Next Article in Journal
Molecular Characterization of Hepatitis B Virus in People Living with HIV in Rural and Peri-Urban Communities in Botswana
Next Article in Special Issue
Exploring Cognitive Impairments Associated with Primary Open-Angle Glaucoma and Exfoliation Glaucoma
Previous Article in Journal
Embryonic Zebrafish as a Model for Investigating the Interaction between Environmental Pollutants and Neurodegenerative Disorders
Previous Article in Special Issue
Platelet Levels of Brain-Derived Neurotrophic Factor in Adults with Autism Spectrum Disorder: Is There a Specific Association with Autism Spectrum Psychopathology?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Changes in the Ischemic Brain as Non-Invasive Brain Stimulation Targets—TMS and tDCS Mechanisms, Therapeutic Challenges, and Combination Therapies

by
Aleksandra Markowska
* and
Beata Tarnacka
Department of Rehabilitation Medicine, Faculty of Medicine, Warsaw Medical University, Spartańska 1, 02-637 Warsaw, Poland
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(7), 1560; https://doi.org/10.3390/biomedicines12071560
Submission received: 4 June 2024 / Revised: 7 July 2024 / Accepted: 10 July 2024 / Published: 13 July 2024

Abstract

:
Ischemic stroke is one of the leading causes of death and disability. As the currently used neurorehabilitation methods present several limitations, the ongoing research focuses on the use of non-invasive brain stimulation (NIBS) techniques such as transcranial magnetic stimulation (TMS) and transcranial direct current stimulation (tDCS). NIBS methods were demonstrated to modulate neural excitability and improve motor and cognitive functioning in neurodegenerative diseases. However, their mechanisms of action are not fully elucidated, and the clinical outcomes are often unpredictable. This review explores the molecular processes underlying the effects of TMS and tDCS in stroke rehabilitation, including oxidative stress reduction, cell death, stimulation of neurogenesis, and neuroprotective phenotypes of glial cells. A highlight is put on the newly emerging therapeutic targets, such as ferroptotic and pyroptotic pathways. In addition, the issue of interindividual variability is discussed, and the role of neuroimaging techniques is investigated to get closer to personalized medicine. Furthermore, translational challenges of NIBS techniques are analyzed, and limitations of current clinical trials are investigated. The paper concludes with suggestions for further neurorehabilitation stroke treatment, putting the focus on combination and personalized therapies, as well as novel protocols of brain stimulation techniques.

1. Introduction

1.1. Stroke Statistics and Current Rehabilitation Methods

Ischemic stroke is one of the major causes of death and disability. There are over 12.2 million new strokes each year. Globally, one in four people over age 25 will have a stroke in their lifetime. Six and a half million people die from stroke annually [1]. Around one in three stroke survivors will develop clinically significant symptoms of depression [2], 30% will develop anxiety [3], and over 70% will suffer from cognitive deficits [4]. As predicted in thirty-year projections of stroke epidemiology, the number of people living with stroke is estimated to increase by 27% between 2017 and 2047 in the European Union [5]. As the therapeutic time window for reperfusion therapy is narrow [6], significantly limiting the percentage of eligible patients [7,8], there is a tremendous need for effective stroke rehabilitation methods. The current options involve mainly physical therapy, speech therapy, and occupational therapy. However, they are time-consuming, require high patient compliance, and neuropsychiatric complications such as post-stroke depression may seriously reduce patients’ willingness to participate in rehabilitation and delay the recovery process [9,10,11]. Non-invasive brain stimulation (NIBS) is an increasingly used [12,13,14,15], novel, safe, and effective technique and may be a valuable alternative to conventional stroke rehabilitation methods.

1.2. TMS and tDCS: Characteristics and Differences

Transcranial magnetic stimulation and transcranial direct current stimulation are two of the most popular non-invasive brain stimulation (NIBS) techniques [16]. They both modulate the excitability of the cortex and synaptic plasticity, being able to induce long-term potentiation (LTP) and long-term depression (LTD) changes, which contributes to the long-term effects of NIBS techniques, outlasting the period of stimulation [17,18,19]. However, there are differences that may prove important when choosing a particular one in research.
TMS uses a magnetic field to induce an electrical current sufficient to depolarize neurons and trigger action potentials in the stimulated cortical area [20,21]. It involves different modes, including repetitive transcranial stimulation (rTMS) and theta burst stimulation (TBS). rTMS involves delivering equal intervals of magnetic pulses of fixed frequency [22,23]. Low-frequency rTMS (LF-rTMS) (0–2 Hz) has been shown to have an inhibitory effect on cortex excitability, whereas high-frequency rTMS (HF-rTMS) (>5 Hz) has been observed to increase it [24,25]. HF-rTMS was found to improve the glutamatergic synaptic transmission in mouse peri-infarct cortex by regulating integrin α3/AMPA receptors signaling pathway and leading to motor function recovery after stroke [26]. Moreover, it was shown to increase dendritic complexity in mouse prefrontal and primary motor cortex, inducing intracortical rearrangement of neural circuits and modifying cortical connectivity [27,28]. Interestingly, apart from structural remodeling in the medial prefrontal cortex, HF-rTMS-treated mice also showed anti-depressant-like activity [28]. Another form of rTMS is theta burst stimulation, which involves the application of three bursts of high-frequency (50 Hz) pulses with an in-between interval of 200 ms that mimic the endogenous theta rhythms and induce LTP and LTD changes [24,25,29,30]. The key feature of TBS is the long-lasting effect of synaptic plasticity despite the short duration of the application period [31,32]; conventional rTMS sessions last up to 40 min, whereas TBS treatments take 1 to 3 min [25,33].
tDCS, on the other hand, uses weak direct electric current (1–2 mA) through the electrodes placed on distant regions of the scalp as anode and cathode. Contrary to TMS, it is not able to trigger the action potential but alters the resting membrane potential of neurons, changing the probability of discharge [16,21,34]. Analogously to TMS, anodal tDCS increases cortex excitability, and cathodal tDCS decreases it. Importantly, stroke causes a decrease in cortical excitability in the affected hemisphere and a compensatory increase in the unaffected hemisphere, which increases interhemispheric transcallosal inhibition and suppresses the activity in the lesioned hemisphere [20]. Therefore, applying different types of tDCS and TMS at different frequencies may exert an excitatory or inhibitory effect on the ischemic cortex. However, research on non-invasive treatment is mainly limited to the chronic phase of stroke [35,36], and brain stimulation techniques are being increasingly investigated in the early stages. In mice models, cathodal tDCS was found to reverse the maladaptive hyperconnectivity in the subacute phase of stroke and promote motor recovery [37]. Similarly, bihemispheric tDCS, with an anode placed over the lesioned motor cortex and a cathode over the contralateral one, applied in the early subacute phase, accelerated the rate of motor recovery in stroke mouse model [38]. As an important note, encouraging results from the use of tDCS in acute and subacute phases of stroke are pivotal for patients who are not able to start physiotherapy in the early stage. However, Peruzzotti-Jametti et al. demonstrated that, although cathodal stimulation applied in the acute phase of stroke preserved cortical neurons from the ischemic damage and decreased cortical glutamate, the anodal stimulation provoked an increase in lesion volume and blood–brain barrier disruption, which may be the result of early hyperreperfusion and hyperemia [36].
Both TMS and tDCS show effectiveness in functional recovery after stroke, and until now, neither technique has been proven to be more beneficial in stroke rehabilitation. However, there are very few studies comparing their outcomes [39], or they have small sample sizes [40], which affects the significance of the results. Notwithstanding, the differences in their mechanisms of action can prove important in the selection of the most appropriate approach for specific therapeutic goals. As TMS induces a more focal electrical field than tDCS and generates action potentials in specific neural circuits, it is presumed to be more effective in stimulating specific white matter tracts and affecting transcallosal neurons than tDCS [41]. Interestingly, the application of LF-rTMS over contralesional M1 was found to be more beneficial in fine hand movement than for large motor function [40]. As the fine movements require greater engagement of the cortical networks of contralateral M1, higher interhemispheric interaction from the non-lesional hemisphere may inhibit the engagement of the lesioned M1, which may, in turn, negatively affect motor recovery in stroke patients [42]. Therefore, applying LF-rTMS over non-lesioned M1, which decreases high interhemispheric inhibition from the contralesional M1, may improve fine movements. Similarly, bihemispheric tDCS has been demonstrated to modulate intracortical inhibitory pathways in the contralesional primary motor cortex [43] and modulate plasticity within ipsilesional and contralesional motor cortices, leading to reorganization of interhemispheric interactions [44]. Both TMS and tDCS have been found to decrease neuropathic pain, including central post-stroke pain, with rTMS having a slightly superior level of pain relief [45]. In a comparative study of TMS and tDCS on cognitive recovery after stroke, HF-rTMS was found to be the most promising therapeutic option in enhancing global cognitive function, particularly over the left dorsolateral prefrontal cortex (DLPFC). Moreover, dual-tDCS over bilateral DLPFC was shown to be superior to other NIBS in improving memory function. On the other hand, the effects of both stimulation techniques on attention, executive function, and activities of daily living (ADL) were found to be insignificant [46]. A different network meta-analysis confirms that DLPFC is the most promising target for cognitive recovery using NIBS techniques and recognizes HF-rTMS as the superior NIBS technique for ameliorating cognitive impairment. Nevertheless, dual-rTMS was found to be most effective in improving ADL functioning and LF-rTMS in alleviating unilateral spatial neglect [47].
In comparison to large and heavy TMS equipment, tDCS devices are simple, small, and portable and can be used at home [Table 1]. Their costs are also lower than those of TMS devices. However, their disadvantages include low spatial resolution and difficulty in precisely localizing the electric field current [48]. TMS, on the other hand, is characterized by high temporal and spatial resolution, which allows for targeting specific neural circuits [49]. However, they are more expensive, are not portable, and do not allow for home therapy. Possible adverse effects of TMS include seizures and syncope [48]. tDCS has been associated with fatigue, headache, skin redness, itching, and burning sensation under the stimulation electrodes [48]. Both techniques mostly target cortical regions and cannot stimulate subcortical areas without affecting the cortex [48].
As NIBS techniques outlast the period of stimulation [52], a variety of processes affecting synaptic plasticity, cell viability, and neurogenesis must occur. In the next chapters, a detailed analysis of the effect of NIBS on cellular and molecular mechanisms will be presented. Neuroinflammation, oxidative stress, apoptosis, pyroptosis, ferroptosis, and neurogenesis will be explored.

