity in elements of the study design: duration (10–30 min), training task (e.g., N-back, Flanker task, Affective Flexibility, etc.), outcome measures, intensity (1–2 mA), and timing (during or after stimulation) [173]. In pooled analyses, anodal, cathodal, or bilateral tDCS did not yield a significant effect on general executive function. However, in evaluation of subdomains of executive function, there was a significant effect of anodal tDCS on updating tasks but not inhibiting or shifting. Other tDCS studies focused on executive function have also targeted the superior temporal gyrus. In Hosseinzadeh et al. [174], 100 participants with chronic ischemic stroke were divided into 4 groups (anodal tDCS to the left superior temporal gyrus, cathodal tDCS to the left superior temporal gyrus, and anode at the contralateral supraorbital region, sham, and control), where participants underwent 3 sessions of 30 min per week for 1 month. The group undergoing anodal tDCS to the left superior temporal gyrus showed significant increases in the Trail Making Test, a measure of executive function, after 1 and 3 months post-stimulation when compared to the three other groups, suggesting durability of the intervention effect.

4.3.4. Perceptual Motor Function

Perceptual motor function involves the acquisition of a motor skill without the awareness of learning using the interpretation of sensory information [175]. Perceptual disorders, though they affect almost 75% of stroke survivors [176], often go undetected. Though many studies have focused on motor strength improvement with tDCS, few studies have focused specifically on perceptual motor function and tDCS, but several have stimulated the ipsilesional primary motor cortex paired with motor exercises of the affected limb [177]. Anodal tDCS may enhance induction for consolidation, but its effects on long-term retention are still unknown [178].

4.3.5. Social Cognition

Social cognition, a domain of cognition, refers to one’s ability to process, judge, and apply information about social behaviors, including that of oneself and others [179], and is mediated by the right temporal parietal junction (TPJ) [180]. Studies of tDCS in stroke patient populations have not focused on amelioration of social cognition, but studies from healthy controls may provide insight into the potential effects in individuals with post-stroke cognitive impairment. One study of healthy controls evaluated anodal, cathodal, or sham tDCS to the right TPJ at 1 mA for 20 min [181]. Anodal stimulation was associated with enhanced imitation control and visual perspective taking, which are components of social cognition, but did not affect attribution of mental states. Conversely, cathodal stimulation of the right TPJ with 1.5 mA current for 20 min was associated with decreased cognitive empathy, further supporting evidence that cortical excitability in this region may be an important stimulation target for impairments in social cognition among individuals with stroke and other acquired brain injuries [182].

4.4. Considerations Unique to Post-Stroke Patient Populations

4.4.1. Medications Acting on the Central Nervous System

Based on the mechanism of action of tDCS, medications acting on the central nervous system can alter the effects of stimulation, including ones commonly used or prescribed during the care of stroke patients, like nicotine, amphetamines, and selective serotonin reuptake inhibitors (SSRIs). Nicotine administration in non-smokers was found to dampen the excitability typically enhanced under anodal tDCS and diminish the excitability, reducing aftereffects under the cathodal tDCS [183,184]. Though the mechanism is not clearly defined, based on animal models, nicotine modulates nicotinic acetylcholine receptors to elicit neuronal depolarization at presynaptic sites, thus modulating glutamate and dopamine release [185,186]. SSRIs are commonly prescribed in individuals with stroke-related hemiparesis to promote motor recovery or to treat post-stroke depression or pain [187]. With anodal stimulation, citalopram can amplify and prolong tDCS-induced excitability after even a single dose, with the effect thought to be NMDA-receptor dependent [188,189]. A similar medication class, selective norepinephrine reuptake inhibitor (SNRI), has also demonstrated enhanced excitability with anodal tDCS [190]. Chronic use of both SSRIs and SNRIs with cathodal stimulation converts the expected long-term depression-like plasticity into facilitation [188,190]. Serotonin can impact the influx of calcium ions through NMDA receptors to enhance neuronal depolarization [191]. Similar effects are seen in amphetamines, which is an NMDA receptor antagonist [192]. Medications blocking dopamine receptors reduced both the typical anodal excitability enhancement and the cathodal excitability reduction [193]. Conversely, administration of a D2 agonist has been shown to eliminate the aftereffects of tDCS under both the anode and cathode [194]. D2/D3 agonists, such as ropinirole, have been demonstrated to reverse the anodal aftereffects of tDCS at low dosages but have no effect with medium doses; at high dosages, ropinirole decreased the magnitude of anodal stimulation aftereffects [193].

4.4.2. Chronicity of Intervention

After an ischemic stroke, destruction of neural networks stimulates network reorganization following injury [195]. The mechanisms involve the activation of cell repair and genesis and the unmasking of pre-existing subthreshold connections or the creation of new connections [196,197]. In cognitive networks, the interactions are interhemispheric, and post-stroke dynamic functional connectivity was significantly increased in the bilateral prefrontal cortices immediately after stroke [198]. Indeed, synaptic connectivity related to complex functions like memory or cognition are distributed throughout the cortex [199]. Human and animal studies have revealed a period of heightened neuroplasticity, often referred to as a “sensitive period”, that ends in the first month after stroke in rodent models [200] and in the first 3 months after stroke in humans [201,202]. During this time, dynamic molecular, electrophysiologic, and structural reorganization occurs [196,201]. Cortical circuit remapping in the peri-infarct region is activity dependent aided by underlying synaptic plasticity, which ensures neurons receive adequate synaptic input [203] and redistribute synaptic strength through neural circuits [196,204]. These changes on a neural level are also reflected clinically; the most spontaneous recovery after stroke is also observed clinically during this sensitive period [205,206].
Thus, neuromodulation through tDCS may augment spontaneous neuroplastic processes occurring during the subacute period after stroke. Further, post-stroke rehabilitation studies indicate that delayed initiation of therapies are associated with worse functional outcomes and longer hospital length of stays [207]. However, the majority of stroke recovery studies, including those involving neuromodulation, involve intervention in the chronic stage, where spontaneous recovery is thought to be static. A recent meta-analysis found that the effects of tDCS were more pronounced when performed <40 days after stroke compared to 40 days or more after stroke: Χ2 = 5.11 and p = 0.02 [23]. There were only 4 studies involving a total of 233 participants that evaluated tDCS in this early subacute period, and there have been no studies directly comparing neuronal effects of tDCS across different timepoints after stroke. Our knowledge of the underlying neuroplastic processes occurring during the first 3 months post-injury suggests that the timing for optimal impact of an intervention, especially one like tDCS that modulates plasticity, may be in the early subacute period.

4.4.3. Neurovascular Effects

In addition to its effect on synaptic plasticity, tDCS can modulate changes in cerebral perfusion. Both anodal and cathodal stimulation increase cerebral blood flow with the magnitude in healthy adults [208]. In animal models of stroke, cathodal tDCS can restore perfusion in ischemic tissue at risk of infarction through several mechanisms, including collateral perfusion enhancement by inducing vasodilation [209]. Direct vascular response to electrical current originates from the activation of perivascular nerves evoked by electrical current and a robust neurovascular response [210]. In studies involving humans, cerebrovascular vasomotor reactivity, the vasodilatory capacity of cerebral vasculature in response to internal molecular signals, was reduced with anodal stimulation and amplified with cathodal stimulation [211]. Pilot studies of tDCS in acute ischemic stroke have shown that cathodal stimulation is associated with increased cerebral blood volume and perfusion through induction of vasodilation [212]. In addition, cathodal stimulation is also postulated to provide direct neuroprotection by inhibiting peri-lesional excitotoxic and inflammatory effects [209]. These findings—cathodal stimulation can have a neuroprotectant effect through the reduction in excitotoxic responses and through increased cerebral perfusion—may appear to contradict the interhemispheric inhibition and bimodal balances models, which would suggest cathodal stimulation of the lesioned region would be detrimental. These models account for neuroplasticity-related changes that occur in the post-acute period after stroke but not the immediate post-injury period, in which a lack of perfusion (e.g., vessel occlusion) causes ischemic injury. The perfusional benefits of cathodal stimulation may provide benefits in the hyperacute period by restoring cerebral blood flow to hypoperfused areas and reducing the apoptotic and inflammatory cascades during neuronal cell death. As such, tDCS may be a promising direction for acute stroke management to alleviate hypoperfused ischemic regions.

5. Conclusions

Though the consistency of study designs has been variable, tDCS appears to be a promising adjunct to cognitive therapy. Patients typically receive the most intensive cognitive therapy during the acute to subacute stages after stroke, and based on previous research on cognitive recovery, the greatest improvement from post-stroke cognitive impairment occurs within the first 6 months [213]. Therefore, it is reasonable that the effects of tDCS would be enriched if delivered during this time period. In the stroke patient population, published studies of tDCS for cognitive recovery are limited by small sample sizes and inconsistent study design, and randomized controlled trials are needed to assess efficacy.
The body of literature currently available is limited by small study samples, heterogenous study designs, and limited long-term follow up. Further research is indicated to determine several key factors to optimize tDCS as an intervention for post-stroke cognitive impairment. Larger, well-designed trials are needed to assess not only the granular effects of tDCS (i.e., improvement on an individual cognitive task) but also generalizability to other functional outcomes. In addition, stimulation parameters need to be identified in order to standardize intervention protocols, which will facilitate pooled analyses across studies. This method of intervention may be personalized based on clinical characteristics and neuroimaging factors, but further research is needed to determine which variables predict responsiveness to tDCS. And equally importantly, research is needed to establish if neuromodulation combined with cognitive therapy affects functional outcomes in ways which are meaningful to individual circumstances and needs, central to the concept of patient-centered care; the implementation of a therapy that could speed the rate of recovery of cognitive skills and may also impact quality of life for individuals with stroke.

