Next Article in Journal
The Antimalarial Drug Artesunate Mediates Selective Cytotoxicity by Upregulating HO-1 in Melanoma Cells
Previous Article in Journal
Chronic Endometritis and Uterine Endometrium Microbiota in Recurrent Implantation Failure and Recurrent Pregnancy Loss
Previous Article in Special Issue
Intracellular cAMP Signaling Pathway via Gs Protein-Coupled Receptor Activation in Rat Primary Cultured Trigeminal Ganglion Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cognitive Functions following Trigeminal Neuromodulation

1
Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
2
AOU Sassari, Unit of Endocrinology, Nutritional and Metabolic Disorders, 07100 Sassari, Italy
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(9), 2392; https://doi.org/10.3390/biomedicines11092392
Submission received: 20 July 2023 / Revised: 13 August 2023 / Accepted: 25 August 2023 / Published: 27 August 2023
(This article belongs to the Special Issue State-of-the-Art Research in Trigeminal Nerve Stimulation)

Abstract

:
Vast scientific effort in recent years have been focused on the search for effective and safe treatments for cognitive decline. In this regard, non-invasive neuromodulation has gained increasing attention for its reported effectiveness in promoting the recovery of multiple cognitive domains after central nervous system damage. In this short review, we discuss the available evidence supporting a possible cognitive effect of trigeminal nerve stimulation (TNS). In particular, we ask that, while TNS has been widely and successfully used in the treatment of various neuropsychiatric conditions, as far as research in the cognitive field is concerned, where does TNS stand? The trigeminal nerve is the largest cranial nerve, conveying the sensory information from the face to the trigeminal sensory nuclei, and from there to the thalamus and up to the somatosensory cortex. On these bases, a bottom-up mechanism has been proposed, positing that TNS-induced modulation of the brainstem noradrenergic system may affect the function of the brain networks involved in cognition. Nevertheless, despite the promising theories, to date, the use of TNS for cognitive empowering and/or cognitive decline treatment has several challenges ahead of it, mainly due to little uniformity of the stimulation protocols. However, as the field continues to grow, standardization of practice will allow for data comparisons across studies, leading to optimized protocols targeting specific brain circuitries, which may, in turn, influence cognition in a designed manner.

1. Introduction

During the last two decades, the evolution and expansion of neuromodulation techniques have made them among the most attractive non-pharmacological tools for selected neurological and neuropsychiatric disorders. In fact, with respect to conventional drug-based approaches, neuromodulation techniques often provide several advantages such as safety, flexibility, and (in some instances) specificity of the treatment [1,2,3,4,5,6,7].
Nowadays, the panorama of non-invasive neurostimulation includes several techniques, which are thought to operate by stimulating the brain directly (e.g., transcranial direct current stimulation, tDCS; repetitive transcranial magnetic stimulation, rTMS; transcranial ultrasound stimulation, TUS, etc.). Interestingly, recently developed TUS allows for a focused application of the stimulus, overcoming a serious limitation of the other non-invasive techniques, namely their low spatial resolution [5,8]. On the other hand, stimulation of peripheral nerves (e.g., vagal nerve stimulation, trigeminal nerve stimulation, etc.) is also an effective non-invasive neurostimulation technique. With respect to direct neurostimulation techniques, peripheral nerve stimulation is often more practical and cheaper, while maintaining a relevant therapeutic power.
In recent times, there has been growing interest in the use of neuromodulation approaches to modulate the activity of the healthy brain, with the general aim of overcoming the physiological limitations of human capabilities, both physical and cognitive [9]. Applications may range from the enhancement of military personnel’s mental and/or physical skills [10,11], to the treatment of cognitive decline, either aging- or neurodegeneration-related, which significantly impacts the daily life of millions of people worldwide, urging the necessity for novel treatments [12,13]. While the majority of the studies in the field have been performed using tDCS, in recent years, it has been shown that cranial nerve stimulation may also affect brain circuits linked to cognitive domains [3,14,15]. Among all peripheral neuromodulation techniques, transcutaneous trigeminal nerve stimulation (TNS) has recently gained increasing success in clinical practice, since it has been consistently proven to exert beneficial effects in the symptomatic treatment of several neuropsychiatric disorders [16,17].
However, the real therapeutic potential, as well as the mechanism(s) of non-invasive neuromodulation methods on cognitive performance in both pathological conditions and healthy individuals, remains unclear. In this short review, we will focus on the current knowledge of the neurobiological mechanisms by which TNS may affect cognition.

2. Trigeminal Nerve Stimulation: Use and Safety

The first evidence of efficacy of TNS in an animal model of pentylenetetrazole-induced convulsions dates back more than twenty years ago [18]. Indeed, TNS was originally developed mainly as an alternative to vagal nerve stimulation (VNS) to overcome its well-known limitations. In particular, these include VNS-induced autonomic side effects due to the presence in the vagus nerve of visceral efferent components affecting cardiac and laryngeal function [19,20], and the exposure to surgery-related risks such as peritracheal hematoma and infections [21,22]. Since then, TNS has been tested in the treatment of different neurological and psychiatric conditions, proving beneficial for seizure control, depression, migraine, attention deficit hyperactivity disorder (ADHD), post-traumatic stress disorder, and tinnitus [23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41]. TNS has now reached a considerable diffusion in clinical practice, allowing for the development of a substantial base of knowledge supporting its possible therapeutic applications. Reported side effects are generally mild and include drowsiness, skin irritation, and headaches [4,7] However, increased appetite, fatigue, trouble sleeping, and teeth clenching have also been reported in ADHD patients [29]. In short, the available evidence shows that TNS administration is well tolerated, and no severe adverse events occur after treatment [27,42]. In April 2019, the U.S. Food and Drug Administration (FDA) allowed the marketing of the first medical TNS device to treat ADHD [36], and, as of today, TNS remains the first non-drug treatment for ADHD granted marketing authorization. The FDA has also approved the use of TNS for the treatment of migraine [43,44], and several clinical trials for drug-resistant epilepsy, depression, and other conditions are in different stages of completion. To date, in Europe, TNS has obtained approval for adjunctive treatment of epilepsy and major depressive disorder for patients over nine years of age [45,46]. Interestingly, besides being approved for epilepsy, depression, and ADHD therapy, TNS has been shown to have the potential to enhance cognition and mood in both healthy subjects and patients with neuropsychiatric disorders [41,47,48].
Along with the favorable safety profile of TNS, its ease of use is certainly an important factor favoring its wide diffusion. In fact, nowadays, technological advancements allow the production of portable, affordable, and user-friendly external stimulators. A TNS device is generally constituted by a programmable pulse generator delivering low-intensity electrical pulses to small electrodes applied to the cutaneous areas overlying specific branches of the trigeminal nerve. This allows great ease and flexibility in the administration of TNS, ranging from the application of disposable patches for overnight stimulation to the use of simple wearable devices. Furthermore, the trigeminal nerve may also be stimulated via the auricular branch, with the advantage of stimulating electrodes being less noticeable. This may allow for longer stimulation protocols, which can also be continuous throughout the day, including working hours. TNS devices are also affordable, and their use does not require trained operators, contrary to the use of other neurostimulation techniques (such as rTMS or tDCS), which often involve a more complex instrumental setting, as well as specialized personnel. Typically, TNS stimulation electrodes are positioned on the forehead branches of the first (V1) or second (V2) division of the trigeminal nerve (i.e., the supraorbital and infraorbital nerves, respectively). Other positioning schemes are possible (such as the anterosuperior and medial areas of the external ear innervated by the auricolo-temporal nerve, or the masseter belly innervated by the trigeminal motor branches), and may even result in other benefits, although we know little about them (Figure 1). In fact, mainly due to the lack of knowledge of the neurobiological bases of the effects of TNS, right now, the choice of stimulation parameters (waveform, amplitude, frequency, pulse duration, duty cycle, and session duration), and electrode placement heavily depends on the device characteristics and the physician’s particular experience for the condition to be treated.

