You are on page 1of 9

Clinical Neuroscience Research 4 (2004) 71–79

www.elsevier.com/locate/clires

Mechanisms of action of vagus nerve stimulation (VNS)


Mark S. Georgea,b,c,*, Ziad Nahasa,b, Daryl E. Bohninga, Qiwen Mua,b,
F. Andrew Kozela,b,c, Jeffrey Borckhardtb, Stewart Denslowa
a
Center for Advanced Imaging Research, Medical University of South Carolina, Charleston, SC, USA
b
Brain Stimulation Laboratory, Department of Psychiatry, Medical University of South Carolina, 67 President Street, Room 502 North, Charleston, SC, USA
c
Ralph H. Johnson Veterans Hospital, Charleston, SC, USA

Abstract
In 1992, Dr Jake Zabara discovered in a canine model that repeated stimulation of the vagus nerve in the neck could stop seizures. Since
that time, repeated stimulation of the vagus nerve has been FDA approved as an anticonvulsant. Zabara’s work is built on a 50-year-old theme
in the literature where scientists had sought to exploit the fact that the vagus nerve is composed of 80% afferent fibers. Thus vagus stimulation
might potentially provide a ‘window’ into the brain. This manuscript reviews the basic science and brain imaging work done to date with
VNS, attempting to understand how VNS affects the brain. This research is crucial to perfecting VNS as an anticonvulsant, and for
determining other neuropsychiatric conditions that might be helped by VNS.
q 2004 Elsevier B.V. All rights reserved.
Keywords: Vagus nerve; VNS; Locus ceruleus; Anticonvulsant; Antidepressant; Brain stimulation; fMRI

1. Introduction the nucleus tractus solitarius (NTS) to diverse brain regions


[2 –4] (see Table 1). As early as 1938, Bailey and Bremer
1.1. Background found that VNS in the cat elicited synchronized EEG
activity in orbital cortex [2]. Later in 1949, MacLean and
Of all the cranial nerves (CN), with the exception of CN I Pribram stimulated the vagus nerve and recorded EEG from
for olfaction, the vagus nerve (CN X), has been the most the cortical surface of anesthetized monkeys. They found
intriguing, and arguably the most misunderstood. The vagus that vagus nerve stimulation caused inconsistent slow waves
nerve helps to regulate the body’s autonomic functions, which generated from the lateral frontal cortex [4, p. 468]. Shortly
are important in a variety of emotional tasks. For reasons that after that, Dell and Olson studied the effects of VNS in
are unclear, most people are more familiar with the vagus awake cats with high cervical spinal lesions [3]. They found
nerve’s efferent functions, where it serves as the messenger that VNS evoked a slow wave response in the anterior rhinal
for signals from the brain to the viscera. Traditionally, the sulcus, as well as in the amygdala. More recently, MacLean
vagus nerve has been considered a parasympathetic efferent continued his 1949 work 30 years later using more
nerve (controlling and regulating autonomic functions such as sophisticated techniques. Single unit recordings showed
heart rate and gastric tone). The afferent role of the vagus has that vagus stimulation produced specific activity in the
been underemphasized in the traditional literature. The vagus
cingulate and other limbic regions. Activation of these cells
is actually a mixed nerve composed of about 80% afferent
was specific to VNS, as rubbing the face and other
sensory fibers carrying information to the brain from the head,
stimulatory controls had no effect on these regions. He
neck, thorax and abdomen [1].
thus demonstrated that the brain effect was through a direct
Over the past 100 years several different researchers have
vagus signal [5].
convincingly demonstrated the extensive projections of
Reasoning from this body of literature, Zabara [6,7]
the vagus nerve via its sensory afferent connections in
demonstrated an anticonvulsant action of VNS on exper-
imental seizures in dogs. Zabara hypothesized that VNS
* Corresponding author. Address: Brain Stimulation Laboratory, could prevent or control the motor and autonomic
Department of Psychiatry, Medical University of South Carolina, 67
components of epilepsy [8]. Penry and others ushered in
President Street, Room 502 North, Charleston, SC, USA. Tel.: þ 1-843-
876-5142; fax: þ 1-843-792-5702. the modern clinical application of VNS in 1988 using an
E-mail address: georgem@musc.edu (M.S. George). implanted device to treat epilepsy [9,10].
1566-2772/$ - see front matter q 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.cnr.2004.06.006
72 M.S. George et al. / Clinical Neuroscience Research 4 (2004) 71–79

Table 1 (one of the primary norepinephrine containing areas of the


Early work involving VNS brain) (see Fig. 1).
VNS behavioral research The PB/LC directly connect with various forebrain
1938—Bailey and Bremer, VNS in cat elicited synchronous orbital cortex structures, including the hypothalamus, amygdala and bed
activity nucleus of the stria terminalis, as well as several thalamic
1949—MacLean and Pribram, VNS in monkeys, lateral frontal changes regions that control the insula, orbitofrontal and prefrontal
1951—Dell and Olson, VNS evoked a slow wave response in the anterior
cortex [20,21]. These brainstem and limbic anatomic
rhinal sulcus, as well as in the amygdala in awake cats with high cervical
spinal section connections can be demonstrated in non-human animals
1980—MacLean, VNS in monkey—marked single unit effects on and in man (using functional neuroimaging). The oncogen
cingulate, thalamus, basal limbic structures C-fos is a general marker for cellular activity. C-fos studies
1985—Zabara—dogs, VNS caused cortical EEG changes, stopped seizures in rats, while the vagus is being stimulated, revealed
1988—Kiffin Penry (Bowman–Gray), human, resistant epilepsy patient
increased activity in the amygdala, cingulate, LC, and
1998—MUSC, UTSW, NYSPI, Baylor—resistant depression
hypothalamus [22]. Further, the signal into the brain from
VNS can be amplified or reduced by modifying the activity
1.2. Relevant neuroanatomy (Fig. 1) in several of these key relay nuclei. In a very important
study that demonstrates the potential for combining VNS
Although the route of entry into the brain is constrained, with general or local pharmacology, Walker and colleagues
VNS offers the potential for modulating and modifying outlined a possible role of the NTS in how VNS reduces
function in many brain regions, through trans-synaptic seizures [23]. By microinjecting the NTS with either GABA
agonists or glutamate antagonists, they found that increased
connections. For a detailed discussion of vagus circuitry
GABA or decreased glutamate in the NTS blocked seizures.
see Refs. [11,12]. As an overview, it is important to
Indirectly, these findings suggest that VNS may change
understand that the sensory afferent cell bodies of the
NTS GABA and glutamate concentrations, with secondary
vagus reside in the nodose ganglion and relay information to
changes in the function of specific limbic structures noted
the nucleus of the solitary tract (NTS). The NTS relays
above (e.g. amygdala, cingulate, insula, and hippocampus).
incoming sensory information to higher brain regions
In sum, incoming sensory (afferent) connections of the
through at least three pathways: (1) an autonomic feedback
vagus nerve provide direct projections to many of the brain
loop; (2) direct projections to the reticular formation in the
regions implicated in neuropsychiatric disorders. These
medulla and; (3) ascending projections to the forebrain
connections provide a basis for understanding how VNS
largely through the parabrachial nucleus (PB) and the locus
might be a portal to the brainstem and connected limbic and
ceruleus (LC) [13,14]. In addition to these routes, the NTS
cortical regions. These pathways likely account for the
sends direct projections to the amygdala [15 – 17] and
neuropsychiatric effects of VNS, and they invite additional
hypothalamus [18]. The PB sits adjacent to the LC [19]
theoretical considerations for potential research and clinical
applications.

1.3. VNS Methods

The broad term, ‘vagus nerve stimulation’ generally


refers to several different techniques used to stimulate the
vagus nerve, including studies in animals where the vagus
was accessed through the abdomen and diaphragm.
However, for virtually all human studies, VNS refers
to stimulation of the left cervical vagus nerve using a
commercial device, the VNS Therapy System [24] (see
Fig. 2). (An important exception to this rule was the recent
work using a modified form of VNS to stimulate both
subdiaphragmatic vagus nerves for the treatment of
obesity.) [25]
VNS has been commercially available for the treatment
of resistant partial onset seizures in Europe since 1994, and
Fig. 1. Vagus neuroanatomy. This simplified cross-sectional sagittal slice of in the US since 1997. About 22,000 people with epilepsy
the brain highlights the vagus nerve afferents, which terminate in the worldwide have been treated with the VNS therapy system
Nucleus Solitary Tract (NTS). From there they project to several important (September 2003). Typically, although not always, epi-
brainstem and cerebellar regulatory nuclei, including the locus ceruleus. It is
thought that the vagus afferents, acting through these nuclei, then influence
lepsy patients considering VNS have had unsatisfactory
higher order limbic and cortical regions. VNS is thus a ‘bottom-up’ seizure control despite treatment with multiple medi-
approach to brain stimulation. cations. In many ways, VNS resembles implanting
M.S. George et al. / Clinical Neuroscience Research 4 (2004) 71–79 73

When the magnet is removed, normal programmed


stimulation resumes. This allows patients to control and
temporarily eliminate stimulation-related side effects during
key behaviors like public speaking (voice tremor) or heavy
exercising (mild shortness of breath). This safety feature
was an impediment to using functional MRI scanning in
conjunction with VNS, but this technical problem has been
partially solved [28,29].
How VNS compares to other brain stimulation methods
(ECT, TMS, DBS). While both VNS and electroconvulsive
therapy (ECT) have anticonvulsant effects, unlike ECT,
VNS does not cause a seizure and seems to be free of
harmful cognitive side effects [30]. VNS differs from
another new brain stimulation technique, transcranial
magnetic stimulation (TMS), in that VNS initially stimulates
deep brain regions (the vagus through the NTS and locus),
with secondary stimulation of limbic cortex. This might be
labeled a ‘bottom-up’ approach to brain stimulation. In
contrast, TMS is limited to direct superficial cortical
stimulation, with secondary effects on subcortical structures
(a ‘top-down’ approach) [31,32]. The other key differences
Fig. 2. The VNS therapy system. The current method of delivering VNS in
between TMS and VNS involve regional specificity and
humans is depicted in this photograph of a model. She is holding a wand
over her chest which interacts with the VNS generator, which is connected chronicity. TMS can be focused anywhere over the skull,
to the vagus nerve in the neck through a subcutaneous wire. The treating while VNS is neuroanatomically limited in its entry pathway
physician can program the VNS generator, using a hand-held PDA, to into the brain (NTS and other brainstem nuclei). However,
deliver stimulation with varying use parameters (frequency, intensity, time VNS delivers constant treatment (i.e. intermittent stimu-
on, time off, pulse width).
lation) for many years, while TMS requires patients to return
to the clinic to receive subsequent courses of TMS.
a cardiac pacemaker. In both VNS and cardiac pacemakers, This difference with TMS brings up the important
a subcutaneous generator sends an electrical signal to an question of whether VNS effects might be regionally
organ through an implanted electrode. In fact, the surgery ‘focused’ with different parameters of stimulation. Animal
for implanting the NCP generator in the chest is much like studies and two human studies suggest that various VNS
inserting a cardiac pacemaker [26]. The two techniques parameters can cause different behaviors (like emotional
differ in the site of stimulation. With VNS, the electrical memory), implying that the secondary circuits affected by
stimulation is delivered through the NCP Pulse Generator, VNS might be altered with different intensities or
an implantable, multi-programmable, bipolar pulse gen- frequencies or other stimulation parameters such as pulse
erator (about the size of a pocket watch) that is implanted width. Recent interleaved VNS/functional magnetic reson-
in the left chest wall to deliver electrical signals to the left ance imaging (fMRI) data suggest that VNS may have
vagus nerve through a bipolar lead. Thus, VNS resembles different regional effects as a function of various use
in some ways deep brain stimulation (DBS) [27], with the parameters, either frequency [33] or pulse width [34].
main differences being the location of the end wire Recent psychophysiological studies further support the
(Vagus—VNS, Deep Brain, DBS), the need for breaching notion that VNS use parameters have biological importance.
the skull with consequent risks in DBS but not VNS, and For example, Borckhardt and colleagues at MUSC have
the nature of the electrical pulse—(intermittent and studied acute pain sensitivity in VNS-implanted depression
relatively low frequency with VNS, constant and high patients. Fig. 3 shows preliminary data from eight subjects,
frequency (. 100 Hz) with DBS). In VNS the electrode showing that different levels of VNS intensity, frequency, or
is wrapped around the vagus nerve in the neck and pulse width all have significant effects on immediately
is connected to the generator subcutaneously. perceived pain. A vitally important area for the further
The VNS implantation surgery is typically an outpatient development of VNS will be determining whether VNS can
procedure, most commonly but not exclusively done by be regionally focused with different use parameters, and
neurosurgeons. The VNS generator can be controlled by a produce different behaviors based on the affected anatomy.
laptop computer (or even a personal digital assistant (PDA)) VNS differs from DBS in that with VNS the surgery is
and an infrared wand. As a safety feature, the VNS therapy simpler and there is no device directly implanted into the
system is designed to shut off in the presence of a constant brain. DBS is used to treat Parkinson’s Disease, and has
magnetic field. Each patient is thus given a magnet that, shown promising open results in obsessive compulsive
when held over the pulse generator, turns off stimulation. disorder (OCD) [35].
74 M.S. George et al. / Clinical Neuroscience Research 4 (2004) 71–79

Fig. 3. Pain thresholds in VNS implanted patients as a function of VNS use parameters. These are pooled data from eight VNS patients who have been
systematically tested for their acute pain response while the VNS is delivered with different frequencies (left), intensities (middle) or pulse width (right). Note
that the left most setting is with no VNS, providing an internal sham control. Acutely, real VNS causes subjects to remove their finger sooner from the hotplate.
Different responses are seen with the different use parameters. Although acutely VNS makes subjects more sensitive to pain, chronic treatment has been shown
to cause a reduction in pain sensitivity. Any understanding of the mechanisms of action of VNS must incorporate the differences between acute effects (minutes
to hours), and the longer term effects (weeks to months to years).

2. VNS as a treatment pulse width) and a low stimulation group (1 Hz, 30 s on,
90– 180 min off, 130 ms pulse width). The majority of the
2.1. Epilepsy VNS epilepsy efficacy and safety data come from trials with
use parameters similar to those used in the high-stimulation
See the article by Labar in this issue for a full discussion group. Similarly, most of the data from other neuropsychia-
of the clinical use of VNS in controlling epilepsy. An tric disorders (depression, anxiety) involve VNS at use
important point from this literature that directly speaks to parameters similar to those used in the initial epilepsy
potential mechanisms of action is that in patients with studies. It is difficult to imagine that these use parameters
epilepsy, the long-term efficacy of VNS is either are the maximally effective choices, or that the same
maintained or improved [36], while the frequency of parameters work equally well in all conditions. Epilepsy
adverse events generally decreases as patients accommo- physicians frequently switch non-responding patients to use
date to stimulation [37]. Data from uncontrolled obser- parameters different from their current settings. However,
vations suggest that, contrary to a tolerance effect, there has been no clear demonstration that changing settings
improvement in seizure control is maintained or may improves efficacy. Further work understanding the transla-
improve over time [37,38]. It appears that there is little or tional neurobiology of these use parameter choices, and how
no tolerance over time to the anticonvulsant actions of they relate to clinical symptoms, is a key area for future
VNS. The patient with the longest exposure to VNS has growth of the field.
had the system operating for 14 years.
There are several different programmable variables in 2.2. Depression
determining how to deliver VNS. These ‘use parameters’
include the pulse width of the electrical signal (130, 250, For a full discussion of the initial reasoning behind
500, 750, 1000 ms), the intensity (0.25 – 4 mA is clinically using VNS in depression in June, 1998, see Ref. [39].
tolerated), the frequency of stimulation (1 –145 Hz), the Dr Marangell in this volume reviews the clinical anti-
length of stimulation (7 –270 s) and the length of time depressant work with VNS. In the initial decision to perform
between trains of stimuli (0.2 s –180 min). In general, the a clinical trial in depression, the knowledge of vagus
initial epilepsy use parameter settings were derived from connections in the brainstem, in particular the role of the LC
the animal studies where anticonvulsant effects were found. was crucial. Additional supporting information came from
The initial epilepsy studies compared efficacy in two (1) mood effects of VNS observed in patients with epilepsy
groups, based on different use parameters. There was a [40,41], (2) evidence from positron emission tomography
high-stimulation group (30 Hz, 30 s on, 5 min off, 500 ms (PET) imaging that VNS affects the function of important
M.S. George et al. / Clinical Neuroscience Research 4 (2004) 71–79 75

Fig. 4. Long term results in Acute VNS depression responders. These are individual plots of each person who was classified as a responder in the original VNS
depression trial, with their acute, 1 year and 2 year change from baseline depicted on the y-axis. In general, those who responded acutely continue to do well
clinically at 2 years of therapy, although some patients suffered relapse.

limbic structures [42], (3) the role of anticonvulsant antidepressant treatment during the acute VNS trial were
medications in mood disorders [43] and (4) neurochemical related to VNS outcome. For example, none of the 13
studies in both animals and humans that revealed that VNS patents who had not responded adequately to more than
alters concentrations of monoamines within the CNS seven research defined adequate antidepressant trials in
(transmission of serotonin [44], norepinephrine [19,45], the current episode responded, while 39% of the remaining
gamma aminobutyric acid (GABA), and glutamate [23]. 46 patients did respond ðP ¼ 0:0057Þ [46].
Most interestingly, VNS as used in the initial open study Results seen at the end of the 10-week acute phase appear
was more effective in depressed patients with lower degrees to largely continue and even improve over 9 months of long-
of treatment resistance. For the group of 59 patients, a term maintenance VNS treatment following exit from the
history of treatment resistance and intensity of concurrent acute study [47], and even persist as long as 2 years out.

Fig. 5. Long-term results in Acute VNS depression non-responders. These are individual plots of each person who was classified as a non-responder in the
original VNS depression trial, with their acute, 1 year and 2 year change from baseline depicted on the y-axis. Many of the subjects who failed to respond
acutely responded later. Understanding the long-term effects of VNS is an important area of clinical and basic science research.
76 M.S. George et al. / Clinical Neuroscience Research 4 (2004) 71–79

Changes in psychotropic medications and in VNS stimulus medications. For the full 10 subjects, there was a 35%
parameters were allowed during this long-term follow-up. decrease in the Hamilton Anxiety Scale at 3 months, and a
In general, initial responders continue to do well, while 26% drop at 6 months. Three of the seven OCD patients met
many initial non-responders later achieve a response response criteria on the YBOC at 3 and 6 months. Further
(see Figs. 4 and 5). Focusing on who responds to VNS, work in this area is warranted.
and when, may provide valuable clues to its mechanisms of Obesity. Information about hunger and satiety from the
action. stomach and small intestine travels through the vagus nerve
to the brain. It seems plausible to ask whether vagus
2.3. Other potential applications stimulation might change eating patterns and other forms of
craving. To investigate this, Roslin and colleagues in NY
Anxiety. As reviewed above, the sensory afferent nerve implanted bilateral subdiagphragmatic vagus stimulators in
fibers that VNS stimulates in the vagus travel to the brain several normal weight mongrel dogs. Over several months,
and terminate in the nucleus solitary tract (NTS). These chronic intermittent VNS in this manner resulted in
fibers are the primary means by which the brain receives substantial weight loss. Interestingly, there was no
information from the organs within the gut [45]. From there, change in the dog’s metabolism. Rather, they took much
information travels to the LC, the primary site of all longer to consume their food, and they even left food on
norepinephrine fibers in the brain. Norepinephrine has long their plate —something mongrel dogs rarely do, if ever [25].
been considered to be a critical neurotransmitter Recently, six subjects with morbid obesity have been
system involved in the pathogenesis and regulation of implanted in this manner [25]. There was a recent abstract
anxiety [48,49]. A device that directly stimulates this from an obesity meeting where a group has implanted an
norepinephrine control site would likely have important electrical stimulator within the gastric mucosa in morbidly
effects on anxiety. obese subjects, with positive results [51]. This procedure
The historical importance of this pathway is that VNS may be simply a different method of vagus nerve
modulates can be seen in the oldest theory about the brain stimulation, with the vagus fibers in the stomach being
origins of fear—the James – Lange theory of emotions. stimulated rather than the actual nerve in the diaphragm or
William James [50] radically argued that all emotion neck.
actually resided in the body, and it was the brains’ Pain. The classical pain pathway involves nerves that
interpretation of this signal through the vagus nerve, that enter the dorsal horn of the spinal cord and then travel
caused someone to be anxious. Interestingly, this theory has through the spine to the brainstem and later thalamus. Pain
been hard to disprove, and most modern anxiety researchers is then perceived in the thalamus, and thalamocingulate
think that the ultimate answer lies in central – peripheral fibers then provide the affective weighting to the pain signal.
loops. However, all agree that the information flowing In addition to this circuit, some information about pain,
through the vagus to the brain is an important part of anxiety especially visceral pain, travels through the vagus nerve.
regulation. Obviously a device that directly affects this A recent study showing changes in pain perception as a
information flow could be a powerful way to alter anxiety. function of different VNS settings hints at the promise of
Animal c-fos studies, and PET, single photon emission using VNS for some form of pain modulation. Acutely,
computed tomography (SPECT) and more recently fMRI VNS increases pain perception [52], but used chronically,
studies of VNS indicate that VNS causes brain activity VNS results in a decrease in pain perception [53]. It is
changes in regions long thought to regulate anxiety. Thus, extremely important to eventually understand these mech-
the function of areas of the brain implicated in anxiety anisms of action, both in terms of the functional and
appears to be altered by VNS. Moreover, there were some translational neurobiology and the dynamic temporal
anti-anxiety effects in the initial open pilot depression study. changes over time.
In these 30 patients, improvements in the HRSD items 9
(agitation), 10 (psychic anxiety) and 11 (somatic anxiety)
were significantly improved (P , 0:001 using signed rank 3. Brain effects of VNS and mechanisms of action
test). The improvement in agitation was from 0.6 to 0.2
(73% improvement), in psychic anxiety from 2.3 to 1.2 Because we still lack full and complete understanding
(50% improvement) and in somatic anxiety from 1.6 to of the pathophysiology of either the depressions or the
1.0 (36% improvement). For all these reasons, a multisite epilepsies, we have only partial clues about how anti-
open treatment trial in resistant anxiety disorder patients convulsant, antidepressant and mood stabilizing treatments
was launched in May 2001. This study has enrolled 10 work. At a simplistic level, antidepressants and anti-
anxiety disorder patients across the several sites with a convulsants can inhibit an area of pathological hyper-
primary diagnosis of either OCD, post-traumatic stress activity, excite an area of pathological hypoactivity, or
disorder (PTSD), or panic disorder. Seven of these 10 had a provide for restored regional balance or synchrony. In
primary diagnosis of OCD. As a group, they were treatment- animal models, there is good evidence that VNS restores
resistant and had tried and failed 18 prior psychotropic neuronal synchrony [7].
M.S. George et al. / Clinical Neuroscience Research 4 (2004) 71–79 77

3.1. Limitations of animal models to involve visceral afferents [42]. Interestingly, they
also showed that this VNS-induced activation varied as a
One of the major limitations of VNS is the relatively small function of the intensity (electrical charge) of stimulation,
body of knowledge about the brain effects of VNS as a and that VNS epilepsy responders had more acute changes
function of the various use parameters. A contributing factor in thalamus activity than did non-responders [55]. There
to this lack of information is the relative difficulty of have been several other VNS SPECT and PET studies.
stimulating the vagus in animals in a manner that resembles
the way VNS is applied in humans. For example, the vagus is 3.2.1. Functional magnetic resonance imaging (fMRI)
a very small nerve in the rat or mouse. Even when someone is fMRI has higher spatial and temporal resolution than
skilled enough to be able to expose the vagus and attach a PET or SPECT, without radiation exposure. However, it is
wire, the coil must be smaller in animals than in humans, not easy to scan VNS patients with fMRI. For example, as
requiring the production of a special small animal VNS a safety measure, the VNS generator is programmed to
electrode. On top of this, the VNS generator is almost as large turn itself off in the presence of a high magnetic field.
as a small rat or mouse and the animal thus cannot wear the Several groups have simultaneously mastered the technical
generator. Thus, in most animal studies to date, VNS has problems involved in performing fMRI in patients
been performed intermittently with the rat restrained in a implanted with VNS generators [28,29,33,56]. At MUSC,
harness or cage, and the VNS generator attached to the wire Bohning and colleagues have now used fMRI to demon-
through a cap-like connection on the head, running to the strate the functional regional anatomy of VNS in patients
vagus in the neck. This results in VNS application that is only with depression. VNS causes acute changes in hypothala-
for several hours a day, unlike in humans where the device mus, orbitofrontal cortex, the medial temporal lobe
cycles on and off 24 h each day. To achieve 24-h stimulation subsuming the amygdala and hippocampus, insula, medial
in a rat, one must restrict the animal’s movements in a way prefrontal cortex, and cingulate gyrus. At a smaller degree
that greatly changes the animal’s behavior. of significance, the thalamus was activated. These are the
Despite these technical hurdles, Krahl and colleagues regions that are consistently implicated in mood and anxiety
have performed several elegant animal studies with VNS. regulation, and are also regions to which norepinephrine
They have demonstrated that LC lesions suppress the fibers from the LC project.
antiepileptic effects of VNS [19]. Most recently they have More recently, this group has begun to use fMRI to
found that intermittent VNS (30 min/day) in the rat in the determine whether different brain effects are associated with
Porsolt Swim test produces ‘antidepressant’ effects that different VNS use parameters, to study the brain changes
equal ECS or desipramine. Finally, Walker and colleagues over time, and to relate these changes to clinical
have shown that changing pharmacology at the NTS can antidepressant or antianxiety effects. In an initial study
enhance or reduce the anti-epilepsy effects of VNS [23]. examining the differences in the brain of VNS at two
More animal studies in epilepsy and depression exploring the different frequencies (5 and 20 Hz), they found that 20 Hz
biological effects of the VNS use parameters (intensity, pulse VNS had markedly more significant activation than did
width, frequency, duty cycle, dosing strategy) are needed. 5 Hz VNS [33]. Studies like this which relate CNS effects
with stimulation parameters and clinical outcomes hold the
3.2. Functional imaging promise of helping to clarify the neurobiology of VNS and
that of the underlying neuropsychiatric condition.
Functional brain imaging is not hampered by the
technical issues surrounding VNS studies in small animals, 3.3. Other neurobiological studies
although there are other complexities and hurdles.
PET and SPECT. The initial brain imaging studies with Early epilepsy studies found that VNS changed CSF
VNS were done in epilepsy patients, using the radiotracer- 5-HIAA, a serotonin metabolite [44]. It was thus surprising
based techniques of SPECT and PET. These techniques that a recent study in depression of CSF changes over time
have a spatial resolution of 5 –8 mm, and they image an failed to confirm this finding, and instead found significant
average of brain activity over relatively long periods of time increases in CSF HVA, a dopamine metabolite [57].
(SPECT 1 min, O15 PET 1 min, fluorodeoxyglucose (FDG)
PET 20 min). These differences in temporal and spatial
resolution make comparisons across these studies difficult, 4. Future directions
as the different scanning techniques provide an average of
different lengths of brain activity. For a full discussion of In conclusion, current knowledge suggests that VNS
these studies and this complex literature, see Ref. [54]. might well have a role in the treatment of several
Henry and colleagues at Emory have performed some of neuropsychiatric disorders, in addition to its current role
the most important and elegant imaging studies to date. in epilepsy. It is an interesting new method of influencing
They have shown that VNS in epilepsy patients causes acute brain activity, with almost 100% compliance and interesting
blood flow changes in the neuroanatomical regions known clinical effects that appear over the course of many months
78 M.S. George et al. / Clinical Neuroscience Research 4 (2004) 71–79

or even years of treatment. However, more knowledge about [12] Henry TR. Therapeutic mechanisms of vagus nerve stimulation.
the neurobiology of VNS would likely substantially aid its Neurology 2002;59(Suppl 4):S15–S20.
[13] Ter Horst GJ, De Boer P, Luiten PGM, Van Willigen JD. Ascending
applications in these new pioneering applications. Further
projection from the solitary tract nucleus to the hypothalamus. A
work is needed involving animal studies, functional brain Phaseolus bulgaris lectin tracing study in the rat. Neuroscience 1989;
imaging and acute psychophysiological studies in VNS 31:785–97.
implanted humans, and then hypothesis driven clinical [14] Ter Horst GJ, Streetland C. Ascending projections of the solitary tract
trials, in order to perfect and refine the technique. Particular nucleus. In: Robin I, Barraco A, editors. Nucleus of the solitary tract.
London: CRC Press; 1994.
importance needs to be paid to both the acute effects, as well
[15] Ricardo JA, Koh ET. Anatomical evidence of direct projections from
as longer term effects that might suggest permanent the nucleus of the solitary tract to the hypothalamus, amygdala, and
neuronal remodeling. other forebrain structures. Brain Res 1978;152:1–10.
VNS is thus a most interesting new approach to therapy [16] Otterson OP. Afferent connection to the amygdaloin complex of the
and understanding of the brain with several theoretical but rat with some observations in the cat. III. Afferents from lower
still unproven mechanisms of action. brainstem. J Comp Neurol 1981;202:335.
[17] Zardetto-Smith AM, Gray TS. Organization of peptidergic and
catecholaminergic efferents from the nucleus of the solitary tract to
the rat amygdala. Brain Res Bull 1990;25:875.
Acknowledgements [18] Van der Kooy D, Koda LY, McGinty JF, Gerfen CR, Bloom FE. The
organization of projections from the cortex, amygdala, and hypo-
thalamus to the nucleus of the solitary tract in the rat. J Comp Neurol
The authors and the Brain Stimulation Laboratory have 1984;224:1–20.
received grant support from the Defense Advanced [19] Krahl SE, Clark KB, Smith DC, Browning RA. Locus coeruleus
Research Projects Agency (DARPA), the Stanley Foun- lesions suppress the seizure attenuating effects of vagus nerve
dation, the National Alliance for Research on Schizophrenia stimulation. Epilepsia 1998;39:709–14.
and Depression (NARSAD), the Borderline Personality [20] Rutecki P. Anatomical, physiological and theoretical basis for the
antiepileptic effect of vagus nerve stimulation. Epilepsia 1990;31:
Disorders Foundation (BPDRF), NINDS grant RO1-
S1 –S36.
AG40956, the Charles A. Dana Foundation, and Cybero- [21] Van Bockstaele EJ, Peoples J, Valentino RJ. Anatomic basis for
nics. Drs George and Nahas have received speakers fees differential regulation of the rostrolateral per-locus coeruleus region
from Cyberonics, and Dr George is a consultant to by limbic afferents. Biol Psychiatry 1999;46:1352 –63.
Cyberonics. [22] Naritoku DK, Terry WJ, Helfert RH. Regional induction of Fos
immunoreactivity in the brain by anticonvulsant stimulation of the
vagus nerve. Epilepsy Res 1995;22:53 –62.
[23] Walker BR, Easton A, Gale K. Regulation of limbic motor seizures by
References GABA and glutamate transmission in nucleus tractus solitarius.
Epilepsia 1999;40:1051– 7.
[1] Foley JO, DuBois F. Quantitative studies of the vagus nerve in the cat. [24] Schachter SC, Saper CB. Vagus nerve stimulation. Epilepsia 1998;
I. The ratio of sensory and motor studies. J Comp Neurol 1937;67: 39(7):677–86.
49–67. [25] Roslin M, Kurian M. The use of electrical stimulation of the vagus
[2] Bailey P, Bremer F. A sensory cortical representation of the vagus nerve to treat morbid obesity. Epilepsy Behav 2001;2:S11 –S16.
nerve. J Neurophysiol 1938;405–12. [26] Amar AP, Heck CN, Levy ML, Smith T, DeGiorgio CM, Oviedo S,
[3] Dell P, Olson R. Projections ‘secondaires’ mesencephaliques, et al. An institutional experience with cervical vagus nerve trunk
diencephaliques et amygdaliennesdes afferences viscerales vagales. stimulation for medically refractory epilepsy: rationale, technique and
C R Soc Biol 1951;145:1088–91. outcome. Neurosurgery 1998;43:1265–80.
[4] Maclean PD. The triune brain in evolution: role in paleocerebral [27] Benazzouz A, Hallett M. Mechanism of action of deep brain
functions. New York: Plenum Press; 1990. stimulation. Neurology 2000;55:S13–S16.
[5] Maclean PD. Culminating developments in the evolution of the limbic [28] Maniker A, Liu W-C, Marks D, Moser K, Kalnin A. Positioning of
system: the thalamocingulate division. In: Doane BK, Livingston KE, vagal nerve stimulators: technical note. Surg Neurol 2000;53:178 –81.
editors. The limbic system: functional organization and clinical [29] Bohning DE, Lomarev MP, Denslow S, Nahas Z, Shastri A, George
disorders. New York: Raven Press; 1986. MS. Feasibility of vagus nerve stimulation-synchronized blood
[6] Zabara J. Time course of seizure control to brief, repetitive stimuli. oxygenation level-dependent functional MRI. Invest Radiol 2001;
Epilepsia 1985;26:518. 36:470–9.
[7] Zabara J. Peripheral control of hypersynchronous discharge in [30] Sackeim HA, Keilp JG, Rush AJ, George MS, Marangell LB, Dormer
epilepsy. Electroencephalogr Clin Neurophysiol 1985;61s:S162. JS, et al. The effects of vagus nerve stimulation on cognitive
[8] Zabara J. Inhibition of experimental seizures in canines by repetitive performance in patients with treatment-resistant depression. Neurop-
vagal stimulation. Epilepsia 1992;33:1005– 12. sychiatry Neuropsychol Behav Neurol 2001;14(1):53–62.
[9] Penry JK, Dean JC. Prevention of intractable partial seizures by [31] George MS, Belmaker RH. Transcranial magnetic stimulation in
intermittent vagal nerve stimulation in humans: preliminary results. neuropsychiatry, 1st ed. Washington, DC: American Psychiatric
Epilepsy 1990;31:S40–3. Press; 2000.
[10] Uthman BM, Wilder BJ, Penry JK, Dean C, Ramsay RE, Reid SA, [32] George MS, Lisanby SH, Sackeim HA. Transcranial magnetic
et al. Treatment of epilepsy by stimulation of the vagus nerve. stimulation: applications in neuropsychiatry. Arch Gen Psychiatry
Neurology 1993;43:1338–45. 1999;56:300–11.
[11] George MS, Sackeim HA, Rush AJ, Marangell LB, Nahas Z, Husain [33] Lomarev M, Denslow S, Nahas Z, Chae JH, George MS, Bohning DE.
MM, et al. Vagus nerve stimulation: a new tool for brain research and Vagus nerve stimulation (VNS) synchronized BOLD fMRI suggests
therapy. Biol Psychiatry 2000;47:287– 95. that VNS in depressed adults has frequency and/or dose dependent
M.S. George et al. / Clinical Neuroscience Research 4 (2004) 71–79 79

effects at rest and during a simple task. J Psychiatry Res 2002;36: [45] Krahl SE, Senanayake SS, Handforth A. Seizure suppression by
219–27. systemic epinephrine is mediated by the vagus nerve. Epilepsy Res
[34] Mu Q, Nahas Z, Bohning DE, Moghadam P, Anderson B, Walker J, 2000;38:171 –5.
et al. Interleaved VNS-fMRI is a new method for understanding [46] Sackeim HA, Rush AJ, George MS, Marangell LB, Husain MM,
different VNS use parameters: effects of varying pulse-width in Nahas Z, et al. Vagus nerve stimulation (VNS) for treatment-resistant
depressed patients. Biol Psychiatry 2003;53:213S. (#603). depression: efficacy, side effects and predictors of outcome.
[35] Nuttin B, Cosyns P, Demeulemeester H, Gybels J, Meyerson B. Neuropsychopharmacology 2001;25:713–28.
Electrical stimulation in anterior limbs of internal capsules in patients [47] Marangell LB, Rush AJ, George MS, Sackeim HA, Johnson CR,
with obsessive–compulsive disorder. Lancet 1999;354:1526. Husain MM, et al. Vagus nerve stimulation (VNS) for major
[36] Morris GL, Mueller WM, (E01-E05) VNSSG. Long-term treatment depressive episodes: longer-term outcome. Biol Psychiatry 2002;51:
with vagus nerve stimulation in patients with refractory epilepsy. 280–7.
Neurology 1999;53:1731–5. [48] Charney DS, Heninger GR. Noradrenergic function and the
[37] Salinsky MC, Uthman BM, Ristanovic RK, Wernicke JF, Tarver WB, mechanism of action of antianxiety treatment. I. The effect of long-
Group at VVSS. Vagus nerve stimulation for the treatment of term alprazolam treatment. Arch Gen Psychiatry 1985;42:458–67.
medically intractable seizures: results of a 1-year open-extension trial. [49] Charney DS, Heninger GR. Noradrenergic function and the
Arch Neurol 1996;53:1176–80. mechanism of action of antianxiety treatment. II. The effect of
[38] George R, Salinsky M, Kuzniecky R, Rosenfeld W, Bergen D, Tarver
long-term imipramine treatment. Arch Gen Psychiatry 1985;42:
WB, et al. Vagus nerve stimulation for treatment of partial seizures. 2.
473 –81.
Long-term follow-up on first 67 patients exiting a controlled study.
[50] James W. What is an emotion? Mind 1884;9:188– 205.
Epilepsia 1994;35:637–43.
[51] Cigaina V. Gastric stimulation as a therapy for morbid obesity:
[39] George MS, Sackeim HA, Rush AJ, Marangell LB, Nahas Z, Husain
preliminary results. Obes Surg 2002;12(1).
MM, et al. Vagus nerve stimulation: a new tool for brain research and
[52] Ness TJ, Fillingim RB, Randich A, Backensto EM, Faught E. Low
therapy. Biol Psychiatry 2000;47(4):287–95.
intensity vagal nerve stimulation lowers human thermal pain
[40] Harden CL, Pulver MC, Ravdin LD, Nikolov B, Halper JP, Labar DR.
thresholds. Pain 2000;86:81– 5.
A pilot study of mood in epilepsy patients treated with vagus nerve
stimulation. Epilepsy Behav 2000;1:93–9. [53] Kirchner A, Birklein F, Stefan H, Handwerker HO. Left vagus nerve
[41] Elger G, Hoppe C, Falkai P, Rush AJ, Elger CE. Vagus nerve stimulation suppresses experimentally induced pain. Neurology 2000;
stimulation is associated with mood improvements in epilepsy 55:1167–71.
patients. Epilepsy Res 2000;42:203– 10. [54] Chae JH, Nahas Z, Lomarev M, Denslow S, Lorberbaum JP, Bohning
[42] Henry TR, Bakay RA, Votaw JR, Pennell PB, Epstein CM, Faber TL, DE, et al. A review of functional neuroimaging studies of vagus nerve
et al. Brain blood flow alterations induced by therapeutic vagus nerve stimulation (VNS). J Psychiatr Res 2003;37:443–55.
stimulation in partial epilepsy. I. Acute effects at high and low levels [55] Henry TR, Votaw JR, Pennell PB, Epstein CM, Bakay RAE, Faber
of stimulation. Epilepsia 1998;39(9):983–90. TL, et al. Acute blood flow changes and efficacy of vagus nerve
[43] Post RM, Ketter TA, Denicoff K, Pazzaglia PJ, Leverich GS, stimulation in partial epilepsy. Neurology 1999;52:1166 –73.
Marangell LB, et al. The place of anticonvulsant therapy in bipolar [56] Narayanan JT, Watts R, Haddad N, Labar DR, Li PM, Filippi CG.
illness. Psychopharmacology 1996;128(2):115 –29. Cerebral activation during vagus nerve stimulation: a functional MRI
[44] Ben-Menachem E, Hamberger A, Hedner T, Hammond EJ, Uthman study. Epilepsia 2002;43(12):1509–14.
BM, Slater J, et al. Effects of vagus nerve stimulation on amino acids [57] Carpenter LL, Moreno F, Kling MA. Effects of vagus nerve
and other metabolites in the CSF of patients with partial seizures. stimulation on cerebrospinal fluid in depressed patients. Biol
Epilepsy Res 1995;20:221–7. Psychiatry 2002;51:152S–449.

You might also like