Author Contributions

Conceptualization, R.H.H. and K.L.S.; methodology, R.H.H. and K.L.S.; investigation, R.H.H. and K.L.S.; data curation, R.H.H. and K.L.S.; writing—original draft preparation, K.L.S.; writing—review and editing, R.H.H. and K.L.S.; visualization, R.H.H. and K.L.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Benjamin, E.J.; Blaha, M.J.; Chiuve, S.E.; Cushman, M.; Das, S.R.; Deo, R.; De Ferranti, S.D.; Floyd, J.; Fornage, M.; Gillespie, C.; et al. Heart Disease and Stroke Statistics’ 2017 Update: A Report from the American Heart Association. Circulation 2017, 135, e146–e603. [Google Scholar] [CrossRef]
  2. Nys, G.M.S.; van Zandvoort, M.J.E.; de Kort, P.L.M.; Jansen, B.P.W.; de Haan, E.H.F.; Kappelle, L.J. Cognitive Disorders in Acute Stroke: Prevalence and Clinical Determinants. Cerebrovasc. Dis. 2007, 23, 408–416. [Google Scholar] [CrossRef]
  3. Jaillard, A.; Naegele, B.; Trabucco-Miguel, S.; LeBas, J.F.; Hommel, M. Hidden Dysfunctioning in Subacute Stroke. Stroke 2009, 40, 2473–2479. [Google Scholar] [CrossRef]
  4. Allan, L.M.; Rowan, E.N.; Firbank, M.J.; Thomas, A.J.; Parry, S.W.; Polvikoski, T.M.; O’Brien, J.T.; Kalaria, R.N. Long Term Incidence of Dementia, Predictors of Mortality and Pathological Diagnosis in Older Stroke Survivors. Brain 2011, 134, 3716–3727. [Google Scholar] [CrossRef]
  5. Levine, D.A.; Wadley, V.G.; Langa, K.M.; Unverzagt, F.W.; Kabeto, M.U.; Giordani, B.; Howard, G.; Howard, V.J.; Cushman, M.; Judd, S.E.; et al. Risk Factors for Poststroke Cognitive Decline: The REGARDS Study (Reasons for Geographic and Racial Differences in Stroke). Stroke 2018, 49, 987–994. [Google Scholar]
  6. Gottesman, R.F.; Hillis, A.E. Predictors and Assessment of Cognitive Dysfunction Resulting from Ischaemic Stroke. Lancet Neurol. 2010, 9, 895–905. [Google Scholar] [CrossRef]
  7. Zietemann, V.; Georgakis, M.K.; Dondaine, T.; Muller, C.; Mendyk, A.M.; Kopczak, A.; Hénon, H.; Bombois, S.; Wollenweber, F.A.; Bordet, R.; et al. Early MoCA Predicts Long-Term Cognitive and Functional Outcome and Mortality after Stroke. Neurology 2018, 91, E1838–E1850. [Google Scholar] [CrossRef]
  8. Wu, C.-Y.; Skidmore, E.R.; Rodakowski, J. Relationship Consensus and Caregiver Burden in Adults with Cognitive Impairments 6 Months Following Stroke. PM&R 2019, 11, 597–603. [Google Scholar] [CrossRef]
  9. Jokinen, H.; Melkas, S.; Ylikoski, R.; Pohjasvaara, T.; Kaste, M.; Erkinjuntti, T.; Hietanen, M. Post-Stroke Cognitive Impairment Is Common Even after Successful Clinical Recovery. Eur. J. Neurol. 2015, 22, 1288–1294. [Google Scholar] [CrossRef]
  10. Lo, J.W.; Crawford, J.D.; Desmond, D.W.; Godefroy, O.; Jokinen, H.; Mahinrad, S.; Bae, H.J.; Lim, J.S.; Köhler, S.; Douven, E.; et al. Profile of and Risk Factors for Poststroke Cognitive Impairment in Diverse Ethnoregional Groups. Neurology 2019, 93, E2257–E2271. [Google Scholar] [CrossRef]
  11. Hyndman, D.; Ashburn, A. People with Stroke Living in the Community: Attention Deficits, Balance, ADL Ability and Falls. Disabil. Rehabil. 2003, 25, 817–822. [Google Scholar] [CrossRef]
  12. Hyndman, D.; Ashburn, A.; Stack, E. Fall Events among People with Stroke Living in the Community: Circumstances of Falls and Characteristics of Fallers. Arch. Phys. Med. Rehabil. 2002, 83, 165–170. [Google Scholar] [CrossRef]
  13. Elgh, E.; Hu, X. Dynamic Trajectory of Long-Term Cognitive Improvement up to 10 Years in Young Community-Dwelling Stroke Survivors: A Cohort Study. Front. Neurol. 2019, 10, 97. [Google Scholar] [CrossRef]
  14. McDowd, J.M.; Filion, D.L.; Pohl, P.S.; Richards, L.G.; Stiers, W. Attentional Abilities and Functional Outcomes Following Stroke. J. Gerontol.—Ser. B Psychol. Sci. Soc. Sci. 2003, 58, 45–53. [Google Scholar] [CrossRef]
  15. das Nair, R.; Cogger, H.; Worthington, E.; Lincoln, N. Cognitive Rehabilitation for Memory Deficits after Stroke. Cochrane Database Syst. Rev. 2016, 2016, CD002293. [Google Scholar] [CrossRef]
  16. Bhogal, S.K.; Teasell, R.; Speechley, M. Intensity of Aphasia Therapy, Impact on Recovery. Stroke 2003, 34, 987–992. [Google Scholar] [CrossRef]
  17. Narasimhalu, K.; Effendy, S.; Sim, C.H.; Lee, J.M.; Chen, I.; Hia, S.B.; Xue, H.L.; Corrales, M.P.; Chang, H.M.; Wong, M.C.; et al. A Randomized Controlled Trial of Rivastigmine in Patients with Cognitive Impairment No Dementia Because of Cerebrovascular Disease. Acta Neurol. Scand. 2010, 121, 217–224. [Google Scholar] [CrossRef]
  18. Iso-Markku, P.; Kujala, U.M.; Knittle, K.; Polet, J.; Vuoksimaa, E.; Waller, K. Physical Activity as a Protective Factor for Dementia and Alzheimer’s Disease: Systematic Review, Meta-Analysis and Quality Assessment of Cohort and Case–Control Studies. Br. J. Sports Med. 2022, 56, 701–709. [Google Scholar] [CrossRef]
  19. Dao, E.; Barha, C.K.; Zou, J.; Wei, N.; Liu-Ambrose, T. Prevention of Vascular Contributions to Cognitive Impairment and Dementia: The Role of Physical Activity and Exercise. Stroke 2024, 55, 812–821. [Google Scholar] [CrossRef]
  20. Moore, S.A.; Hallsworth, K.; Jakovljevic, D.G.; Blamire, A.M.; He, J.; Ford, G.A.; Rochester, L.; Trenell, M.I. Effects of Community Exercise Therapy on Metabolic, Brain, Physical, and Cognitive Function Following Stroke: A Randomized Controlled Pilot Trial. Neurorehabil. Neural Repair 2015, 29, 623–635. [Google Scholar] [CrossRef]
  21. Cicerone, K.D.; Langenbahn, D.M.; Braden, C.; Malec, J.F.; Kalmar, K.; Fraas, M.; Felicetti, T.; Laatsch, L.; Harley, J.P.; Bergquist, T.; et al. Evidence-Based Cognitive Rehabilitation: Updated Review of the Literature from 2003 through 2008. Arch. Phys. Med. Rehabil. 2011, 92, 519–530. [Google Scholar] [CrossRef]
  22. Cicerone, K.D.; Dahlberg, C.; Kalmar, K.; Langenbahn, D.M.; Malec, J.F.; Bergquist, T.F.; Felicetti, T.; Giacino, J.T.; Harley, J.P.; Harrington, D.E.; et al. Evidence-Based Cognitive Rehabilitation: Recommendations for Clinical Practice. Arch. Phys. Med. Rehabil. 2000, 81, 1596–1615. [Google Scholar] [CrossRef]
  23. Yan, R.B.; Zhang, X.L.; Li, Y.H.; Hou, J.M.; Chen, H.; Liu, H.L. Effect of Transcranial Direct-Current Stimulation on Cognitive Function in Stroke Patients: A Systematic Review and Meta-Analysis. PLoS ONE 2020, 15, e0233903. [Google Scholar] [CrossRef]
  24. Biou, E.; Cassoudesalle, H.; Cogné, M.; Sibon, I.; De Gabory, I.; Dehail, P.; Aupy, J.; Glize, B. Transcranial Direct Current Stimulation in Post-Stroke Aphasia Rehabilitation: A Systematic Review. Ann. Phys. Rehabil. Med. 2019, 62, 104–121. [Google Scholar] [CrossRef]
  25. Ferrucci, R.; Mameli, F.; Guidi, I.; Mrakic-Sposta, S.; Vergari, M.; Marceglia, S.; Cogiamanian, F.; Barbieri, S.; Scarpini, E.; Priori, A. Transcranial Direct Current Stimulation Improves Recognition Memory in Alzheimer Disease. Neurology 2008, 71, 493–498. [Google Scholar] [CrossRef]
  26. Norise, C.; Sacchetti, D.; Hamilton, R. Transcranial Direct Current Stimulation in Post-Stroke Chronic Aphasia: The Impact of Baseline Severity and Task Specificity in a Pilot Sample. Front. Hum. Neurosci. 2017, 11, 260. [Google Scholar] [CrossRef]
  27. Dubreuil-Vall, L.; Chau, P.; Ruffini, G.; Widge, A.S.; Camprodon, J.A. TDCS to the Left DLPFC Modulates Cognitive and Physiological Correlates of Executive Function in a State-Dependent Manner. Brain Stimul. 2019, 12, 1456–1463. [Google Scholar] [CrossRef]
  28. Mesulam, M.-M. Large-Scale Neurocognitive Networks and Distributed Processing for Attention, Language, and Memory. Ann. Neurol. 1990, 28, 597–613. [Google Scholar] [CrossRef]
  29. Stolwyk, R.J.; Mihaljcic, T.; Wong, D.K.; Chapman, J.E.; Rogers, J.M. Poststroke Cognitive Impairment Negatively Impacts Activity and Participation Outcomes. Stroke 2021, 52, 748–760. [Google Scholar] [CrossRef]
  30. Blackburn, D.J.; Bafadhel, L.; Randall, M.; Harkness, K.A. Cognitive Screening in the Acute Stroke Setting. Age Ageing 2013, 42, 113–116. [Google Scholar] [CrossRef]
  31. Aam, S.; Einstad, M.S.; Munthe-Kaas, R.; Lydersen, S.; Ihle-Hansen, H.; Knapskog, A.B.; Ellekjær, H.; Seljeseth, Y.; Saltvedt, I. Post-Stroke Cognitive Impairment—Impact of Follow-Up Time and Stroke Subtype on Severity and Cognitive Profile: The Nor-COAST Study. Front. Neurol. 2020, 11, 551524. [Google Scholar]