3. How Trigeminal Nerve Stimulation May Affect Cognitive Function

Cognition has been classically defined as “all the processes by which the sensory input is transformed, reduced, elaborated, stored, recovered and used” [49]. While more recent efforts are still trying to reach a better definition of cognition [50], it is now largely accepted that cognitive processes emerge from the coordinated activity of hundreds of brain regions [51,52,53]. Brain network activity is dynamically reconfigured according to contingent sensory inputs, and the resulting state of the network involved is thought to produce a specific result [54,55]. This allows the brain to perform a large variety of cognitive and motor tasks, in order to adapt to changing environments.
Cranial nerves are a specialized part of the peripheral nervous system conveying crucial sensory information from and to the brain. Their inputs can modulate regional brain activity, potentially affecting the respective cognitive functions [3], as well as the activity of the brain networks they are involved in. Depending on the stimulated structure and stimulation modality, the result can either be inhibitory or facilitatory, thus influencing brain network activity in different ways. All these effects have been attributed to a “bottom-up” mechanism of modulation of the signal processing performed by the central nervous system (CNS), which is also thought to be the basis of the effects of TNS [6,56,57]. The trigeminal nerve is the largest cranial nerve, conveying the sensory information from the face, a major part of the scalp, the teeth, the oral and nasal cavity, as well as providing the motor supply to the masticatory muscles, the mylohyoid, the tensor tympani and tensor veli palatini muscles [58]. Trigeminal sensory nuclei send sensory information to the thalamus and from here to the somatosensory cortex [59], but also project to multiple nearby brainstem nuclei [60] including the nucleus solitary tract (NST), the locus coeruleus (LC), and the raphe nucleus (RN). As a whole, the output of the trigeminal nuclei can influence the activity of several higher-level structures. The influence on the NST appears particularly relevant since this structure integrates visceral inputs and relays signals to various structures such as the LC, RN, the reticular activating system hypothalamus, thalamus, amygdala, hippocampus, limbic forebrain, anterior cingulate cortex, and insula [60]. These pathways also open the possibility of regulating attention, cognition, and sensorimotor behaviors using the TNS via a bottom-up mechanism [48]. However, so far, the effects of TNS on cognitive processes have been explored only within neurological and neuropsychiatric populations, and only a few studies specifically investigated TNS effects on cognition as a stand-alone phenomenon.
In laboratory animals, the neuroanatomical basis and possible sites for action of TNS in physiological conditions have been poorly investigated so far. In the work by Mercante and colleagues [61], it has been shown that acute TNS administration in male adult Sprague Dawley rats increases c-Fos-like immunoreactivity in selected brainstem and forebrain regions. In further detail, following 3-hour-long unilateral stimulation of the infraorbital nerve (IoN, V2 division), a significant increase in c-Fos-like immunoreactivity was observed not only in the trigeminal sensory nuclei and in the somatosensory cortex, as expected, but also in the NST, RN, LC, amygdala, endopiriform nucleus, hippocampus, and entorhinal cortex. Such a diffuse effect, even after acute short-term TNS administration, supports the postulate that TNS can have a widespread influence on higher subcortical and cortical structures. The same study demonstrated that cell proliferation in the dentate gyrus of the hippocampus (as indexed by bromodeoxyuridine cell incorporation) was consistently increased after 24 h from acute TNS administration, in comparison to sham and naïve rats. Interestingly, since it has been proposed that the increase in hippocampal cellular proliferation may be due to an increase in noradrenergic activity [62,63,64], the marked c-Fos-like immunoreactivity observed in the LC after TNS may be causally related to it. Furthermore, in a subchronic pilocarpine-based epilepsy model (4 weeks of treatment, percutaneous stimulation of the IoN) TNS reduced hippocampal apoptosis and inflammation, showing protective effects on cognitive functions such as learning and memory [65].
The evidence provided in experimental animals gives support to the idea that the trigeminal system has the full potential to influence the activity of cortical and subcortical structures involved in cognitive, affective, and behavioral control [61,66]. Nevertheless, available human studies on TNS effects on cognition and behavior are still rather inconclusive (Table 1).
The effect(s) of TNS on CNS activity in healthy human subjects have been investigated using different non-invasive methods such as transcranial magnetic stimulation (TMS), electroencephalography (EEG), magnetic resonance spectroscopy, and brainstem reflex recordings. Paired-pulse TMS was used to evaluate the activity of inhibitory and excitatory intracortical circuits in the hand representation area of the primary motor cortex, during and after acute TNS administration in healthy subjects [67,68]. These studies failed to demonstrate significant changes in any of the neurophysiological outcomes tested (i.e., motor threshold, amplitude of the TMS-evoked motor potential, short-latency intracortical inhibition, and intracortical facilitation), suggesting that TNS effects may be exerted mainly at the brainstem level [67,68]. This hypothesis was also supported by other studies showing that short-term TNS administration significantly depressed brainstem circuits mediating the R2 component of the blink reflex [68], with a long-lasting action resembling long-term depression-like plasticity [69]. In agreement with this hypothesis, a recent paper has shown that short-term TNS was able to depress the amplitude of the defensive hand blink reflex [70], which is mediated by a brainstem circuit that is continuously modulated by cortical areas devoted to encoding the peripersonal space [71]. Ginatempo and colleagues [72] evaluated the effects of acute TNS administration at the cortical level using EEG, showing that TNS induces a spatially diffused, non-specific desynchronization of fast EEG rhythms, with a significant decrease in the intra- and inter-hemispheric synchronization of beta frequencies. In addition, a trend of increase in the gamma band power and in mean EEG frequency total power and a trend of decrease in interhemispheric gamma coherence was also observed during and after TNS. These results were interpreted as expressions at the cortical level of modifications originating in the brainstem, such as a change in the activity of the noradrenergic nuclei, namely LC [72]. Interestingly, a recent paper using ultrahigh-field magnetic resonance spectroscopy showed that acute TNS administration slightly decreased total creatine concentrations in the dorsolateral prefrontal cortex (DLPFC) ~60 min post-TNS [73]. However, while creatine CNS concentration has been shown to negatively correlate with measures of cognitive function [74], whether relatively small changes (4–5% decrease) in the DLPFC could provide a cognitive benefit is unknown. Vasomotor centers in the brainstem are directly connected to the trigeminal nerve [28,75,76], and its stimulation has a significant impact on cerebral perfusion [77,78,79]. Neurogenic control of cerebral blood flow via TNS may be a way to induce cerebral vasodilation and improve cerebral perfusion in both normal conditions and pathologic states [80,81]
On the behavioral side, studies investigating TNS effects in healthy subjects often reported a state of relaxation or sedation, decreased attention and vigilance, with a tendency to fall asleep during and/or immediately after acute TNS administration. Piquet et al. 2011 [82] investigated the sedative effects of TNS and reported a decrease in vigilance and arousal following acute high-frequency TNS. The authors hypothesized that a change in activity levels of the orexin-arcuate-periaqueductal gray matter circuit could occur during supraorbital neurostimulation and might explain the decrease in vigilance [82]. A subsequent study tested whether the sedative effects of TNS were associated with hypnotic effects, such as sleep latency reduction, and found no association with a reduction in sleep latency as indexed by the Multiple Sleep Latency Test [83].
Long-term treatment of TNS using low-frequency stimulation may also have a calming effect and improve sleep quality and mood [48]. A potential explanation for the ability of trigeminal modulation to influence sleep is the modulation of the ascending reticular activating system [84], a network of nuclei that control attention and the sleep/wake cycle including the LC and pedunculopontine nucleus [85].
Recently, the TNS effects on cortical activity related to modulation of the function of the noradrenergic system were also investigated using event-related potentials (ERP). This approach seems particularly relevant since many ERP components have been operationally related to several specific neurocognitive processes [86,87], including attention, working memory, and decision-making [88,89,90]. Both studies used short-term TNS administration but, interestingly, one study [91] used bilateral transcutaneous stimulation of the trigeminal motor branches (over the masseter belly, incisura sigmoidea), while in the other [92], the IoN sensitive branch was bilaterally stimulated.
In the first study, Fantozzi and colleagues [91] showed in 13 healthy subjects a reduction of the amplitude of the P300 wave elicited by an acoustic oddball paradigm in several cortical areas, and a positive correlation between P300 amplitude in frontal and median cortical region and pupil size. The authors ascribed these results to increased cortical norepinephrine (NE) levels due to a TNS-augmented activity of LC neurons. High cortical NE levels may improve sensory processing, suggesting that TNS could be used for improving cognitive performance in subjects with cognitive disorders or arousal dysfunction [91,93].
The second study focused on several major ERP components recorded during the administration of a simple visual oddball task (P200 and P300), and a paired-click paradigm (P50, N100, and P200), to investigate in healthy subjects the possible mechanism(s) of action of TNS related to high-level neurocognitive processes [92]. These paradigms are largely used to study information processing and cognitive brain functions [90,94], which are modulated by multiple brain systems and, in particular, by the LC and the reticular formation [95,96], both targeted by TNS [61]. Results show that all parameters measured were unaffected by TNS, as shown by the non-significant differences between the effects of sham and real TNS administration.
Table 1. Studies on the effects of trigeminal nerve stimulation report specific cognitive measures. Parameters and data as stated in the original publication.
Table 1. Studies on the effects of trigeminal nerve stimulation report specific cognitive measures. Parameters and data as stated in the original publication.
CitationStudy PopulationStimulated NerveTNS ParametersTime of AssessmentAssessment MethodologyOutcomeSide Effects
Piquet et al., 2011 [82]HSSupraorbital nerve, bilateral
(A)
120 Hz pulsed, 250 μs pulse width.
(B)
<14 mA
(C)
20 min
After stimulationPsychological testModulation of vigilanceNo major side effects reported
Boasso et al., 2016 [48]HSRight supraorbital nerve,
Cervical nerve
(A)
biphasic pulse-modulated (3–11 kHz)
(B)
5–7 mA
(C)
20 min per day
After one week of treatmentPsychological test
Biochemical measurements
Improved sleep quality, stress reductionNo major side effects reported
Trevizol et al., 2016 [40]NPDSupraorbital nerve, bilateral
(A)
120 Hz pulsed, 200 μs pulse width
(B)
N/A
(C)
30 min per day
After 10 days of treatmentPsychological testNo significant difference in cognitive performancesNo major side effects reported
Loo et al., 2020 [36]ADHDSupraorbital nerve, bilateral
(A)
120 Hz pulsed, 250 μs pulse width
(B)
2–4 mA
(C)
8 h per night
After 4 weeks of treatmentPsychological test
Resting state EEG
Improved/normalized executive functions,
modulation of right frontal brain activity
Increase in fatigue, headache, appetite. Skin whitening/discoloration under electrode patch
Tramonti Fantozzi et al., 2020 [93]HSMandibular motor branch
(A)
biphasic, 0.618 Hz, 540 μs pulse width,
(B)
21–25 mA
(C)
15 min
After stimulationPsychological test
Pupil size
Trigeminal sensory-motor imbalance may affect cognitive performanceNo major side effects reported
Tramonti Fantozzi et al., 2021 [91]HSMandibular motor branch
(A)
biphasic, 0.618 Hz, 540 μs pulse width,
(B)
21–25 mA
(C)
10 min
After stimulationPupil size
EEG power change
ERP (auditory oddball)
Reduced P300 amplitude, positive correlation between P300 and pupil sizeNo major side effects reported
Mercante et al., 2023 [92]HSInfraorbital nerve, bilateral
(A)
biphasic,120 Hz, 250 μs pulse width,
(B)
1–20 mA
(C)
20 min
After stimulationERP (visual oddball, sensory gating)No changes in ERP parameters measuredNo major effects reported
Healthy subjects: HS; Neuro-Psychiatric Disorders: NPD; Attention Deficit Hyperactivity Disorder: ADHD. TNS parameters: A: Stimulus characteristics; B: Stimulus intensity; C: Treatment duration. Electroencephalography: EEG; Event-Related Potentials: ERP.

