You are on page 1of 6

Autonomic Neuroscience: Basic and Clinical 243 (2022) 103038

Contents lists available at ScienceDirect

Autonomic Neuroscience: Basic and Clinical


journal homepage: www.elsevier.com/locate/autneu

Review

Transcutaneous vagus nerve stimulation - A brief introduction


and overview
Max J. Hilz a, b, *
a
Department of Neurology, University of Erlangen-Nuremberg, Erlangen, Germany
b
Icahn School of Medicine at Mount Sinai, New York, NY, USA

A R T I C L E I N F O A B S T R A C T

Keywords: Invasive cervical vagus nerve stimulation (VNS) is approved for the treatment of epilepsies, depression, obesity,
Transcutaneous vagus nerve stimulation and for stroke-rehabilitation. The procedure requires surgery, has side-effects, is expensive and not readily
Nucleus tractus solitarii available. Consequently, transcutaneous VNS (tVNS) has been developed 20 years ago as non-invasive, less
Locus coeruleus
expensive, and easily applicable alternative. Since the vagus nerve reaches the skin at the outer acoustic canal
Auricular branch of the vagus nerve
and ear, and reflex-responses such as the ear-cough-reflex or the auriculo-cardiac reflex have been observed upon
Concha cymba
Tragus auricular stimulation, the ear seems well suited for tVNS. However, several sensory nerves with variable fiber-
density and significant overlap innervate the outer ear: the auricular branch of the vagus nerve (ABVN), the
auriculotemporal nerve, greater auricular nerve, and to some extent the lesser occipital nerve. VNS requires
activation of Aβ-fibers which are far less present in the ABVN than the cervical vagus nerve. Thus, optimal
stimulation sites and parameters, and tVNS-algorithms need to be further explored. Unravelling central pathways
and structures that mediate tVNS-effects is another challenge. tVNS impulses reach the nucleus of the solitary
tract and activate the locus-coeruleus-norepinephrine system. However, many more brain areas are activated or
deactivated upon VNS, including structures of the central autonomic network and the limbic system. Still, the
realm of therapeutic tVNS applications grows rapidly and includes medication-refractory epilepsies, depressive
mood disorders, headaches including migraine, pain, heart failure, gastrointestinal inflammatory diseases and
many more. tVNS might become a standard tool to enhance autonomic balance and function in various auto­
nomic, neurological, psychiatric, rheumatologic, as well as other diseases.

1. Invasive vagus nerve stimulation in epilepsy, depression, vagus nerve fibers (Garamendi-Ruiz and Gomez-Esteban, 2019). The
obesity, and heart failure electrodes must be surgically positioned around the left cervical vagus
nerve using an approach similar to that for anterior cervical discectomy
Effects of Vagus Nerve stimulation (VNS) on brain activity have been (Reid, 1990; Giordano et al., 2017). Left-sided stimulation is preferred to
studied already in 1938 by Bailey and Bremer (1938) and in 1951 by reduce the risk of bradycardia or asystole that may be induced by
Dell and Olson (1951). Based on the observation of antiepileptic VNS stimulating the sinoatrial node via right-sided cervical VNS (Giordano
effects, an implantable VNS was developed in 1987, first implanted in et al., 2017).
1988, and approved by the United States Food and Drug Administration The observation of mood improvement in epilepsy patients receiving
(FDA) in 1997 as adjunctive treatment tool for adult and adolescent VNS treatment prompted studies that showed not only improved quality
patients with partial onset seizures that are refractory to antiepileptic of life, emotional adjustment, concentration, and cognitive functioning
drugs (Gonzalez et al., 2019). Nowadays, the implantable VNS is also upon VNS (Austelle et al., 2022); in furtherance of potential antide­
approved for patients aged four years and older suffering from partial pressant VNS effects, Ben-Menachem et al. also found that VNS treat­
onset seizures refractory to antiepileptic medication drugs (Gonzalez ment of 16 patients with complex partial seizures had effects on amino
et al., 2019). In epilepsy patients, a pulse generator implanted in a acid levels in the patients’ cerebrospinal fluid: levels of inhibitory
subcutaneous pocket below the clavicle delivers stimuli via electrodes, gamma-aminobutyric acid were higher while levels of excitatory
usually to the left cervical vagus nerve, in order to activate afferent glutamate were somewhat lower after three months of VNS than before

* Corresponding author at: University of Erlangen-Nuremberg, Schlossplatz 4, D-91054 Erlangen, Germany.


E-mail address: max.hilz@outlook.com.

https://doi.org/10.1016/j.autneu.2022.103038
Received 23 July 2022; Received in revised form 25 September 2022; Accepted 25 September 2022
Available online 27 September 2022
1566-0702/© 2022 Elsevier B.V. All rights reserved.
M.J. Hilz Autonomic Neuroscience: Basic and Clinical 243 (2022) 103038

VNS, supporting the antiseizure effects of VNS. However, the study also stimulation of the recurrent laryngeal nerve while surgical removal,
showed a trend towards increased levels of 5-hydroxyindoleacetic acid, revision or replacement of the device is considered in 4–16.8 % of cases
the main serotonin metabolite, upon VNS (Ben-Menachem et al., 1995) due to device malfunction, failure of VNS therapy, severe side effects, or
The increase of 5-hydroxyindoleacetic acid suggests that VNS might based on the patient’s request (Giordano et al., 2017).
have effects on serotoninergic modulation and thus antidepressant ef­
fects (Austelle et al., 2022). 3. The approach towards noninvasive VNS
Further studies confirmed that VNS yields improvements in the pa­
tient’s sense of well-being and quality of life, concentration, cognitive While invasive VNS is widely used in epilepsy centers (Gonzalez
and social functioning, and emotional adjustment (Dodrill and Morris, et al., 2019; Toffa et al., 2020; Ryvlin et al., 2021), the above mentioned
2001), improved mood scores (Harden et al., 2000), as well as signifi­ side-effects and risk factors as well as the cost of the device and its im­
cant improvement of mild depressive mood disorders (Elger et al., plantation have limited the use of invasive VNS in psychiatry (Austelle
2000). Promising results of many successive studies (e.g., Sackeim et al., et al., 2022).
2001; Marangell et al., 2002; Nahas et al., 2005; Rush et al., 2005a; Rush More than 20 years ago, an alternative concept was developed based
et al., 2005b) in 2005 finally led to the FDA approval of VNS for the on the positive results of invasive VNS, on experience with the already
treatment of major depressive disorders (Austelle et al., 2022). accepted use of transcutaneous electrical nerve stimulation, TENS, e.g.
In 2015, VNS also obtained FDA approval for the treatment of pa­ for pain control or muscle healing, and on the fact that the vagus nerve
tients with moderate to severe obesity (Garamendi-Ruiz and Gomez- reaches the skin in the so called Ramsay Hunt zone of the external
Esteban, 2019). For obesity treatment, a pulse generator again had to auditory canal (Ventureyra, 2000). In many persons, tactile stimulation
be positioned subcutaneously to induce intermittent vagal blockade via of the posterior and inferior quadrants of the external auditory canal
two leads that must be positioned around the anterior and posterior triggers coughing, i.e. the so called Arnold’s ear-cough reflex, but might
vagal trunks close to the gastroesophageal junction (Ikramuddin et al., also induce tear-flow, via an auriculo-lacrimal reflex (Engel, 1979;
2014). In the so-called ReCharge trial, patients with a body mass index Gupta et al., 1986; Ellrich, 2019). There were also reports on physio­
of at least 35 underwent intermittent vagus nerve blockade with the logical interactions between stimuli applied to the external auditory
stimulator delivering charges between 6 and 8 mA for at least 12 h per canal and internal organs, such as the stomach, esophagus, lungs, heart,
day (Ikramuddin et al., 2014). Compared to a control group that uterus, and male or female sexual organs with the vagus nerve as
received sham stimulation, the patients who had received intermittent mediator between auricular impulses and the internal organ responses;
vagus nerve blockade had lost 8.5 % more of their excess weight some of the interactions with organs occurring upon stimulation of the
(Ikramuddin et al., 2014). Side-effects were rare and mainly consisted of external auditory canal are known as ear-vomiting reflex, auriculo-
mild or moderate heartburn, dyspepsia, and abdominal pain (Ikra­ cardiac reflex with cardiac inhibition and subsequent syncope, or gas­
muddin et al., 2014). In 2016, a two-year follow-up study confirmed that troauricular phenomenon; in cats stimulation of the external auditory
the technique induces a medically beneficial weight loss with a mean canal may induce the auriculo-uterine reflex or auriculo-genital reflex
decrease in excess weight of 21 % and significant improvements in lipid consisting of muscle contractions around the cat’s vaginal orifice
levels, HbA1c, systolic and diastolic blood pressure, and quality of life, respectively cutaneous muscle contractions in the penile and perineal
but also corroborated a favorable safety profile (Apovian et al., 2017). area of male cats (Engel, 1979; Ellrich, 2019).
Even in heart failure patients, implanted Vagus nerve stimulators Moreover, there were conference reports suggesting that acupunc­
have been used – but are not yet officially approved - to ameliorate the ture in the region of the auricular branch of the vagus nerve had bene­
imbalance between augmented sympathetic and diminished para­ ficial effects on seizure control (Ventureyra, 2000). Based on these
sympathetic activity in heart failure and thus amend ventricular reports and the known interactions between various organs and sensory
remodeling of the failing heart (Klein and Ferrari, 2010; Garamendi- sites of the external acoustic canal and outer ear, transcutaneous elec­
Ruiz and Gomez-Esteban, 2019). Different from the stimulation in epi­ trical stimulation was suggested as a readily and widely available non-
lepsy patients, the stimulating electrodes are surgically positioned invasive treatment option in epilepsy patients (Ventureyra, 2000).
around the right cervical vagus nerve with the cathode distal to the
anode in order to activate the sinoatrial node in the right cardiac atrium 4. Innervation of the external ear
(Garamendi-Ruiz and Gomez-Esteban, 2019).
While the technique yielded quality-of-life improvement in symp­ In fact, the sensory innervation of the external ear is intricate and not
tomatic heart failure patients, it so far has not reached study end-points only provided by the auricular branch of the vagus nerve (ABVN) but also
such as improvement in left ventricular end-systolic diameter or other by the auriculotemporal nerve, a branch of the mandibular and thus
echocardiographic indices (Zannad et al., 2015). trigeminal nerve, and by the greater auricular nerve (Fig. 1) (Butt et al.,
2020).
2. Possible side-effects of invasive VNS The auriculotemporal nerve has connections to the facial nerve and to
the ganglion oticum where it receives parasympathetic fibers from the
In general, the application of implanted vagus nerve stimulators is glossopharyngeal nerve which then supply the parotid gland (Butt et al.,
associated with a variety of drawbacks and side-effects (Giordano et al., 2020).
2017). The implantation requires general anesthesia with endotracheal The greater auricular nerve branches off the cervical plexus and arises
intubation which might cause vocal cord damage (Giordano et al., from the spinal nerves C2 and C3 (Ginsberg and Eicher, 2000). More­
2017). During intraoperative impedance testing there is a risk of over, there may be contributions from the lesser occipital nerve that also
bradycardia or asystole. According to Giordano et al. further surgery arises from the cervical plexus and contains C2 and C3 fibers (Butt et al.,
related risks include peritracheal hematoma, infections, vagus nerve 2020).
injury causing left vocal cord paralysis with hoarseness, dyspnea, and
dysphagia; related to the stimulating system, there may be postsurgical 5. A beta fibers, not C-fibers mediate afferent VNS impulses to
or later infections and wound healing difficulties as well as later injuries the nucleus tractus solitarii
of the vagus nerve; the cervical vagus nerve stimulation itself may cause
arrhythmias, hoarseness, dyspnea, or coughing; even obstructive sleep In human patients and in animals, invasive cervical VNS studies
apnea may occur; there might be stimulation of the adjacent phrenic provided evidence that therapeutic effects are mediated via A beta nerve
nerve, and tonsillar pain (Giordano et al., 2017). 66 % of patients fiber activation (Vonck et al., 2007; Ellrich, 2019). For example, in rats
experience - usually transient - laryngopharyngeal dysfunction with capsaicin-induced destruction of C-fibers did not compromise VNS-