  32. El Husseini, N.; Katzan, I.L.; Rost, N.S.; Blake, M.L.; Byun, E.; Pendlebury, S.T.; Aparicio, H.J.; Marquine, M.J.; Gottesman, R.F.; Smith, E.E. Cognitive Impairment After Ischemic and Hemorrhagic Stroke: A Scientific Statement from the American Heart Association/American Stroke Association. Stroke 2023, 54, E272–E291. [Google Scholar] [CrossRef]
  33. Stephens, S.; Kenny, R.A.; Rowan, E.; Allan, L.; Kalaria, R.N.; Bradbury, M.; Ballard, C.G. Neuropsychological Characteristics of Mild Vascular Cognitive Impairment and Dementia after Stroke. Int. J. Geriatr. Psychiatry 2004, 19, 1053–1057. [Google Scholar] [CrossRef]
  34. Lees, R.; Fearon, P.; Harrison, J.K.; Broomfield, N.M.; Quinn, T.J. Cognitive and Mood Assessment in Stroke Research: Focused Review of Contemporary Studies. Stroke 2012, 43, 1678–1680. [Google Scholar] [CrossRef]
  35. Pendlebury, S.T.; Rothwell, P.M. Prevalence, Incidence, and Factors Associated with Pre-Stroke and Post-Stroke Dementia: A Systematic Review and Meta-Analysis. Lancet Neurol. 2009, 8, 1006–1018. [Google Scholar] [CrossRef]
  36. Douiri, A.; Rudd, A.G.; Wolfe, C.D.A. Prevalence of Poststroke Cognitive Impairment: South London Stroke Register 1995–2010. Stroke 2013, 44, 138–145. [Google Scholar] [CrossRef]
  37. Corbetta, M.; Ramsey, L.; Callejas, A.; Baldassarre, A.; Hacker, C.D.; Siegel, J.S.; Astafiev, S.V.; Rengachary, J.; Zinn, K.; Lang, C.E.; et al. Common Behavioral Clusters and Subcortical Anatomy in Stroke. Neuron 2015, 85, 927–941. [Google Scholar] [CrossRef]
  38. Puy, L.; Barbay, M.; Roussel, M.; Canaple, S.; Lamy, C.; Arnoux, A.; Leclercq, C.; Mas, J.L.; Tasseel-Ponche, S.; Constans, J.M.; et al. Neuroimaging Determinants of Poststroke Cognitive Performance: The GRECogVASC Study. Stroke 2018, 49, 2666–2673. [Google Scholar] [CrossRef]
  39. Mullick, A.A.; Subramanian, S.K.; Levin, M.F. Emerging Evidence of the Association between Cognitive Deficits and Arm Motor Recovery after Stroke: A Meta-Analysis. Restor. Neurol. Neurosci. 2015, 33, 389–403. [Google Scholar] [CrossRef]
  40. Dignam, J.; Copland, D.; O’Brien, K.; Burfein, P.; Khan, A.; Rodriguez, A.D. Influence of Cognitive Ability on Therapy Outcomes for Anomia in Adults with Chronic Poststroke Aphasia. J. Speech Lang. Hear. Res. 2017, 60, 406–421. [Google Scholar]
  41. Barker-Collo, S.; Feigin, V.; Feigin, V. The Impact of Neuropsychological Deficits on Functional Stroke Outcomes. Neuropsychol. Rev. 2006, 16, 53–64. [Google Scholar] [CrossRef]
  42. Gilmore, N.; Meier, E.L.; Johnson, J.P.; Kiran, S. Nonlinguistic Cognitive Factors Predict Treatment-Induced Recovery in Chronic Poststroke Aphasia. Arch. Phys. Med. Rehabil. 2019, 100, 1251–1258. [Google Scholar] [CrossRef]
  43. Hyndman, D.; Pickering, R.M.; Ashburn, A. The Influence of Attention Deficits on Functional Recovery Post Stroke during the First 12 Months after Discharge from Hospital. J. Neurol. Neurosurg. Psychiatry 2008, 79, 656–663. [Google Scholar] [CrossRef]
  44. Cirstea, M.C.; Levin, M.F. Improvement of Arm Movement Patterns and Endpoint Control Depends on Type of Feedback during Practice in Stroke Survivors. Neurorehabil. Neural Repair 2007, 21, 398–411. [Google Scholar] [CrossRef]
  45. Chen, C.; Leys, D.; Esquenazi, A. The Interaction between Neuropsychological and Motor Deficits in Patients after Stroke. Neurology 2013, 80, S27–S34. [Google Scholar] [CrossRef]
  46. Einstad, M.S.; Saltvedt, I.; Lydersen, S.; Ursin, M.H.; Munthe-Kaas, R.; Ihle-Hansen, H.; Knapskog, A.B.; Askim, T.; Beyer, M.K.; Næss, H.; et al. Associations between Post-Stroke Motor and Cognitive Function: A Cross-Sectional Study. BMC Geriatr. 2021, 21, 1–10. [Google Scholar] [CrossRef]
  47. Cramer, S.C. Improving Outcomes after Stroke by LEAPS (Locomotor Experience Applied Post-Stroke) and Bounds. Stroke 2011, 42, 3659–3660. [Google Scholar] [CrossRef]
  48. Rice, J.; Corp, D.T.; Swarowsky, A.; Cahalin, L.P.; Cabral, D.F.; Nunez, C.; Koch, S.; Rundek, T.; Gomes-Osman, J. Greater Cognitive-Motor Interference in Individuals Post-Stroke during More Complex Motor Tasks. J. Neurol. Phys. Ther. 2022, 46, 26–33. [Google Scholar] [CrossRef]
  49. Yagi, M.; Yasunaga, H.; Matsui, H.; Morita, K.; Fushimi, K.; Fujimoto, M.; Koyama, T.; Fujitani, J. Impact of Rehabilitation on Outcomes in Patients with Ischemic Stroke: A Nationwide Retrospective Cohort Study in Japan. Stroke 2017, 48, 740–746. [Google Scholar] [CrossRef]
  50. Cogan, A.M.; Weaver, J.A.; Davidson, L.F.; Khromouchkine, N.; Mallinson, T. Association of Therapy Time and Cognitive Recovery in Stroke Patients in Post-Acute Rehabilitation. J. Am. Med. Dir. Assoc. 2021, 22, 453–458.e3. [Google Scholar] [CrossRef]
  51. West, T.; Bernhardt, J. Physical Activity in Hospitalised Stroke Patients. Stroke Res. Treat. 2012, 2012, 13. [Google Scholar] [CrossRef]
  52. Dorsch, A.; Thomas, S.; Xu, X.; Kaiser, W.; Dobkin, B.H. SIRRACT: An International Randomized Clinical Trial of Activity Feedback During Inpatient Stroke Rehabilitation Enabled by Wireless Sensing. Neurorehabil. Neural Repair 2015, 29, 407–415. [Google Scholar] [CrossRef]
  53. Skolarus, L.E.; Burke, J.F.; Morgenstern, L.B.; Meurer, W.J.; Adelman, E.E.; Kerber, K.A.; Callaghan, B.C.; Lisabeth, L.D. Impact of State Medicaid Coverage on Utilization of Inpatient Rehabilitation Facilities among Patients with Stroke. Stroke 2014, 45, 2472–2474. [Google Scholar] [CrossRef]
  54. Chan, L.; Wang, H.; Terdiman, J.; Hoffman, J.; Ciol, M.A.; Lattimore, B.F.; Sidney, S.; Quesenberry, C.; Lu, Q.; Sandel, M.E. Disparities in Outpatient and Home Health Service Utilization Following Stroke: Results of a 9-Year Cohort Study in Northern California. PM&R 2009, 1, 997–1003. [Google Scholar] [CrossRef]
  55. Young, B.M.; Holman, E.A.; Cramer, S.C.; Shah, S.; Griessenauer, C.J.; Patel, N.; Lin, D.J.; Gee, J.; Moon, J.; Schwertfeger, J.; et al. Rehabilitation Therapy Doses Are Low After Stroke and Predicted by Clinical Factors. Stroke 2023, 54, 831–839. [Google Scholar] [CrossRef]
  56. Nicholson, S.; Sniehotta, F.F.; Van Wijck, F.; Greig, C.A.; Johnston, M.; Mcmurdo, M.E.T.; Dennis, M.; Mead, G.E. A Systematic Review of Perceived Barriers and Motivators to Physical Activity after Stroke. Int. J. Stroke 2013, 8, 357–364. [Google Scholar] [CrossRef]
  57. Black, S.; Román, G.C.; Geldmacher, D.S.; Salloway, S.; Hecker, J.; Burns, A.; Perdomo, C.; Kumar, D.; Pratt, R. Efficacy and Tolerability of Donepezil in Vascular Dementia Positive Results of a 24-Week, Multicenter, International, Randomized, Placebo-Controlled Clinical Trial. Stroke 2003, 34, 2323–2330. [Google Scholar] [CrossRef]
  58. Jorge, R.E.; Acion, L.; Moser, D.; Adams, H.P.; Robinson, R.G. Escitalopram and Enhancement of Cognitive Recovery Following Stroke. Arch. Gen. Psychiatry 2010, 67, 187–196. [Google Scholar] [CrossRef]
  59. Gagnon, D.J.; Leclerc, A.M.; Riker, R.R.; Brown, C.S.; May, T.; Nocella, K.; Cote, J.; Eldridge, A.; Seder, D.B. Amantadine and Modafinil as Neurostimulants During Post-Stroke Care: A Systematic Review. Neurocrit. Care 2020, 33, 283–297. [Google Scholar]
  60. Kim, B.R.; Chun, M.H.; Kim, L.S.; Park, J.Y. Effect of Virtual Reality on Cognition in Stroke Patients. Ann. Rehabil. Med. 2011, 35, 450. [Google Scholar] [CrossRef]
  61. Zhou, Y.; Feng, H.; Li, G.; Xu, C.; Wu, Y.; Li, H. Efficacy of Computerized Cognitive Training on Improving Cognitive Functions of Stroke Patients: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Int. J. Nurs. Pract. 2022, 28, e12966. [Google Scholar] [CrossRef]
  62. Zheng, G.; Xia, R.; Zhou, W.; Tao, J.; Chen, L. Aerobic Exercise Ameliorates Cognitive Function in Older Adults with Mild Cognitive Impairment: A Systematic Review and Meta-Analysis of Randomised Controlled Trials. Br. J. Sports Med. 2016, 50, 1443–1450. [Google Scholar] [CrossRef]
  63. Alonso-Alonso, M.; Fregni, F.; Pascual-Leone, A. Brain Stimulation in Poststroke Rehabilitation. Cerebrovasc. Dis. 2007, 24, 157–166. [Google Scholar] [CrossRef]
  64. Yamada, Y.; Sumiyoshi, T. Neurobiological Mechanisms of Transcranial Direct Current Stimulation for Psychiatric Disorders; Neurophysiological, Chemical, and Anatomical Considerations. Front. Hum. Neurosci. 2021, 15, 631838. [Google Scholar]