2. Discussion

2.1. Cell Death: Apoptosis, Pyroptosis, Ferroptosis, Necroptosis

The mitochondrial dysfunction and oxidative stress that follow brain ischemic injury lead to apoptotic and non-apoptotic programmed cell death [53,54,55]. After the TLR4/DR (toll-like receptor/death receptor) on the cell membrane receives the inflammatory signal, it triggers apoptosis, pyroptosis, necroptosis, and ferroptosis [56,57].
Contrary to necrosis occurring immediately in the core of ischemic injury, apoptosis occurs in the penumbra zone, around the core, within several hours or days [58,59,60]. In the ischemic brain, two main apoptotic pathways can be activated. In the intrinsic pathway, pro-apoptotic Bcl-2 (B-cell lymphoma 2) family proteins Bax (Bcl-2-associated protein X) and Bak (Bcl-2 homologous antagonist/killer) form pores in the outer mitochondrial membrane which results in the release of intermembrane space proteins such as cytochrome c and apoptosis-inducing factor (AIF) into cytosol which in turn activates the caspases (caspase-3, caspase-9) that cause DNA fragmentation, chromatin condensation and cell destruction [53,54,61]. It is reported that the balance between Bcl-2 and Bax plays a key role in apoptotic mechanisms determining whether the cell will survive or undergo programmed death [55]. Importantly, the decreased Bcl-2/Bax ratio is described in animal stroke models [62]. In the extrinsic pathway, extracellular ligands (such as tumor necrosis factor (TNF)-α, Fas ligand, TRAIL, glucocorticoids) bind to the death receptors on the cell membrane and activate the intracellular caspase-8 and caspase-3 leading to apoptotic cell death [53,54,61].
Importantly, Guo et al. demonstrated that rTMS significantly increased the expression of Bcl-2 and decreased the expression of Bax. Moreover, TUNEL staining detecting DNA breakage during early and late stages of apoptosis [63] showed that rTMS downregulated neuronal apoptosis [64]. Similarly, tDCS treatment was found to increase the Bcl-2/Bax ratio in MCAO rats as well as decrease the caspase-3 level [65]. Furthermore, Zong et al. reported that rTMS inhibited the intrinsic mitochondrial caspase-9/3 apoptotic pathway and attenuated delayed apoptotic cell death in the peri-infarct area in the photothrombotic (PT) rat model of ischemic stroke [66]. Zhou et al. demonstrated that tDCS upregulated BDNF–TrkB and its downstream PI3K/Akt signaling, which antagonizes the pro-apoptotic activity of the Bcl-2 family, such as Bad and Bax [67], eventually protecting neurons from apoptosis [68].
Pyroptosis is a recently discovered pro-inflammatory form of programmed cell death that is triggered by inflammasome induction and mediated by gasdermin (GSDM) family proteins [69,70]. The inflammasome converts procaspase-1 into caspase-1, which cleaves GSDM, releasing an N-terminal fragment (N-GSDM) [56,71]. N-GSDM binds to the cell membrane and perforates it, which eventually leads to the cell rupture and release of pro-inflammatory substances (IL-1β, IL-18). Released inflammatory factors contribute to the pyroptosis of neural cells, glial cells, and endothelial cells, which results in blood–brain barrier (BBB) disruption and irreversible brain damage [56,72]. Luo et al. showed that theta-burst rTMS (iTBS) inhibited the expression of proteins associated with pyroptosis, such as caspase-1, IL-1β, IL-18, ASC, GSDMD, and NLRP1 in the peri-infarcted area and inhibited TLR4/NFκB/NLRP3 signaling pathway modulating microglial activation and inhibiting neuronal pyroptosis [73].
Ferroptosis is an iron-dependent programmed cell death. In an ischemic brain, microglia release substantial amounts of metalloproteinases that disrupt the extracellular matrix and dysregulate the blood–brain barrier. Increased BBB permeability results in iron ions influx into brain tissue and iron overload [74]. Iron is involved in a number of detrimental processes occurring after ischemia, i.e., release of free radicals, excitotoxicity, and inflammatory response [75]. Iron-dependent lipid peroxidation that takes place during ischemic stroke results in oxidative membrane damage and, consequently, cell death [76,77]. The inactivation of glutathione peroxidase 4 (GPX4) that converts toxic lipid hydroperoxides into non-toxic lipid alcohols, protecting against membrane lipid peroxidation, leads to ferroptotic cell death, and its expression tends to be decreased in animal ischemic models [78]. Interestingly, Zhou et al. found that HF-rTMS increased GPX4 levels and decreased ASCL4 and TFRC, key components of ferroptotic processes reversing the reduced GPX4 level and the elevated ASCL4 and TFRC levels in MCAO rats. In addition, it reduced the concentrations of pro-inflammatory factors such as IL-1β, IL-6, and TNF-α in the cerebrospinal fluid [79]. Similarly, Shen et al. observed that intermittent theta burst stimulation increased GPX4 levels [80].

2.2. Oxidative Stress

Lipid degradation of the cell membrane and mitochondrial dysfunction lead to excessive production of free radicals, which cause damage to DNA structure, protein denaturation, and lipid peroxidation [81], thereby causing cell death. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase is the main source of oxygen-reactive species [82]. Following ischemic stroke, its expression rapidly increases in neurons and brain vessels [83]. In experimental stroke models, it has been demonstrated that blocking NADPH activation protects against focal ischemic injury, reducing superoxide generation and improving neurological functioning [82,84]. Following cerebral ischemia, the main mechanism by which ischemic brain damage is reduced is through superoxide dismutase (SOD) enzymes that eliminate O2 by converting it into H2O2 and O2 [85]. Manganese superoxide dismutase (MnSOD) is one of the most important antioxidant cell components [86]. MnSOD deficiency exacerbates cerebral infarction, and the reperfusion after cerebral ischemia reduces the expression of MnSOD [85]. Therefore, increasing its concentration, and thereby, neuroprotective effects, has become the subject of current research.
It was demonstrated that rTMS administration reduced NADPH oxidase activation and superoxide production in the peri-infarct cortical region in the photothrombotic stroke model and increased the MnSOD production, which attenuated oxidative neuronal damage measured by labeling lipid peroxidation, DNA double-strand breaks, and oxidized DNA damage [66]. Administrating tDCS caused an increase in SOD levels in the cerebral ischemia/reperfusion (I/R) model, especially in the c/a-tDCS mode, which is a combination mode of the cathodal current in the ischemia stage and anodal current in the reperfusion period [87].

2.3. Glial Cells

Although it has been previously thought that brain stimulation techniques primarily affect nerve cells [88], glial cells have now become a critical NIBS target.
It was demonstrated that TMS and tDCS can affect the morphology and activation of astrocytes and microglia. In response to tDCS, astrocytes displayed elongated cell bodies with cellular filopodia that were oriented perpendicularly to the direct current electric field [89]. Studies on deep brain stimulation suggest that the stimulation of astrocytes can set in motion the release of gliotransmitters, which can trigger axonal activation [90]. Moreover, tDCS was also shown to enlarge microglial soma size in an adrenergic receptor-dependent manner [91]. However, the role of soma enlargement in positive outcomes of tDCS is not fully understood. Microglial soma enlargement has been associated with inflammatory processes [92], and it is suggested that the pro-inflammatory molecules released by microglia may be involved in synaptic plasticity induced by tDCS [91].
In in vitro models of ischemia, high-frequency repetitive magnetic stimulation (HF-rMS) was shown to have a direct modulatory effect on astrocytes and stimulate the release of trophic factors, including GDNF and PDGF-BB from their secretome, which promoted neuronal survival after ischemic period [93]. Importantly, the presence of astrocytes was shown to be crucial to the beneficial effects induced by HF-rMS after ischemia [94]. tDCS was demonstrated to directly modulate gene expression in astrocytes upregulating BDNF, playing a key role in neuronal plasticity, survival, and growth [95], as well as FOS, the marker of cell activation, differentiation, and proliferation in isolated astrocytes in vitro [96]. Moreover, DCS was shown to promote microglial phagocytic activity, responsible not only for debris clearance but also network remodeling and engulfment of excess synapses, being a sign of neuroplasticity [97,98,99]. Interestingly, microglia can respond to DCS indirectly through neuron–microglia communication as well as can directly perceive weak electrical fields [98].
In the study on rats with middle cerebral artery occlusion (MCAO), rTMS administration inhibited the neurotoxic polarization of astrocytes, maintaining their neuroprotective phenotype. It was shown that rTMS reduced the expression of neurotoxic markers (iNOS), increased the expression of neuroprotective markers (arginase 1), promoted astrocytic synaptic formation, and alleviated neuronal apoptosis, which eventually promoted neurological functional recovery in vivo [100]. Similarly, tDCS was also found to positively influence the recovery of function. Administrating cathodal-tDCS (c-tDCS) in mice with PT stroke was demonstrated to enhance the ramification of microglia at the perilesional region and modulate the phenotype of microglia, shifting its activation towards anti-inflammatory response, indicated by higher expression of anti-inflammatory markers in the ischemic core [101]. Similarly, Walter et al. showed that tDCS received daily by mice with experimental focal cerebral ischemia enhanced neurogenesis in the subventricular zone, diminished microglia polarization toward the neurotoxic CD16/32+ M1 phenotype and stabilized microglia polarization toward neuroprotective CD206+ M2-phenotype [102]. Braun et al. found that c-tDCS promoted the recruitment of oligodendrocyte precursors towards the lesion; however, contrary to the current literature findings on the effects of NIBS on microglia polarization [101,102,103,104], they observed that c-tDCS promoted microglia M1 pro-inflammatory phenotype [105] which suggests that spatiotemporal dynamics of microglia are much more complex and at different phases of ischemic stroke the tDCS effect may differ. Single-cell RNA sequencing and cell–cell communication analysis confirm that within the early stage of acute ischemic stroke, microglia exhibit distinct heterogeneity rather than M1/M2 polarization [106].

2.4. Neurogenesis

Neurogenesis involves the proliferation of neural stem cells, migration of neuroblasts to the infarct zone, and differentiation into neurons [107]. In an adult brain, neurogenesis takes place primarily in two regions: in the subventricular zone (SVZ) located along the lateral ventricles and in the subgranular zone of the dentate gyrus [107,108]. Ischemia can trigger neurogenesis in an adult brain; however, it is insufficient to restore brain function after a stroke [109]. In the ischemic brain, the survival of new neurons is reduced due to the lack of neurotrophic factors and chronic inflammation [110]. Stroke-induced hypoxia was reported to increase Notch signaling in neural stem cells (NSCs), which initiated an irreversible switch from neurogenesis to gliosis [111,112], hindering brain repair by engulfing synapses [113].
In animal stroke models, non-invasive brain stimulation techniques have been shown to facilitate endogenous neural stem cell regeneration. Guo et al. demonstrated that rTMS upregulated the BDNF signaling pathway and promoted neurogenesis as well as suppressed apoptosis in the ipsilateral hippocampus of adult rats with cerebral focal ischemia [64]. Luo et al. found that rTMS promoted the proliferation of neural stem cells in the ischemic penumbra through a Ca2+ influx-dependent phosphorylated AKT/glycogen synthase kinase 3β/β-catenin signaling pathway [114]. Zong et al. reported that continuous theta-burst stimulation (cTBS), a modality of transcranial magnetic stimulation (TMS), significantly expanded the pool of neural progenitor cells and newly generated immature neurons, attenuated their apoptotic death and maintained their survival in the peri-infarct region in a photothrombotic stroke rat model [115]. Furthermore, Peng et al. demonstrated that the combination of human neural stem cells (hNSCs) transplantation and rTMS in a middle cerebral artery occlusion (MCAO) rat model accelerated the functional recovery after ischemic stroke. rTMS promoted the neural differentiation of hNSCs after transplantation in rats, and the combined therapy synergistically enhanced neurogenesis in the SVZ through the BDNF-TrkB signaling pathway and increased the expression of neurotrophin BDNF [116].
Studies using tDCS demonstrated similar outcomes. Lei et al. showed that bilateral tDCS promoted the migration of NSC-derived neuroblasts from SVZ toward the cathode direction into the post-stroke striatum, protects against neuronal death, and improves the functional recovery of rats subjected to ischemia-reperfusion injury [117]. Zhang et al. found that tDCS promoted the proliferation of NSCs in the subventricular zone in the MCAO rat model, accelerated NSC migration from the SVZ to the ischemic site, and promoted NSC differentiation to oligodendrocytes and neurons by inhibiting Notch 1 signaling pathway [118]. Braun et al. demonstrated that tDCS, independently of polarity, increased the area covered by neuroblasts in the SVZ of the ipsilateral hemisphere but had no effect on the dentate gyrus of the hippocampus or the contralateral hemisphere [105]. Pikhovych et al. found that cathodal, more than anodal, tDCS induced neurogenesis in the mouse brain [119].
Importantly, applying transcranial magnetic stimulation on the stroke hemisphere of patients with sub-acute stroke was demonstrated to modulate endogenous neuroplasticity. Plasma levels of neurogenesis and axonogenesis biomarkers such as miR-25 and netrin-1 were significantly increased in the rTMS-treated group [120]. Netrin-1 facilitates synaptic formation and axonal regeneration, and miR-25 promotes adult neural stem cell proliferation. However, it was also found to downregulate the level of BDNF, contradicting the results of the aforementioned studies. Interestingly, the level of BDNF was found to be both increased [64,116,121,122] and decreased [123,124] in the research investigating the effect of TMS, which suggests that different frequencies and durations of this technique, as well as population characteristics, may play a pivotal role in the final plasma BDNF levels [124]. Nonetheless, the outcome of the study showed an association between rTMS and neurogenesis/axonogenesis biomarker enhancement, indicating that HF-rTMS may modulate endogenous neurogenesis and axonal sprouting after ischemic stroke in humans.

2.5. Combination Therapies

As the TMS and tDCS have been shown to enhance endogenous neural stem cell proliferation, migration, survival, and differentiation [114,115,116], NIBS techniques are increasingly used in combination with stem cell transplantations in animal models to improve the differentiation of exogenous stem cells into mature neurons and improve their integration into neural networks in the lesioned brain. Combining TMS with BMSCs displayed a more efficient recovery in rats with spine injury in comparison with monotherapy by reducing neuronal apoptosis, increasing neurotrophic expression levels (GAP-43, NGF, BDNF), and downregulating the expression of glial fibrillary acidic protein (GFAP) [125], the marker of astrocytic activation mediating glial scar formation which is also released into the bloodstream after brain tissue damage and is associated with stroke severity [126,127]. Similarly, combination of rTMS with hMSCs in Parkinson’s disease rat model was shown to create a favorable microenvironment by elevating the expression of neurotrophic factors (BDNF, GDNF, NGF, PDGF), enhancing the expression of pro-inflammatory cytokines (IL-10) and suppressing pro-inflammatory cytokines (TNF-α, IFN-γ) to amplify immune modulation effects in a synergistic manner and the combination treatment was more effective than monotherapies [128]. The mechanisms investigated in the aforementioned neurological disorders, such as preventing glial scar formation, neuroinflammation, and cell death, as well as enhancing the expression of neurotrophic factors, are also important in stroke treatment. So far, the combination therapy in stroke animal models involved the use of human neural stem cells (hNSCs) in rats after ischemic stroke. rTMS was shown to promote the neural differentiation of hNSCs after transplantation in rats, and the combined therapy synergistically enhanced neurogenesis in the SVZ through the BDNF-TrkB signaling pathway and increased the expression of neurotrophin BDNF [116]. Most importantly, combination therapy has also demonstrated improvements in clinical outcomes in a stroke patient. A case report involving administrating MSCs and rTMS following the acute phase of the ischemic stroke has been shown to improve patient’s motor strength and cognitive functions, which was assessed by the National Institutes of Health Stroke Scale (NIHSS), Fugl-Meyer Assessment and Montreal Cognitive Assessment-Indonesian version (MoCA-INA) [129].

2.6. Personalised Therapy

Although non-invasive brain stimulation techniques have been increasingly used in stroke rehabilitation research, there is considerable variability in their clinical effects, often making their outcomes unpredictable. The source of variability can be divided into two groups; the first one is the result of different stimulation protocols and inconsistent methodology of studies, and the second one is caused by interindividual variations such as brain anatomy, connectivity, cortical excitability, severity of stroke lesions, extent of corticospinal tract damage, baseline level of function, neurochemistry, genetics and age [130] [Figure 1]. Quantification of these factors can be achieved through neuroimaging techniques such as mainly MRI and its modalities, including fractional anisotropy (FA) of diffusion tensor imaging (DTI), structural MRI (sMRI), and functional MRI (fMRI) [131]. A study analyzing motor networks in responder and non-responder groups using fMRI showed that responders to rTMS and tDCS presented an increased involvement of contralesional M1, greater interhemispheric connectivity, and higher motor network efficiency before the stimulation [132]. The outcome suggests that brain stimulation techniques may be effective in patients with disrupted network balance but with functional interhemispheric connectivity. Similarly, the integrity of interhemispheric connections was shown to be crucial in post-stroke neglect recovery and response to cTBS [133]. The stroke severity, depending on the extent of white matter tract damage, was also found to be a predictor of NIBS effectiveness [131,134,135,136]. tDCS was demonstrated to improve limb control for patients with mild impairment and worsen it for patients with moderate to severe impairment [134]. Fractional anisotropy was used to measure the asymmetry between the posterior limbs of the internal capsule, determining the CST integrity, making the neuroimaging techniques an important tool in identifying non-responder groups. Moreover, gene polymorphisms may also affect the neuroplastic response to brain stimulation techniques in stroke patients. Val66Val carriers of the BDNF gene showed a decrease in cortical excitability, while Val66Met carriers presented an initial increase in cortical excitability followed by delayed inhibitory response 30 min after the stimulation [137]. Val66Met polymorphism was associated with less improvement of post-stroke aphasia after cTBS treatment in comparison with Val66Val, which suggests that genotype plays an important role in responsiveness to brain stimulation [138]. Current clinical studies focus on personalized tDCS using individual electrical field models [139,140]. As the brain and cranial structure affect the distribution of current density [130], simulation models based on anatomical data are created to investigate optimal current parameters. Individual electrical field models are researched to personalize the current strength and placement of the electrode grid on the scalp [139]. New software is being developed to analyze magnetic resonance images of stroke patients to generate brain models and calculate the magnitude of the electric field generated by tDCS. Importantly, lesion location and brain atrophy are taken into consideration in order to maximize the effects of tDCS [140].

3. Conclusion, Limitations, and Directions for Further Research

Although applying transcranial magnetic stimulation and transcranial direct current stimulation have been associated with various neuroprotective and neuroregenerative effects, still many challenges of non-invasive brain stimulation therapies need to be overcome.
There is a large heterogeneity of stimulation parameters in the studies, which makes it difficult to accurately compare the results in meta-analyses and further apply them in clinical practice. Developing protocols with exact parameters involving optimal frequencies, spatial distribution, pulse numbers, stimulation time, and intervals personalized to different groups of patients need to be established in order to ensure their maximum safety and efficacy.
The majority of studies involve in vitro or animal models, which present several translational challenges. In vitro studies cannot accurately mirror the complex brain environments in the ischemic brain. As an example, the application of the electrical current across the monolayer of astrocytes in vitro may have different effects than across the brain [96]. Moreover, the intricate interactions between glial cells, neural cells, and endothelial cells cannot be properly investigated in vitro. Furthermore, in vivo studies apply rTMS coils made for humans to animals, and different head-to-coil size ratios may lead to reduced stimulation efficiency [141]. Importantly, the brain structure of rodents, used most frequently in stroke rehabilitation research, is different from that of humans. Mice and rats have smooth brain structures [142] with a different geometry than the folded human cortex, which may affect the properties of the electric field of rTMS [141]. In addition, in the research on microglia polarization, it is important to note that the microglia distribution profile is different in the human and rodent brains [143]. The translational issues and limitations of current clinical studies make it imperative to conduct more randomized controlled trials.
What is important from the clinical point of view is that current trials have short treatment duration [144], involve small samples not reaching statistical significance [145,146,147], and are not followed by long-term assessments to investigate permanent changes in the brain after the end of the stimulation period [144,147,148]. The results are mixed, showing as well no significant changes in functional connectivity [149]. Furthermore, there are very limited dose comparison studies, and largely varying stimulation doses between research papers may count for the mixed outcomes [150]. Meta-analysis and meta-regression results suggest a dose–response relationship with electrode size, charge density, and current density, although the lack of detailed dosages and administration methods in the analyzed studies were the main limitations [150]. Moreover, they lack neurophysiological studies exploring the neuroplasticity mechanisms and assessing the clinical improvement after the intervention [147]. Importantly, publication bias [151] and low quality of studies [152] with methodological limitations [153] of NIBS clinical trials are common outcomes of current meta-analyses, leaving the most effective stimulation mode still to be determined [154].
The underlying mechanisms of not only non-invasive brain stimulation techniques but also molecular changes occurring in the ischemic brain are still not fully elucidated, and more research should focus on newly emerging therapeutic targets such as ferroptotic and pyroptotic cell death.
In addition, as novel protocols of TMS, such as intermittent (iTBS) and continuous theta burst stimulation (cTBS), present several advantages over standard TMS, i.e., long-lasting effects despite short administration periods, more research should focus on their clinical applications.
Noteworthy are the numerous studies indicating the synergistic effects of combining brain stimulation techniques with other neuroregenerative and neurorehabilitation strategies [Figure 2]. Encouraging results have been obtained using the combined therapy with stem cells transplantations [116,128,129], nanomaterials such as superparamagnetic iron oxide nanoparticles (SPIONs) [155], acupuncture [156], pharmacotherapy [157,158,159], botulinum toxin [160,161], physical exercise and movement therapy [162,163,164,165,166,167,168], virtual reality [165,169,170], working memory tasks [171], mindfulness-based stress reduction [160], occupational therapy [161,172,173], music therapy [174], as well as combining TMS with tDCS [175].
Although more research is still necessary to link the therapeutic effects of NIBS with molecular mechanisms occurring in the ischemic brain, recent progress in preclinical and clinical studies, and growing understanding of factors affecting responsiveness to brain stimulation techniques provide a promising basis for finding an effective form of stroke rehabilitation method.

Author Contributions

Conceptualization, literature search, and writing of the manuscript, A.M.; work revision and “limitations” chapter suggestions, B.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AKT(=PKB) protein kinase B
ASCL4acyl-coa synthetase long-chain family member 4
Bakbcl-2 homologous antagonist/killer
Baxbcl-2-associated protein X
BBBblood-brain barrier
Bcl-2b-cell lymphoma 2
BDNFbrain-derived neurotrophic factor
BMSCbone marrow-derived mesenchymal stem cells
COX2cyclooxygenase 2
DLPFCdorsolateral prefrontal cortex
DRdeath receptor
DTIdiffusion tensor imaging
FAfractional anisotropy
GAP-43growth-associated protein 43
GDNFglial cell line-derived neurotrophic factor
GFAPglial fibrillary acidic protein
GPX4glutathione peroxidase 4
GSDMgasdermin
IFNinterferon
iNOSinducible nitric oxide synthase
LTDlong-term depression
LTPlong-term potentiation
MCAOmiddle cerebral artery occlusion
MRImagnetic resonance imaging
MSCsmesenchymal stem cells
NADPHnicotinamide adenine dinucleotide phosphate
NGFnerve growth factor
NIBSnon-invasive brain stimulation
NIHSSNational Institutes of Health Stroke Scale
NLRPnucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing
NSCneural stem cells
PDGFplatelet-derived growth factor
PDGF-BBplatelet-derived growth factor—two b subunits
PI3Kphosphoinositide 3-kinase
PTphotothrombotic
SODsuperoxide dismutase
SPIONsuperparamagnetic iron oxide nanoparticles
SVZsubventricular zone
TBStheta burst stimulation
tDCStranscranial direct current stimulation
TFRCtransferrin receptor
TLR4toll-like receptor
TMStranscranial magnetic stimulation
TNFtumor necrosis factor
TRAILTNF-related apoptosis-inducing ligand
TUNELterminal deoxynucleotidyl transferase dUTP nick end labeling

References

  1. Feigin, V.L.; Brainin, M.; Norrving, B.; Martins, S.; Sacco, R.L.; Hacke, W.; Fisher, M.; Pandian, J.; Lindsay, P. World Stroke Organization (WSO): Global Stroke Fact Sheet 2022. Int. J. Stroke 2022, 17, 18–29. [Google Scholar] [CrossRef] [PubMed]
  2. Almeida, O.P. Stroke, depression, and self-harm in later life. Curr. Opin. Psychiatry 2023, 36, 371–375. [Google Scholar] [CrossRef] [PubMed]
  3. Rafsten, L.; Danielsson, A.; Sunnerhagen, K.S. Anxiety after stroke: A systematic review and meta-analysis. J. Rehabil. Med. 2018, 50, 769–778. [Google Scholar] [CrossRef]
  4. Rost, N.S.; Brodtmann, A.; Pase, M.P.; Veluw, S.J.v.; Biffi, A.; Duering, M.; Hinman, J.D.; Dichgans, M. Post-Stroke Cognitive Impairment and Dementia. Circ. Res. 2022, 130, 1252–1271. [Google Scholar] [CrossRef] [PubMed]
  5. Wafa, H.A.; Wolfe, C.D.A.; Emmett, E.; Roth, G.A.; Johnson, C.O.; Wang, Y. Burden of Stroke in Europe: Thirty-Year Projections of Incidence, Prevalence, Deaths, and Disability-Adjusted Life Years. Stroke 2020, 51, 2418–2427. [Google Scholar] [CrossRef] [PubMed]
  6. Scheldeman, L.; Wouters, A.; Lemmens, R. Imaging selection for reperfusion therapy in acute ischemic stroke beyond the conventional time window. J. Neurol. 2022, 269, 1715–1723. [Google Scholar] [CrossRef] [PubMed]
  7. Fang, M.C.; Cutler, D.M.; Rosen, A.B. Trends in thrombolytic use for ischemic stroke in the United States. J. Hosp. Med. 2010, 5, 406–409. [Google Scholar] [CrossRef] [PubMed]
  8. Acherqui, M.; Khattab, H.; Habtany, Y.; Amzil, R.; Bellakhdar, S.; Otmani, H.E.; Moutawakil, B.E.; Rafai, M.A. Assessment of eligibility for thrombolysis in acute ischaemic stroke patients in Morocco. Pan Afr. Med. J. 2020, 36, 351. [Google Scholar] [CrossRef] [PubMed]
  9. Xie, J.; Geng, X.; Fan, F.; Fu, X.; He, S.; Li, T. The efficacy of therapies for post-stroke depression in aging: An umbrella review. Front. Aging Neurosci. 2022, 14, 993250. [Google Scholar] [CrossRef] [PubMed]
  10. Tombak, Y.; Karaahmet, O.Z.; Umay, E.; Tombak, A.; Gurcay, E. Factors influencing the willingness to participate in rehabilitation in patients with subacute stroke. J. Clin. Neurosci. 2023, 116, 99–103. [Google Scholar] [CrossRef] [PubMed]
  11. Sheng, R.; Chen, C.; Chen, H.; Yu, P. Repetitive transcranial magnetic stimulation for stroke rehabilitation: Insights into the molecular and cellular mechanisms of neuroinflammation. Front. Immunol. 2023, 14, 1197422. [Google Scholar] [CrossRef] [PubMed]
  12. Battaglia, S.; Nazzi, C.; Fullana, M.A.; di Pellegrino, G.; Borgomaneri, S. ‘Nip it in the bud’: Low-frequency rTMS of the prefrontal cortex disrupts threat memory consolidation in humans. Behav. Res. Ther. 2024, 178, 104548. [Google Scholar] [CrossRef] [PubMed]
  13. Battaglia, S.; Avenanti, A.; Vécsei, L.; Tanaka, M. Neurodegeneration in Cognitive Impairment and Mood Disorders for Experimental, Clinical and Translational Neuropsychiatry. Biomedicines 2024, 12, 574. [Google Scholar] [CrossRef] [PubMed]
  14. Battaglia, S.; Nazzi, C.; Di Fazio, C.; Borgomaneri, S. The role of pre-supplementary motor cortex in action control with emotional stimuli: A repetitive transcranial magnetic stimulation study. Ann. N. Y. Acad. Sci. 2024, 1536, 151–166. [Google Scholar] [CrossRef] [PubMed]
  15. Tanaka, M.; Vécsei, L. A Decade of Dedication: Pioneering Perspectives on Neurological Diseases and Mental Illnesses. Biomedicines 2024, 12, 1083. [Google Scholar] [CrossRef] [PubMed]
  16. Sanches, C.; Stengel, C.; Godard, J.; Mertz, J.; Teichmann, M.; Migliaccio, R.; Valero-Cabré, A. Past, Present, and Future of Non-invasive Brain Stimulation Approaches to Treat Cognitive Impairment in Neurodegenerative Diseases: Time for a Comprehensive Critical Review. Front. Aging Neurosci. 2020, 12, 578339. [Google Scholar] [CrossRef] [PubMed]
  17. Cirillo, G.; Di Pino, G.; Capone, F.; Ranieri, F.; Florio, L.; Todisco, V.; Tedeschi, G.; Funke, K.; Di Lazzaro, V. Neurobiological after-effects of non-invasive brain stimulation. Brain Stimul. 2017, 10, 1–18. [Google Scholar] [CrossRef] [PubMed]
  18. Antal, A.; Luber, B.; Brem, A.K.; Bikson, M.; Brunoni, A.R.; Cohen Kadosh, R.; Dubljević, V.; Fecteau, S.; Ferreri, F.; Flöel, A.; et al. Non-invasive brain stimulation and neuroenhancement. Clin. Neurophysiol. Pract. 2022, 7, 146–165. [Google Scholar] [CrossRef] [PubMed]
  19. Chisari, C.; Fanciullacci, C.; Lamola, G.; Rossi, B.; Cohen, L.G. NIBS-driven brain plasticity. Arch. Ital. Biol. 2014, 152, 247–258. [Google Scholar] [CrossRef]
  20. Kesikburun, S. Non-invasive brain stimulation in rehabilitation. Turk. J. Phys. Med. Rehabil. 2022, 68, 1–8. [Google Scholar] [CrossRef] [PubMed]
  21. Xiong, H.Y.; Zheng, J.J.; Wang, X.Q. Non-invasive Brain Stimulation for Chronic Pain: State of the Art and Future Directions. Front. Mol. Neurosci. 2022, 15, 888716. [Google Scholar] [CrossRef] [PubMed]
  22. Cole, E.; O’Sullivan, S.J.; Tik, M.; Williams, N.R. Accelerated Theta Burst Stimulation: Safety, Efficacy, and Future Advancements. Biol. Psychiatry 2024, 95, 523–535. [Google Scholar] [CrossRef] [PubMed]
  23. Bai, Y.W.; Yang, Q.H.; Chen, P.J.; Wang, X.Q. Repetitive transcranial magnetic stimulation regulates neuroinflammation in neuropathic pain. Front. Immunol. 2023, 14, 1172293. [Google Scholar] [CrossRef] [PubMed]
  24. Schambra, H.M. Repetitive Transcranial Magnetic Stimulation for Upper Extremity Motor Recovery: Does It Help? Curr. Neurol. Neurosci. Rep. 2018, 18, 97. [Google Scholar] [CrossRef] [PubMed]
  25. Chung, S.W.; Hoy, K.E.; Fitzgerald, P.B. Theta-burst stimulation: A new form of TMS treatment for depression? Depress. Anxiety 2015, 32, 182–192. [Google Scholar] [CrossRef] [PubMed]
  26. Liu, L.; Hu, H.; Wu, J.; Koleske, A.J.; Chen, H.; Wang, N.; Yu, K.; Wu, Y.; Xiao, X.; Zhang, Q. Integrin α3 is required for high-frequency repetitive transcranial magnetic stimulation-induced glutamatergic synaptic transmission in mice with ischemia. CNS Neurosci. Ther. 2024, 30, e14498. [Google Scholar] [CrossRef] [PubMed]
  27. Cambiaghi, M.; Cherchi, L.; Masin, L.; Infortuna, C.; Briski, N.; Caviasco, C.; Hazaveh, S.; Han, Z.; Buffelli, M.; Battaglia, F. High-frequency repetitive transcranial magnetic stimulation enhances layer II/III morphological dendritic plasticity in mouse primary motor cortex. Behav. Brain Res. 2021, 410, 113352. [Google Scholar] [CrossRef] [PubMed]
  28. Cambiaghi, M.; Infortuna, C.; Gualano, F.; Elsamadisi, A.; Malik, W.; Buffelli, M.; Han, Z.; Solhkhah, R.; Thomas, F.P.; Battaglia, F. High-frequency rTMS modulates emotional behaviors and structural plasticity in layers II/III and V of the mPFC. Front. Cell Neurosci. 2022, 16, 1082211. [Google Scholar] [CrossRef] [PubMed]
  29. Solomon, E.A.; Sperling, M.R.; Sharan, A.D.; Wanda, P.A.; Levy, D.F.; Lyalenko, A.; Pedisich, I.; Rizzuto, D.S.; Kahana, M.J. Theta-burst stimulation entrains frequency-specific oscillatory responses. Brain Stimul. 2021, 14, 1271–1284. [Google Scholar] [CrossRef] [PubMed]
  30. Khan, F.; Chevidikunnan, F. Theta burst stimulation a new paradigm of non-invasive brain stimulation for post-stroke upper limb motor rehabilitation. Turk. J. Phys. Med. Rehabil. 2017, 63, 193–196. [Google Scholar] [CrossRef]
  31. William, M.; McDonald, M.D. Theta Burst TMS Technology: Great Promise and a Lot to Learn. Am. J. Psychiatry 2024, 181, 14–15. [Google Scholar] [CrossRef]
  32. Huang, Y.-Z.; Edwards, M.J.; Rounis, E.; Bhatia, K.P.; Rothwell, J.C. Theta Burst Stimulation of the Human Motor Cortex. Neuron 2005, 45, 201–206. [Google Scholar] [CrossRef] [PubMed]
  33. Voigt, J.D.; Leuchter, A.F.; Carpenter, L.L. Theta burst stimulation for the acute treatment of major depressive disorder: A systematic review and meta-analysis. Transl. Psychiatry 2021, 11, 330. [Google Scholar] [CrossRef] [PubMed]
  34. Woods, A.J.; Antal, A.; Bikson, M.; Boggio, P.S.; Brunoni, A.R.; Celnik, P.; Cohen, L.G.; Fregni, F.; Herrmann, C.S.; Kappenman, E.S.; et al. A technical guide to tDCS, and related non-invasive brain stimulation tools. Clin. Neurophysiol. 2016, 127, 1031–1048. [Google Scholar] [CrossRef] [PubMed]
  35. Bornheim, S.; Croisier, J.L.; Maquet, P.; Kaux, J.F. Transcranial direct current stimulation associated with physical-therapy in acute stroke patients—A randomized, triple blind, sham-controlled study. Brain Stimul. 2020, 13, 329–336. [Google Scholar] [CrossRef] [PubMed]
  36. Peruzzotti-Jametti, L.; Cambiaghi, M.; Bacigaluppi, M.; Gallizioli, M.; Gaude, E.; Mari, S.; Sandrone, S.; Cursi, M.; Teneud, L.; Comi, G.; et al. Safety and efficacy of transcranial direct current stimulation in acute experimental ischemic stroke. Stroke 2013, 44, 3166–3174. [Google Scholar] [CrossRef] [PubMed]
  37. Blaschke, S.J.; Vlachakis, S.; Pallast, N.; Walter, H.L.; Volz, L.J.; Wiedermann, D.; Fink, G.R.; Hoehn, M.; Aswendt, M.; Schroeter, M.; et al. Transcranial Direct Current Stimulation Reverses Stroke-Induced Network Alterations in Mice. Stroke 2023, 54, 2145–2155. [Google Scholar] [CrossRef] [PubMed]
  38. Longo, V.; Barbati, S.A.; Re, A.; Paciello, F.; Bolla, M.; Rinaudo, M.; Miraglia, F.; Alù, F.; Di Donna, M.G.; Vecchio, F.; et al. Transcranial Direct Current Stimulation Enhances Neuroplasticity and Accelerates Motor Recovery in a Stroke Mouse Model. Stroke 2022, 53, 1746–1758. [Google Scholar] [CrossRef] [PubMed]
  39. Calderón, M.A.F.; Jiménez, L.O.; Ledesma, M.J.S. Transcranial Magnetic Stimulation versus Transcranial Direct Current Stimulation as neuromodulatory techniques in stroke rehabilitation. In Proceedings of the Sixth International Conference on Technological Ecosystems for Enhancing Multiculturality, Salamanca, Spain, 24–26 October 2018; pp. 422–427. [Google Scholar]
  40. Doris Miu, K.Y.; Kok, C.; Leung, S.S.; Chan, E.Y.L.; Wong, E. Comparison of Repetitive Transcranial Magnetic Stimulation and Transcranial Direct Current Stimulation on Upper Limb Recovery among Patients with Recent Stroke. Ann. Rehabil. Med. 2020, 44, 428–437. [Google Scholar] [CrossRef] [PubMed]
  41. Nicolo, P.; Magnin, C.; Pedrazzini, E.; Plomp, G.; Mottaz, A.; Schnider, A.; Guggisberg, A.G. Comparison of Neuroplastic Responses to Cathodal Transcranial Direct Current Stimulation and Continuous Theta Burst Stimulation in Subacute Stroke. Arch. Phys. Med. Rehabil. 2018, 99, 862–872.e861. [Google Scholar] [CrossRef] [PubMed]
  42. 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] [PubMed]
  43. Goodwill, A.M.; Teo, W.P.; Morgan, P.; Daly, R.M.; Kidgell, D.J. Bihemispheric-tDCS and Upper Limb Rehabilitation Improves Retention of Motor Function in Chronic Stroke: A Pilot Study. Front. Hum. Neurosci. 2016, 10, 258. [Google Scholar] [CrossRef] [PubMed]
  44. Bolognini, N.; Vallar, G.; Casati, C.; Latif, L.A.; El-Nazer, R.; Williams, J.; Banco, E.; Macea, D.D.; Tesio, L.; Chessa, C.; et al. Neurophysiological and Behavioral Effects of tDCS Combined with Constraint-Induced Movement Therapy in Poststroke Patients. Neurorehabilit. Neural Repair 2011, 25, 819–829. [Google Scholar] [CrossRef] [PubMed]
  45. André-Obadia, N.; Hodaj, H.; Hodaj, E.; Simon, E.; Delon-Martin, C.; Garcia-Larrea, L. Better Fields or Currents? A Head-to-Head Comparison of Transcranial Magnetic (rTMS) Versus Direct Current Stimulation (tDCS) for Neuropathic Pain. Neurotherapeutics 2023, 20, 207–219. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, Y.; Liu, W.; Chen, J.; Bai, J.; Yu, H.; Ma, H.; Rao, J.; Xu, G. Comparative efficacy of different noninvasive brain stimulation therapies for recovery of global cognitive function, attention, memory, and executive function after stroke: A network meta-analysis of randomized controlled trials. Ther. Adv. Chronic Dis. 2023, 14, 20406223231168754. [Google Scholar] [CrossRef] [PubMed]
  47. Yan, M.; Liu, J.; Guo, Y.; Hou, Q.; Song, J.; Wang, X.; Yu, W.; Lü, Y. Comparative efficacy of non-invasive brain stimulation for post-stroke cognitive impairment: A network meta-analysis. Aging Clin. Exp. Res. 2024, 36, 37. [Google Scholar] [CrossRef] [PubMed]
  48. Bhattacharya, A.; Mrudula, K.; Sreepada, S.S.; Sathyaprabha, T.N.; Pal, P.K.; Chen, R.; Udupa, K. An Overview of Noninvasive Brain Stimulation: Basic Principles and Clinical Applications. Can. J. Neurol. Sci./J. Can. Des Sci. Neurol. 2022, 49, 479–492. [Google Scholar] [CrossRef] [PubMed]
  49. Priori, A.; Hallett, M.; Rothwell, J.C. Repetitive transcranial magnetic stimulation or transcranial direct current stimulation? Brain Stimul. 2009, 2, 241–245. [Google Scholar] [CrossRef] [PubMed]
  50. Wick, S. What Are TMS Machines & How They Work. Available online: https://www.neuraliatms.com.au/tms-therapy/machine/ (accessed on 4 May 2024).
  51. What Is Transcranial Direct Current Stimulation? Available online: https://neuromodec.org/what-is-transcranial-direct-current-stimulation-tdcs/ (accessed on 4 May 2024).
  52. Huang, Y.Z.; Lu, M.K.; Antal, A.; Classen, J.; Nitsche, M.; Ziemann, U.; Ridding, M.; Hamada, M.; Ugawa, Y.; Jaberzadeh, S.; et al. Plasticity induced by non-invasive transcranial brain stimulation: A position paper. Clin. Neurophysiol. 2017, 128, 2318–2329. [Google Scholar] [CrossRef]
  53. Nguyen, T.T.; Wei, S.; Nguyen, T.H.; Jo, Y.; Zhang, Y.; Park, W.; Gariani, K.; Oh, C.-M.; Kim, H.H.; Ha, K.-T.; et al. Mitochondria-associated programmed cell death as a therapeutic target for age-related disease. Exp. Mol. Med. 2023, 55, 1595–1619. [Google Scholar] [CrossRef]
  54. Bayir, H.; Kagan, V.E. Bench-to-bedside review: Mitochondrial injury, oxidative stress and apoptosis—There is nothing more practical than a good theory. Crit. Care 2008, 12, 206. [Google Scholar] [CrossRef] [PubMed]
  55. Zhang, B.; Zhang, H.-X.; Shi, S.-T.; Bai, Y.-L.; Zhe, X.; Zhang, S.-J.; Li, Y.-J. Interleukin-11 treatment protected against cerebral ischemia/reperfusion injury. Biomed. Pharmacother. 2019, 115, 108816. [Google Scholar] [CrossRef] [PubMed]
  56. Long, J.; Sun, Y.; Liu, S.; Yang, S.; Chen, C.; Zhang, Z.; Chu, S.; Yang, Y.; Pei, G.; Lin, M.; et al. Targeting pyroptosis as a preventive and therapeutic approach for stroke. Cell Death Discov. 2023, 9, 155. [Google Scholar] [CrossRef] [PubMed]
  57. Gao, W.; Wang, X.; Zhou, Y.; Wang, X.; Yu, Y. Autophagy, ferroptosis, pyroptosis, and necroptosis in tumor immunotherapy. Signal Transduct. Target. Ther. 2022, 7, 196. [Google Scholar] [CrossRef] [PubMed]
  58. Radak, D.; Katsiki, N.; Resanovic, I.; Jovanovic, A.; Sudar-Milovanovic, E.; Zafirovic, S.; Mousad, A.S.; Isenovic, R.E. Apoptosis and Acute Brain Ischemia in Ischemic Stroke. Curr. Vasc. Pharmacol. 2017, 15, 115–122. [Google Scholar] [CrossRef] [PubMed]
  59. Hou, Y.; Wang, K.; Wan, W.; Cheng, Y.; Pu, X.; Ye, X. Resveratrol provides neuroprotection by regulating the JAK2/STAT3/PI3K/AKT/mTOR pathway after stroke in rats. Genes Dis. 2018, 5, 245–255. [Google Scholar] [CrossRef] [PubMed]
  60. Li, Z.; Xiao, G.; Wang, H.; He, S.; Zhu, Y. A preparation of Ginkgo biloba L. leaves extract inhibits the apoptosis of hippocampal neurons in post-stroke mice via regulating the expression of Bax/Bcl-2 and Caspase-3. J. Ethnopharmacol. 2021, 280, 114481. [Google Scholar] [CrossRef] [PubMed]
  61. Uzdensky, A.B. Apoptosis regulation in the penumbra after ischemic stroke: Expression of pro- and antiapoptotic proteins. Apoptosis 2019, 24, 687–702. [Google Scholar] [CrossRef] [PubMed]
  62. Xu, D.; Kong, T.; Shao, Z.; Liu, M.; Zhang, R.; Zhang, S.; Kong, Q.; Chen, J.; Cheng, B.; Wang, C. Orexin-A alleviates astrocytic apoptosis and inflammation via inhibiting OX1R-mediated NF-κB and MAPK signaling pathways in cerebral ischemia/reperfusion injury. Biochim. Biophys. Acta BBA-Mol. Basis Dis. 2021, 1867, 166230. [Google Scholar] [CrossRef] [PubMed]
  63. Mirzayans, R.; Murray, D. Do TUNEL and Other Apoptosis Assays Detect Cell Death in Preclinical Studies? Int. J. Mol. Sci. 2020, 21, 9090. [Google Scholar] [CrossRef]
  64. Guo, F.; Lou, J.; Han, X.; Deng, Y.; Huang, X. Repetitive Transcranial Magnetic Stimulation Ameliorates Cognitive Impairment by Enhancing Neurogenesis and Suppressing Apoptosis in the Hippocampus in Rats with Ischemic Stroke. Front. Physiol. 2017, 8, 559. [Google Scholar] [CrossRef] [PubMed]
  65. Zhang, K.Y.; Rui, G.; Zhang, J.P.; Guo, L.; An, G.Z.; Lin, J.J.; He, W.; Ding, G.R. Cathodal tDCS exerts neuroprotective effect in rat brain after acute ischemic stroke. BMC Neurosci. 2020, 21, 21. [Google Scholar] [CrossRef] [PubMed]
  66. Zong, X.; Dong, Y.; Li, Y.; Yang, L.; Li, Y.; Yang, B.; Tucker, L.; Zhao, N.; Brann, D.W.; Yan, X.; et al. Beneficial Effects of Theta-Burst Transcranial Magnetic Stimulation on Stroke Injury via Improving Neuronal Microenvironment and Mitochondrial Integrity. Transl. Stroke Res. 2020, 11, 450–467. [Google Scholar] [CrossRef] [PubMed]
  67. Sussman, M.A. Mitochondrial integrity: Preservation through Akt/Pim-1 kinase signaling in the cardiomyocyte. Expert Rev. Cardiovasc. Ther. 2009, 7, 929–938. [Google Scholar] [CrossRef] [PubMed]
  68. Zhou, Q.; Chen, Y.; Tang, H.; Zhang, L.; Ma, Y.; Bai, D.; Kong, Y. Transcranial direct current stimulation alleviated ischemic stroke induced injury involving the BDNF-TrkB signaling axis in rats. Heliyon 2023, 9, e14946. [Google Scholar] [CrossRef] [PubMed]
  69. Li, W.; Shen, N.; Kong, L.; Huang, H.; Wang, X.; Zhang, Y.; Wang, G.; Xu, P.; Hu, W. STING mediates microglial pyroptosis via interaction with NLRP3 in cerebral ischaemic stroke. Stroke Vasc. Neurol. 2024, 9, 153–164. [Google Scholar] [CrossRef] [PubMed]
  70. Li, J.; Xu, P.; Hong, Y.; Xie, Y.; Peng, M.; Sun, R.; Guo, H.; Zhang, X.; Zhu, W.; Wang, J.; et al. Lipocalin-2-mediated astrocyte pyroptosis promotes neuroinflammatory injury via NLRP3 inflammasome activation in cerebral ischemia/reperfusion injury. J. Neuroinflamm. 2023, 20, 148. [Google Scholar] [CrossRef] [PubMed]
  71. Hu, R.; Liang, J.; Ding, L.; Zhang, W.; Liu, X.; Song, B.; Xu, Y. Edaravone dexborneol provides neuroprotective benefits by suppressing NLRP3 inflammasome-induced microglial pyroptosis in experimental ischemic stroke. Int. Immunopharmacol. 2022, 113, 109315. [Google Scholar] [CrossRef] [PubMed]
  72. Zhao, H.; Yang, Y.; Si, X.; Liu, H.; Wang, H. The Role of Pyroptosis and Autophagy in Ischemia Reperfusion Injury. Biomolecules 2022, 12, 1010. [Google Scholar] [CrossRef] [PubMed]
  73. Luo, L.; Liu, M.; Fan, Y.; Zhang, J.; Liu, L.; Li, Y.; Zhang, Q.; Xie, H.; Jiang, C.; Wu, J.; et al. Intermittent theta-burst stimulation improves motor function by inhibiting neuronal pyroptosis and regulating microglial polarization via TLR4/NFκB/NLRP3 signaling pathway in cerebral ischemic mice. J. Neuroinflamm. 2022, 19, 141. [Google Scholar] [CrossRef] [PubMed]
  74. Zhang, L.; Bai, X.Y.; Sun, K.Y.; Li, X.; Zhang, Z.Q.; Liu, Y.D.; Xiang, Y.; Liu, X.L. A New Perspective in the Treatment of Ischemic Stroke: Ferroptosis. Neurochem. Res. 2024, 49, 815–833. [Google Scholar] [CrossRef] [PubMed]
  75. Guo, J.; Tuo, Q.-Z.; Lei, P. Iron, ferroptosis, and ischemic stroke. J. Neurochem. 2023, 165, 487–520. [Google Scholar] [CrossRef] [PubMed]
  76. Li, J.; Cao, F.; Yin, H.-L.; Huang, Z.-J.; Lin, Z.-T.; Mao, N.; Sun, B.; Wang, G. Ferroptosis: Past, present and future. Cell Death Dis. 2020, 11, 88. [Google Scholar] [CrossRef] [PubMed]
  77. Xu, Y.; Li, K.; Zhao, Y.; Zhou, L.; Liu, Y.; Zhao, J. Role of Ferroptosis in Stroke. Cell. Mol. Neurobiol. 2023, 43, 205–222. [Google Scholar] [CrossRef] [PubMed]
  78. Fu, C.; Wu, Y.; Liu, S.; Luo, C.; Lu, Y.; Liu, M.; Wang, L.; Zhang, Y.; Liu, X. Rehmannioside A improves cognitive impairment and alleviates ferroptosis via activating PI3K/AKT/Nrf2 and SLC7A11/GPX4 signaling pathway after ischemia. J. Ethnopharmacol. 2022, 289, 115021. [Google Scholar] [CrossRef] [PubMed]
  79. Zhou, G.-J.; Liu, D.-N.; Huang, X.-R.; Wu, Q.; Feng, W.-B.; Zeng, Y.-H.; Liu, H.-Y.; Yu, J.; Xiao, Z.-J.; Zhou, J. High-frequency repetitive transcranial magnetic stimulation protects against cerebral ischemia/reperfusion injury in rats: Involving the mitigation of ferroptosis and inflammation. Brain Behav. 2023, 13, e2988. [Google Scholar] [CrossRef] [PubMed]
  80. Shen, X.Y.; Zhang, X.Y.; Han, P.P.; Zhao, Y.N.; Xu, G.H.; Bi, X. Mechanisms of intermittent theta-burst stimulation attenuating nerve injury after ischemic reperfusion in rats through endoplasmic reticulum stress and ferroptosis. Mol. Biol. Rep. 2024, 51, 377. [Google Scholar] [CrossRef] [PubMed]
  81. Jakubczyk, K.; Dec, K.; Kałduńska, J.; Kawczuga, D.; Kochman, J.; Janda, K. Reactive oxygen species—Sources, functions, oxidative damage. Pol. Merkur. Lek. 2020, 48, 124–127. [Google Scholar]
  82. Yao, H.; Ago, T.; Kitazono, T.; Nabika, T. NADPH Oxidase-Related Pathophysiology in Experimental Models of Stroke. Int. J. Mol. Sci. 2017, 18, 2123. [Google Scholar] [CrossRef] [PubMed]
  83. Duan, J.; Gao, S.; Tu, S.; Lenahan, C.; Shao, A.; Sheng, J. Pathophysiology and Therapeutic Potential of NADPH Oxidases in Ischemic Stroke-Induced Oxidative Stress. Oxid. Med. Cell Longev. 2021, 2021, 6631805. [Google Scholar] [CrossRef] [PubMed]
  84. Tang, X.N.; Cairns, B.; Kim, J.Y.; Yenari, M.A. NADPH oxidase in stroke and cerebrovascular disease. Neurol. Res. 2012, 34, 338–345. [Google Scholar] [CrossRef] [PubMed]
  85. Huang, H.F.; Guo, F.; Cao, Y.Z.; Shi, W.; Xia, Q. Neuroprotection by manganese superoxide dismutase (MnSOD) mimics: Antioxidant effect and oxidative stress regulation in acute experimental stroke. CNS Neurosci. Ther. 2012, 18, 811–818. [Google Scholar] [CrossRef] [PubMed]
  86. Rahman, I.; Biswas, S.K. Oxidants and Antioxidants|Antioxidants, Enzymatic. In Encyclopedia of Respiratory Medicine; Laurent, G.J., Shapiro, S.D., Eds.; Academic Press: Oxford, UK, 2006; pp. 258–266. [Google Scholar]
  87. Kaviannejad, R.; Karimian, S.M.; Riahi, E.; Ashabi, G. Using dual polarities of transcranial direct current stimulation in global cerebral ischemia and its following reperfusion period attenuates neuronal injury. Metab. Brain Dis. 2022, 37, 1503–1516. [Google Scholar] [CrossRef] [PubMed]
  88. Ruohonen, J.; Karhu, J. tDCS possibly stimulates glial cells. Clin. Neurophysiol. 2012, 123, 2006–2009. [Google Scholar] [CrossRef] [PubMed]
  89. Pelletier, S.J.; Lagacé, M.; St-Amour, I.; Arsenault, D.; Cisbani, G.; Chabrat, A.; Fecteau, S.; Lévesque, M.; Cicchetti, F. The morphological and molecular changes of brain cells exposed to direct current electric field stimulation. Int. J. Neuropsychopharmacol. 2014, 18, pyu090. [Google Scholar] [CrossRef]
  90. Fenoy, A.J.; Goetz, L.; Chabardès, S.; Xia, Y. Deep brain stimulation: Are astrocytes a key driver behind the scene? CNS Neurosci. Ther. 2014, 20, 191–201. [Google Scholar] [CrossRef] [PubMed]
  91. Mishima, T.; Nagai, T.; Yahagi, K.; Akther, S.; Oe, Y.; Monai, H.; Kohsaka, S.; Hirase, H. Transcranial Direct Current Stimulation (tDCS) Induces Adrenergic Receptor-Dependent Microglial Morphological Changes in Mice. eNeuro 2019, 6, 1–12. [Google Scholar] [CrossRef] [PubMed]
  92. Woodburn, S.C.; Bollinger, J.L.; Wohleb, E.S. The semantics of microglia activation: Neuroinflammation, homeostasis, and stress. J. Neuroinflamm. 2021, 18, 258. [Google Scholar] [CrossRef] [PubMed]
  93. Gava-Junior, G.; Ferreira, S.A.; Roque, C.; Mendes-Oliveira, J.; Serrenho, I.; Pinto, N.; Patto, M.V.; Baltazar, G. High-frequency repetitive magnetic stimulation rescues ischemia-injured neurons through modulation of glial-derived neurotrophic factor present in the astrocyte’s secretome. J. Neurochem. 2023, 164, 813–828. [Google Scholar] [CrossRef] [PubMed]
  94. Roque, C.; Pinto, N.; Vaz Patto, M.; Baltazar, G. Astrocytes contribute to the neuronal recovery promoted by high-frequency repetitive magnetic stimulation in in vitro models of ischemia. J. Neurosci. Res. 2021, 99, 1414–1432. [Google Scholar] [CrossRef] [PubMed]
  95. Bathina, S.; Das, U.N. Brain-derived neurotrophic factor and its clinical implications. Arch. Med. Sci. 2015, 11, 1164–1178. [Google Scholar] [CrossRef]
  96. Cancel, L.M.; Silas, D.; Bikson, M.; Tarbell, J.M. Direct current stimulation modulates gene expression in isolated astrocytes with implications for glia-mediated plasticity. Sci. Rep. 2022, 12, 17964. [Google Scholar] [CrossRef]
  97. Paolicelli, R.C.; Bolasco, G.; Pagani, F.; Maggi, L.; Scianni, M.; Panzanelli, P.; Giustetto, M.; Ferreira, T.A.; Guiducci, E.; Dumas, L.; et al. Synaptic Pruning by Microglia Is Necessary for Normal Brain Development. Science 2011, 333, 1456–1458. [Google Scholar] [CrossRef]
  98. Gellner, A.K.; Reis, J.; Fiebich, B.L.; Fritsch, B. Electrified microglia: Impact of direct current stimulation on diverse properties of the most versatile brain cell. Brain Stimul. 2021, 14, 1248–1258. [Google Scholar] [CrossRef]
  99. Qiao, C.; Liu, Z.; Qie, S. The Implications of Microglial Regulation in Neuroplasticity-Dependent Stroke Recovery. Biomolecules 2023, 13, 571. [Google Scholar] [CrossRef]
  100. Hong, Y.; Liu, Q.; Peng, M.; Bai, M.; Li, J.; Sun, R.; Guo, H.; Xu, P.; Xie, Y.; Li, Y.; et al. High-frequency repetitive transcranial magnetic stimulation improves functional recovery by inhibiting neurotoxic polarization of astrocytes in ischemic rats. J. Neuroinflamm. 2020, 17, 150. [Google Scholar] [CrossRef]
  101. Cherchi, L.; Anni, D.; Buffelli, M.; Cambiaghi, M. Early Application of Ipsilateral Cathodal-tDCS in a Mouse Model of Brain Ischemia Results in Functional Improvement and Perilesional Microglia Modulation. Biomolecules 2022, 12, 588. [Google Scholar] [CrossRef]
  102. Walter, H.L.; Pikhovych, A.; Endepols, H.; Rotthues, S.; Bärmann, J.; Backes, H.; Hoehn, M.; Wiedermann, D.; Neumaier, B.; Fink, G.R.; et al. Transcranial-Direct-Current-Stimulation Accelerates Motor Recovery after Cortical Infarction in Mice: The Interplay of Structural Cellular Responses and Functional Recovery. Neurorehabilit. Neural Repair 2022, 36, 701–714. [Google Scholar] [CrossRef]
  103. Hong, Y.; Lyu, J.; Zhu, L.; Wang, X.; Peng, M.; Chen, X.; Deng, Q.; Gao, J.; Yuan, Z.; Wang, D.; et al. High-frequency repetitive transcranial magnetic stimulation (rTMS) protects against ischemic stroke by inhibiting M1 microglia polarization through let-7b-5p/HMGA2/NF-κB signaling pathway. BMC Neurosci. 2022, 23, 49. [Google Scholar] [CrossRef]
  104. Chen, J.; Zeng, Y.; Hong, J.; Li, C.; Zhang, X.; Wen, H. Effects of HF-rTMS on microglial polarization and white matter integrity in rats with poststroke cognitive impairment. Behav. Brain Res. 2023, 439, 114242. [Google Scholar] [CrossRef]
  105. Braun, R.; Klein, R.; Walter, H.L.; Ohren, M.; Freudenmacher, L.; Getachew, K.; Ladwig, A.; Luelling, J.; Neumaier, B.; Endepols, H.; et al. Transcranial direct current stimulation accelerates recovery of function, induces neurogenesis and recruits oligodendrocyte precursors in a rat model of stroke. Exp. Neurol. 2016, 279, 127–136. [Google Scholar] [CrossRef] [PubMed]
  106. Ma, H.; Li, H.; Zhang, Y.; Zhou, Y.; Liu, H.; Xu, H.; Zhu, L.; Zhang, G.; Wang, J.; Li, Z.; et al. Microglia Exhibit Distinct Heterogeneity Rather than M1/M2 Polarization within the Early Stage of Acute Ischemic Stroke. Aging Dis. 2023, 14, 2284–2302. [Google Scholar] [CrossRef] [PubMed]
  107. Rahman, A.A.; Amruta, N.; Pinteaux, E.; Bix, G.J. Neurogenesis after Stroke: A Therapeutic Perspective. Transl. Stroke Res. 2021, 12, 1–14. [Google Scholar] [CrossRef] [PubMed]
  108. Markowska, A.; Koziorowski, D.; Szlufik, S. Microglia and Stem Cells for Ischemic Stroke Treatment—Mechanisms, Current Status, and Therapeutic Challenges. Front. Biosci. Landmark Ed. 2023, 28, 269. [Google Scholar] [CrossRef] [PubMed]
  109. Marques, B.L.; Carvalho, G.A.; Freitas, E.M.M.; Chiareli, R.A.; Barbosa, T.G.; Di Araújo, A.G.P.; Nogueira, Y.L.; Ribeiro, R.I.; Parreira, R.C.; Vieira, M.S.; et al. The role of neurogenesis in neurorepair after ischemic stroke. Semin. Cell Dev. Biol. 2019, 95, 98–110. [Google Scholar] [CrossRef] [PubMed]
  110. Lindvall, O.; Kokaia, Z. Neurogenesis following Stroke Affecting the Adult Brain. Cold Spring Harb. Perspect. Biol. 2015, 7, a019034. [Google Scholar] [CrossRef] [PubMed]
  111. Koh, S.-H.; Park, H.-H. Neurogenesis in Stroke Recovery. Transl. Stroke Res. 2017, 8, 3–13. [Google Scholar] [CrossRef] [PubMed]
  112. Li, Z.; Song, Y.; He, T.; Wen, R.; Li, Y.; Chen, T.; Huang, S.; Wang, Y.; Tang, Y.; Shen, F.; et al. M2 microglial small extracellular vesicles reduce glial scar formation via the miR-124/STAT3 pathway after ischemic stroke in mice. Theranostics 2021, 11, 1232–1248. [Google Scholar] [CrossRef] [PubMed]
  113. Shi, X.; Luo, L.; Wang, J.; Shen, H.; Li, Y.; Mamtilahun, M.; Liu, C.; Shi, R.; Lee, J.H.; Tian, H.; et al. Stroke subtype-dependent synapse elimination by reactive gliosis in mice. Nat. Commun. 2021, 12, 6943. [Google Scholar] [CrossRef] [PubMed]
  114. Luo, J.; Feng, Y.; Hong, Z.; Yin, M.; Zheng, H.; Zhang, L.; Hu, X. High-frequency repetitive transcranial magnetic stimulation promotes neural stem cell proliferation after ischemic stroke. Neural Regen. Res. 2024, 19, 1772–1780. [Google Scholar] [CrossRef] [PubMed]
  115. Zong, X.; Gu, J.; Zhou, S.; Ding, D.; Hu, Y.; Tucker, L.; Huang, Z.; Geng, D.; Gao, D. Continuous theta-burst stimulation enhances and sustains neurogenesis following ischemic stroke. Theranostics 2022, 12, 5710–5726. [Google Scholar] [CrossRef] [PubMed]
  116. Peng, J.-J.; Sha, R.; Li, M.-X.; Chen, L.-T.; Han, X.-H.; Guo, F.; Chen, H.; Huang, X.-L. Repetitive transcranial magnetic stimulation promotes functional recovery and differentiation of human neural stem cells in rats after ischemic stroke. Exp. Neurol. 2019, 313, 1–9. [Google Scholar] [CrossRef] [PubMed]
  117. Lei, R.; Wang, S.; Liu, A.; Cheng, J.; Zhang, Z.; Ren, J.; Yao, X.; Kong, X.; Ma, W.; Che, F.; et al. Bilateral transcranial direct-current stimulation promotes migration of subventricular zone-derived neuroblasts toward ischemic brain. FASEB Bioadv. 2023, 5, 277–286. [Google Scholar] [CrossRef] [PubMed]
  118. Zhang, K.; Guo, L.; Zhang, J.; Rui, G.; An, G.; Zhou, Y.; Lin, J.; Xing, J.; Zhao, T.; Ding, G. tDCS Accelerates the Rehabilitation of MCAO-Induced Motor Function Deficits via Neurogenesis Modulated by the Notch1 Signaling Pathway. Neurorehabil. Neural Repair 2020, 34, 640–651. [Google Scholar] [CrossRef] [PubMed]
  119. Pikhovych, A.; Stolberg, N.P.; Jessica Flitsch, L.; Walter, H.L.; Graf, R.; Fink, G.R.; Schroeter, M.; Rueger, M.A. Transcranial Direct Current Stimulation Modulates Neurogenesis and Microglia Activation in the Mouse Brain. Stem Cells Int. 2016, 2016, 2715196. [Google Scholar] [CrossRef] [PubMed]
  120. De Michele, M.; Piscopo, P.; Costanzo, M.; Lorenzano, S.; Crestini, A.; Rivabene, R.; Manzini, V.; Petraglia, L.; Iacobucci, M.; Berto, I.; et al. Can Repetitive Transcranial Magnetic Stimulation (rTMS) Promote Neurogenesis and Axonogenesis in Subacute Human Ischemic Stroke? Biomedicines 2024, 12, 670. [Google Scholar] [CrossRef] [PubMed]
  121. Niimi, M.; Hashimoto, K.; Kakuda, W.; Miyano, S.; Momosaki, R.; Ishima, T.; Abo, M. Role of Brain-Derived Neurotrophic Factor in Beneficial Effects of Repetitive Transcranial Magnetic Stimulation for Upper Limb Hemiparesis after Stroke. PLoS ONE 2016, 11, e0152241. [Google Scholar] [CrossRef] [PubMed]
  122. Li, H.; Shang, J.; Zhang, C.; Lu, R.; Chen, J.; Zhou, X. Repetitive Transcranial Magnetic Stimulation Alleviates Neurological Deficits after Cerebral Ischemia through Interaction Between RACK1 and BDNF exon IV by the Phosphorylation-Dependent Factor MeCP2. Neurotherapeutics 2020, 17, 651–663. [Google Scholar] [CrossRef] [PubMed]
  123. Lu, H.; Zhang, T.; Wen, M.; Sun, L. Impact of repetitive transcranial magnetic stimulation on post-stroke dysmnesia and the role of BDNF Val66Met SNP. Med. Sci. Monit. 2015, 21, 761–768. [Google Scholar] [CrossRef] [PubMed]
  124. Jiang, B.; He, D. Repetitive transcranial magnetic stimulation (rTMS) fails to increase serum brain-derived neurotrophic factor (BDNF). Neurophysiol. Clin. 2019, 49, 295–300. [Google Scholar] [CrossRef]
  125. Feng, S.; Wang, S.; Sun, S.; Su, H.; Zhang, L. Effects of combination treatment with transcranial magnetic stimulation and bone marrow mesenchymal stem cell transplantation or Raf inhibition on spinal cord injury in rats. Mol. Med. Rep. 2021, 23, 294. [Google Scholar] [CrossRef] [PubMed]
  126. Jæger, H.S.; Tranberg, D.; Larsen, K.; Valentin, J.B.; Blauenfeldt, R.A.; Luger, S.; Bache, K.G.; Gude, M.F. Diagnostic performance of Glial Fibrillary Acidic Protein and Prehospital Stroke Scale for identification of stroke and stroke subtypes in an unselected patient cohort with symptom onset < 4.5 h. Scand. J. Trauma Resusc. Emerg. Med. 2023, 31, 1. [Google Scholar] [CrossRef] [PubMed]
  127. Puspitasari, V.; Gunawan, P.Y.; Wiradarma, H.D.; Hartoyo, V. Glial Fibrillary Acidic Protein Serum Level as a Predictor of Clinical Outcome in Ischemic Stroke. Open Access Maced. J. Med. Sci. 2019, 7, 1471–1474. [Google Scholar] [CrossRef] [PubMed]
  128. Lee, J.Y.; Kim, H.S.; Kim, S.H.; Kim, H.S.; Cho, B.P. Combination of Human Mesenchymal Stem Cells and Repetitive Transcranial Magnetic Stimulation Enhances Neurological Recovery of 6-Hydroxydopamine Model of Parkinsonian’s Disease. Tissue Eng. Regen. Med. 2020, 17, 67–80. [Google Scholar] [CrossRef] [PubMed]
  129. Danuaji, R.; Hambarsari, Y.; Hamidi, B.L.; Hutabarat, E.A.J.; Tedjo, R.A.A.; Fairuzya, A.F.; Savitri, M.O.D. Combination of stem cell and repetitive transcranial magnetic stimulation in acute ischaemic stroke as a promising treatment: A case report. JKKI J. Kedokt. Dan Kesehat. Indones. 2024, 15, 132–140. [Google Scholar] [CrossRef]
  130. Li, L.M.; Uehara, K.; Hanakawa, T. The contribution of interindividual factors to variability of response in transcranial direct current stimulation studies. Front. Cell Neurosci. 2015, 9, 181. [Google Scholar] [CrossRef] [PubMed]
  131. Salazar, C.A.; Feng, W.; Bonilha, L.; Kautz, S.; Jensen, J.H.; George, M.S.; Rowland, N.C. Transcranial Direct Current Stimulation for Chronic Stroke: Is Neuroimaging the Answer to the Next Leap Forward? J. Clin. Med. 2023, 12, 2601. [Google Scholar] [CrossRef]
  132. Lee, J.; Lee, A.; Kim, H.; Shin, M.; Yun, S.M.; Jung, Y.; Chang, W.H.; Kim, Y.H. Different Brain Connectivity between Responders and Nonresponders to Dual-Mode Noninvasive Brain Stimulation over Bilateral Primary Motor Cortices in Stroke Patients. Neural Plast. 2019, 2019, 3826495. [Google Scholar] [CrossRef] [PubMed]
  133. Nyffeler, T.; Vanbellingen, T.; Kaufmann, B.C.; Pflugshaupt, T.; Bauer, D.; Frey, J.; Chechlacz, M.; Bohlhalter, S.; Müri, R.M.; Nef, T.; et al. Theta burst stimulation in neglect after stroke: Functional outcome and response variability origins. Brain 2019, 142, 992–1008. [Google Scholar] [CrossRef] [PubMed]
  134. Bradnam, L.V.; Stinear, C.M.; Barber, P.A.; Byblow, W.D. Contralesional hemisphere control of the proximal paretic upper limb following stroke. Cereb. Cortex 2012, 22, 2662–2671. [Google Scholar] [CrossRef] [PubMed]
  135. Lindenberg, R.; Zhu, L.L.; Rüber, T.; Schlaug, G. Predicting functional motor potential in chronic stroke patients using diffusion tensor imaging. Hum. Brain Mapp. 2012, 33, 1040–1051. [Google Scholar] [CrossRef] [PubMed]
  136. Zheng, X.; Schlaug, G. Structural white matter changes in descending motor tracts correlate with improvements in motor impairment after undergoing a treatment course of tDCS and physical therapy. Front. Hum. Neurosci. 2015, 9, 229. [Google Scholar] [CrossRef] [PubMed]
  137. Parchure, S.; Harvey, D.Y.; Shah-Basak, P.P.; DeLoretta, L.; Wurzman, R.; Sacchetti, D.; Faseyitan, O.; Lohoff, F.W.; Hamilton, R.H. Brain-Derived Neurotrophic Factor Gene Polymorphism Predicts Response to Continuous Theta Burst Stimulation in Chronic Stroke Patients. Neuromodulation 2022, 25, 569–577. [Google Scholar] [CrossRef] [PubMed]
  138. Dresang, H.C.; Harvey, D.Y.; Xie, S.X.; Shah-Basak, P.P.; DeLoretta, L.; Wurzman, R.; Parchure, S.Y.; Sacchetti, D.; Faseyitan, O.; Lohoff, F.W.; et al. Genetic and Neurophysiological Biomarkers of Neuroplasticity Inform Post-Stroke Language Recovery. Neurorehabil. Neural Repair 2022, 36, 371–380. [Google Scholar] [CrossRef] [PubMed]
  139. Kolmos, M.; Madsen, M.J.; Liu, M.L.; Karabanov, A.; Johansen, K.L.; Thielscher, A.; Gandrup, K.; Lundell, H.; Fuglsang, S.; Thade, E.; et al. Patient-tailored transcranial direct current stimulation to improve stroke rehabilitation: Study protocol of a randomized sham-controlled trial. Trials 2023, 24, 216. [Google Scholar] [CrossRef] [PubMed]
  140. Yoo, Y.J.; Park, H.J.; Kim, T.Y.; Yoon, M.J.; Oh, H.M.; Lee, Y.J.; Hong, B.Y.; Kim, D.; Kim, T.W.; Lim, S.H. MRI-Based Personalized Transcranial Direct Current Stimulation to Enhance the Upper Limb Function in Patients with Stroke: Study Protocol for a Double-Blind Randomized Controlled Trial. Brain Sci. 2022, 12, 1673. [Google Scholar] [CrossRef] [PubMed]
  141. Choung, J.S.; Bhattacharjee, S.; Son, J.P.; Kim, J.M.; Cho, D.S.; Cho, C.S.; Kim, M. Development and application of rTMS device to murine model. Sci. Rep. 2023, 13, 5490. [Google Scholar] [CrossRef] [PubMed]
  142. Sun, T.; Hevner, R.F. Growth and folding of the mammalian cerebral cortex: From molecules to malformations. Nat. Rev. Neurosci. 2014, 15, 217–232. [Google Scholar] [CrossRef] [PubMed]
  143. Yu, F.; Huang, T.; Ran, Y.; Li, D.; Ye, L.; Tian, G.; Xi, J.; Liu, Z. New Insights Into the Roles of Microglial Regulation in Brain Plasticity-Dependent Stroke Recovery. Front. Cell Neurosci. 2021, 15, 727899. [Google Scholar] [CrossRef] [PubMed]
  144. Tai, J.; Hu, R.; Fan, S.; Wu, Y.; Wang, T.; Wu, J. Theta-burst transcranial magnetic stimulation for dysphagia patients during recovery stage of stroke: A randomized controlled trial. Eur. J. Phys. Rehabil. Med. 2023, 59, 543–553. [Google Scholar] [CrossRef]
  145. Tedesco Triccas, L.; Burridge, J.H.; Hughes, A.M.; Pickering, R.M.; Desikan, M.; Rothwell, J.C.; Verheyden, G. Multiple sessions of transcranial direct current stimulation and upper extremity rehabilitation in stroke: A review and meta-analysis. Clin. Neurophysiol. 2016, 127, 946–955. [Google Scholar] [CrossRef] [PubMed]
  146. Hordacre, B.; Moezzi, B.; Ridding, M.C. Neuroplasticity and network connectivity of the motor cortex following stroke: A transcranial direct current stimulation study. Hum. Brain Mapp. 2018, 39, 3326–3339. [Google Scholar] [CrossRef] [PubMed]
  147. Kang, J.H.; Kim, M.W.; Park, K.H.; Choi, Y.A. The effects of additional electrical stimulation combined with repetitive transcranial magnetic stimulation and motor imagery on upper extremity motor recovery in the subacute period after stroke: A preliminary study. Medicine 2021, 100, e27170. [Google Scholar] [CrossRef] [PubMed]
  148. Chu, M.; Zhang, Y.; Chen, J.; Chen, W.; Hong, Z.; Zhang, Y.; Yu, H.; Zhang, F.; Ye, X.; Li, J.; et al. Efficacy of Intermittent Theta-Burst Stimulation and Transcranial Direct Current Stimulation in Treatment of Post-Stroke Cognitive Impairment. J. Integr. Neurosci. 2022, 21, 130. [Google Scholar] [CrossRef] [PubMed]
  149. Gröhn, H.; Gillick, B.T.; Tkáč, I.; Bednařík, P.; Mascali, D.; Deelchand, D.K.; Michaeli, S.; Meekins, G.D.; Leffler-McCabe, M.J.; MacKinnon, C.D.; et al. Influence of Repetitive Transcranial Magnetic Stimulation on Human Neurochemistry and Functional Connectivity: A Pilot MRI/MRS Study at 7 T. Front. Neurosci. 2019, 13, 1260. [Google Scholar] [CrossRef] [PubMed]
  150. Chhatbar, P.Y.; Ramakrishnan, V.; Kautz, S.; George, M.S.; Adams, R.J.; Feng, W. Transcranial Direct Current Stimulation Post-Stroke Upper Extremity Motor Recovery Studies Exhibit a Dose-Response Relationship. Brain Stimul. 2016, 9, 16–26. [Google Scholar] [CrossRef] [PubMed]
  151. Bai, Z.; Zhang, J.; Fong, K.N.K. Effects of transcranial magnetic stimulation in modulating cortical excitability in patients with stroke: A systematic review and meta-analysis. J. Neuroeng. Rehabil. 2022, 19, 24. [Google Scholar] [CrossRef] [PubMed]
  152. Guo, J.; Chen, X.; Lyu, Z.; Xiu, H.; Lin, S.; Liu, F. Repetitive transcranial magnetic stimulation (rTMS) for post-stroke sleep disorders: A systematic review of randomized controlled trials. Neurol. Sci. 2022, 43, 6783–6794. [Google Scholar] [CrossRef]
  153. Veldema, J.; Gharabaghi, A. Non-invasive brain stimulation for improving gait, balance, and lower limbs motor function in stroke. J. Neuroeng. Rehabil. 2022, 19, 84. [Google Scholar] [CrossRef] [PubMed]
  154. Gómez-García, N.; Álvarez-Barrio, L.; Leirós-Rodríguez, R.; Soto-Rodríguez, A.; Andrade-Gómez, E.; Hernández-Lucas, P. Transcranial direct current stimulation for post-stroke dysphagia: A meta-analysis. J. Neuroeng. Rehabil. 2023, 20, 165. [Google Scholar] [CrossRef]
  155. Li, R.; Wang, J.; Yu, X.; Xu, P.; Zhang, S.; Xu, J.; Bai, Y.; Dai, Z.; Sun, Y.; Ye, R.; et al. Enhancing the effects of transcranial magnetic stimulation with intravenously injected magnetic nanoparticles. Biomater. Sci. 2019, 7, 2297–2307. [Google Scholar] [CrossRef] [PubMed]
  156. Xie, Y.; Pan, J.; Chen, J.; Zhang, D.; Jin, S. Acupuncture combined with repeated transcranial magnetic stimulation for upper limb motor function after stroke: A systematic review and meta-analysis. NeuroRehabilitation 2023, 53, 423–438. [Google Scholar] [CrossRef] [PubMed]
  157. Peng, Y.; Lin, Y.; Yu, N.W.; Liao, X.L.; Shi, L. The Clinical Efficacy and Possible Mechanism of Combination Treatment of Cerebral Ischemic Stroke with Ginkgo Biloba Extract and Low-Frequency Repetitive Transcranial Magnetic Stimulation. Sichuan Da Xue Xue Bao Yi Xue Ban 2021, 52, 883–889. [Google Scholar] [CrossRef] [PubMed]
  158. Bonin Pinto, C.; Morales-Quezada, L.; de Toledo Piza, P.V.; Zeng, D.; Saleh Vélez, F.G.; Ferreira, I.S.; Lucena, P.H.; Duarte, D.; Lopes, F.; El-Hagrassy, M.M.; et al. Combining Fluoxetine and rTMS in Poststroke Motor Recovery: A Placebo-Controlled Double-Blind Randomized Phase 2 Clinical Trial. Neurorehabil. Neural Repair 2019, 33, 643–655. [Google Scholar] [CrossRef] [PubMed]
  159. Hu, W.; Wang, X.; Li, X.; Wang, Q. Effect of Transcranial Direct Current Stimulation Combined with Donepezil on stroke patients with memory impairment. Pak. J. Med. Sci. 2023, 39, 898–901. [Google Scholar] [CrossRef] [PubMed]
  160. Hirakawa, Y.; Koyama, S.; Tanabe, S.; Takeda, K.; Ueda, T.; Motoya, I.; Sakurai, H.; Kanada, Y.; Kawamura, N.; Kawamura, M.; et al. Combined effects of botulinum toxin type A and repetitive transcranial magnetic stimulation with intensive motor training immediately after injection in a patient with chronic stroke: A case report. J. Hand Ther. 2019, 32, 519–524. [Google Scholar] [CrossRef] [PubMed]
  161. Yamada, N.; Kakuda, W.; Kondo, T.; Mitani, S.; Shimizu, M.; Abo, M. Local muscle injection of botulinum toxin type a synergistically improves the beneficial effects of repetitive transcranial magnetic stimulation and intensive occupational therapy in post-stroke patients with spastic upper limb hemiparesis. Eur. Neurol. 2014, 72, 290–298. [Google Scholar] [CrossRef] [PubMed]
  162. Graef, P.; Dadalt, M.L.R.; Rodrigués, D.; Stein, C.; Pagnussat, A.S. Transcranial magnetic stimulation combined with upper-limb training for improving function after stroke: A systematic review and meta-analysis. J. Neurol. Sci. 2016, 369, 149–158. [Google Scholar] [CrossRef] [PubMed]
  163. Ross, R.E.; VanDerwerker, C.J.; George, M.S.; Gregory, C.M. Feasibility of performing a multi-arm clinical trial examining the novel combination of repetitive transcranial magnetic stimulation and aerobic exercise for post-stroke depression. Top. Stroke Rehabil. 2023, 30, 649–662. [Google Scholar] [CrossRef]
  164. Garrido, M.M.; Álvarez, E.E.; Acevedo, P.F.; Moyano, V.Á.; Castillo, N.N.; Cavada Ch, G. Early transcranial direct current stimulation with modified constraint-induced movement therapy for motor and functional upper limb recovery in hospitalized patients with stroke: A randomized, multicentre, double-blind, clinical trial. Brain Stimul. 2023, 16, 40–47. [Google Scholar] [CrossRef] [PubMed]
  165. Lee, J.H.; Jeun, Y.J.; Park, H.Y.; Jung, Y.J. Effect of Transcranial Direct Current Stimulation Combined with Rehabilitation on Arm and Hand Function in Stroke Patients: A Systematic Review and Meta-Analysis. Healthcare 2021, 9, 1705. [Google Scholar] [CrossRef] [PubMed]
  166. Navarro-López, V.; Molina-Rueda, F.; Jiménez-Jiménez, S.; Alguacil-Diego, I.M.; Carratalá-Tejada, M. Effects of Transcranial Direct Current Stimulation Combined with Physiotherapy on Gait Pattern, Balance, and Functionality in Stroke Patients. A Systematic Review. Diagnostics 2021, 11, 656. [Google Scholar] [CrossRef] [PubMed]
  167. Navarro-López, V.; Del Valle-Gratacós, M.; Fernández-Matías, R.; Carratalá-Tejada, M.; Cuesta-Gómez, A.; Molina-Rueda, F. The Long-Term Maintenance of Upper Limb Motor Improvements Following Transcranial Direct Current Stimulation Combined with Rehabilitation in People with Stroke: A Systematic Review of Randomized Sham-Controlled Trials. Sensors 2021, 21, 5216. [Google Scholar] [CrossRef] [PubMed]
  168. Sivaramakrishnan, A.; Madhavan, S. Combining transcranial direct current stimulation with aerobic exercise to optimize cortical priming in stroke. Appl. Physiol. Nutr. Metab. 2021, 46, 426–435. [Google Scholar] [CrossRef] [PubMed]
  169. Meng, J.; Yan, Z.; Gu, F.; Tao, X.; Xue, T.; Liu, D.; Wang, Z. Transcranial direct current stimulation with virtual reality versus virtual reality alone for upper extremity rehabilitation in stroke: A meta-analysis. Heliyon 2023, 9, e12695. [Google Scholar] [CrossRef] [PubMed]
  170. Llorens, R.; Fuentes, M.A.; Borrego, A.; Latorre, J.; Alcañiz, M.; Colomer, C.; Noé, E. Effectiveness of a combined transcranial direct current stimulation and virtual reality-based intervention on upper limb function in chronic individuals post-stroke with persistent severe hemiparesis: A randomized controlled trial. J. Neuroeng. Rehabil. 2021, 18, 108. [Google Scholar] [CrossRef] [PubMed]
  171. San Agustín, A.; Crevillén, D.; Soto-León, V.; Moreno, J.C.; Oliviero, A.; Pons, J.L. Transcranial magnetic stimulation combined with endogenous human hippocampal and motor cortical activity enhances memory. PLoS ONE 2023, 18, e0295413. [Google Scholar] [CrossRef] [PubMed]
  172. Tatsuno, H.; Hamaguchi, T.; Sasanuma, J.; Kakita, K.; Okamoto, T.; Shimizu, M.; Nakaya, N.; Abo, M. Does a combination treatment of repetitive transcranial magnetic stimulation and occupational therapy improve upper limb muscle paralysis equally in patients with chronic stroke caused by cerebral hemorrhage and infarction?: A retrospective cohort study. Medicine 2021, 100, e26339. [Google Scholar] [CrossRef] [PubMed]
  173. Kakuda, W.; Abo, M.; Kobayashi, K.; Momosaki, R.; Yokoi, A.; Fukuda, A.; Ito, H.; Tominaga, A. Combination treatment of low-frequency rTMS and occupational therapy with levodopa administration: An intensive neurorehabilitative approach for upper limb hemiparesis after stroke. Int. J. Neurosci. 2011, 121, 373–378. [Google Scholar] [CrossRef] [PubMed]
  174. Liu, Q.; Li, W.; Chen, Y.; Zhang, S.; Sun, Z.; Yang, Y.; Lv, P.; Yin, Y. Effects of repetitive transcranial magnetic stimulation combined with music therapy in non-fluent aphasia after stroke: A randomised controlled study. Int. J. Lang. Commun. Disord. 2024, 59, 1211–1222. [Google Scholar] [CrossRef] [PubMed]
  175. Gong, Y.; Long, X.M.; Xu, Y.; Cai, X.Y.; Ye, M. Effects of repetitive transcranial magnetic stimulation combined with transcranial direct current stimulation on motor function and cortex excitability in subacute stroke patients: A randomized controlled trial. Clin. Rehabil. 2021, 35, 718–727. [Google Scholar] [CrossRef]
Figure 1. Interindividual variability in non-invasive brain stimulation effects.
Figure 1. Interindividual variability in non-invasive brain stimulation effects.
Biomedicines 12 01560 g001
Figure 2. Combination therapies used with NIBS in stroke rehabilitation research.
Figure 2. Combination therapies used with NIBS in stroke rehabilitation research.
Biomedicines 12 01560 g002
Table 1. Characteristics and differences between TMS and tDCS.
Table 1. Characteristics and differences between TMS and tDCS.
TMStDCS
CharacteristicsUses a magnetic field to induce electrical currents in the brain to modulate the excitability of the cortexDelivers weak direct electric current (1–2 mA) through the electrodes placed on the scalp as anode and cathode to modulate the excitability of the cortex
Mechanism of interhemispheric modulationInduces more focal electrical field and generates action potentials in a specific neural circuit [40]Causes weak polarization of a larger number of neurons, which modulates synaptic activity during motor activation [40]
Possible adverse effectsSeizure and syncope [48]Fatigue, headache, skin redness, itching, and burning sensation under the stimulation electrodes [48]
Size and portability of TMS and tDCS devicesLarge, heavy, not portable [49] Light, small, portable, and can be used at home [49]
Power supply requirementsRequires power supplyBattery driven
CostsHigher cost (up to around USD 80,000) [50]Lower cost (from around USD 100 to thousands of dollars) [51]
Neurophysiologic specificity High temporal and spatial resolution allows for targeting specific neural circuits [49]Low spatial resolution and difficulty in precisely localizing the electric field current [48]
Target regionsMostly targets cortical regions and cannot stimulate subcortical areas without affecting the cortex [48]Mostly targets cortical regions and cannot stimulate subcortical areas without affecting the cortex [48]
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

Markowska, A.; Tarnacka, B. Molecular Changes in the Ischemic Brain as Non-Invasive Brain Stimulation Targets—TMS and tDCS Mechanisms, Therapeutic Challenges, and Combination Therapies. Biomedicines 2024, 12, 1560. https://doi.org/10.3390/biomedicines12071560

AMA Style

Markowska A, Tarnacka B. Molecular Changes in the Ischemic Brain as Non-Invasive Brain Stimulation Targets—TMS and tDCS Mechanisms, Therapeutic Challenges, and Combination Therapies. Biomedicines. 2024; 12(7):1560. https://doi.org/10.3390/biomedicines12071560

Chicago/Turabian Style

Markowska, Aleksandra, and Beata Tarnacka. 2024. "Molecular Changes in the Ischemic Brain as Non-Invasive Brain Stimulation Targets—TMS and tDCS Mechanisms, Therapeutic Challenges, and Combination Therapies" Biomedicines 12, no. 7: 1560. https://doi.org/10.3390/biomedicines12071560

APA Style

Markowska, A., & Tarnacka, B. (2024). Molecular Changes in the Ischemic Brain as Non-Invasive Brain Stimulation Targets—TMS and tDCS Mechanisms, Therapeutic Challenges, and Combination Therapies. Biomedicines, 12(7), 1560. https://doi.org/10.3390/biomedicines12071560

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