4. New Possibilities

Ease of use and safety of TNS allows its administration in conjunction with other interventions. In particular, in recent years, the association of TNS with auricular or tongue stimulation has seen a growing interest and development [97,98,99].
As of today, auricular stimulation is mainly used for transcutaneous VNS (tVNS) via the auricular branch of the vagus nerve. However, the external ear is also innervated by trigeminal nerve fibers, partially overlapping with the vagus territory [100,101]; therefore, an appropriate electrode positioning easily results in the stimulation of both trigeminal and vagal afferents [6,102]. The study of auricular tVNS has seen great development in recent years, especially with regard to improved cognitive performance, not only in the clinical population but also in healthy subjects [103,104,105,106,107]. While the neurobiological mechanisms are still unclear, the available literature mostly seek to explain the positive effects of tVNS on cognitive performance in terms of a change in brain NE levels [64,108]. Thus, vagal afferents arriving at the NTS and then at the LC may cause an increase in NE levels, which would lead to an increase in brain activation and consequently to increased cognitive performance [109,110,111,112]. Consistent evidence shows that both trigeminal and vagal nerve systems share common relay stations in the brainstem, the spinal trigeminal nucleus and the NST in particular. From here, there is potential to activate four core neuromodulatory networks, including the noradrenergic (LC), cholinergic (nucleus basalis of Meynert), serotonergic (RN), and the substantia nigra and ventral tegmental area dopaminergic neurotransmitter systems [57]. These structures, in turn, can profoundly affect the function of different brain networks involved in cognitive functioning, including the thalamus, hypothalamus, hippocampus, amygdala, and cortical areas.
TNS cognitive effects on healthy subjects are much less characterized, both in features and magnitude. However, based on the anatomical and functional data reported above, it can be assumed that the possible effects on the cognitive function of TNS might share the same mediators and CNS targets used by VNS. On this basis, it is possible to hypothesize that paired stimulation of both trigeminal and vagal afferents at the external ear may result in improved results.
Another modality used for activating the trigeminal system is translingual nerve stimulation (TLNS), a neuromodulation approach often used in combination with neurological rehabilitation procedures, to improve outcomes for subjects with neurological conditions possibly by facilitating neuroplasticity-related changes in the brain [98,113,114]. So far, most studies demonstrated the feasibility and safety of TLNS, as well as its usefulness mainly in the treatment of balance and gait deficits in subjects with traumatic brain injury and multiple sclerosis [113,115]. TLNS has also been recently applied, paired with sound stimulation, for tinnitus treatment with ambiguous results [25,116,117]. Interestingly, it has recently been shown that a single 20 min TLNS administration in healthy subjects can affect resting brain activity, as indexed by high-density EEG [118]. This observation, despite substantial differences in the study methods, is also in line with the results of a study performed with fMRI in traumatic brain injury (TBI) patients [119]. These effects on resting brain activity, together with the improvements in cognition demonstrated in TBI patients, suggest that TLNS may be an interesting technique to be evaluated for cognitive effects. With regard to the underlying neurobiological mechanisms, by using TLNS, it is possible to stimulate two cranial nerves: trigeminal (lingual branch) and facial (chorda tympani branch). It is hypothesized that the stimulation of these structures modulates the activity of their respective brainstem nuclei, including the NST [115]. In a bottom-up fashion, connected regions are also subsequently affected, modifying corresponding neural network activity.

5. Conclusions

The increasing evidence from clinical research of the cognitive effects of TNS has stimulated basic studies aimed at unraveling sites and mechanisms of TNS action at the CNS level. However, these are yet to be understood in both physiological and pathological conditions. Available neurophysiological and behavioral evidence confirms an effect of TNS over CNS activity in healthy subjects. However, with regard to any activity at the cortical level, it cannot be excluded that these effects may be, at least in part, mediated by TNS action on brainstem structures. Since the neurobiological mechanisms by which TNS may act on brainstem structures are not well characterized, the nature of the resulting modulatory effects on supraspinal nuclei and structures remains largely speculative.
One of the most important factors hampering our understanding of TNS effects is the heterogeneity of the methodology of stimulation in both animal and human studies, which makes it difficult to compare results between studies. For instance, rodents have generally been found to be adequate for mechanistic studies of TNS. V1 and V2 divisions (purely sensory branches), in particular, have largely been used as viable access points for either invasive or non-invasive stimulation [120]. One striking disparity in the available literature is the multiple surgical approaches to access the IoN in rodents, which has the potential to become an unnecessary variable [18,61,121,122]. This aspect has been extensively discussed by Dingle and colleagues (2019) [122], where different accesses to the IoN and supraorbital nerve (SoN) are shown, in an effort to standardize the surgical approach as an integral part of the reproducibility of future TNS studies. Initial TNS studies have been performed by implanting cuff electrodes on IoN [18], which is relatively large and easy to access, making it an ideal target either for mechanistic studies or electrode/stimulation procedures development. However, since IoN stimulation is also commonly used to study chronic neuropathic pain in rats [123,124,125], it is essential to ensure that diligent consideration is given to both electrode design and implantation methodology.
In fact, constriction injury to the IoN could result in a trigeminal neuropathic pain syndrome with profound effects on behavior [125]. It has been suggested that trigeminal neuropathic pain may induce profound CNS modification at different levels, inducing reorganization of cortical sensory areas and negatively affecting the function of the dopaminergic mesolimbic system, one of the four core neuromodulatory networks possibly involved in TNS cognitive effects [126,127].
Similarly, in human studies, TNS may be administered in different ways (using subcutaneous, percutaneous, or cutaneous electrodes), for different amounts of time (acute or chronic treatment), and with different stimulation parameters (waveform, amplitude, frequency, etc.). As of today, stimulation parameters are mainly chosen depending on the used device characteristics and the physician’s (or experimenter’s) particular experience. Together with our lack of basic knowledge of the neurobiological effects of TNS, this heterogeneity in administration protocols seriously hampers the comparison of data by different researchers. Another important confounding factor may result from differences in study populations, since the neurobiology of aging-induced cognitive decline may profoundly differ from neurodegeneration-induced cognitive decline, let alone results from healthy subjects. Lastly, the methodology used for cognitive function assessment should also be adequately sensitive to properly define the population under study, as well as specifically designed to allow comparison of the results obtained. To this end, the time-course of the TNS administration protocol must be considered in order to allow a clear distinction between acute effects measured during stimulation, subacute effects measured immediately after, and long-term effects measured after a certain time period. This may be particularly important to differentiate results that could reflect functional changes from structural neuroplastic modifications. With regard to cognitive function assessment, several different approaches can be used, ranging from pen-and-paper tests to more objective neurophysiological methods. In our view, an appropriate combination of these approaches should be used in future studies. In fact, new analysis methods allow combining cognitive test results with data from objective methods, such as functional magnetic resonance, magnetic encephalography, or EEG, in order to monitor different cognitive domains [128,129]. So far, the large heterogeneity of the available data, either in terms of TNS administration protocols and/or populations under study, is both a cause and consequence of our lack of knowledge about the true relevance of TNS effects on cognition. However, new studies aimed to measure specific psychobiological markers after TNS in homogeneous populations should be able to produce relevant results.
Thus, despite being a safe, well-tolerated, and versatile potential intervention, TNS application for cognitive empowering and/or cognitive decline may remain an underrated or overrated approach until, as the field continues to grow, the standardization of practice allows for data comparisons across studies.