  65. Iyer, P.C.; Madhavan, S. Non-Invasive Brain Stimulation in the Modulation of Cerebral Blood Flow after Stroke: A Systematic Review of Transcranial Doppler Studies. Clin. Neurophysiol. 2018, 129, 2544–2551. [Google Scholar] [CrossRef]
  66. Nitsche, M.A.; Paulus, W. Sustained Excitability Elevations Induced by Transcranial DC Motor Cortex Stimulation in Humans. Neurology 2001, 57, 1899–1901. [Google Scholar] [CrossRef]
  67. Batsikadze, G.; Moliadze, V.; Paulus, W.; Kuo, M.F.; Nitsche, M.A. Partially Non-Linear Stimulation Intensity-Dependent Effects of Direct Current Stimulation on Motor Cortex Excitability in Humans. J. Physiol. 2013, 591, 1987–2000. [Google Scholar] [CrossRef]
  68. Nitsche, M.A.; Paulus, W. Excitability Changes Induced in the Human Motor Cortex by Weak Transcranial Direct Current Stimulation. J. Physiol. 2000, 527, 633–639. [Google Scholar] [CrossRef]
  69. Baeken, C.; Remue, J.; Vanderhasselt, M.A.; Brunoni, A.R.; De Witte, S.; Duprat, R.; Koster, E.H.W.; De Raedt, R.; Wu, G.R. Increased Left Prefrontal Brain Perfusion after MRI Compatible TDCS Attenuates Momentary Ruminative Self-Referential Thoughts. Brain Stimul. 2017, 10, 1088–1095. [Google Scholar] [CrossRef]
  70. Bikson, M.; Grossman, P.; Thomas, C.; Zannou, A.L.; Jiang, J.; Adnan, T.; Mourdoukoutas, A.P.; Kronberg, G.; Truong, D.; Boggio, P.; et al. Safety of Transcranial Direct Current Stimulation: Evidence Based Update 2016. Brain Stimul. 2016, 9, 641–661. [Google Scholar] [CrossRef]
  71. Knotkova, H.; Riggs, A.; Berisha, D.; Borges, H.; Bernstein, H.; Patel, V.; Truong, D.Q.; Unal, G.; Arce, D.; Datta, A.; et al. Automatic M1-SO Montage Headgear for Transcranial Direct Current Stimulation (TDCS) Suitable for Home and High-Throughput In-Clinic Applications. Neuromodul. Technol. Neural Interface 2018, 22, 904–910. [Google Scholar] [CrossRef]
  72. Charvet, L.E.; Kasschau, M.; Datta, A.; Knotkova, H.; Stevens, M.C.; Alonzo, A.; Loo, C.; Krull, K.R.; Bikson, M. Remotely-Supervised Transcranial Direct Current Stimulation (TDCS) for Clinical Trials: Guidelines for Technology and Protocols. Front. Syst. Neurosci. 2015, 9, 26. [Google Scholar] [CrossRef]
  73. Shaw, M.; Pilloni, G.; Charvet, L. Delivering Transcranial Direct Current Stimulation Away from Clinic: Remotely Supervised TDCS. Mil. Med. 2020, 185, 319–325. [Google Scholar] [CrossRef]
  74. Dobbs, B.; Pawlak, N.; Biagioni, M.; Agarwal, S.; Shaw, M.; Pilloni, G.; Bikson, M.; Datta, A.; Charvet, L. Generalizing Remotely Supervised Transcranial Direct Current Stimulation (TDCS): Feasibility and Benefit in Parkinson’s Disease. J. Neuroeng. Rehabil. 2018, 15, 114. [Google Scholar] [CrossRef]
  75. Pilloni, G.; Vogel-Eyny, A.; Lustberg, M.; Best, P.; Malik, M.; Walton-Masters, L.; George, A.; Mirza, I.; Zhovtis, L.; Datta, A.; et al. Tolerability and Feasibility of At-Home Remotely Supervised Transcranial Direct Current Stimulation (RS-TDCS): Single-Center Evidence from 6779 Sessions. Brain Stimul. 2022, 15, 707–716. [Google Scholar] [CrossRef]
  76. Koutsomitros, T.; Schwarz, S.A.; van der Zee, K.T.; Schuhmann, T.; Sack, A.T. Home-Administered Transcranial Direct Current Stimulation with Asynchronous Remote Supervision in the Treatment of Depression: Feasibility, Tolerability, and Clinical Effectiveness. Front. Psychiatry 2023, 14, 1206805. [Google Scholar] [CrossRef]
  77. Alonzo, A.; Fong, J.; Ball, N.; Martin, D.; Chand, N.; Loo, C. Pilot Trial of Home-Administered Transcranial Direct Current Stimulation for the Treatment of Depression. J. Affect. Disord. 2019, 252, 475–483. [Google Scholar] [CrossRef]
  78. Ko, M.H.; Yoon, J.Y.; Jo, Y.J.; Son, M.N.; Kim, D.S.; Kim, G.W.; Won, Y.H.; Park, S.H.; Seo, J.H.; Kim, Y.H. Home-Based Transcranial Direct Current Stimulation to Enhance Cognition in Stroke: Randomized Controlled Trial. Stroke 2022, 53, 2992–3001. [Google Scholar] [CrossRef]
  79. Villamar, M.F.; Volz, M.S.; Bikson, M.; Datta, A.; Dasilva, A.F.; Fregni, F. Technique and Considerations in the Use of 4x1 Ring High-Definition Transcranial Direct Current Stimulation (HD-TDCS). J. Vis. Exp. 2013, e50309. [Google Scholar] [CrossRef]
  80. Murase, N.; Duque, J.; Mazzocchio, R.; Cohen, L.G. Influence of Interhemispheric Interactions on Motor Function in Chronic Stroke. Ann. Neurol. 2004, 55, 400–409. [Google Scholar] [CrossRef]
  81. Feng, W.; Kautz, S.A.; Schlaug, G.; Meinzer, C.; George, M.S.; Chhatbar, P.Y. Transcranial Direct Current Stimulation for Post-Stroke Motor Recovery: Challenges and Opportunities. PM&R 2018, 10, S157–S164. [Google Scholar] [CrossRef]
  82. Schlaug, G.; Renga, V.; Nair, D. Transcranial Direct Current Stimulation in Stroke Recovery. Arch. Neurol. 2008, 65, 1571–1576. [Google Scholar] [CrossRef]
  83. González-Rodriguez, B.; Serradell-Ribé, N.; Viejo-Sobera, R.; Romero-Muñoz, J.P.; Marron, E.M. Transcranial Direct Current Stimulation in Neglect Rehabilitation after Stroke: A Systematic Review. J. Neurol. 2022, 269, 6310–6329. [Google Scholar] [CrossRef]
  84. Cappon, D.; Jahanshahi, M.; Bisiacchi, P. Value and Efficacy of Transcranial Direct Current Stimulation in the Cognitive Rehabilitation: A Critical Review since 2000. Front. Neurosci. 2016, 10, 157. [Google Scholar] [CrossRef]
  85. You, D.S.; Kim, D.Y.; Chun, M.H.; Jung, S.E.; Park, S.J. Cathodal Transcranial Direct Current Stimulation of the Right Wernicke’s Area Improves Comprehension in Subacute Stroke Patients. Brain Lang. 2011, 119, 1–5. [Google Scholar] [CrossRef]
  86. Elsner, B.; Kugler, J.; Mehrholz, J. Transcranial Direct Current Stimulation (TDCS) for Improving Aphasia after Stroke: A Systematic Review with Network Meta-Analysis of Randomized Controlled Trials. J. Neuroeng. Rehabil. 2020, 17, 88. [Google Scholar]
  87. Fridriksson, J.; Rorden, C.; Elm, J.; Sen, S.; George, M.S.; Bonilha, L. Transcranial Direct Current Stimulation vs Sham Stimulation to Treat Aphasia After Stroke: A Randomized Clinical Trial. JAMA Neurol. 2018, 75, 1470–1476. [Google Scholar] [CrossRef]
  88. Di Pino, G.; Pellegrino, G.; Assenza, G.; Capone, F.; Ferreri, F.; Formica, D.; Ranieri, F.; Tombini, M.; Ziemann, U.; Rothwell, J.C.; et al. Modulation of Brain Plasticity in Stroke: A Novel Model for Neurorehabilitation. Nat. Rev. Neurol. 2014, 10, 597–608. [Google Scholar] [CrossRef]
  89. Fox, M.D. Mapping Symptoms to Brain Networks with the Human Connectome. N. Engl. J. Med. 2018, 379, 2237–2245. [Google Scholar] [CrossRef]
  90. Bahmani, Z.; Clark, K.; Merrikhi, Y.; Mueller, A.; Pettine, W.; Isabel Vanegas, M.; Moore, T.; Noudoost, B. Prefrontal Contributions to Attention and Working Memory. Curr. Top. Behav. Neurosci. 2019, 41, 129–153. [Google Scholar] [CrossRef]
  91. Curtis, C.E.; D’Esposito, M. Persistent Activity in the Prefrontal Cortex during Working Memory. Trends Cogn. Sci. 2003, 7, 415–423. [Google Scholar] [CrossRef]
  92. Shulman, G.L.; Tansy, A.P.; Kincade, M.; Petersen, S.E.; McAvoy, M.P.; Corbetta, M. Reactivation of Networks Involved in Preparatory States. Cereb. Cortex 2002, 12, 590–600. [Google Scholar] [CrossRef]
  93. Chao, L.L.; Knight, R.T. Contribution of Human Prefrontal Cortex to Delay Performance. J. Cogn. Neurosci. 1998, 10, 161–177. [Google Scholar] [CrossRef]
  94. Stagg, C.J.; Lin, R.L.; Mezue, M.; Segerdahl, A.; Kong, Y.; Xie, J.; Tracey, I. Widespread Modulation of Cerebral Perfusion Induced during and after Transcranial Direct Current Stimulation Applied to the Left Dorsolateral Prefrontal Cortex. J. Neurosci. 2013, 33, 11425–11431. [Google Scholar] [CrossRef]
  95. Jamil, A.; Batsikadze, G.; Kuo, H.I.; Labruna, L.; Hasan, A.; Paulus, W.; Nitsche, M.A. Systematic Evaluation of the Impact of Stimulation Intensity on Neuroplastic After-Effects Induced by Transcranial Direct Current Stimulation. J. Physiol. 2017, 595, 1273–1288. [Google Scholar] [CrossRef]
  96. Artola, A.; Bröcher, S.; Singer, W. Different Voltage-Dependent Thresholds for Inducing Long-Term Depression and Long-Term Potentiation in Slices of Rat Visual Cortex. Nature 1990, 347, 69–72. [Google Scholar] [CrossRef]
  97. Sjöström, P.J.; Häusser, M. A Cooperative Switch Determines the Sign of Synaptic Plasticity in Distal Dendrites of Neocortical Pyramidal Neurons. Neuron 2006, 51, 227–238. [Google Scholar] [CrossRef]