2
M.J. Hilz Autonomic Neuroscience: Basic and Clinical 243 (2022) 103038

Fig. 1. The outer ear and its cutaneous innervation: mainly three nerves
contribute to the innervation of the outer ear: the auricular branch of the vagus
nerve (ABVN), the greater auricular nerve (GAN), and the auriculotemporal
nerve (ATN) (Murray et al., 2016). Fig. 2. Distribution and overlap of the ABVN (green) with the auriculotemporal
[Reprinted from Autonomic Neurosciences: Basic & Clinical, 2016, Vol 199, nerve (red), greater auricular nerve (yellow), and minor occipital nerve (blue)
Murray, A. R., L. Atkinson, M. K. Mahadi, S. A. Deuchars and J. Deuchars. The according to He et al. (2012). (For interpretation of the references to color in
strange case of the ear and the heart: The auricular vagus nerve and its influ­ this figure legend, the reader is referred to the web version of this article.)
ence on cardiac control. Pages 48–53, Copyright 2016, with permission [Reprinted from Evidence-Based Complementary and Alternative Medicine,
from Elsevier]. Volume 2012, Article ID 786839, Hindawi Publishing Corporation; He, W., X.
Wang, H. Shi, H. Shang, L. Li, X. Jing and B. Zhu (2012). Auricular acupuncture
mediated suppression of pentylenetetrazol triggered seizures (Krahl and vagal regulation. Pages 1–6; Copyright © 2012 Wei He et al. This is an open
et al., 2001). Yet, afferent A beta fibers are five or six times less common access article distributed under the Creative Commons Attribution License; no
formal permissions required].
in the ABVN than in the cervical vagus nerve, and the fiber count may
even vary more between individuals which may hamper transcutaneous
VNS in some persons (Butt et al., 2020). studies evaluating functional magnetic resonance imaging (fMRI) re­
The facts that there are relatively less A beta fibers in the ABVN than sponses or somatosensory evoked potentials induced by auricular VNS
in the cervical vagus nerve, that there are interconnections and overlap also used different frequencies, intensities, and stimulus duration as
of the ABVN with the various aforementioned nerves (Fig. 2), and that summarized by Butt et al. (2020). Side-effects are minor with these
cadaveric studies of the ABVN distribution and fiber density show het­ auricular tVNS parameters and mainly include skin reddening and irri­
erogeneous results explain why the determination of the optimal tation (Badran et al., 2019).
auricular stimulation site is still ongoing (Badran et al., 2018a; Badran
et al., 2019). Still, it seems that the concha, particularly the cymba 7. The complex pathways mediating autonomic and other
conchae are sites of adequate ABVN presentation and thus particularly responses to auricular tVNS
suitable for noninvasive auricular VNS (Yakunina et al., 2017; Butt et al.,
2020). In contrast, there is an overlap of nerves at various other areas, e. While VNS is not considered to induce its physiological effects by C-
g. the ear lobe (He et al., 2012). fiber depolarization but via A beta fiber activation (Butt et al., 2020),
autonomic effects still occur with transcutaneous auricular VNS. [For
6. Auricular stimulation parameters reviews see Butt et al., 2020]. However, cardiovascular autonomic ef­
fects vary and seem to depend on the auricular stimulation site and
Mostly, auricular stimulation is delivered at the left ear as this side varying stimulation parameters (Butt et al., 2020). For example, Badran
has been thought to be safer although Badran et al. report that data from et al. tested effects of inner tragus stimulation on heart rate using nine
a large trial do not show an increased risk of adverse events with right varying stimulation blocks with frequencies set at 1 Hz, 10 Hz, and 25
ear stimulation (Badran et al., 2019). Hz and pulse widths at 100 μs, 200 μs, and 500 μs and found that only
Stimulation parameters also vary between studies (Badran et al., stimuli of 500 μs width delivered at 10 Hz and at 20 Hz significantly
2019; Butt et al., 2020). Activation of the thickly myelinated A beta fi­ reduced heart rate (Badran et al., 2018b). Clancy and coworkers induced
bers that mediate proprioceptive sensation such as touch or vibration a shift in spectral powers of sympathetically and parasympathetically
perception, is assured by electrical stimulus intensities slightly above the mediated heart rate modulation towards relatively less sympathetic and
lowest stimulus intensity evoking a perceivable, somewhat tingling more parasympathetic activity, and moreover recorded a decrease in
sensation (Ellrich, 2019). Stimulus intensities are kept below levels that muscle sympathetic nerve activity (MSNA) upon 15 min of tragus
reach or exceed the pain threshold, i.e. the threshold at which stimuli stimulation with 30 Hz pulses of 200 μs width and intensities of 10-50
induce unpleasant or pricking sensations via C fiber activation (Ellrich, mA adjusted to the level of sensory thresholds (Clancy et al., 2014).
2019). Since nerve fiber depolarization not only depends on stimulus With adequate stimulation parameters, tVNS not only enhances
intensity but also on stimulus duration and its frequency, parameters parasympathetic activity but also attenuates sympathetic activity as
vary between studies but are mostly delivered at a stimulus frequency of shown not only by the MSNA decrease recorded by Clancy et al. (2014)
10–15 Hz with a stimulus pulse-width between 250 μs and 500 μs, and but also in a tVNS study conducted in dogs with surgically induced
constant current intensities below 5 mA (Badran et al., 2019). However, myocardial infarction. After 90 days, the dogs who had received 4 h per

3
M.J. Hilz Autonomic Neuroscience: Basic and Clinical 243 (2022) 103038

diem of ABVN stimulation had a significantly better infarct-size reduc­ as the dorsal raphe nuclei that provide the serotonergic innervation of
tion and significantly lower noradrenaline levels than the dogs without central autonomic regions and further transfer stimuli to the spinal cord,
ABVN stimulation (Wang et al., 2014). hypothalamus, and brainstem areas, including the rostroventrolateral
Murray et al. assume that the parasympathetic upregulation and medulla where the serotonergic impulses inhibit sympathetic RVLM
sympathetic withdrawal upon ABNV stimulation is mediated via outflow (Benarroch, 1997).
afferent impulses travelling along the ABNV towards the nucleus of the Furthermore, the NTS directly or indirectly projects to many central
solitary tract (NTS) which receives around 95 % of afferent vagus nerve structures [for review see (Butt et al., 2020) and (Yakunina et al.,
impulses (Murray et al., 2016; Butt et al., 2020). Upon receipt of these 2017)]. Among the direct projections are for example the spinal tri­
stimuli, the NTS sends impulses to the dorsal vagal nucleus (DVN) and geminal nucleus, the parabrachial region, periaqueductal gray, the
the nucleus ambiguus which both generate efferent impulses that reach thalamus, amygdala, insula, bed nucleus of the stria terminalis, or the
the heart via cardiovagal fibers and thus slow heart rate. Simulta­ hypothalamus (Komisaruk and Frangos, 2022). Indirectly, the NTS
neously, the NTS might send activating impulses to the caudal ventro­ projects onto further structures: the parabrachial nuclei reach the par­
lateral medulla (CVLM) which in turn inhibits the rostroventrolateral aventricular nucleus of the hypothalamus, the lateral hypothalamus,
medulla (RVLM), i.e. the main source of activating preganglionic sym­ and the central amygdala (Komisaruk and Frangos, 2022). Indirect
pathetic neurons in the intermediolateral column (Fig. 3). The subse­ projections may reach of the cingulate gyrus, nucleus accumbens, the
quently reduced sympathetic outflow could explain the decrease in limbic system - which is also rich in serotoninergic receptors (Benarroch,
MSNA or noradrenaline levels upon ABVN stimulation (Murray et al., 1997), and thus might hold therapeutic promise, e.g. in tVNS treatment
2016). of depressive disorders - and forebrain regions [for review see (Butt
et al., 2020) and (Yakunina et al., 2017)]. Additional details regarding
tVNS related central projections are beyond the scope of this review and
7.1. Widespread central nervous system involvement upon auricular tVNS
subject to further research.
However, auricular tVNS activates not only the NTS. In rats, the use
8. Clinical tVNS proof-of-concept studies encouraged further
of C-Fos immunochemistry as marker of neuronal activity upon left
therapeutic approaches
cavum conchae stimulation demonstrated bilateral C-Fos staining in the
NTS and loci coerulei, i.e. the major noradrenergic cerebral areas (Ay
Early proof of concept studies, e.g., in patients with pharmaco-
et al., 2015). In animals, retrograde tracing studies of the ABVN by
resistant epilepsies showed that transcutaneous VNS is suitable to
means of transganglionic horseradish peroxidase (HRP) moreover
reduce seizure frequency and is moreover safe and well-tolerated (Stefan
showed HRP staining of the NTS, nucleus cuneatus, and the caudal tri­
et al., 2012).
geminal nucleus [for review see (Butt et al., 2020)].
Similarly, tVNS benefits were demonstrated in chronic migraine
Various fMRI studies confirmed that vagus nerve stimulation acti­
patients, particularly with 1 Hz stimulation but also - albeit to a lesser
vates the NTS which projects not only to the nucleus coeruleus, the area
degree - with 25 Hz stimuli (Straube et al., 2015).
assumed to be significantly involved in therapeutic VNS effects, partic­
In healthy persons, an early tVNS study showed that left outer
ularly in seizure control (Fig. 4) (Fornai et al., 2011; Butt et al., 2020).
auditory canal stimulation improved the sense of well-being and
Depending on the auricular stimulation site and stimulation parameters,
decreased fMRI BOLD-signals in limbic brain areas, including the
the NTS projects to various other brain regions (Yakunina et al., 2017;
amygdala, hippocampus, parahippocampal gyrus, and the middle and
Ellrich, 2019; Butt et al., 2020). Among these are brainstem areas such

Fig. 3. Possible pathways of ABVN stimulation


inducing cardiovascular effects via nucleus tractus
solitarii (NTS) stimulation. Upon ABVN stimulation,
the NTS activates the dorsal vagal nucleus (DVN) and
the nucleus ambiguus (NA) in the medulla which
both generate cardiovagal outflow. With ABVN
stimulation, the NTS may also activate the caudal
ventrolateral medulla (CVLM) which will in turn
inhibit the rostroventrolateral medulla (RVLM) and
thus sympathetic outflow to the neurons of the
intermediolateral cell column (IML) in the spinal cord
(Murray et al., 2016).
[Reprinted from Autonomic Neurosciences: Basic &
Clinical, 2016, Vol 199, Murray, A. R., L. Atkinson,
M. K. Mahadi, S. A. Deuchars and J. Deuchars. The
strange case of the ear and the heart: The auricular
vagus nerve and its influence on cardiac control.
Pages 48–53, Copyright 2016, with permission from
Elsevier.]

4
M.J. Hilz Autonomic Neuroscience: Basic and Clinical 243 (2022) 103038

review see Li et al., 2022). For example, Redgrave et al. evaluated tVNS
effects in patients with post-stroke upper limb dysfunction and showed a
significant improvement of the upper limb Fugl-Meyer score after the
patients had completed 18 one-hour sessions of repetitive arm move­
ments combined with auricular tVNS (Redgrave et al., 2018).
Finally, non-invasive VNS might be used to evaluate or predict the
responsiveness e.g. of epilepsy patients to invasive VNS (Mertens et al.,
2018).
So far, various stimulators have been developed for auricular trans­
cutaneous stimulation (Mertens et al., 2018). There is also a hand-held,
transcutaneous stimulator used to stimulate the cervical branch of the
vagus nerve; this GammaCore device (Electrocore LLC, Basking Ridge,
USA) has received FDA approval for the treatment of episodic cluster
headache (Holle-Lee and Gaul, 2016; Mwamburi et al., 2017).
As of July 2022, a Pubmed search for “transcutaneous vagus nerve
stimulation” shows 547 publications most of which have been published
within the last three years. While the field of noninvasive VNS is rapidly
expanding questions regarding optimal stimulus frequencies, pulse
widths, intensities, and best suited treatment duration and intervals,
central pathways, as well as successful areas of application still require
further studies. Yet, tVNS may become a useful and readily available
therapeutic tool for many different neurological and other indications.

Dedication

This paper is dedicated to Professor Dr. Bernhard Neundörfer,


Chairman Emeritus of the Department of Neurology at the University of
Erlangen-Nuremberg, Germany, on the occasion of his 85th birthday
and in gratitude to an enthusiastic physician, academic teacher, and
mentor who promoted clinical autonomic research and served as
Fig. 4. Main pathways that may result from cervical VNS activation of the founding President of the German Autonomic Society and co-founder of
nucleus tractus solitarii (NTS) according to Fornai et al. (2011). the European Federation of Autonomic Societies.
NTS impulses, elicited by cervical VNS, reach the locus coeruleus (LC) via the
nucleus paragiganto-cellularis (PGi) and the nucleus prepositus hypoglossi
(Prep). The locus coeruleus conveys noradrenergic (NE) impulses to the cere­ Declaration of competing interest
bral cortex, thalamus, and dorsal raphe nuclei (DRN). The thalamus projects
glutamatergic signals (GLU) to the cortex; the dorsal raphe nuclei convey The author has no conflict of interest.
serotoninergic (5HT) impulses to the cortex but also to central autonomic re­
gions, the spinal cord, hypothalamus, and brainstem areas, including the ros­ Data availability
troventrolateral medulla (Benarroch, 1997; Fornai et al., 2011).
[Reprinted from European Journal of Neuroscience 2011: 33(12); Fornai, F., R. No data was used for the research described in the article.
Ruffoli, F. S. Giorgi and A. Paparelli. The role of locus coeruleus in the anti­
epileptic activity induced by vagus nerve stimulation. Pages 2169–2178;
References
Copyright 2011, with permission from John Wiley & Sons].
Apovian, C.M., Shah, S.N., Wolfe, B.M., Ikramuddin, S., Miller, C.J., Tweden, K.S.,
superior temporal gyrus, while the stimulation increased BOLD signals Billington, C.J., Shikora, S.A., 2017. Two-year outcomes of vagal nerve blocking
in the insula, precentral gyrus, and thalamus, i.e. tVNS generated brain (vBloc) for the treatment of obesity in the ReCharge trial. Obes. Surg. 27 (1),
169–176.
activation patterns similar to those induced by invasive vagus nerve Austelle, C.W., O’Leary, G.H., Thompson, S., Gruber, E., Kahn, A., Manett, A.J., Short, B.,
stimulation (Kraus et al., 2007). Badran, B.W., 2022. A comprehensive review of vagus nerve stimulation for
Several studies in patients with major depression support the hy­ depression. Neuromodulation 25 (3), 309–315. https://doi.org/10.1111/ner.13528.
Ay, I., Napadow, V., Ay, H., 2015. Electrical stimulation of the vagus nerve dermatome in
pothesis that tVNS ameliorates depression in patients with major the external ear is protective in rat cerebral ischemia. Brain Stimul. 8 (1), 7–12.
depressive disorder and modulates the resting state functional connec­ Badran, B.W., Brown, J.C., Dowdle, L.T., Mithoefer, O.J., LaBate, N.T., Coatsworth, J.,
tivity between the right amygdala and left dorsolateral prefrontal cortex DeVries, W.H., Austelle, C.W., McTeague, L.M., Yu, A., Bikson, M., Jenkins, D.D.,
George, M.S., 2018. Tragus or cymba conchae? Investigating the anatomical
(Liu et al., 2016), and also suggest that tVNS might be a feasible treat­ foundation of transcutaneous auricular vagus nerve stimulation (taVNS). Brain
ment option for patients with treatment-resistant depression (e.g. (Rong Stimul. 11 (4), 947–948.
et al., 2016, Trevizol et al., 2016, Evensen et al., 2022)). Badran, B.W., Mithoefer, O.J., Summer, C.E., LaBate, N.T., Glusman, C.E., Badran, A.W.,
DeVries, W.H., Summers, P.M., Austelle, C.W., McTeague, L.M., Borckardt, J.J.,
By now, transcutaneous vagus nerve stimulation has become a
George, M.S., 2018. Short trains of transcutaneous auricular vagus nerve stimulation
promising alternative to invasive VNS. The transcutaneous approach is (taVNS) have parameter-specific effects on heart rate. Brain Stimul. 11 (4), 699–708.
used not only in patients with epilepsy, depression or anxiety disorders Badran, B.W., Yu, A.B., Adair, D., Mappin, G., DeVries, W.H., Jenkins, D.D., George, M.S.,
but tVNS has also been tested for the treatment of migraine, chronic Bikson, M., 2019. Laboratory Administration of Transcutaneous Auricular Vagus
Nerve Stimulation (taVNS): technique, targeting, and considerations. J. Vis. Exp.
pain, tinnitus, Alzheimer’s disease, heart failure, obesity, inflammatory (143).
diseases, or to improve cardiovascular autonomic modulation and bar­ Bailey, P., Bremer, F., 1938. A sensory cortical representation of the vagus nerve: with a
oreflex sensitivity, to mention only a few indications (Clancy et al., note on the effects of low blood pressure on the cortical electrogram.
J. Neurophysiol. 1 (5), 405–412.
2014; Hyvarinen et al., 2015; Mertens et al., 2018; Badran et al., 2019; Benarroch, E.E., 1997. Central Autonomic Network: Functional Organization and
Butt et al., 2020). Clinical Correlations. Futura Publishing Company, Inc, Armonk, NY.
Several studies use tVNS for rehabilitation in post-stroke patients (for Ben-Menachem, E., Hamberger, A., Hedner, T., Hammond, E.J., Uthman, B.M., Slater, J.,
Treig, T., Stefan, H., Ramsay, R.E., Wernicke, J.F., et al., 1995. Effects of vagus nerve

5
M.J. Hilz Autonomic Neuroscience: Basic and Clinical 243 (2022) 103038

stimulation on amino acids and other metabolites in the CSF of patients with partial Mertens, A., Raedt, R., Gadeyne, S., Carrette, E., Boon, P., Vonck, K., 2018. Recent
seizures. Epilepsy Res. 20 (3), 221–227. advances in devices for vagus nerve stimulation. Expert Rev. Med. Devices 15 (8),
Butt, M.F., Albusoda, A., Farmer, A.D., Aziz, Q., 2020. The anatomical basis for 527–539.
transcutaneous auricular vagus nerve stimulation. J. Anat. 236 (4), 588–611. Murray, A.R., Atkinson, L., Mahadi, M.K., Deuchars, S.A., Deuchars, J., 2016. The strange
Clancy, J.A., Mary, D.A., Witte, K.K., Greenwood, J.P., Deuchars, S.A., Deuchars, J., case of the ear and the heart: the auricular vagus nerve and its influence on cardiac
2014. Non-invasive vagus nerve stimulation in healthy humans reduces sympathetic control. Auton. Neurosci. 199, 48–53.
nerve activity. Brain Stimul. 7 (6), 871–877. Mwamburi, M., Liebler, E.J., Tenaglia, A.T., 2017. Review of non-invasive vagus nerve
Dell, P., Olson, R., 1951. Thalamic, cortical and cerebellar projections of vagal visceral stimulation (gammaCore): efficacy, safety, potential impact on comorbidities, and
afferences. C. R. Seances Soc. Biol. Fil. 145 (13–14), 1084–1088. economic burden for episodic and chronic cluster headache. Am. J. Manag. Care 23
Dodrill, C.B., Morris, G.L., 2001. Effects of vagal nerve stimulation on cognition and (17 Suppl), S317–S325.
quality of life in epilepsy. Epilepsy Behav. 2 (1), 46–53. Nahas, Z., Marangell, L.B., Husain, M.M., Rush, A.J., Sackeim, H.A., Lisanby, S.H.,
Elger, G., Hoppe, C., Falkai, P., Rush, A.J., Elger, C.E., 2000. Vagus nerve stimulation is Martinez, J.M., George, M.S., 2005. Two-year outcome of vagus nerve stimulation
associated with mood improvements in epilepsy patients. Epilepsy Res. 42 (2–3), (VNS) for treatment of major depressive episodes. J. Clin. Psychiatry 66 (9),
203–210. 1097–1104.
Ellrich, J., 2019. Transcutaneous auricular vagus nerve stimulation. J. Clin. Redgrave, J.N., Moore, L., Oyekunle, T., Ebrahim, M., Falidas, K., Snowdon, N., Ali, A.,
Neurophysiol. 36 (6), 437–442. Majid, A., 2018. Transcutaneous auricular vagus nerve stimulation with concurrent
Engel, D., 1979. The gastroauricular phenomenon and related vagus reflexes. Arch. upper limb repetitive task practice for poststroke motor recovery: a pilot study.
Psychiatr. Nervenkr. (1970) 227 (3), 271–277. J. Stroke Cerebrovasc. Dis. 27 (7), 1998–2005.
Evensen, K., Jorgensen, M.B., Sabers, A., Martiny, K., 2022. Transcutaneous vagal nerve Reid, S.A., 1990. Surgical technique for implantation of the neurocybernetic prosthesis.
stimulation in treatment-resistant depression: a feasibility study. Neuromodulation Epilepsia 31 (Suppl. 2), S38–S39.
25 (3), 443–449. Rong, P., Liu, J., Wang, L., Liu, R., Fang, J., Zhao, J., Zhao, Y., Wang, H., Vangel, M.,
Fornai, F., Ruffoli, R., Giorgi, F.S., Paparelli, A., 2011. The role of locus coeruleus in the Sun, S., Ben, H., Park, J., Li, S., Meng, H., Zhu, B., Kong, J., 2016. Effect of
antiepileptic activity induced by vagus nerve stimulation. Eur. J. Neurosci. 33 (12), transcutaneous auricular vagus nerve stimulation on major depressive disorder: a
2169–2178. nonrandomized controlled pilot study. J. Affect. Disord. 195, 172–179.
Garamendi-Ruiz, I., Gomez-Esteban, J.C., 2019. Cardiovascular autonomic effects of Rush, A.J., Marangell, L.B., Sackeim, H.A., George, M.S., Brannan, S.K., Davis, S.M.,
vagus nerve stimulation. Clin. Auton. Res. 29 (2), 183–194. Howland, R., Kling, M.A., Rittberg, B.R., Burke, W.J., Rapaport, M.H., Zajecka, J.,
Ginsberg, L.E., Eicher, S.A., 2000. Great auricular nerve: anatomy and imaging in a case Nierenberg, A.A., Husain, M.M., Ginsberg, D., Cooke, R.G., 2005. Vagus nerve
of perineural tumor spread. AJNR Am. J. Neuroradiol. 21 (3), 568–571. stimulation for treatment-resistant depression: a randomized, controlled acute phase
Giordano, F., Zicca, A., Barba, C., Guerrini, R., Genitori, L., 2017. Vagus nerve trial. Biol. Psychiatry 58 (5), 347–354.
stimulation: surgical technique of implantation and revision and related morbidity. Rush, A.J., Sackeim, H.A., Marangell, L.B., George, M.S., Brannan, S.K., Davis, S.M.,
Epilepsia 58 (Suppl. 1), 85–90. Lavori, P., Howland, R., Kling, M.A., Rittberg, B., Carpenter, L., Ninan, P.,
Gonzalez, H.F.J., Yengo-Kahn, A., Englot, D.J., 2019. Vagus nerve stimulation for the Moreno, F., Schwartz, T., Conway, C., Burke, M., Barry, J.J., 2005. Effects of 12
treatment of epilepsy. Neurosurg. Clin. N. Am. 30 (2), 219–230. months of vagus nerve stimulation in treatment-resistant depression: a naturalistic
Gupta, D., Verma, S., Vishwakarma, S.K., 1986. Anatomic basis of Arnold’s ear-cough study. Biol. Psychiatry 58 (5), 355–363.
reflex. Surg. Radiol. Anat. 8 (4), 217–220. Ryvlin, P., Rheims, S., Hirsch, L.J., Sokolov, A., Jehi, L., 2021. Neuromodulation in
Harden, C.L., Pulver, M.C., Ravdin, L.D., Nikolov, B., Halper, J.P., Labar, D.R., 2000. epilepsy: state-of-the-art approved therapies. Lancet Neurol. 20 (12), 1038–1047.
A pilot study of mood in epilepsy patients treated with vagus nerve stimulation. Sackeim, H.A., Rush, A.J., George, M.S., Marangell, L.B., Husain, M.M., Nahas, Z.,
Epilepsy Behav. 1 (2), 93–99. Johnson, C.R., Seidman, S., Giller, C., Haines, S., Simpson Jr., R.K., Goodman, R.R.,
He, W., Wang, X., Shi, H., Shang, H., Li, L., Jing, X., Zhu, B., 2012. Auricular acupuncture 2001. Vagus nerve stimulation (VNS) for treatment-resistant depression: efficacy,
and vagal regulation. Evid. Based Complement. Alternat. Med. 2012, 786839. side effects, and predictors of outcome. Neuropsychopharmacology 25 (5), 713–728.
Holle-Lee, D., Gaul, C., 2016. Noninvasive vagus nerve stimulation in the management of Stefan, H., Kreiselmeyer, G., Kerling, F., Kurzbuch, K., Rauch, C., Heers, M., Kasper, B.S.,
cluster headache: clinical evidence and practical experience. Ther. Adv. Neurol. Hammen, T., Rzonsa, M., Pauli, E., Ellrich, J., Graf, W., Hopfengartner, R., 2012.
Disord. 9 (3), 230–234. Transcutaneous vagus nerve stimulation (t-VNS) in pharmacoresistant epilepsies: a
Hyvarinen, P., Yrttiaho, S., Lehtimaki, J., Ilmoniemi, R.J., Makitie, A., Ylikoski, J., proof of concept trial. Epilepsia 53 (7), e115–e118.
Makela, J.P., Aarnisalo, A.A., 2015. Transcutaneous vagus nerve stimulation Straube, A., Ellrich, J., Eren, O., Blum, B., Ruscheweyh, R., 2015. Treatment of chronic
modulates tinnitus-related beta- and gamma-band activity. Ear Hear. 36 (3), migraine with transcutaneous stimulation of the auricular branch of the vagal nerve
e76–e85. (auricular t-VNS): a randomized, monocentric clinical trial. J. Headache Pain 16,
Ikramuddin, S., Blackstone, R.P., Brancatisano, A., Toouli, J., Shah, S.N., Wolfe, B.M., 543.
Fujioka, K., Maher, J.W., Swain, J., Que, F.G., Morton, J.M., Leslie, D.B., Toffa, D.H., Touma, L., El Meskine, T., Bouthillier, A., Nguyen, D.K., 2020. Learnings
Brancatisano, R., Kow, L., O’Rourke, R.W., Deveney, C., Takata, M., Miller, C.J., from 30 years of reported efficacy and safety of vagus nerve stimulation (VNS) for
Knudson, M.B., Tweden, K.S., Shikora, S.A., Sarr, M.G., Billington, C.J., 2014. Effect epilepsy treatment: a critical review. Seizure 83, 104–123.
of reversible intermittent intra-abdominal vagal nerve blockade on morbid obesity: Trevizol, A.P., Shiozawa, P., Taiar, I., Soares, A., Gomes, J.S., Barros, M.D., Liquidato, B.
the ReCharge randomized clinical trial. JAMA 312 (9), 915–922. M., Cordeiro, Q., 2016. Transcutaneous vagus nerve stimulation (taVNS) for major
Klein, H.U., Ferrari, G.M., 2010. Vagus nerve stimulation: a new approach to reduce depressive disorder: an open label proof-of-concept trial. Brain Stimul. 9 (3),
heart failure. Cardiol. J. 17 (6), 638–644. 453–454.
Komisaruk, B.R., Frangos, E., 2022. Vagus nerve afferent stimulation: projection into the Ventureyra, E.C., 2000. Transcutaneous vagus nerve stimulation for partial onset seizure
brain, reflexive physiological, perceptual, and behavioral responses, and clinical therapy. A new concept. Childs Nerv. Syst. 16 (2), 101–102.
relevance. Auton. Neurosci. 237, 102908. Vonck, K., Boon, P., Van Roost, D., 2007. Anatomical and physiological basis and
Krahl, S.E., Senanayake, S.S., Handforth, A., 2001. Destruction of peripheral C-fibers mechanism of action of neurostimulation for epilepsy. Acta Neurochir. Suppl. 97 (Pt
does not alter subsequent vagus nerve stimulation-induced seizure suppression in 2), 321–328.
rats. Epilepsia 42 (5), 586–589. Wang, Z., Yu, L., Wang, S., Huang, B., Liao, K., Saren, G., Tan, T., Jiang, H., 2014.
Kraus, T., Hosl, K., Kiess, O., Schanze, A., Kornhuber, J., Forster, C., 2007. BOLD fMRI Chronic intermittent low-level transcutaneous electrical stimulation of auricular
deactivation of limbic and temporal brain structures and mood enhancing effect by branch of vagus nerve improves left ventricular remodeling in conscious dogs with
transcutaneous vagus nerve stimulation. J. Neural Transm. (Vienna) 114 (11), healed myocardial infarction. Circ. Heart Fail. 7 (6), 1014–1021.
1485–1493. Yakunina, N., Kim, S.S., Nam, E.C., 2017. Optimization of transcutaneous vagus nerve
Li, L., Wang, D., Pan, H., Huang, L., Sun, X., He, C., Wei, Q., 2022. Non-invasive vagus stimulation using functional MRI. Neuromodulation 20 (3), 290–300.
nerve stimulation in cerebral stroke: current status and future perspectives. Front. Zannad, F., De Ferrari, G.M., Tuinenburg, A.E., Wright, D., Brugada, J., Butter, C.,
Neurosci. 16, 820665. Klein, H., Stolen, C., Meyer, S., Stein, K.M., Ramuzat, A., Schubert, B., Daum, D.,
Liu, J., Fang, J., Wang, Z., Rong, P., Hong, Y., Fan, Y., Wang, X., Park, J., Jin, Y., Liu, C., Neuzil, P., Botman, C., Castel, M.A., D’Onofrio, A., Solomon, S.D., Wold, N.,
Zhu, B., Kong, J., 2016. Transcutaneous vagus nerve stimulation modulates Ruble, S.B., 2015. Chronic vagal stimulation for the treatment of low ejection
amygdala functional connectivity in patients with depression. J. Affect. Disord. 205, fraction heart failure: results of the NEural cardiac TherApy foR heart failure
319–326. (NECTAR-HF) randomized controlled trial. Eur. Heart J. 36 (7), 425–433.
Marangell, L.B., Rush, A.J., George, M.S., Sackeim, H.A., Johnson, C.R., Husain, M.M.,
Nahas, Z., Lisanby, S.H., 2002. Vagus nerve stimulation (VNS) for major depressive
episodes: one year outcomes. Biol. Psychiatry 51 (4), 280–287.

You might also like