Author Contributions

Conceptualization, B.M., P.E. and F.D.; writing—original draft preparation, B.M., P.E. and F.D.; writing—review and editing, B.M., P.E. and F.D.; supervision, F.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Van Rooij, S.J.H.; Riva-Posse, P.; McDonald, W.M. The Efficacy and Safety of Neuromodulation Treatments in Late-Life Depression. Curr. Treat. Options Psychiatry 2020, 7, 337–348. [Google Scholar] [CrossRef] [PubMed]
  2. Sluka, K.A.; Walsh, D. Transcutaneous Electrical Nerve Stimulation: Basic Science Mechanisms and Clinical Effectiveness. J. Pain 2003, 4, 109–121. [Google Scholar] [CrossRef]
  3. Adair, D.; Truong, D.; Esmaeilpour, Z.; Gebodh, N.; Borges, H.; Ho, L.; Bremner, J.D.; Badran, B.W.; Napadow, V.; Clark, V.P.; et al. Electrical Stimulation of Cranial Nerves in Cognition and Disease. Brain Stimul. 2020, 13, 717–750. [Google Scholar] [CrossRef] [PubMed]
  4. Pop, J.; Murray, D.; Markovic, D.; DeGiorgio, C.M. Acute and Long-Term Safety of External Trigeminal Nerve Stimulation for Drug-Resistant Epilepsy. Epilepsy Behav. 2011, 22, 574–576. [Google Scholar] [CrossRef]
  5. Darmani, G.; Bergmann, T.O.; Pauly, K.B.; Caskey, C.F.; de Lecea, L.; Fomenko, A.; Fouragnan, E.; Legon, W.; Murphy, K.R.; Nandi, T.; et al. Non-Invasive Transcranial Ultrasound Stimulation for Neuromodulation. Clin. Neurophysiol. 2022, 135, 51–73. [Google Scholar] [CrossRef] [PubMed]
  6. Mercante, B.; Ginatempo, F.; Manca, A.; Melis, F.; Enrico, P.; Deriu, F. Anatomo-Physiologic Basis for Auricular Stimulation. Med. Acupunct. 2018, 30, 141–150. [Google Scholar] [CrossRef]
  7. Magis, D.; Sava, S.; d’Elia, T.S.; Baschi, R.; Schoenen, J. Safety and Patients’ Satisfaction of Transcutaneous Supraorbital NeuroStimulation (TSNS) with the Cefaly® Device in Headache Treatment: A Survey of 2313 Headache Sufferers in the General Population. J. Headache Pain 2013, 14, 95. [Google Scholar] [CrossRef]
  8. di Biase, L.; Falato, E.; Lazzaro, V.D. Transcranial Focused Ultrasound (TFUS) and Transcranial Unfocused Ultrasound (TUS) Neuromodulation: From Theoretical Principles to Stimulation Practices. Front. Neurol. 2019, 10, 549. [Google Scholar] [CrossRef]
  9. Marois, A.; Lafond, D. Augmenting Cognitive Work: A Review of Cognitive Enhancement Methods and Applications for Operational Domains. Cogn. Technol. Work 2022, 24, 589–608. [Google Scholar] [CrossRef]
  10. Davis, S.E.; Smith, G.A. Transcranial Direct Current Stimulation Use in Warfighting: Benefits, Risks, and Future Prospects. Front. Hum. Neurosci. 2019, 13, 114. [Google Scholar] [CrossRef]
  11. Clark, V.P.; Coffman, B.A.; Mayer, A.R.; Weisend, M.P.; Lane, T.D.R.; Calhoun, V.D.; Raybourn, E.M.; Garcia, C.M.; Wassermann, E.M. TDCS Guided Using FMRI Significantly Accelerates Learning to Identify Concealed Objects. NeuroImage 2012, 59, 117–128. [Google Scholar] [CrossRef] [PubMed]
  12. 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. 2021, 12, 578339. [Google Scholar] [CrossRef]
  13. Siegert, A.; Diedrich, L.; Antal, A. New Methods, Old Brains—A Systematic Review on the Effects of TDCS on the Cognition of Elderly People. Front. Hum. Neurosci. 2021, 15, 730134. [Google Scholar] [CrossRef] [PubMed]
  14. Ghacibeh, G.A.; Shenker, J.I.; Shenal, B.; Uthman, B.M.; Heilman, K.M. Effect of Vagus Nerve Stimulation on Creativity and Cognitive Flexibility. Epilepsy Behav. 2006, 8, 720–725. [Google Scholar] [CrossRef]
  15. Ghacibeh, G.A.; Shenker, J.I.; Shenal, B.; Uthman, B.M.; Heilman, K.M. The Influence of Vagus Nerve Stimulation on Memory. Cogn. Behav. Neurol. 2006, 19, 119–122. [Google Scholar] [CrossRef]
  16. Goellner, E.; Rocha, C.E. Anatomy of Trigeminal Neuromodulation Targets: From Periphery to the Brain. Neuromodul. Facial Pain 2020, 35, 18–34. [Google Scholar] [CrossRef]
  17. Salles, A.D.; Gorgulho, A. Influence of the Brain Function through the Peripheral Nerves. Surg. Neurol. Int. 2012, 3, 245. [Google Scholar] [CrossRef] [PubMed]
  18. Fanselow, E.E.; Reid, A.P.; Nicolelis, M.A.L. Reduction of Pentylenetetrazole-Induced Seizure Activity in Awake Rats by Seizure-Triggered Trigeminal Nerve Stimulation. J. Neurosci. 2000, 20, 8160–8168. [Google Scholar] [CrossRef]
  19. Liporace, J.; Hucko, D.; Morrow, R.; Barolat, G.; Nei, M.; Schnur, J.; Sperling, M. Vagal Nerve Stimulation&colon; Adjustments to Reduce Painful Side Effects. Neurology 2001, 57, 885–886. [Google Scholar] [CrossRef]
  20. Redgrave, J.; Day, D.; Leung, H.; Laud, P.J.; Ali, A.; Lindert, R.; Majid, A. Safety and Tolerability of Transcutaneous Vagus Nerve Stimulation in Humans; a Systematic Review. Brain Stimul. 2018, 11, 1225–1238. [Google Scholar] [CrossRef]
  21. Giordano, F.; Zicca, A.; Barba, C.; Guerrini, R.; Genitori, L. Vagus Nerve Stimulation: Surgical Technique of Implantation and Revision and Related Morbidity. Epilepsia 2017, 58, 85–90. [Google Scholar] [CrossRef] [PubMed]
  22. Spuck, S.; Tronnier, V.; Orosz, I.; Schönweiler, R.; Sepehrnia, A.; Nowak, G.; Sperner, J. Operative and Technical Complications of Vagus Nerve Stimulator Implantation. Oper. Neurosurg. 2010, 67, ons489–ons494. [Google Scholar] [CrossRef] [PubMed]
  23. Chou, D.E.; Yugrakh, M.S.; Winegarner, D.; Rowe, V.; Kuruvilla, D.; Schoenen, J. Acute Migraine Therapy with External Trigeminal Neurostimulation (ACME): A Randomized Controlled Trial. Cephalalgia 2018, 39, 3–14. [Google Scholar] [CrossRef] [PubMed]
  24. Clark, O.; Mahjoub, A.; Osman, N.; Surmava, A.-M.; Jan, S.; Lagman-Bartolome, A.M. Non-Invasive Neuromodulation in the Acute Treatment of Migraine: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Neurol. Sci. 2021, 43, 153–165. [Google Scholar] [CrossRef]
  25. Conlon, B.; Hamilton, C.; Hughes, S.; Meade, E.; Hall, D.A.; Vanneste, S.; Langguth, B.; Lim, H.H. Noninvasive Bimodal Neuromodulation for the Treatment of Tinnitus: Protocol for a Second Large-Scale Double-Blind Randomized Clinical Trial to Optimize Stimulation Parameters. Jmir. Res. Protoc. 2019, 8, e13176. [Google Scholar] [CrossRef]
  26. Cook, I.A.; Abrams, M.; Leuchter, A.F. Trigeminal Nerve Stimulation for Comorbid Posttraumatic Stress Disorder and Major Depressive Disorder. Neuromodul. Technol. Neural Interface 2016, 19, 299–305. [Google Scholar] [CrossRef]
  27. Cook, I.A.; Schrader, L.M.; DeGiorgio, C.M.; Miller, P.R.; Maremont, E.R.; Leuchter, A.F. Trigeminal Nerve Stimulation in Major Depressive Disorder: Acute Outcomes in an Open Pilot Study. Epilepsy Behav. 2013, 28, 221–226. [Google Scholar] [CrossRef]