  98. Kronberg, G.; Bridi, M.; Abel, T.; Bikson, M.; Parra, L.C. Direct Current Stimulation Modulates LTP and LTD: Activity Dependence and Dendritic Effects. Brain Stimul. 2017, 10, e23–e24. [Google Scholar] [CrossRef]
  99. Li, L.M.; Violante, I.R.; Leech, R.; Ross, E.; Hampshire, A.; Opitz, A.; Rothwell, J.C.; Carmichael, D.W.; Sharp, D.J. Brain State and Polarity Dependent Modulation of Brain Networks by Transcranial Direct Current Stimulation. Hum. Brain Mapp. 2019, 40, 904. [Google Scholar] [CrossRef]
  100. Bergmann, T.O. Brain State-Dependent Brain Stimulation. Front. Psychol. 2018, 9, 422698. [Google Scholar]
  101. Hulme, S.R.; Jones, O.D.; Abraham, W.C. Emerging Roles of Metaplasticity in Behaviour and Disease. Trends Neurosci. 2013, 36, 353–362. [Google Scholar] [CrossRef]
  102. Saleem, M.; Herrmann, N.; Dinoff, A.; Mielke, M.M.; Oh, P.I.; Shammi, P.; Cao, X.; Venkata, S.L.V.; Haughey, N.J.; Lanctôt, K.L. A Lipidomics Approach to Assess the Association between Plasma Sphingolipids and Verbal Memory Performance in Coronary Artery Disease Patients Undertaking Cardiac Rehabilitation: A C18:0 Signature for Cognitive Response to Exercise. J. Alzheimers Dis. 2017, 60, 829. [Google Scholar] [CrossRef]
  103. Singh, A.M.; Neva, J.L.; Staines, W.R. Acute Exercise Enhances the Response to Paired Associative Stimulation-Induced Plasticity in the Primary Motor Cortex. Exp. Brain Res. 2014, 232, 3675–3685. [Google Scholar] [CrossRef]
  104. Steinberg, F.; Pixa, N.H.; Fregni, F. A Review of Acute Aerobic Exercise and Transcranial Direct Current Stimulation Effects on Cognitive Functions and Their Potential Synergies. Front. Hum. Neurosci. 2018, 12, 534. [Google Scholar] [CrossRef]
  105. Liu, C.S.; Herrmann, N.; Song, B.X.; Ba, J.; Gallagher, D.; Oh, P.I.; Marzolini, S.; Rajji, T.K.; Charles, J.; Papneja, P.; et al. Exercise Priming with Transcranial Direct Current Stimulation: A Study Protocol for a Randomized, Parallel-Design, Sham-Controlled Trial in Mild Cognitive Impairment and Alzheimer’s Disease. BMC Geriatr. 2021, 21, 677. [Google Scholar] [CrossRef]
  106. Sathappan, A.V.; Luber, B.M.; Lisanby, S.H. The Dynamic Duo: Combining Noninvasive Brain Stimulation with Cognitive Interventions. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 89, 347–360. [Google Scholar] [CrossRef]
  107. Hermans, E.J.; Van Marle, H.J.F.; Ossewaarde, L.; Henckens, M.J.A.G.; Qin, S.; Van Kesteren, M.T.R.; Schoots, V.C.; Cousijn, H.; Rijpkema, M.; Oostenveld, R.; et al. Stress-Related Noradrenergic Activity Prompts Large-Scale Neural Network Reconfiguration. Science 2011, 334, 1151–1153. [Google Scholar] [CrossRef]
  108. De Smet, S.; Razza, L.B.; Pulopulos, M.M.; De Raedt, R.; Baeken, C.; Brunoni, A.R.; Vanderhasselt, M.A. Stress Priming Transcranial Direct Current Stimulation (TDCS) Enhances Updating of Emotional Content in Working Memory. Brain Stimul. 2024, 17, 434–443. [Google Scholar] [CrossRef]
  109. Christova, M.; Rafolt, D.; Gallasch, E. Cumulative Effects of Anodal and Priming Cathodal TDCS on Pegboard Test Performance and Motor Cortical Excitability. Behav. Brain Res. 2015, 287, 27–33. [Google Scholar] [CrossRef]
  110. Müller-Dahlhaus, F.; Ziemann, U. Metaplasticity in Human Cortex. Neuroscientist 2015, 21, 185–202. [Google Scholar] [CrossRef]
  111. Zhu, F.; Xu, X.; Jin, M.; Chen, J.; Feng, X.; Wang, J.; Yu, D.; Wang, R.; Lian, Y.; Huai, B.; et al. Priming Transcranial Direct Current Stimulation for Improving Hemiparetic Upper Limb in Patients with Subacute Stroke: Study Protocol for a Randomised Controlled Trial. BMJ Open 2024, 14, e079372. [Google Scholar] [CrossRef]
  112. Wagner, T.; Valero-Cabre, A.; Pascual-Leone, A. Noninvasive Human Brain Stimulation. Annu. Rev. Biomed. Eng. 2007, 9, 527–565. [Google Scholar] [CrossRef]
  113. Datta, A.; Bansal, V.; Diaz, J.; Patel, J.; Reato, D.; Bikson, M. Gyri–Precise Head Model of Transcranial DC Stimulation: Improved Spatial Focality Using a Ring Electrode versus Conventional Rectangular Pad. Brain Stimul. 2009, 2, 201. [Google Scholar] [CrossRef]
  114. Fregni, F.; El-Hagrassy, M.M.; Pacheco-Barrios, K.; Carvalho, S.; Leite, J.; Simis, M.; Brunelin, J.; Nakamura-Palacios, E.M.; Marangolo, P.; Venkatasubramanian, G.; et al. Evidence-Based Guidelines and Secondary Meta-Analysis for the Use of Transcranial Direct Current Stimulation in Neurological and Psychiatric Disorders. Int. J. Neuropsychopharmacol. 2021, 24, 256–313. [Google Scholar] [CrossRef]
  115. Pascual-leone, A.; Valls-solé, J.; Wassermann, E.M.; Hallett, M. Responses to Rapid-Rate Transcranial Magnetic Stimulation of the Human Motor Cortex. Brain 1994, 117 Pt 4, 847–858. [Google Scholar] [CrossRef]
  116. Schrader, L.M.; Stern, J.M.; Koski, L.; Nuwer, M.R.; Engel, J. Seizure Incidence during Single- and Paired-Pulse Transcranial Magnetic Stimulation (TMS) in Individuals with Epilepsy. Clin. Neurophysiol. 2004, 115, 2728–2737. [Google Scholar] [CrossRef]
  117. Lerner, A.J.; Wassermann, E.M.; Tamir, D.I. Seizures from Transcranial Magnetic Stimulation 2012–2016: Results of a Survey of Active Laboratories and Clinics. Clin. Neurophysiol. 2019, 130, 1409–1416. [Google Scholar] [CrossRef]
  118. Chou, Y.-H.; Ton That, V.; Chen, A.Y.C.; Sundman, M.; Huang, Y.Z. TMS-Induced Seizure Cases Stratified by Population, Stimulation Protocol, and Stimulation Site: A Systematic Literature Search. Clin. Neurophysiol. 2020, 131, 1019–1020. [Google Scholar] [CrossRef]
  119. Rossi, S.; Antal, A.; Bestmann, S.; Bikson, M.; Brewer, C.; Brockmöller, J.; Carpenter, L.L.; Cincotta, M.; Chen, R.; Daskalakis, J.D.; et al. Safety and Recommendations for TMS Use in Healthy Subjects and Patient Populations, with Updates on Training, Ethical and Regulatory Issues: Expert Guidelines. Clin. Neurophysiol. 2021, 132, 269–306. [Google Scholar] [CrossRef]
  120. Passera, B.; Chauvin, A.; Raffin, E.; Bougerol, T.; David, O.; Harquel, S. Exploring the Spatial Resolution of TMS-EEG Coupling on the Sensorimotor Region. NeuroImage 2022, 259, 119419. [Google Scholar] [CrossRef]
  121. Cattaneo, L. Fancies and Fallacies of Spatial Sampling with Transcranial Magnetic Stimulation (TMS). Front. Psychol. 2018, 9, 373783. [Google Scholar]
  122. Gao, Y.; Qiu, Y.; Yang, Q.; Tang, S.; Gong, J.; Fan, H.; Wu, Y.; Lu, X. Repetitive Transcranial Magnetic Stimulation Combined with Cognitive Training for Cognitive Function and Activities of Daily Living in Patients with Post-Stroke Cognitive Impairment: A Systematic Review and Meta-Analysis. Ageing Res. Rev. 2023, 87, 101919. [Google Scholar] [CrossRef]
  123. Hofmeijer, J.; Ham, F.; Kwakkel, G. Evidence of RTMS for Motor or Cognitive Stroke Recovery: Hype or Hope? Stroke 2023, 54, 2500–2511. [Google Scholar] [CrossRef]
  124. O’Brien, W.D. Ultrasound—Biophysics Mechanisms. Prog. Biophys. Mol. Biol. 2007, 93, 212. [Google Scholar] [CrossRef]
  125. Kim, H.; Taghados, S.J.; Fischer, K.; Maeng, L.S.; Park, S.; Yoo, S.S. Noninvasive Transcranial Stimulation of Rat Abducens Nerve by Focused Ultrasound. Ultrasound Med. Biol. 2012, 38, 1568–1575. [Google Scholar] [CrossRef]
  126. Tufail, Y.; Yoshihiro, A.; Pati, S.; Li, M.M.; Tyler, W.J. Ultrasonic Neuromodulation by Brain Stimulation with Transcranial Ultrasound. Nat. Protoc. 2011, 6, 1453–1470. [Google Scholar] [CrossRef]
  127. Shin, J.; Kong, C.; Cho, J.S.; Lee, J.; Koh, C.S.; Yoon, M.S.; Na, Y.C.; Chang, W.S.; Chang, J.W. Focused Ultrasound-Mediated Noninvasive Blood-Brain Barrier Modulation: Preclinical Examination of Efficacy and Safety in Various Sonication Parameters. Neurosurg. Focus. 2018, 44, E15. [Google Scholar] [CrossRef]
  128. Yoo, S.S.; Bystritsky, A.; Lee, J.H.; Zhang, Y.; Fischer, K.; Min, B.K.; McDannold, N.J.; Pascual-Leone, A.; Jolesz, F.A. Focused Ultrasound Modulates Region-Specific Brain Activity. NeuroImage 2011, 56, 1267–1275. [Google Scholar] [CrossRef]
  129. Guo, J.; Lo, W.L.A.; Hu, H.; Yan, L.; Li, L. Transcranial Ultrasound Stimulation Applied in Ischemic Stroke Rehabilitation: A Review. Front. Neurosci. 2022, 16, 964060. [Google Scholar] [CrossRef]
  130. Ciullo, V.; Spalletta, G.; Caltagirone, C.; Banaj, N.; Vecchio, D.; Piras, F.; Piras, F. Transcranial Direct Current Stimulation and Cognition in Neuropsychiatric Disorders: Systematic Review of the Evidence and Future Directions. Neuroscientist 2021, 27, 285–309. [Google Scholar] [CrossRef]
  131. Zaninotto, A.L.; El-Hagrassy, M.M.; Green, J.R.; Babo, M.; Paglioni, V.M.; Benute, G.G.; Paiva, W.S. Transcranial Direct Current Stimulation (TDCS) Effects on Traumatic Injury (TBI) Recovery: A Systematic Review. Dement. Neuropsychol. 2019, 13, 172. [Google Scholar] [CrossRef]
  132. Gill, J.; Shah-Basak, P.P.; Hamilton, R. It’s the Thought That Counts: Examining the Task-Dependent Effects of Transcranial Direct Current Stimulation on Executive Function. Brain Stimul. 2015, 8, 253–259. [Google Scholar] [CrossRef]
  133. Nissim, N.R.; O’Shea, A.; Indahlastari, A.; Kraft, J.N.; von Mering, O.; Aksu, S.; Porges, E.; Cohen, R.; Woods, A.J. Effects of Transcranial Direct Current Stimulation Paired with Cognitive Training on Functional Connectivity of the Working Memory Network in Older Adults. Front. Aging Neurosci. 2019, 11, 340. [Google Scholar] [CrossRef]
  134. Hsu, W.-Y.; Ku, Y.; Zanto, T.P.; Gazzaley, A. Effects of Non-Invasive Brain Stimulation on Cognitive Function in Healthy Aging and Alzheimer’s Disease: A Systematic Review and Meta-Analysis. Neurobiol. Aging 2015, 36, 2348–2359. [Google Scholar]
  135. Mattioli, F.; Bellomi, F.; Stampatori, C.; Capra, R.; Miniussi, C. Neuroenhancement through Cognitive Training and Anodal TDCS in Multiple Sclerosis. Mult. Scler. J. 2016, 22, 222–230. [Google Scholar] [CrossRef]
  136. Ashrafi, A.; Mohseni-Bandpei, M.A.; Seydi, M. The Effect of TDCS on the Fatigue in Patients with Multiple Sclerosis: A Systematic Review of Randomized Controlled Clinical Trials. J. Clin. Neurosci. 2020, 78, 277–283. [Google Scholar] [CrossRef]
  137. Ahorsu, D.K.; Adjaottor, E.S.; Yin, B.; Lam, H.; Borgomaneri, S.; Sellaro, R. Intervention Effect of Non-Invasive Brain Stimulation on Cognitive Functions among People with Traumatic Brain Injury: A Systematic Review and Meta-Analysis. Brain Sci. 2021, 11, 840. [Google Scholar] [CrossRef]
  138. Dhaliwal, S.K.; Meek, B.P.; Modirrousta, M.M. Non-Invasive Brain Stimulation for the Treatment of Symptoms Following Traumatic Brain Injury. Front. Psychiatry 2015, 6, 119. [Google Scholar] [CrossRef]
  139. Monti, A.; Ferrucci, R.; Fumagalli, M.; Mameli, F.; Cogiamanian, F.; Ardolino, G.; Priori, A. Transcranial Direct Current Stimulation (TDCS) and Language. J. Neurol. Neurosurg. Psychiatry 2013, 84, 832–842. [Google Scholar] [CrossRef]
  140. Baker, J.M.; Rorden, C.; Fridriksson, J. Using Transcranial Direct-Current Stimulation to Treat Stroke Patients with Aphasia. Stroke 2010, 41, 1229–1236. [Google Scholar] [CrossRef]
  141. Fiori, V.; Nitsche, M.A.; Cucuzza, G.; Caltagirone, C.; Marangolo, P. High-Definition Transcranial Direct Current Stimulation Improves Verb Recovery in Aphasic Patients Depending on Current Intensity. Neuroscience 2019, 406, 159–166. [Google Scholar] [CrossRef]
  142. Fridriksson, J.; Richardson, J.D.; Baker, J.M.; Rorden, C. Transcranial Direct Current Stimulation Improves Naming Reaction Time in Fluent Aphasia: A Double-Blind, Sham-Controlled Study. Stroke 2011, 42, 819–821. [Google Scholar] [CrossRef]
  143. Stockbridge, M.D.; Faria, A.V.; Fridriksson, J.; Rorden, C.; Bonilha, L.; Hillis, A.E. Subacute Aphasia Recovery Is Associated with Resting-State Connectivity within and beyond the Language Network. Ann. Clin. Transl. Neurol. 2023, 10, 1525–1532. [Google Scholar] [CrossRef]
  144. Turkeltaub, P.E.; Coslett, H.B.; Thomas, A.L.; Faseyitan, O.; Benson, J.; Norise, C.; Hamilton, R.H. The Right Hemisphere Is Not Unitary in Its Role in Aphasia Recovery. Cortex 2012, 48, 1179–1186. [Google Scholar] [CrossRef]
  145. Turkeltaub, P.E.; Messing, S.; Norise, C.; Hamilton, R.H. Are Networks for Residual Language Function and Recovery Consistent across Aphasic Patients? Neurology 2011, 76, 1726–1734. [Google Scholar] [CrossRef]
  146. Fridriksson, J.; Elm, J.; Stark, B.C.; Basilakos, A.; Rorden, C.; Sen, S.; George, M.S.; Gottfried, M.; Bonilha, L. BDNF Genotype and TDCS Interaction in Aphasia Treatment. Brain Stimul. 2018, 11, 1276–1281. [Google Scholar] [CrossRef]
  147. Vestito, L.; Rosellini, S.; Mantero, M.; Bandini, F. Long-Term Effects of Transcranial Direct-Current Stimulation in Chronic Post-Stroke Aphasia: A Pilot Study. Front. Hum. Neurosci. 2014, 8, 785. [Google Scholar] [CrossRef]
  148. Campana, S.; Caltagirone, C.; Marangolo, P. Combining Voxel-Based Lesion-Symptom Mapping (VLSM) with A-TDCS Language Treatment: Predicting Outcome of Recovery in Nonfluent Chronic Aphasia. Brain Stimul. 2015, 8, 769–776. [Google Scholar] [CrossRef]
  149. Fiori, V.; Coccia, M.; Marinelli, C.V.; Vecchi, V.; Bonifazi, S.; Gabriella Ceravolo, M.; Provinciali, L.; Tomaiuolo, F.; Marangolo, P. Transcranial Direct Current Stimulation Improves Word Retrieval in Healthy and Nonfluent Aphasic Subjects. J. Cogn. Neurosci. 2011, 23, 2309–2323. [Google Scholar] [CrossRef]
  150. Marangolo, P.; Fiori, V.; Calpagnano, M.A.; Campana, S.; Razzano, C.; Caltagirone, C.; Marini, A. TDCS over the Left Inferior Frontal Cortex Improves Speech Production in Aphasia. Front. Hum. Neurosci. 2013, 7, 61793. [Google Scholar] [CrossRef]
  151. Woodhead, Z.V.J.; Kerry, S.J.; Aguilar, O.M.; Ong, Y.H.; Hogan, J.S.; Pappa, K.; Leff, A.P.; Crinion, J.T. Randomized Trial of IReadMore Word Reading Training and Brain Stimulation in Central Alexia. Brain 2018, 141, 2127–2141. [Google Scholar] [CrossRef]
  152. Kang, E.K.; Kim, Y.K.; Sohn, H.M.; Cohen, L.G.; Paik, N.J. Improved Picture Naming in Aphasia Patients Treated with Cathodal TDCS to Inhibit the Right Broca’s Homologue Area. Restor. Neurol. Neurosci. 2011, 29, 141. [Google Scholar] [CrossRef]
  153. Cheng, W.; Li, Y.; Cheng, B.; Chen, Y.; Chen, Z.; Cui, L.; Chen, X.; Chen, Z. Effects of Transcranial Direct Current Stimulation over the Right Hemisphere on Naming Ability in Patients with Poststroke Aphasia: A Meta-Analysis. J. Neurolinguist. 2021, 58, 100986. [Google Scholar] [CrossRef]
  154. Mariën, P.; Ackermann, H.; Adamaszek, M.; Barwood, C.H.S.; Beaton, A.; Desmond, J.; De Witte, E.; Fawcett, A.J.; Hertrich, I.; Küper, M.; et al. Consensus Paper: Language and the Cerebellum: An Ongoing Enigma. Cerebellum 2014, 13, 386. [Google Scholar] [CrossRef]
  155. Wessel, M.J.; Hummel, F.C. Non-Invasive Cerebellar Stimulation: A Promising Approach for Stroke Recovery? Cerebellum 2018, 17, 359–371. [Google Scholar] [CrossRef]
  156. Sebastian, R.; Kim, J.H.; Brenowitz, R.; Tippett, D.C.; Desmond, J.E.; Celnik, P.A.; Hillis, A.E. Cerebellar Neuromodulation Improves Naming in Post-Stroke Aphasia. Brain Commun. 2020, 2, fcaa179. [Google Scholar] [CrossRef]
  157. van Dun, K.; Bodranghien, F.C.A.A.; Mariën, P.; Manto, M.U. TDCS of the Cerebellum: Where Do We Stand in 2016? Technical Issues and Critical Review of the Literature. Front. Hum. Neurosci. 2016, 10, 199. [Google Scholar] [CrossRef]
  158. Vallar, G.; Perani, D. The Anatomy of Unilateral Neglect after Right-Hemisphere Stroke Lesions. A Clinical/CT-Scan Correlation Study in Man. Neuropsychologia 1986, 24, 609–622. [Google Scholar] [CrossRef]
  159. Malhotra, P.; Coulthard, E.J.; Husain, M. Role of Right Posterior Parietal Cortex in Maintaining Attention to Spatial Locations over Time. Brain 2009, 132, 645. [Google Scholar] [CrossRef]
  160. Yi, Y.G.; Chun, M.H.; Do, K.H.; Sung, E.J.; Kwon, Y.G.; Kim, D.Y. The Effect of Transcranial Direct Current Stimulation on Neglect Syndrome in Stroke Patients. Ann. Rehabil. Med. 2016, 40, 223–229. [Google Scholar] [CrossRef]
  161. da Silva, T.R.; de Carvalho Nunes, H.R.; Martins, L.G.; da Costa, R.D.M.; de Souza, J.T.; Winckler, F.C.; Sartor, L.C.A.; Modolo, G.P.; Ferreira, N.C.; da Silva Rodrigues, J.C.; et al. Non-Invasive Brain Stimulation Can Reduce Unilateral Spatial Neglect after Stroke: ELETRON Trial. Ann. Neurol. 2022, 92, 400–410. [Google Scholar] [CrossRef]
  162. Sunwoo, H.; Kim, Y.H.; Chang, W.H.; Noh, S.; Kim, E.J.; Ko, M.H. Effects of Dual Transcranial Direct Current Stimulation on Post-Stroke Unilateral Visuospatial Neglect. Neurosci. Lett. 2013, 554, 94–98. [Google Scholar] [CrossRef]