  28. DeGiorgio, C.M.; Fanselow, E.E.; Schrader, L.M.; Cook, I.A. Trigeminal Nerve Stimulation: Seminal Animal and Human Studies for Epilepsy and Depression. Neurosurg. Clin. N. Am. 2011, 22, 449–456. [Google Scholar] [CrossRef]
  29. McGough, J.J.; Sturm, A.; Cowen, J.; Tung, K.; Salgari, G.C.; Leuchter, A.F.; Cook, I.A.; Sugar, C.A.; Loo, S.K. Double-Blind, Sham-Controlled, Pilot Study of Trigeminal Nerve Stimulation for Attention-Deficit/Hyperactivity Disorder. J. Am. Acad. Child Adolesc. Psychiatry 2019, 58, 403–411.e3. [Google Scholar] [CrossRef]
  30. Generoso, M.B.; Taiar, I.T.; Garrocini, L.P.; Bernardon, R.; Cordeiro, Q.; Uchida, R.R.; Shiozawa, P. Effect of a 10-Day Transcutaneous Trigeminal Nerve Stimulation (TNS) Protocol for Depression Amelioration: A Randomized, Double Blind, and Sham-Controlled Phase II Clinical Trial. Epilepsy Behav. 2019, 95, 39–42. [Google Scholar] [CrossRef]
  31. Gil-López, F.; Boget, T.; Manzanares, I.; Donaire, A.; Conde-Blanco, E.; Baillés, E.; Pintor, L.; Setoaín, X.; Bargalló, N.; Navarro, J.; et al. External Trigeminal Nerve Stimulation for Drug Resistant Epilepsy: A Randomized Controlled Trial. Brain Stimul. 2020, 13, 1245–1253. [Google Scholar] [CrossRef]
  32. Ginatempo, F.; Fois, C.; Carli, F.D.; Todesco, S.; Mercante, B.; Sechi, G.; Deriu, F. Effect of Short-Term Transcutaneous Trigeminal Nerve Stimulation on EEG Activity in Drug-Resistant Epilepsy. J. Neurol. Sci. 2019, 400, 90–96. [Google Scholar] [CrossRef] [PubMed]
  33. Olivié-García, L.; Giraldez, B.G.; Sierra-Marcos, A.; Díaz-Gómez, E.; Serratosa, J.M. External Trigeminal Nerve Stimulation: A Long Term Follow up Study. Seizure 2019, 69, 218–220. [Google Scholar] [CrossRef] [PubMed]
  34. Shiozawa, P.; da Silva, M.E.; de Carvalho, T.C.; Cordeiro, Q.; Brunoni, A.R.; Fregni, F. Transcutaneous Vagus and Trigeminal Nerve Stimulation for Neuropsychiatric Disorders: A Systematic Review. Arq. Neuro-Psiquiat. 2014, 72, 542–547. [Google Scholar] [CrossRef]
  35. Koek, R.J.; Roach, J.; Athanasiou, N.; van’t Wout-Frank, M.; Philip, N.S. Neuromodulatory Treatments for Post-Traumatic Stress Disorder (PTSD). Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2019, 92, 148–160. [Google Scholar] [CrossRef] [PubMed]
  36. Loo, S.K.; Salgari, G.C.; Ellis, A.; Cowen, J.; Dillon, A.; McGough, J.J. Trigeminal Nerve Stimulation for Attention-Deficit/Hyperactivity Disorder: Cognitive and Electrophysiological Predictors of Treatment Response. J. Am. Acad. Child Adolesc. Psychiatry 2020, 60, 856–864.e1. [Google Scholar] [CrossRef]
  37. Slaght, S.J.; Nashef, L. An Audit of External Trigeminal Nerve Stimulation (ETNS) in Epilepsy. Seizure 2017, 52, 60–62. [Google Scholar] [CrossRef] [PubMed]
  38. Soleymani, T.; Pieton, D.; Pezeshkian, P.; Miller, P.; Gorgulho, A.A.; Pouratian, N.; Salles, A.A.F.D. Surgical Approaches to Tinnitus Treatment: A Review and Novel Approaches. Surg. Neurol. Int. 2011, 2, 154. [Google Scholar] [CrossRef]
  39. Soss, J.; Heck, C.; Murray, D.; Markovic, D.; Oviedo, S.; Corrale-Leyva, G.; Gordon, S.; Kealey, C.; DeGiorgio, C. A Prospective Long-Term Study of External Trigeminal Nerve Stimulation for Drug-Resistant Epilepsy. Epilepsy Behav. 2015, 42, 44–47. [Google Scholar] [CrossRef]
  40. Trevizol, A.; Bonadia, B.; Gomes, J.S.; Cordeiro, Q.; Shiozawa, P. Integrity of Cognitive Functions in Trigeminal Nerve Stimulation Trials in Neuropsychiatry. Trends Psychiatry Psychother. 2016, 38, 60–61. [Google Scholar] [CrossRef]
  41. Trevizol, A.P.; Sato, I.A.; Cook, I.A.; Shiozawa, P.; Lowenthal, R.; Cordeiro, Q. Trigeminal Nerve Stimulation (TNS) for Posttraumatic Stress Disorder and Major Depressive Disorder: An Open-Label Proof-of-Concept Trial. Epilepsy Behav. 2016, 60, 240–241. [Google Scholar] [CrossRef] [PubMed]
  42. Canady, V.A. FDA Approves Marketing of First Nondrug Treatment for ADHD. Ment. Health Wkly. 2019, 29, 3–4. [Google Scholar] [CrossRef]
  43. Lauritsen, C.G.; Silberstein, S.D. Rationale for Electrical Parameter Determination in External Trigeminal Nerve Stimulation (ETNS) for Migraine: A Narrative Review. Cephalalgia 2018, 39, 750–760. [Google Scholar] [CrossRef] [PubMed]
  44. Schoenen, J.; Coppola, G. Efficacy and Mode of Action of External Trigeminal Neurostimulation in Migraine. Expert Rev. Neurother. 2018, 18, 545–555. [Google Scholar] [CrossRef]
  45. Konstantinos, S.; Vikelis, M.; Rapoport, A. Acute Care and Treatment of Migraine. J. Neuro-Ophthalmol. 2020, 40, 472–484. [Google Scholar] [CrossRef] [PubMed]
  46. Nune, G.; DeGiorgio, C.; Heck, C. Neuromodulation in the Treatment of Epilepsy. Curr. Treat. Option Neurol. 2015, 17, 43. [Google Scholar] [CrossRef]
  47. Colzato, L.S.; Vonck, K. Theory-Driven Approaches to Cognitive Enhancement; Springer International Publishing: Cham, Switzerland, 2017; pp. 115–126. [Google Scholar] [CrossRef]
  48. Boasso, A.M.; Mortimore, H.; Silva, R.; Aven, L.; Tyler, W.J. Transdermal Electrical Neuromodulation of the Trigeminal Sensory Nuclear Complex Improves Sleep Quality and Mood. Biorxiv 2016, 043901. [Google Scholar] [CrossRef]
  49. Neisser. Cognition and Reality: Principles and Implications of Cognitive Psychology; W.H. Freeman and Co.: San Francisco, CA, USA, 1976. [Google Scholar]
  50. Bayne, T.; Brainard, D.; Byrne, R.W.; Chittka, L.; Clayton, N.; Heyes, C.; Mather, J.; Ölveczky, B.; Shadlen, M.; Suddendorf, T.; et al. What Is Cognition? Curr. Biol. 2019, 29, R608–R615. [Google Scholar] [CrossRef]
  51. Beaty, R.E.; Benedek, M.; Silvia, P.J.; Schacter, D.L. Creative Cognition and Brain Network Dynamics. Trends Cogn. Sci. 2016, 20, 87–95. [Google Scholar] [CrossRef]
  52. Park, H.-J.; Friston, K. Structural and Functional Brain Networks: From Connections to Cognition. Science 2013, 342, 1238411. [Google Scholar] [CrossRef]
  53. Petersen, S.E.; Sporns, O. Brain Networks and Cognitive Architectures. Neuron 2015, 88, 207–219. [Google Scholar] [CrossRef]
  54. Cohen, J.R.; D’Esposito, M. The Segregation and Integration of Distinct Brain Networks and Their Relationship to Cognition. J. Neurosci. 2016, 36, 12083–12094. [Google Scholar] [CrossRef]
  55. Hearne, L.J.; Cocchi, L.; Zalesky, A.; Mattingley, J.B. Reconfiguration of Brain Network Architectures between Resting-State and Complexity-Dependent Cognitive Reasoning. J. Neurosci. 2017, 37, 8399–8411. [Google Scholar] [CrossRef]
  56. Luckey, A.M.; McLeod, L.S.; Huang, Y.; Mohan, A.; Vanneste, S. Making Memories Last Using the Peripheral Effect of Direct Current Stimulation. eLife 2023, 12, e75586. [Google Scholar] [CrossRef]
  57. Luckey, A.M.; Adcock, K.; Vanneste, S. Peripheral Nerve Stimulation: A Neuromodulation-Based Approach. Neurosci. Biobehav. Rev. 2023, 149, 105180. [Google Scholar] [CrossRef]
  58. Joo, W.; Yoshioka, F.; Funaki, T.; Mizokami, K.; Rhoton, A.L. Microsurgical Anatomy of the Trigeminal Nerve. Clin. Anat. 2014, 27, 61–88. [Google Scholar] [CrossRef] [PubMed]
  59. Haines, D.E.; Mihailoff, G.A. Chapter 14—A Synopsis of Cranial Nerves of the Brainstem. In Fundamental Neuroscience for Basic and Clinical Applications, 5th ed.; Elsevier: Amsterdam, The Netherlands, 2018; pp. 195–211. [Google Scholar] [CrossRef]
  60. Ricardo, J.A.; Koh, E.T. Anatomical Evidence of Direct Projections from the Nucleus of the Solitary Tract to the Hypothalamus, Amygdala, and Other Forebrain Structures in the Rat. Brain Res. 1978, 153, 1–26. [Google Scholar] [CrossRef] [PubMed]
  61. Mercante, B.; Enrico, P.; Floris, G.; Quartu, M.; Boi, M.; Serra, M.P.; Follesa, P.; Deriu, F. Trigeminal Nerve Stimulation Induces Fos Immunoreactivity in Selected Brain Regions, Increases Hippocampal Cell Proliferation and Reduces Seizure Severity in Rats. Neuroscience 2017, 361, 69–80. [Google Scholar] [CrossRef] [PubMed]
  62. Jhaveri, D.J.; Mackay, E.W.; Hamlin, A.S.; Marathe, S.V.; Nandam, L.S.; Vaidya, V.A.; Bartlett, P.F. Norepinephrine Directly Activates Adult Hippocampal Precursors via 3-Adrenergic Receptors. J. Neurosci. 2010, 30, 2795–2806. [Google Scholar] [CrossRef] [PubMed]
  63. Roosevelt, R.W.; Smith, D.C.; Clough, R.W.; Jensen, R.A.; Browning, R.A. Increased Extracellular Concentrations of Norepinephrine in Cortex and Hippocampus Following Vagus Nerve Stimulation in the Rat. Brain Res. 2006, 1119, 124–132. [Google Scholar] [CrossRef]
  64. Follesa, P.; Biggio, F.; Gorini, G.; Caria, S.; Talani, G.; Dazzi, L.; Puligheddu, M.; Marrosu, F.; Biggio, G. Vagus Nerve Stimulation Increases Norepinephrine Concentration and the Gene Expression of BDNF and BFGF in the Rat Brain. Brain Res. 2007, 1179, 28–34. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, Q.-Q.; Zhu, L.-J.; Wang, X.-H.; Zuo, J.; He, H.-Y.; Tian, M.-M.; Wang, L.; Liang, G.-L.; Wang, Y. Chronic Trigeminal Nerve Stimulation Protects Against Seizures, Cognitive Impairments, Hippocampal Apoptosis, and Inflammatory Responses in Epileptic Rats. J. Mol. Neurosci. 2016, 59, 78–89. [Google Scholar] [CrossRef] [PubMed]
  66. Xu, J.; Wu, S.; Huo, L.; Zhang, Q.; Liu, L.; Ye, Z.; Cao, J.; Ma, H.; Shang, C.; Ma, C. Trigeminal Nerve Stimulation Restores Hippocampal Dopamine Deficiency to Promote Cognitive Recovery in Traumatic Brain Injury. Prog. Neurobiol. 2023, 227, 102477. [Google Scholar] [CrossRef]
  67. Axelson, H.W.; Isberg, M.; Flink, R.; Amandusson, Å. Trigeminal Nerve Stimulation Does Not Acutely Affect Cortical Excitability in Healthy Subjects. Brain Stimul. 2014, 7, 613–617. [Google Scholar] [CrossRef]
  68. Mercante, B.; Pilurzi, G.; Ginatempo, F.; Manca, A.; Follesa, P.; Tolu, E.; Deriu, F. Trigeminal Nerve Stimulation Modulates Brainstem More than Cortical Excitability in Healthy Humans. Exp. Brain Res. 2015, 233, 3301–3311. [Google Scholar] [CrossRef] [PubMed]
  69. Pilurzi, G.; Mercante, B.; Ginatempo, F.; Follesa, P.; Tolu, E.; Deriu, F. Transcutaneous Trigeminal Nerve Stimulation Induces a Long-Term Depression-like Plasticity of the Human Blink Reflex. Exp. Brain Res. 2016, 234, 453–461. [Google Scholar] [CrossRef]
  70. Mercante, B.; Loi, N.; Ginatempo, F.; Biggio, M.; Manca, A.; Bisio, A.; Enrico, P.; Bove, M.; Deriu, F. Transcutaneous Trigeminal Nerve Stimulation Modulates the Hand Blink Reflex. Sci. Rep. 2020, 10, 21116. [Google Scholar] [CrossRef] [PubMed]
  71. Sambo, C.F.; Forster, B.; Williams, S.C.; Iannetti, G.D. To Blink or Not to Blink: Fine Cognitive Tuning of the Defensive Peripersonal Space. J. Neurosci. 2012, 32, 12921–12927. [Google Scholar] [CrossRef]
  72. Ginatempo, F.; Carli, F.D.; Todesco, S.; Mercante, B.; Sechi, G.P.; Deriu, F. Effects of Acute Trigeminal Nerve Stimulation on Rest EEG Activity in Healthy Adults. Exp. Brain Res. 2018, 236, 2839–2845. [Google Scholar] [CrossRef]
  73. Ritland, B.M.; Neumeier, W.H.; Jiang, S.H.; Smith, C.D.; Heaton, K.J.; Hildebrandt, A.M.; Jabbar, M.A.; Liao, H.J.; Coello, E.; Zhao, W.; et al. Short-term Neurochemical Effects of Transcutaneous Trigeminal Nerve Stimulation Using 7T Magnetic Resonance Spectroscopy. J. Neuroimaging 2022, 33, 279–288. [Google Scholar] [CrossRef]
  74. Ferguson, K.J.; MacLullich, A.M.J.; Marshall, I.; Deary, I.J.; Starr, J.M.; Seckl, J.R.; Wardlaw, J.M. Magnetic Resonance Spectroscopy and Cognitive Function in Healthy Elderly Men. Brain 2002, 125, 2743–2749. [Google Scholar] [CrossRef]
  75. GOADSBY, P.J.; HOSKIN, K.L. The Distribution of Trigeminovascular Afferents in the Nonhuman Primate Brain Macaca Nemestrina: A C-fos Immunocytochemical Study. J. Anat. 1997, 190, 367–375. [Google Scholar] [CrossRef]
  76. Kumada, M.; Dampney, R.A.L.; Reis, D.J. The Trigeminal Depressor Response: A Novel Vasodepressor Response Originating from the Trigeminal System. Brain Res. 1977, 119, 305–326. [Google Scholar] [CrossRef]
  77. Chiluwal, A.; Narayan, R.K.; Chaung, W.; Mehan, N.; Wang, P.; Bouton, C.E.; Golanov, E.V.; Li, C. Neuroprotective Effects of Trigeminal Nerve Stimulation in Severe Traumatic Brain Injury. Sci. Rep. 2017, 7, 6792. [Google Scholar] [CrossRef]
  78. Golanov, E.V. Forehead Stimulation Decreases Volume of the Infarction Triggered by Permanent Occlusion of Middle Cerebral Artery in Rats. J. Neurol. Stroke 2015, 2, 15406. [Google Scholar] [CrossRef]
  79. Li, C.; Chiluwal, A.; Afridi, A.; Chaung, W.; Powell, K.; Yang, W.-L.; Wang, P.; Narayan, R.K. Trigeminal Nerve Stimulation: A Novel Method of Resuscitation for Hemorrhagic Shock. Crit. Care Med. 2019, 47, e478–e484. [Google Scholar] [CrossRef]
  80. Mercante, B.; Nuvoli, S.; Sotgiu, M.A.; Manca, A.; Todesco, S.; Melis, F.; Spanu, A.; Deriu, F. SPECT Imaging of Cerebral Blood Flow Changes Induced by Acute Trigeminal Nerve Stimulation in Drug-Resistant Epilepsy. A Pilot Study. Clin. Neurophysiol. 2021, 132, 1274–1282. [Google Scholar] [CrossRef]
  81. White, T.G.; Powell, K.; Shah, K.A.; Woo, H.H.; Narayan, R.K.; Li, C. Trigeminal Nerve Control of Cerebral Blood Flow: A Brief Review. Front. Neurosci. 2021, 15, 649910. [Google Scholar] [CrossRef]
  82. Piquet, M.; Balestra, C.; Sava, S.L.; Schoenen, J.E. Supraorbital Transcutaneous Neurostimulation Has Sedative Effects in Healthy Subjects. BMC Neurol. 2011, 11, 135. [Google Scholar] [CrossRef]
  83. Ginatempo, F.; Pirina, P.; Melis, F.; Deriu, F. Short-Term Trigeminal Neuromodulation Does Not Alter Sleep Latency in Healthy Subjects: A Pilot Study. Neurol. Sci. 2018, 39, 145–147. [Google Scholar] [CrossRef]
  84. Cicco, V.D.; Fantozzi, M.P.T.; Cataldo, E.; Barresi, M.; Bruschini, L.; Faraguna, U.; Manzoni, D. Trigeminal, Visceral and Vestibular Inputs May Improve Cognitive Functions by Acting through the Locus Coeruleus and the Ascending Reticular Activating System: A New Hypothesis. Front. Neuroanat. 2018, 11, 130. [Google Scholar] [CrossRef] [PubMed]
  85. Tyler, W.J.; Boasso, A.M.; Mortimore, H.M.; Silva, R.S.; Charlesworth, J.D.; Marlin, M.A.; Aebersold, K.; Aven, L.; Wetmore, D.Z.; Pal, S.K. Transdermal Neuromodulation of Noradrenergic Activity Suppresses Psychophysiological and Biochemical Stress Responses in Humans. Sci. Rep. 2015, 5, 13865. [Google Scholar] [CrossRef] [PubMed]
  86. Helfrich, R.F.; Knight, R.T. Chapter 36 Cognitive Neurophysiology: Event-Related Potentials. Handb. Clin. Neurol. 2019, 160, 543–558. [Google Scholar] [CrossRef] [PubMed]
  87. Luck, S.J. An Introduction to the Event-Related Potential Technique, 2nd ed.; The MIT Press: Cambridge, MA, USA, 2014. [Google Scholar]
  88. Wongupparaj, P.; Sumich, A.; Wickens, M.; Kumari, V.; Morris, R.G. Individual Differences in Working Memory and General Intelligence Indexed by P200 and P300: A Latent Variable Model. Biol. Psychol. 2018, 139, 96–105. [Google Scholar] [CrossRef]
  89. Verleger, R. Effects of Relevance and Response Frequency on P3b Amplitudes: Review of Findings and Comparison of Hypotheses about the Process Reflected by P3b. Psychophysiology 2020, 57, e13542. [Google Scholar] [CrossRef]
  90. Herrmann, C.S.; Knight, R.T. Mechanisms of Human Attention: Event-Related Potentials and Oscillations. Neurosci. Biobehav. Rev. 2001, 25, 465–476. [Google Scholar] [CrossRef]
  91. Fantozzi, M.P.T.; Artoni, F.; Galante, M.D.; Briscese, L.; Cicco, V.D.; Bruschini, L.; d’Ascanio, P.; Manzoni, D.; Faraguna, U.; Carboncini, M.C. Effect of the Trigeminal Nerve Stimulation on Auditory Event-Related Potentials. Cereb. Cortex Commun. 2021, 2, tgab012. [Google Scholar] [CrossRef]
  92. Mercante, B.; Enrico, P.; Ginatempo, F.; Loi, N.; Deriu, F. Short-Term Transcutaneous Trigeminal Nerve Stimulation Does Not Affect Visual Oddball Task and Paired-Click Paradigm ERP Responses in Healthy Volunteers. Exp. Brain Res. 2022, 241, 327–339. [Google Scholar] [CrossRef]
  93. Fantozzi, M.P.T.; Cicco, V.D.; Argento, S.; Cicco, D.D.; Barresi, M.; Cataldo, E.; Bruschini, L.; d’Ascanio, P.; Faraguna, U.; Manzoni, D. Trigeminal Input, Pupil Size and Cognitive Performance: From Oral to Brain Matter. Brain Res. 2020, 1751, 147194. [Google Scholar] [CrossRef]
  94. Dunn, B.R.; Dunn, D.A.; Languis, M.; Andrews, D. The Relation of ERP Components to Complex Memory Processing. Brain Cogn. 1998, 36, 355–376. [Google Scholar] [CrossRef]
  95. Sara, S.J. The Locus Coeruleus and Noradrenergic Modulation of Cognition. Nat. Rev. Neurosci. 2009, 10, 211–223. [Google Scholar] [CrossRef] [PubMed]
  96. Wijdicks, E.F.M. The Ascending Reticular Activating System. Neurocrit. Care 2019, 31, 419–422. [Google Scholar] [CrossRef]
  97. Monaco, A.; Cattaneo, R.; Smurra, P.; Nicolantonio, S.D.; Cipriano, F.; Pietropaoli, D.; Ortu, E. Trigeminal Electrical Stimulation with ULFTENS of the Dorsal Anterior Mucosal Surface of the Tongue: Effects on Heart Rate Variability (HRV). PLoS ONE 2023, 18, e0285464. [Google Scholar] [CrossRef]
  98. Danilov, Y.P.; Tyler, M.E.; Kaczmarek, K.A.; Skinner, K.L. New Approach to Neurorehabilitation: Cranial Nerve Noninvasive Neuromodulation (CN-NINM) Technology. In Sensing Technologies for Global Health, Military Medicine, and Environmental Monitoring IV; SPIE: Bellingham, UK, 2014; pp. 91120L–91120L-10. [Google Scholar] [CrossRef]
  99. Daniel, O.; Sharon, R.; Tepper, S.J. A Device Review of Relivion®: An External Combined Occipital and Trigeminal Neurostimulation (ECOT-NS) System for Self-Administered Treatment of Migraine and Major Depressive Disorder. Expert Rev. Med. Devic. 2021, 18, 333–342. [Google Scholar] [CrossRef]
  100. Butt, M.F.; Albusoda, A.; Farmer, A.D.; Aziz, Q. The Anatomical Basis for Transcutaneous Auricular Vagus Nerve Stimulation. J. Anat. 2020, 236, 588–611. [Google Scholar] [CrossRef] [PubMed]
  101. Peuker, E.T.; Filler, T.J. The Nerve Supply of the Human Auricle. Clin. Anat. 2002, 15, 35–37. [Google Scholar] [CrossRef] [PubMed]
  102. Mercante, B.; Deriu, F.; Rangon, C.-M. Auricular Neuromodulation: The Emerging Concept beyond the Stimulation of Vagus and Trigeminal Nerves. Medicines 2018, 5, 10. [Google Scholar] [CrossRef]
  103. Chen, Y.; Lu, X.; Hu, L. Transcutaneous Auricular Vagus Nerve Stimulation Facilitates Cortical Arousal and Alertness. Int. J. Environ. Res. Public Health 2023, 20, 1402. [Google Scholar] [CrossRef]
  104. Gurtubay, I.G.; Perez-Rodriguez, D.R.; Fernandez, E.; Librero-Lopez, J.; Calvo, D.; Bermejo, P.; Pinin-Osorio, C.; Lopez, M. Immediate Effects and Duration of a Short and Single Application of Transcutaneous Auricular Vagus Nerve Stimulation on P300 Event Related Potential. Front. Neurosci. 2023, 17, 1096865. [Google Scholar] [CrossRef]
  105. Konjusha, A.; Yu, S.; Mückschel, M.; Colzato, L.; Ziemssen, T.; Beste, C. Auricular Transcutaneous Vagus Nerve Stimulation Specifically Enhances Working Memory Gate Closing Mechanism: A System Neurophysiological Study. J. Neurosci. 2023, 43, 4709–4724. [Google Scholar] [CrossRef]
  106. Smet, S.D.; Ottaviani, C.; Verkuil, B.; Kappen, M.; Baeken, C.; Vanderhasselt, M. Effects of Non-invasive Vagus Nerve Stimulation on Cognitive and Autonomic Correlates of Perseverative Cognition. Psychophysiology 2023, 60, e14250. [Google Scholar] [CrossRef] [PubMed]
  107. Sommer, A.; Fischer, R.; Borges, U.; Laborde, S.; Achtzehn, S.; Liepelt, R. The Effect of Transcutaneous Auricular Vagus Nerve Stimulation (TaVNS) on Cognitive Control in Multitasking. Neuropsychologia 2023, 187, 108614. [Google Scholar] [CrossRef] [PubMed]
  108. Hassert, D.L.; Miyashita, T.; Williams, C.L. The Effects of Peripheral Vagal Nerve Stimulation at a Memory-Modulating Intensity on Norepinephrine Output in the Basolateral Amygdala. Behav. Neurosci. 2004, 118, 79–88. [Google Scholar] [CrossRef] [PubMed]
  109. Giraudier, M.; Ventura-Bort, C.; Weymar, M. Transcutaneous Vagus Nerve Stimulation (TVNS) Improves High-Confidence Recognition Memory but Not Emotional Word Processing. Front. Psychol. 2020, 11, 1276. [Google Scholar] [CrossRef] [PubMed]
  110. Giraudier, M.; Ventura-Bort, C.; Burger, A.M.; Claes, N.; D’Agostini, M.; Fischer, R.; Franssen, M.; Kaess, M.; Koenig, J.; Liepelt, R.; et al. Evidence for a Modulating Effect of Transcutaneous Auricular Vagus Nerve Stimulation (TaVNS) on Salivary Alpha-Amylase as Indirect Noradrenergic Marker: A Pooled Mega-Analysis. Brain Stimul. 2022, 15, 1378–1388. [Google Scholar] [CrossRef]
  111. Sharon, O.; Fahoum, F.; Nir, Y. Transcutaneous Vagus Nerve Stimulation in Humans Induces Pupil Dilation and Attenuates Alpha Oscillations. J. Neurosci. 2020, 41, 320–330. [Google Scholar] [CrossRef]
  112. Ventura-Bort, C.; Wirkner, J.; Genheimer, H.; Wendt, J.; Hamm, A.O.; Weymar, M. Effects of Transcutaneous Vagus Nerve Stimulation (TVNS) on the P300 and Alpha-Amylase Level: A Pilot Study. Front. Hum. Neurosci. 2018, 12, 202. [Google Scholar] [CrossRef]
  113. Diep, D.; Lam, A.C.L.; Ko, G. A Review of the Evidence and Current Applications of Portable Translingual Neurostimulation Technology. Neuromodul. Technol. Neural Interface 2021, 24, 1377–1387. [Google Scholar] [CrossRef]
  114. Wildenberg, J.C.; Tyler, M.E.; Danilov, Y.P.; Kaczmarek, K.A.; Meyerand, M.E. Sustained Cortical and Subcortical Neuromodulation Induced by Electrical Tongue Stimulation. Brain Imaging Behav. 2010, 4, 199–211. [Google Scholar] [CrossRef]
  115. Danilov, Y.P. Translingual Neurostimulation (TLNS): A Novel Approach to Neurorehabilitation. J. Neurol. Neurophysiol. 2017, 8, 1117. [Google Scholar] [CrossRef]
  116. Spencer, S.; Mielczarek, M.; Olszewski, J.; Sereda, M.; Joossen, I.; Vermeersch, H.; Gilles, A.; Michiels, S. Effectiveness of Bimodal Auditory and Electrical Stimulation in Patients with Tinnitus: A Feasibility Study. Front. Neurosci. 2022, 16, 971633. [Google Scholar] [CrossRef]
  117. Conlon, B.; Langguth, B.; Hamilton, C.; Hughes, S.; Meade, E.; Connor, C.O.; Schecklmann, M.; Hall, D.A.; Vanneste, S.; Leong, S.L.; et al. Bimodal Neuromodulation Combining Sound and Tongue Stimulation Reduces Tinnitus Symptoms in a Large Randomized Clinical Study. Sci. Transl. Med. 2020, 12, eabb2830. [Google Scholar] [CrossRef]
  118. Frehlick, Z.; Lakhani, B.; Fickling, S.D.; Livingstone, A.C.; Danilov, Y.; Sackier, J.M.; D’Arcy, R.C.N. Human Translingual Neurostimulation Alters Resting Brain Activity in High-Density EEG. J. Neuroeng. Rehabil. 2019, 16, 60. [Google Scholar] [CrossRef]
  119. Hou, J.; Mohanty, R.; Chu, D.; Nair, V.A.; Danilov, Y.; Kaczmarek, K.A.; Meyerand, B.; Tyler, M.; Prabhakaran, V. Translingual Neural Stimulation Affects Resting-state Functional Connectivity in Mild-moderate Traumatic Brain Injury. J. Neuroimaging 2022, 32, 1193–1200. [Google Scholar] [CrossRef] [PubMed]
  120. Sanders, R.D. The Trigeminal (V) and Facial (VII) Cranial Nerves: Head and Face Sensation and Movement. Psychiatry (Edgmont) 2010, 7, 13–16. [Google Scholar] [PubMed]
  121. Alhelal, M.A.; Palaska, I.; Panagiotidou, S.; Letourneau, R.; Theoharides, T.C. Trigeminal Nerve Stimulation Triggers Oral Mast Cell Activation and Vascular Permeability. Ann. Allergy Asthma Immunol. 2014, 112, 40–45. [Google Scholar] [CrossRef] [PubMed]
  122. Dingle, A.; Zeng, W.; Ness, J.P.; Albano, N.; Minor, R.L.; Feldman, C.; Austin, M.; Brodnick, S.K.; Shulzhenko, N.; Sanchez, R.; et al. Strategies for Interfacing with the Trigeminal Nerves in Rodents for Bioelectric Medicine. J. Neurosci. Meth. 2019, 324, 108321. [Google Scholar] [CrossRef] [PubMed]
  123. Ding, W.; You, Z.; Shen, S.; Yang, J.; Lim, G.; Doheny, J.T.; Chen, L.; Zhu, S.; Mao, J. An Improved Rodent Model of Trigeminal Neuropathic Pain by Unilateral Chronic Constriction Injury of Distal Infraorbital Nerve. J. Pain 2017, 18, 899–907. [Google Scholar] [CrossRef]
  124. Pitcher, G.M.; Ritchie, J.; Henry, J.L. Nerve Constriction in the Rat: Model of Neuropathic, Surgical and Central Pain. PAIN 1999, 83, 37–46. [Google Scholar] [CrossRef]
  125. Vos, B.; Strassman, A.; Maciewicz, R. Behavioral Evidence of Trigeminal Neuropathic Pain Following Chronic Constriction Injury to the Rat’s Infraorbital Nerve. J. Neurosci. 1994, 14, 2708–2723. [Google Scholar] [CrossRef]
  126. Araya, E.I.; Carvalho, E.C.; Andreatini, R.; Zamponi, G.W.; Chichorro, J.G. Trigeminal Neuropathic Pain Causes Changes in Affective Processing of Pain in Rats. Mol. Pain 2022, 18, 17448069211057750. [Google Scholar] [CrossRef] [PubMed]
  127. Benoist, J.-M.; Gautron, M.; Guilbaud, G. Experimental Model of Trigeminal Pain in the Rat by Constriction of One Infraorbital Nerve: Changes in Neuronal Activities in the Somatosensory Cortices Corresponding to the Infraorbital Nerve. Exp. Brain Res. 1999, 126, 383–398. [Google Scholar] [CrossRef] [PubMed]
  128. Shen, X.; Finn, E.S.; Scheinost, D.; Rosenberg, M.D.; Chun, M.M.; Papademetris, X.; Constable, R.T. Using Connectome-Based Predictive Modeling to Predict Individual Behavior from Brain Connectivity. Nat. Protoc. 2017, 12, 506–518. [Google Scholar] [CrossRef]
  129. Yang, H.; Zhang, J.; Jin, Z.; Bashivan, P.; Li, L. Using Modular Connectome-Based Predictive Modeling to Reveal Brain-Behavior Relationships of Individual Differences in Working Memory. Brain Struct. Funct. 2023, 228, 1479–1492. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Trigeminal nerve branches shown in three different colors: V1—Ophthalmic branch (green); V2—Maxillary branch (pink); and V3—Mandibular branch (blue). Numbers show the electrode placement over the peripheral branches of the trigeminal nerve used in key studies on trigeminal nerve stimulation and cognition: 1—Supraorbital nerve; 2—Infraorbital nerve; 3—Motor branch; 4—Lingual nerve; and 5—Auricolo-temporal nerve.
Figure 1. Trigeminal nerve branches shown in three different colors: V1—Ophthalmic branch (green); V2—Maxillary branch (pink); and V3—Mandibular branch (blue). Numbers show the electrode placement over the peripheral branches of the trigeminal nerve used in key studies on trigeminal nerve stimulation and cognition: 1—Supraorbital nerve; 2—Infraorbital nerve; 3—Motor branch; 4—Lingual nerve; and 5—Auricolo-temporal nerve.
Biomedicines 11 02392 g001
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

Mercante, B.; Enrico, P.; Deriu, F. Cognitive Functions following Trigeminal Neuromodulation. Biomedicines 2023, 11, 2392. https://doi.org/10.3390/biomedicines11092392

AMA Style

Mercante B, Enrico P, Deriu F. Cognitive Functions following Trigeminal Neuromodulation. Biomedicines. 2023; 11(9):2392. https://doi.org/10.3390/biomedicines11092392

Chicago/Turabian Style

Mercante, Beniamina, Paolo Enrico, and Franca Deriu. 2023. "Cognitive Functions following Trigeminal Neuromodulation" Biomedicines 11, no. 9: 2392. https://doi.org/10.3390/biomedicines11092392

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