  163. Richardson, A.G.; Overduin, S.A.; Valero-Cabré, A.; Padoa-Schioppa, C.; Pascual-Leone, A.; Bizzi, E.; Press, D.Z. Disruption of Primary Motor Cortex before Learning Impairs Memory of Movement Dynamics. J. Neurosci. 2006, 26, 12466. [Google Scholar] [CrossRef]
  164. O’Shea, J.; Revol, P.; Cousijn, H.; Near, J.; Petitet, P.; Jacquin-Courtois, S.; Berg, H.J.; Rode, G.; Rossetti, Y. Induced Sensorimotor Cortex Plasticity Remediates Chronic Treatment-Resistant Visual Neglect. eLife 2017, 6, e26602. [Google Scholar] [CrossRef]
  165. Kazuta, T.; Takeda, K.; Osu, R.; Tanaka, S.; Oishi, A.; Kondo, K.; Liu, M. Transcranial Direct Current Stimulation Improves Audioverbal Memory in Stroke Patients. Am. J. Phys. Med. Rehabil. 2017, 96, 565–571. [Google Scholar] [CrossRef]
  166. Yun, G.J.; Chun, M.H.; Kim, B.R. The Effects of Transcranial Direct-Current Stimulation on Cognition in Stroke Patients. J. Stroke 2015, 17, 354–358. [Google Scholar] [CrossRef]
  167. Emch, M.; von Bastian, C.C.; Koch, K. Neural Correlates of Verbal Working Memory: An FMRI Meta-Analysis. Front. Hum. Neurosci. 2019, 13, 447322. [Google Scholar]
  168. Jo, J.M.; Kim, Y.H.; Ko, M.H.; Ohn, S.H.; Joen, B.; Lee, K.H. Enhancing the Working Memory of Stroke Patients Using TDCS. Am. J. Phys. Med. Rehabil. 2009, 88, 404–409. [Google Scholar] [CrossRef]
  169. Park, S.-H.; Koh, E.-J.; Choi, H.-Y.; Ko, M.-H. A Double-Blind, Sham-Controlled, Pilot Study to Assess the Effects of the Concomitant Use of Transcranial Direct Current Stimulation with the Computer Assisted Cognitive Rehabilitation to the Prefrontal Cortex on Cognitive Functions in Patients with Strok. J. Korean Neurosurg. Soc. 2013, 54, 484–488. [Google Scholar] [CrossRef]
  170. Chiou, R.; Lambon Ralph, M.A. The Anterior-Ventrolateral Temporal Lobe Contributes to Boosting Visual Working Memory Capacity for Items Carrying Semantic Information. NeuroImage 2018, 169, 453–461. [Google Scholar] [CrossRef]
  171. Takeuchi, H.; Taki, Y.; Sassa, Y.; Hashizume, H.; Sekiguchi, A.; Fukushima, A.; Kawashima, R. Brain Structures Associated with Executive Functions during Everyday Events in a Non-Clinical Sample. Brain Struct. Funct. 2013, 218, 1017. [Google Scholar] [CrossRef]
  172. Rusnáková, Š.; Daniel, P.; Chládek, J.; Jurák, P.; Rektor, I. The Executive Functions in Frontal and Temporal Lobes: A Flanker Task Intracerebral Recording Study. J. Clin. Neurophysiol. 2011, 28, 30–35. [Google Scholar] [CrossRef]
  173. Imburgio, M.J.; Orr, J.M. Effects of Prefrontal TDCS on Executive Function: Methodological Considerations Revealed by Meta-Analysis. Neuropsychologia 2018, 117, 156–166. [Google Scholar] [CrossRef]
  174. Hosseinzadeh, S.A.; Mazhari, S.; Najafi, K.; Ahmadi, M.; Aghaei, I.; Khaksarian, M. Anodal Transcranial Direct Current Stimulation Enhances Positive Changes in Movement Functions, Visual Attention and Depression of Patients with Chronic Ischemic Stroke: A Clinical Trial. Biomed. Res. Ther. 2018, 5, 2841–2849. [Google Scholar] [CrossRef]
  175. Kleynen, M.; Braun, S.M.; Bleijlevens, M.H.; Lexis, M.A.; Rasquin, S.M.; Halfens, J.; Wilson, M.R.; Beurskens, A.J.; Masters, R.S.W. Using a Delphi Technique to Seek Consensus Regarding Definitions, Descriptions and Classification of Terms Related to Implicit and Explicit Forms of Motor Learning. PLoS ONE 2014, 9, e100227. [Google Scholar] [CrossRef]
  176. Meyer, S.; De Bruyn, N.; Krumlinde-Sundholm, L.; Peeters, A.; Feys, H.; Thijs, V.; Verheyden, G. Associations between Sensorimotor Impairments in the Upper Limb at 1 Week and 6 Months after Stroke. J. Neurol. Phys. Ther. 2016, 40, 186–195. [Google Scholar] [CrossRef]
  177. Buch, E.R.; Santarnecchi, E.; Antal, A.; Born, J.; Celnik, P.A.; Classen, J.; Gerloff, C.; Hallett, M.; Hummel, F.C.; Nitsche, M.A.; et al. Effects of TDCS on Motor Learning and Memory Formation: A Consensus and Critical Position Paper. Clin. Neurophysiol. 2017, 128, 589–603. [Google Scholar] [CrossRef]
  178. Reis, J.; Schambra, H.M.; Cohen, L.G.; Buch, E.R.; Fritsch, B.; Zarahn, E.; Celnik, P.A.; Krakauer, J.W. Noninvasive Cortical Stimulation Enhances Motor Skill Acquisition over Multiple Days through an Effect on Consolidation. Proc. Natl. Acad. Sci. USA 2009, 106, 1590. [Google Scholar] [CrossRef]
  179. Arioli, M.; Crespi, C.; Canessa, N. Social Cognition through the Lens of Cognitive and Clinical Neuroscience. BioMed Res. Int. 2018, 2018, 4283427. [Google Scholar] [CrossRef]
  180. Mars, R.B.; Sallet, J.; Schüffelgen, U.; Jbabdi, S.; Toni, I.; Rushworth, M.F.S. Connectivity-Based Subdivisions of the Human Right “Temporoparietal Junction Area”: Evidence for Different Areas Participating in Different Cortical Networks. Cereb. Cortex 2012, 22, 1894–1903. [Google Scholar] [CrossRef]
  181. Santiesteban, I.; Banissy, M.J.; Catmur, C.; Bird, G. Enhancing Social Ability by Stimulating Right Temporoparietal Junction. Curr. Biol. 2012, 22, 2274–2277. [Google Scholar] [CrossRef]
  182. Mai, X.; Zhang, W.; Hu, X.; Zhen, Z.; Xu, Z.; Zhang, J.; Liu, C. Using TDCS to Explore the Role of the Right Temporo-Parietal Junction in Theory of Mind and Cognitive Empathy. Front. Psychol. 2016, 7, 177459. [Google Scholar]
  183. Thirugnanasambandam, N.; Grundey, J.; Adam, K.; Drees, A.; Skwirba, A.C.; Lang, N.; Paulus, W.; Nitsche, M.A. Nicotinergic Impact on Focal and Non-Focal Neuroplasticity Induced by Non-Invasive Brain Stimulation in Non-Smoking Humans. Neuropsychopharmacology 2011, 36, 879. [Google Scholar] [CrossRef]
  184. Di Marcello Valladão Lugon, M.; Batsikadze, G.; Fresnoza, S.; Grundey, J.; Kuo, M.F.; Paulus, W.; Nakamura-Palacios, E.M.; Nitsche, M.A. Mechanisms of Nicotinic Modulation of Glutamatergic Neuroplasticity in Humans. Cereb. Cortex 2017, 27, 544–553. [Google Scholar] [CrossRef]
  185. Turner, T.J. Nicotine Enhancement of Dopamine Release by a Calcium-Dependent Increase in the Size of the Readily Releasable Pool of Synaptic Vesicles. J. Neurosci. 2004, 24, 11328–11336. [Google Scholar] [CrossRef]
  186. Wang, F.; Chen, H.; Steketee, J.D.; Sharp, B.M. Upregulation of Ionotropic Glutamate Receptor Subunits within Specific Mesocorticolimbic Regions during Chronic Nicotine Self-Administration. Neuropsychopharmacology 2006, 32, 103–109. [Google Scholar] [CrossRef]
  187. Chollet, F.; Tardy, J.; Albucher, J.F.; Thalamas, C.; Berard, E.; Lamy, C.; Bejot, Y.; Deltour, S.; Jaillard, A.; Niclot, P.; et al. Fluoxetine for Motor Recovery after Acute Ischaemic Stroke (FLAME): A Randomised Placebo-Controlled Trial. Lancet Neurol. 2011, 10, 123–130. [Google Scholar] [CrossRef]
  188. Nitsche, M.A.; Kuo, M.F.; Karrasch, R.; Wächter, B.; Liebetanz, D.; Paulus, W. Serotonin Affects Transcranial Direct Current-Induced Neuroplasticity in Humans. Biol. Psychiatry 2009, 66, 503–508. [Google Scholar] [CrossRef]
  189. Kuo, H.I.; Paulus, W.; Batsikadze, G.; Jamil, A.; Kuo, M.F.; Nitsche, M.A. Chronic Enhancement of Serotonin Facilitates Excitatory Transcranial Direct Current Stimulation-Induced Neuroplasticity. Neuropsychopharmacology 2016, 41, 1223–1230. [Google Scholar] [CrossRef]
  190. Kuo, H.I.; Paulus, W.; Batsikadze, G.; Jamil, A.; Kuo, M.F.; Nitsche, M.A. Acute and Chronic Effects of Noradrenergic Enhancement on Transcranial Direct Current Stimulation-induced Neuroplasticity in Humans. J. Physiol. 2017, 595, 1305. [Google Scholar] [CrossRef]
  191. Normann, C.; Clark, K. Selective Modulation of Ca2+ Influx Pathways by 5-HT Regulates Synaptic Long-Term Plasticity in the Hippocampus. Brain Res. 2005, 1037, 187–193. [Google Scholar] [CrossRef]
  192. Nitsche, M.A.; Grundey, J.; Liebetanz, D.; Lang, N.; Tergau, F.; Paulus, W. Catecholaminergic Consolidation of Motor Cortical Neuroplasticity in Humans. Cereb. Cortex 2004, 14, 1240–1245. [Google Scholar] [CrossRef]
  193. Monte-Silva, K.; Liebetanz, D.; Grundey, J.; Paulus, W.; Nitsche, M.A. Dosage-Dependent Non-Linear Effect of l-Dopa on Human Motor Cortex Plasticity. J. Physiol. 2010, 588 Pt 18, 3415. [Google Scholar] [CrossRef]
  194. Fresnoza, S.; Stiksrud, E.; Klinker, F.; Liebetanz, D.; Paulus, W.; Kuo, M.F.; Nitsche, M.A. Dosage-Dependent Effect of Dopamine D2 Receptor Activation on Motor Cortex Plasticity in Humans. J. Neurosci. 2014, 34, 10701. [Google Scholar] [CrossRef]
  195. Li, S.; Carmichael, S.T. Growth-Associated Gene and Protein Expression in the Region of Axonal Sprouting in the Aged Brain after Stroke. Neurobiol. Dis. 2006, 23, 362–373. [Google Scholar] [CrossRef]
  196. Murphy, T.H.; Corbett, D. Plasticity during Stroke Recovery: From Synapse to Behaviour. Nat. Rev. Neurosci. 2009, 10, 861–872. [Google Scholar] [CrossRef]
  197. Wieloch, T.; Nikolich, K. Mechanisms of Neural Plasticity Following Brain Injury. Curr. Opin. Neurobiol. 2006, 16, 258–264. [Google Scholar] [CrossRef]
  198. Liu, H.; Tian, T.; Qin, W.; Li, K.; Yu, C.; Sousa, N.; Patrick Gilman, C.; Hayashi, T. Contrasting Evolutionary Patterns of Functional Connectivity in Sensorimotor and Cognitive Regions after Stroke. Front. Behav. Neurosci. 2016, 10, 72. [Google Scholar] [CrossRef]
  199. Lashley, K.S. In Search of the Engram. In Physiological Mechanisms in Animal Behavior (Society’s Symposium IV); Academic Press: Oxford, UK, 1950; pp. 454–482. [Google Scholar]
  200. Krakauer, J.W.; Carmichael, S.T.; Corbett, D.; Wittenberg, G.F. Getting Neurorehabilitation Right—What Can We Learn from Animal Models? Neurorehabil. Neural Repair 2012, 26, 923. [Google Scholar] [CrossRef]
  201. Zeiler, S.R.; Krakauer, J.W. The Interaction between Training and Plasticity in the Post-Stroke Brain. Curr. Opin. Neurol. 2013, 26, 609–616. [Google Scholar] [CrossRef]
  202. Prabhakaran, S.; Zarahn, E.; Riley, C.; Speizer, A.; Chong, J.Y.; Lazar, R.M.; Marshall, R.S.; Krakauer, J.W. Inter-Individual Variability in the Capacity for Motor Recovery after Ischemic Stroke. Neurorehabil. Neural Repair 2008, 22, 64–71. [Google Scholar] [CrossRef]
  203. Turrigiano, G.G.; Nelson, S.B. Homeostatic Plasticity in the Developing Nervous System. Nat. Rev. Neurosci. 2004, 5, 97–107. [Google Scholar] [CrossRef]
  204. Butts, D.A.; Kanold, P.O.; Shatz, C.J. A Burst-Based “Hebbian” Learning Rule at Retinogeniculate Synapses Links Retinal Waves to Activity-Dependent Refinement. PLoS Biol. 2007, 5, e61. [Google Scholar] [CrossRef]
  205. Kwakkel, G.; Kollen, B.; Twisk, J. Impact of Time on Improvement of Outcome after Stroke. Stroke 2006, 37, 2348–2353. [Google Scholar] [CrossRef]
  206. Bernhardt, J.; Borschmann, K.N.; Kwakkel, G.; Burridge, J.H.; Eng, J.J.; Walker, M.F.; Bird, M.L.; Cramer, S.C.; Hayward, K.S.; O’Sullivan, M.J.; et al. Setting the Scene for the Second Stroke Recovery and Rehabilitation Roundtable. Int. J. Stroke 2019, 14, 450–456. [Google Scholar] [CrossRef]
  207. Teasell, R.W.; Foley, N.C.; Salter, K.L.; Jutai, J.W. A Blueprint for Transforming Stroke Rehabilitation Care in Canada: The Case for Change. Arch. Phys. Med. Rehabil. 2008, 89, 575–578. [Google Scholar] [CrossRef]
  208. Zheng, X.; Alsop, D.C.; Schlaug, G. Effects of Transcranial Direct Current Stimulation (TDCS) on Human Regional Cerebral Blood Flow. NeuroImage 2011, 58, 26. [Google Scholar] [CrossRef]
  209. Bahr-Hosseini, M.; Bikson, M. Neurovascular-Modulation: A Review of Primary Vascular Responses to Transcranial Electrical Stimulation as a Mechanism of Action. Brain Stimul. 2021, 14, 837–847. [Google Scholar] [CrossRef]
  210. Gozalov, A.; Jansen-Olesen, I.; Klaerke, D.; Olesen, J. Role of K ATP Channels in Cephalic Vasodilatation Induced by Calcitonin Gene-Related Peptide, Nitric Oxide, and Transcranial Electrical Stimulation in the Rat. Headache 2008, 48, 1202–1213. [Google Scholar] [CrossRef]
  211. Vernieri, F.; Assenza, G.; Maggio, P.; Tibuzzi, F.; Zappasodi, F.; Altamura, C.; Corbetto, M.; Trotta, L.; Palazzo, P.; Ercolani, M.; et al. Cortical Neuromodulation Modifies Cerebral Vasomotor Reactivity. Stroke 2010, 41, 2087–2090. [Google Scholar] [CrossRef]
  212. Bahr-Hosseini, M.; Nael, K.; Unal, G.; Iacoboni, M.; Liebeskind, D.S.; Bikson, M.; Saver, J.L.; Group, T.T.; Sanossian, N.; Wu, A.; et al. High-Definition Cathodal Direct Current Stimulation for Treatment of Acute Ischemic Stroke: A Randomized Clinical Trial. JAMA Netw. Open 2023, 6, e2319231. [Google Scholar] [CrossRef]
  213. Turunen, K.E.A.; Laari, S.P.K.; Kauranen, T.V.; Uimonen, J.; Mustanoja, S.; Tatlisumak, T.; Poutiainen, E. Domain-Specific Cognitive Recovery after First-Ever Stroke: A 2-Year Follow-Up. J. Int. Neuropsychol. Soc. 2018, 24, 117–127. [Google Scholar] [CrossRef]
Figure 1. Transcranial direct current stimulation (tDCS) setup including battery operated device, electrodes (cathode and anode), head strap on which electrodes are placed. Electrode placement is based on 10–20 electroencephalogram (EEG) montage. Panel (A) is conventional tDCS set up where the cathode is placed on the right supraorbital region and the anode is placed on the left dorsolateral prefrontal cortex, a montage commonly used for treatment of post-stroke cognitive impairment. Panel (B) is a high definition tDCS in a 4 × 1 set up where 4 cathodes surround the anode, which is placed in the left dorsolateral prefrontal cortex. Created with BioRender.com.
Figure 1. Transcranial direct current stimulation (tDCS) setup including battery operated device, electrodes (cathode and anode), head strap on which electrodes are placed. Electrode placement is based on 10–20 electroencephalogram (EEG) montage. Panel (A) is conventional tDCS set up where the cathode is placed on the right supraorbital region and the anode is placed on the left dorsolateral prefrontal cortex, a montage commonly used for treatment of post-stroke cognitive impairment. Panel (B) is a high definition tDCS in a 4 × 1 set up where 4 cathodes surround the anode, which is placed in the left dorsolateral prefrontal cortex. Created with BioRender.com.
Brainsci 14 00614 g001
Figure 2. Models of stroke recovery. Panel (A) illustrates the Interhemispheric Inhibition model, in which perilesional areas increase activity to compensate for the stroke lesion. The transcallosal inhibitory influence from the contralesional hemisphere leads to an imbalanced inhibitory force on the ipsilesional hemisphere. Under this model, excitatory stimulation is applied to the ipsilesional hemisphere or inhibitory stimulation is applied to the contralesional hemisphere. Panel (B) illustrates the Bimodal Balance model, in which contralesional hemisphere structures form a compensatory reserve network resulting in enhanced cortical excitability of the contralesional hemisphere with the effects (beneficial or detrimental to recovery) dependent on the degree of structural reserve. Under this model, excitatory stimulation is applied to both hemispheres. Created with BioRender.com.
Figure 2. Models of stroke recovery. Panel (A) illustrates the Interhemispheric Inhibition model, in which perilesional areas increase activity to compensate for the stroke lesion. The transcallosal inhibitory influence from the contralesional hemisphere leads to an imbalanced inhibitory force on the ipsilesional hemisphere. Under this model, excitatory stimulation is applied to the ipsilesional hemisphere or inhibitory stimulation is applied to the contralesional hemisphere. Panel (B) illustrates the Bimodal Balance model, in which contralesional hemisphere structures form a compensatory reserve network resulting in enhanced cortical excitability of the contralesional hemisphere with the effects (beneficial or detrimental to recovery) dependent on the degree of structural reserve. Under this model, excitatory stimulation is applied to both hemispheres. Created with BioRender.com.
Brainsci 14 00614 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sloane, K.L.; Hamilton, R.H. Transcranial Direct Current Stimulation to Ameliorate Post-Stroke Cognitive Impairment. Brain Sci. 2024, 14, 614. https://doi.org/10.3390/brainsci14060614

AMA Style

Sloane KL, Hamilton RH. Transcranial Direct Current Stimulation to Ameliorate Post-Stroke Cognitive Impairment. Brain Sciences. 2024; 14(6):614. https://doi.org/10.3390/brainsci14060614

Chicago/Turabian Style

Sloane, Kelly L., and Roy H. Hamilton. 2024. "Transcranial Direct Current Stimulation to Ameliorate Post-Stroke Cognitive Impairment" Brain Sciences 14, no. 6: 614. https://doi.org/10.3390/brainsci14060614

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop