4,685
Views
258
CrossRef citations to date
0
Altmetric
Review

A review of vagus nerve stimulation as a therapeutic intervention

&
Pages 203-213 | Published online: 16 May 2018

Abstract

In this review, we provide an overview of the US Food and Drug Administration (FDA)-approved clinical uses of vagus nerve stimulation (VNS) as well as information about the ongoing studies and preclinical research to expand the use of VNS to additional applications. VNS is currently FDA approved for therapeutic use in patients aged >12 years with drug-resistant epilepsy and depression. Recent studies of VNS in in vivo systems have shown that it has anti-inflammatory properties which has led to more preclinical research aimed at expanding VNS treatment across a wider range of inflammatory disorders. Although the signaling pathway and mechanism by which VNS affects inflammation remain unknown, VNS has shown promising results in treating chronic inflammatory disorders such as sepsis, lung injury, rheumatoid arthritis (RA), and diabetes. It is also being used to control pain in fibromyalgia and migraines. This new preclinical research shows that VNS bears the promise of being applied to a wider range of therapeutic applications.

Introduction

Vagus nerve stimulation (VNS) is US Food and Drug Administration (FDA) approved for use in the treatment of epilepsy and depression in patients aged >12 years and is currently being explored as treatment for a variety of other autoimmune and chronic inflammatory disorders, due to its demonstrated anti-inflammatory properties.Citation1Citation6 In this review, we provide an overview of the evidence and diverse applications of VNS in clinical practice, clinical trials, and preclinical research. In addition, we provide a rationale for expanding the use of VNS to a wider range of patients across a wider range of diseases, including sepsis, lung injury, rheumatoid arthritis (RA), stroke, traumatic brain injury (TBI), obesity, diabetes, cardiovascular control, and pain management.

The vagus nerve provides an extensive afferent and efferent network of innervation for the viscera and plays a key role as an interface between higher central nervous system (CNS) circuits and the autonomic control circuitry of the brain stem. It is a mixed autonomic nerve originating at the medulla oblongata and projecting from the brain stem bilaterally along the neck (bundled with the carotid artery rostrally) and esophagus before branching diffusely to innervate the viscera. Most information about the anatomy of the vagus nerve and its projections has been discovered through tract tracing of the rat vagus and is generally assumed to be similar to humans.Citation7 The complete innervation extent of the vagus remains incompletely known, but we have included a brief, simplistic overview of the primary branches and targets of the vagus. The branches that extend off the cervical vagus innervate the bronchi, lungs, heart, and esophagus.Citation7 The subdiaphragmatic vagus has five primary branches, including the dorsal and ventral gastric branches (innervating the stomach) as well as the dorsal and ventral celiac branches (innervating the proximal and descending colon). The hepatic branch divides into the hepatic branch proper (innervating the liver) and the gastroduodenal branch (innervating the duodenum and pancreas).Citation1 The ventral trunk branches into the common hepatic, ventral gastric, and ventral celiac branches. The vagus consists of ~80% sensory afferent and 20% motor efferent fibers.Citation8 Further branching and tertiary targets for the vagus nerve are largely unknown. A fine wire electrode extends from the device and is typically wrapped around the left cervical vagus. Case reports suggest that the right vagus can be used in circumstances where approaching the left vagus is inadvisable. Since the right vagus innervates the sinoatrial node, stimulating on the right is best done with ECG monitoring.

The afferent projections of the vagus nerve are integrated at the level of the autonomic brain stem within the nucleus tractus solitarii (NTS) before projecting to other regions of the CNS. We have incomplete knowledge of how VNS modulates the CNS but the brain stem plays a critical role in integrating and gating signals between the CNS and peripheral organs (). Descending efferents from these regions are responsible for driving cardiorespiratory and gastrointestinal autonomic tone as well as other autonomic functions.Citation1,Citation9 Stimulation of the vagus nerve provides a way to regulate the autonomic tone. Because the vagus nerve is easily accessible at the neck, it is a convenient access point for the implantation of stimulating or surface electrodes for chronic or acute stimulation.

Figure 1 Overview of vagal circuitry linking the central and peripheral nervous system.

Notes: Visceral afferents converge on the NTS in the brain stem, the first point of integration between the peripheral autonomic nervous system and the central nervous system. Visceral afferents project from the dorsal motor nucleus of the vagus and are key to exerting autonomic control in the periphery.

Abbreviations: NA, nucleus ambiguus; NTS, nucleus tractus solitarii; pBC, preBötzinger complex.

Figure 1 Overview of vagal circuitry linking the central and peripheral nervous system.Notes: Visceral afferents converge on the NTS in the brain stem, the first point of integration between the peripheral autonomic nervous system and the central nervous system. Visceral afferents project from the dorsal motor nucleus of the vagus and are key to exerting autonomic control in the periphery.Abbreviations: NA, nucleus ambiguus; NTS, nucleus tractus solitarii; pBC, preBötzinger complex.

Current clinical uses of VNS

Treatment of epilepsy

Epilepsy affects 1% of the US population but costs $12 billion US dollars to treat (figures from 2008).Citation10 VNS to treat epilepsy was first used in the early 1880s by JL Corning, who believed that seizures were caused by changing cerebral blood flow.Citation11 In 1988, the first chronic implantable stimulator was used to treat drug-resistant epilepsy.Citation1 The stimulator was approved by the FDA in 1997 to treat partial onset seizures that were resistant to pharmacological control.Citation1 The current Livanova© (formerly Cyberonics) implantable treatment device consists of a small battery-powered stimulator that requires battery removal and replacement approximately every 6 years.Citation12 A fine wire electrode extends from the device and is typically wrapped around the left cervical vagus. Case reports suggest that the right vagus can be used in circumstances where approaching the left vagus is inadvisable. Since the right vagus innervates the sinoatrial node, stimulating on the right is best done with ECG monitoring.Citation13Citation15 The device can be turned on for 30–90 seconds to provide a brief stimulus to the vagus.Citation16 Once the device is implanted, it is programmed by a physician using a microcomputer, but patients can alter the stimulus program as needed when they feel the onset of a seizure.Citation17 Over 100,000 VNS devices have been implanted in patients worldwide (as of 2015).Citation18 The most common side effects reported are dysphonia, hoarseness, and cough, all of which may be mitigated by changing stimulus parameters.Citation19 Stimulation parameters vary widely, but typical treatment for epilepsy and depression uses a range of stimulation of 20–30 Hz, a pulse duration of up to 500 microseconds, and stimulation on-time of 30–90 seconds followed by off-time of 5 minutes, although stimulus intensity is usually decreased over time.Citation16

Data gathered over the first decade of clinical VNS showed efficacy in patients who had pharmacoresistant seizures. Approximately 40% of patients using VNS showed a 50% reduction in seizures after 2–3 years of treatment.Citation20 The mechanisms by which VNS causes changes in neurochemistry and prevents epileptic seizures are not yet known, although some evidence suggests the vagus nerve plays a role in quenching kindling of seizures in regions susceptible to heightened excitability. These regions include the limbic system, thalamus, and thalamocortical projections.Citation1 VNS may also affect structures in the midbrain and hindbrain, which can contribute to seizure suppression, although the specific changes in these cortical circuits remain unknown. VNS also increases activity in the locus coeruleus and the raphe nuclei and moderates the downstream release of norepinephrine and serotonin, both of which have been shown to have antiepileptic effects.Citation21 VNS success in treating refractory epilepsy with few side effects provides justification for its expansion to both additional conditions and wider populations. shows an overview of the CNS regions likely affected by VNS.

Figure 2 Putative pathways involved in vagus nerve stimulation.

Notes: Stimulation of the vagus activates ascending pathways that alter neural circuits in the brain stem, midbrain, and cortex. Regions that are impacted by vagus nerve stimulation based on past research are included in this diagram.

Abbreviations: NTS, nucleus tractus solitarii; DMX, dorsal motor nucleus of the vagus; LC, locus coeruleus; THAL, thalamus; HypoTHAL, hypothalamus; RTN, retrotrapezoid nucleus; BC, Bötzinger complex; pBC, preBötzinger complex; VRG, ventral respiratory group.

Figure 2 Putative pathways involved in vagus nerve stimulation.Notes: Stimulation of the vagus activates ascending pathways that alter neural circuits in the brain stem, midbrain, and cortex. Regions that are impacted by vagus nerve stimulation based on past research are included in this diagram.Abbreviations: NTS, nucleus tractus solitarii; DMX, dorsal motor nucleus of the vagus; LC, locus coeruleus; THAL, thalamus; HypoTHAL, hypothalamus; RTN, retrotrapezoid nucleus; BC, Bötzinger complex; pBC, preBötzinger complex; VRG, ventral respiratory group.

VNS may also be useful as a treatment for expecting mothers with treatment-resistant epilepsy. One study showed that women with epilepsy had a significantly higher risk of mortality during delivery when compared to women without epilepsy.Citation22 The goal of current epilepsy treatment is to optimize seizure control and minimize in utero fetal exposure to antiepileptic drugs which, during the perinatal period, are associated with major congenital malformation, growth retardation, and neurocognitive developmental deficits.Citation23 VNS has been used successfully as a treatment for medically refractory epilepsy in pregnant women, and physicians have concluded that VNS is a viable option for treatment during pregnancy.Citation24,Citation25 As a non-pharmacological treatment, VNS seems to be beneficial for seizure control in the expecting mother, and there is no evidence of harm to the developing fetus. To date, no large clinical trials have been performed to assess whether VNS has a long-term effect on the developing fetus.

Epilepsy affects 0.5%–1% of the pediatric populationCitation19 or ~470,000 children.Citation26 Chronic epileptic seizures can have a profound impact on children’s long-term neurodevelopmental and social outcome, as well as a lasting impact upon their families.Citation27 Finding an effective treatment can improve the children’s quality of life since children with epilepsy often experience psychiatric and cognitive difficulties and have poor social outcomes as adults.Citation27 Antiepileptic medications have high side effect profiles and can adversely affect the behavior in susceptible children.Citation27 This means that even in children with more easily controlled epilepsy, there is a high risk of psychological and psychiatric disturbance. Children with benign rolandic epilepsy and absence epilepsy have exhibited more aggressive behavior, depression, and anxiety than children without epilepsy.Citation28 Ongoing studies are focused on noninvasive methods to treat epilepsy in pediatric patients. This includes a study by the Chinese Academy of Chinese Medical Sciences testing a non-implant, less invasive transcutaneous auricular vagus nerve stimulator as an effective treatment of pediatric epilepsy.Citation29 The study examines the change in frequency of seizures as well as heart rate variability, quality of life, and electroencephalogram recordings at 2, 4, and 6 months after the start of stimulation.Citation29 In a retrospective cohort study by Elliott et al,Citation19 the effects of VNS on 141 children were analyzed, 61% of whom were <12 years of age. They concluded that VNS was just as effective and relatively complication free in children aged <12 years as it was in older pediatric patients. This study also showed a significant decline in the frequency of seizures in these children, from an average of 10 per week down to three per week. In 41% of patients, there was a 75% reduction in the frequency of seizures.Citation19 Side effects of VNS occurred in a small percentage of children and included hoarseness (0.7%), cough (0.7%), and minor arm pain (0.5%).Citation19 Hallböök et alCitation30 found that pediatric responses to VNS were similar to those of adults. In 40% of children implanted with VNS stimulators, there was a 50% decrease in seizure frequency.Citation30 A retrospective study on 75 children with epilepsy showed that side effects such as hoarseness, cough, and drooling were reported in only 5.4% of patients, in which all were reversible with adjustments to the stimulation parameters.Citation31

Neonates could also benefit from a non-pharmacological approach to controlling epilepsy. Current pharmaceutical treatment options for epilepsy in neonates include phenobarbital and levetiracetam, although each can have detrimental side effects. Phenobarbital is the most widely used anti-epileptic in neonates but can have respiratory and cognitive side effects.Citation32 Levetiracetam has psychological side effects.Citation33 Although VNS is only FDA approved in children aged >12 years, it has been used, along with antiepileptic medication, in children as young as 1 year of age.Citation34 A study conducted by Fernandez et al on children aged <3 years showed that VNS was effective in children with medically intractable epilepsy.Citation34 Their study showed that VNS led to decreased seizure frequency in 33% of patients, and status epilepticus was no longer a symptom after 1 year of treatment. In addition, normal MRIs were associated with decreased seizure incidence.Citation34

Children with developmental disabilities or autism are more likely to exhibit medically refractory epilepsy. One study estimated that between 5% and 38% of children with autism have epilepsy.Citation35 A study by Kirchberger et alCitation85 reported that VNS caused a 50% reduction in seizure frequency in 61% of patients with developmental delays.Citation35 Levy et al also found that there was no statistically significant difference in seizure reduction benefits and quality of life improvements between patients with refractory epilepsy and autism and those without autism.Citation35 Based on these results, VNS seems to be a safe and effective treatment for epilepsy in pediatric patients.

Treatment of depression

Chronic or severe depression affects up to 1.5% of the general population,Citation8 and many of these patients obtain little relief from pharmaceutical treatment. In 2000, depression was estimated to cost $83.1 billion in the United States. Of this cost, $26.1 billion was in direct medical costs, $51.5 billion in indirect workplace costs, and $5.4 billion in suicide-related mortality costs.Citation36 Although VNS was not originally developed to treat depression, patients using VNS to treat epilepsy experienced mood improvement; thus, VNS was expanded to include the treatment of depression. The FDA approved VNS for the treatment of chronic or recurring depression in 2005.Citation37 VNS treatment has been approved for patients aged ≥18 years who have experienced at least one major depressive episode and did not respond well to any of four different pharmaceutical antidepressant treatments.Citation8 A major depressive episode is defined by the Diagnostic and Statistician Manual of Mental Disorders (DSM-IV)Citation38 as having five out of the nine depressive symptoms, including depressed mood or lack of interest in normal day-to-day activities occurring almost daily for at least 2 weeks.Citation39 The goal of treatment is to restore day-to-day function and prevent relapses and remission, as well as alleviate current symptoms, in which VNS has been proven effective in a wide range of patients.Citation8 In a study conducted by Bajbouj et al, patients suffering from chronic “treatment refractory depression” received VNS, in which 53.1% of patients met the response criteria of a 50% reduction in the Hamilton Rating Scale for Depression (HRSD28) (the most commonly used symptom severity scale).Citation37,Citation40 In addition, 38.9% fulfilled the remission criteria with HRSD scores <10.Citation37

Depression is often difficult to treat because patients experiencing recurrent depressive episodes treated with conventional pharmaceuticals often experience relapses or do not experience full remission. A study conducted by Nahas et al showed patients with chronic or recurrent major depressive disorder receiving VNS may have beneficial long-term outcomes.Citation41 In their study, 42% of their patients experienced a positive effect and 22% saw remission after 2 years. Both Bajbouj et al and Nahas et al use the same criteria for clinically significant remission, defined as the absence of clinically significant depressive symptoms.Citation42 Results from neuroimaging studies suggest that the mood-enhancing benefits are caused by VNS stimulation altering medial and prefrontal cortical transmission (). These regions comprise neurons that release neurotransmitters such as serotonin and norepinephrine which have both anticonvulsive and antidepressant effects.Citation37 Although VNS will likely never be recommended as a “first-order” or sole treatment for depression, current clinical evidence shows success with its use as a supplemental treatment for chronic refractory depression.

Between 6% and 13% of pregnant women report symptoms of depression during and post pregnancy.Citation43 The most commonly prescribed drug family for pregnant women with depression is selective serotonin reuptake inhibitors (SSRIs), although there are still unanswered questions about the safety of SSRI treatment for the fetus.Citation43 Antidepressant use during pregnancy may lead to low birth weight and preterm delivery, since they can pass through the placenta.Citation44 A case report by Husain et al showed that VNS was an effective treatment for depression during pregnancy and delivery with no adverse outcomes for the mother or fetus.Citation45 A recent work in a rat model of VNS shows no significant effect of VNS on pups born to a dam with an implanted VNS stimulator.Citation46 Preliminary research suggests that VNS can be a beneficial treatment for both mother and fetus, although more research is required for a clearer picture of the outcome.

Although depression also affects many adolescents, treatment options for pediatric patients are limited. Longitudinal studies on children with major depressive disorders have also shown that the relapse rate is 40% within 2 years and 70% within 5 years.Citation47 Many children with major depression are treated using psychotherapy, but if their depressive symptoms persist, they are typically prescribed antidepressant medications in addition to therapy.Citation48 As with pregnant mothers, the most common antidepressant medications prescribed in the pediatric population are SSRIs.Citation48 As in adults treated with VNS, studies of pediatric patients with VNS implants to treat epilepsy have shown mood improvement. A study by Hallböök et alCitation30 showed that, in children with epilepsy treated using VNS, not only were seizures reduced, but behavior and mood improved while depressive symptoms decreased. Twelve of the 15 children examined had improvement in their quality of life.Citation30 Further studies are needed to examine the impact of VNS on pediatric depression, but the preliminary data show that it remains a promising treatment option and may provide long-term benefit for children with depression.

Potential uses and mechanisms of VNS

An exciting new application of VNS is as an anti-inflammatory treatment. Inflammation is implicated in many chronic diseases including cardiovascular disease, arthritis, and Alzheimer’s disease. Preliminary preclinical evidence suggests that VNS may attenuate the inflammatory response through activation of the cholinergic anti-inflammatory pathway (CAP) – a long loop from the vagus afferents, through the autonomic brain stem and forebrain cortical structures, and then back through the descending vagus efferents (). The CAP upregulates HMGB1, which may regulate cytokine expression, leading to anti-inflammatory effects. In recent years, Tracey et al have devoted significant efforts to quantifying the role that VNS plays as an anti-inflammatory regulator primarily through altered regulation of acetylcholine.Citation2Citation6 These findings provide strong evidence that stimulation of the vagus nerve plays a key role in peripheral cholinergic release and its putative role in suppressing inflammation. The CAP also affects the levels of acetylcholine through nicotinic acetylcholine receptors (nAChRs).Citation2 Several recently completed and ongoing studies are focused on the effects of VNS on inflammatory disorders such as RA, Crohn’s disease, irritable bowel syndrome, and fibromyalgia. Other studies are focused on how VNS affects brain trauma and stroke. Because these are ongoing studies, the efficacy of VNS treatment for these disorders is currently unknown. Many of the inflammatory disorders that VNS may potentially treat also affect the pediatric and neonatal population. Since VNS has been shown to be effective for adult and pediatric populations for epilepsy and depression, it stands to reason that VNS treatment may be beneficial to younger patients for a variety of disorders; however, there is limited data on pediatric applications. We summarize the results of studies related to these disorders below; however, these preliminary data provide a strong rationale for expanding research on the applications of VNS as an anti-inflammatory treatment across a range of different inflammatory diseases.

Sepsis

Sepsis is a multibillion dollar health care burden typically due to systemic bacterial infection and chronic activation of the pro-inflammatory cytokine cascade. Sepsis is estimated to cost $22,000 per patient and affects up to 18 million individuals each year.Citation49 Kessler et al used vagotomized mice to show that lack of vagus input to the CNS can lead to adverse outcomes in a murine model of colon ascendens stent peritonitis.Citation50 In an ex vivo culture of Kupffer cells, tumor necrosis factor a (TNFa) levels were decreased in vagotomized mice compared to controls, even when stimulated with lipopolysaccharide (LPS).Citation50 Huang et al showed that VNS helped to attenuate inflammation by restoring the balance between parasympathetic and sympathetic tone and thus arresting the progression to sepsis.Citation51 These investigators used an intravenous LPS injection model of inflammation to induce sepsis and found that in addition to a decrease in ACh release, heart rate variability was decreased back to baseline levels in the LPS + VNS treatment group, compared to the elevated levels in the LPS-only group.Citation51 In a similar model of LPS-induced endotoxemia, Borovikova et al used VNS and found decreased mortality that they attributed to vagally induced release of acetylcholine.Citation3

Limiting inflammation in pediatric patients without the use of pharmaceuticals is important because neonates, particularly preterm infants, are more susceptible to developing sepsis due to their underdeveloped immune systems and susceptibility to perinatal infection (chorioamnionitis, etc). Since VNS seems to regulate inflammation by modulating the cytokine cascade, our laboratory is looking at the effect of VNS on the early pro-inflammatory cytokines, interleukin-6 (IL-6), TNFa, and IL-1b, in respiratory control regions of the brain stem. We looked at the NTS and hypoglossal motor nucleus (XII), which are regions of critical importance for the control of breathing and implicated in breathing problems in neonatal rats as a model for preterm infants. In the present study, we show that VNS reduces the expression of IL-6 and TNFa in response to a brief (30 minute) bout of high-frequency VNS stimuli.Citation52 Our hope for this preclinical translational work is that it will eventually lead to minimally invasive VNS treatment to provide early intervention and reduce the likelihood of sepsis in preterm infants.

Two major concerns in extending VNS to neonatal practice are the length of time that VNS takes to be effective and the invasive nature of the implantation procedure. Although VNS as currently used for epilepsy and depression can take months to show dramatic effect, short-term stimulation has been used to reduce inflammation quickly, and our experiments have shown that a single bout of high-frequency stimulation (30 minutes) can be effective as an anti-inflammatory treatment.Citation52 In addition, transcutaneous stimulation has been used to treat depression and headaches showing efficacy for even short-term applications using surface electrodes.Citation29,Citation53 A study by He et al uses transcutaneous stimulation at the cervical or auricular vagus to effectively treat epilepsy.Citation29 Further research will be needed to determine if high-frequency stimulation can be paired with transcutaneous stimulation to be an effective treatment for neonatal inflammatory disorders. Preliminary work performed by the Wilson laboratory and others suggests that this may be possible and useful for implementation of VNS treatment in neonates.Citation52,Citation54,Citation55

Pain management

The applications of VNS also extend to widespread inflammatory disorders associated with chronic or intermittent bouts of pain such as fibromyalgia and migraines. Lange et al conducted a Phase I/II clinical trial to assess VNS as an adjunct treatment for patients with fibromyalgia due to its effects on serotonergic and noradrenergic neural circuits – both of which are implicated in pain sensation.Citation56 Their theory was based on results from patients with depression treated with VNS who reported decreased sensation of pain.Citation57 Lange et al’s study included 12 women with fibromyalgia and used the same stimulus parameters as for treatment of epilepsy. After 11 months, 7 of the patients had the minimum clinical difference (MCID+) in their pain symptoms for VNS to be considered effective.Citation56

Another chronic pain ailment that VNS shows promise in treating is migraine headaches. In a study conducted by Barbanti et al, 50 patients with migraine were given VNS treatment applied externally at the neck in two 120-second intervals with 3 minutes in-between. Of those patients, 56% reported pain relief at 1 hour and 64% reported pain relief at 2 hours.Citation53 Silberstein et al performed the ACT1 study (NCT01792817), a clinical trial to use a noninvasive VNS treatment at the neck to treat cluster headaches.Citation58 Their findings suggest that noninvasive VNS can be successfully used to treat episodic cluster headaches.Citation58 Although these results need further research and larger multicenter randomized trials, they present hopeful evidence that VNS can be used to control fibromyalgia and migraines.

Obesity

Although VNS would not likely be recommended as a first-line defense against obesity, research on the effects of VNS on diet and weight have been performed to evaluate VNS for its use as an adjunct treatment in controlling obesity. Finding alternative treatments to obesity is especially important considering 69% of adults and 20% of adolescents in the United States are overweight or obese.Citation59 Burneo et al found that in patients implanted with VNS to control epilepsy, 62% experienced significant weight loss.Citation60 Bodenlos et alCitation61 conducted a study looking at the association between VNS and food cravings in depressed adults and found that left cervical VNS resulted in attenuated food craving.Citation61 Recent work by Val-Laillet et al has shown that bilateral constant current stimulation of the vagus nerve led to lower food intake and sweet cravings in obese minipigs.Citation62 This study did not suggest that VNS actually caused weight loss but rather that VNS helped prevent excess weight gain. The implications of these studies are compelling, but the mechanism by which VNS influences weight loss is still unknown. Some hypotheses include changes in metabolism, decreases in fat stores, or changes in satiety signaling.Citation61 Another potential proposed mechanism for the effect of VNS on weight is reduced intestinal caloric absorption, which can be hypothesized based on the findings that vagal tone can alter peptides that can change gut motility and absorption.Citation63 These preliminary results encourage more extensive research into VNS, and the impact of autonomic control modulation, and hypothalamic signaling and its interactions with the enteric nervous system.

A study performed by Ikramuddin et al showed the effect of VNS on morbid obesity. Since bariatric surgery presents some major risks, investigators are looking for alternative, less-invasive methods of controlling obesity. Their trial showed that weight loss was higher by a statistically significant margin in patients who underwent VNS when compared to sham patients.Citation64 More research is needed, but implantation of VNS devices may be an attractive option for controlling weight and managing obesity for patients who have not had success with more traditional methods of weight control.

Cardiovascular disease

VNS must alter cardiovascular control due to the convergence of inputs in the autonomic control centers of the brain stem, but for how long and to what extent is unknown. The descending cardiac branch of the vagus is key for normal cardiac function.Citation65 Atherosclerosis, which often predisposes one to coronary heart disease, is believed to be due to low-grade systemic inflammation.Citation66 Since there is growing evidence that VNS is anti-inflammatory, it may provide another avenue for treating cardiac dysfunction and atherosclerosis. In the 2007 CARDIA study, Sloan et al showed that there was an inverse relationship between inflammatory markers and vagus nerve activity, measured by heart rate variability, suggesting that VNS is key to anti-inflammatory tone. They also suggest that high levels of pro-inflammatory markers such as IL-6 and C-reactive protein may indicate a predisposition to coronary artery disease.Citation67 VNS may also provide a therapeutic application for preventing heart failure. Zhang et al used a canine model to show that chronic VNS helps to regulate heart rate and improves heart function in a high-rate ventricular pacing model.Citation68 In a rat ischemia/reperfusion (I/R) model, Zhao et al showed that VNS improved cardiac function and reduced infarct size.Citation69 Their results also showed that VNS reduced mesenteric artery pathology and vasodilation caused by the I/R model and that lower levels of TNFa and IL-1b were found in serum. This is likely due to VNS effects on acetylcholine release and systemic levels and upregulation of M3AChR/a7nAChr expressionCitation69 which have been implicated in inflammatory modulation (see “Potential uses and mechanisms of VNS” section), but the precise mechanism is still unknown and needs further work. Chapleau et al used a high salt, spontaneously hypertensive rat model to show that right VNS prevented aortic stiffening and slowed the progression of endothelial dysfunction.Citation70 In addition, they saw significantly higher serum IL-6 levels in VNS rats, which may indicate that VNS modulates inflammatory function in this severe hypertension model.Citation70 Based on these studies, there is a relationship between cardiovascular disease, inflammation, and vagal activity that may be altered by VNS.

Lung injury

VNS is being considered as a treatment for ventilator-induced lung injury (VILI) caused by pressure-induced damage to lung alveoli. Inflammation has been shown to increase the likelihood of VILI, which is often the result of severe lung infection. Other respiratory disorders such as acute lung injury and acute respiratory distress syndrome, both of which can be complicated by sepsis, can also result in pronounced pulmonary inflammation.Citation71Citation73 Experiments by dos Santos et al showed that the vagus nerve plays an important role in pulmonary inflammation. Interruption of the CAP by vagotomy leads to worsening VILI.Citation71 Vagotomized animals with mechanical ventilation had increased alveolar damage and levels of IL-6 and hemorrhage compared to control animals.Citation71 Later experiments showed that VNS, both electrical and pharmacological, attenuated the lung injury in a “two-hit” model of VILI (I/R injury followed by high tidal volume ventilation, which can be further injurious to the lung) by decreasing the pro-inflammatory and pro-apoptotic responses.Citation71

VNS may also be useful in treating gut and lung injuries together. In a study of lung injury caused by hemorrhagic shock, Reys et al showed that VNS prevents intestinal barrier failure and protects against lung injury.Citation74 In addition, pharmacologic blockade of nicotinic cholinergic receptors in an in vitro culture model of pulmonary endothelial cells suggests that VNS acts through the CAP to prevent lung injury and gut–barrier breakdown.Citation73 A study by Levy et al also showed that VNS alleviated lung injury caused by trauma hemorrhagic shock through the reduction of gut permeability.Citation72 These studies suggest that vagus nerve activity is crucial for normal lung function, and the possibility of using VNS to improve outcome in lung injury has great promise for further research. In addition, preventing gut–barrier interaction that contributes to visceral inflammation also deserves further study.

Stroke and TBI

Stroke and TBI are also causes widespread neural inflammation which VNS may be able to alleviate. In a study conducted by Bansal et al, the effect of VNS on TBI was evaluated by measuring tissue and serum ghrelin and serum TNFa.Citation75 Their study was based on the hypothesis that preventing the inflammatory surge after TBI could prevent sepsis, multi-organ failure, and other adverse effects. Since ghrelin is mediated through acetylcholine levels, it is reasonable to assume that VNS may have an application in treating TBI through a ghrelin or other hypothalamic-gated mechanism.Citation75 VNS decreased the serum levels of TNFa, an early cytokine marker for damage in trauma and ischemic injury. Regulation of cytokine expression by VNS may provide significant therapeutic value in these patients. Because VNS is known to have anti-inflammatory properties and affect the levels of acetylcholine, these changes in cytokine upregulation and rebalancing of neurotransmitter release may provide an immediate and controllable way to modulate injury due to stroke, ischemia, or trauma.

Diabetes

Diabetes is another inflammatory-related disorder that may benefit from treatment with VNS. Recent work has shown the role of the vagus nerve in the pathophysiology of diabetes and other related diseases, which may, in turn, suggest that VNS could be useful in treating such disorders. Cardiovascular risk has long been associated with diabetes, but the exact mechanism by which increased risk and diabetes synergize to exacerbate morbidity is not known. Pal et al found that relatives of type 2 diabetics had increased risk for cardiovascular diseases, which they attributed to sympathovagal imbalance.Citation76 Changes in sympathovagal tone may underlie the increased autoinflammation that could be the foundation of this increased cardiovascular risk. Woie and Reed showed a relationship between changes in tracheal edema in control, diabetic, and insulin-treated diabetic rats which suggests that barrier breakdown was significantly greater in control animals but was attenuated in diabetic rats.Citation77 Changes in airway secretion are vagally mediated, and the altered vagus tone in diabetes may exacerbate susceptibility to chronic inflammation. A broader concern is the role vagal tone plays in metabolic disease and obesity. Vagal afferents and projections to the hypothalamus play a significant role in satiety and feeding behavior, and disruption of vagal afferent traffic may contribute to obesity and downregulation of cholinergic descending tone to arrest inflammation.Citation77 Meyers et al used selective efferent stimulation to significantly lower blood glucose, which may be a potential treatment for type 2 diabetes.Citation78 While the interaction of inflammation and metabolic disorders is becoming clear, the potential role of VNS in treating diabetes needs further investigation. This study by Meyers et al uses a crude method of selective efferent stimulation, by cutting the vagus nerve above the stimulating electrode. However, selective blockade of vagal fibers using different electrical stimulation parameters may provide the answer for using VNS to affect metabolism. Since cutting the vagus nerve is not an ideal solution for human patients, similar results may be obtained by using different parameters that can selectively stimulate fiber types or afferent/efferent traffic. Patel and Butera have achieved such results by using high-frequency stimulation in both the vagus and the sciatic nerves in rats.Citation54,Citation79 Whether parameters for selective stimulation such as used by Patel et al could be used to replicate Meyers et al’s results without the need to sever the vagus nerve remains to be determined.

RA

RA is a chronic, inflammatory autoimmune disease of unknown origin, which results in chronic synovial inflammation and damage to cartilage and bone due to release of cytokines and progressive inflammatory damage.Citation80,Citation81 Alpha 7 (a7) nicotinic receptors are present in cells recovered from synovial fluid and from synovial tissue, particularly in cells with a macrophage-like morphology.Citation82 The general suppression of anti-inflammatory cholinergic pathways plays a critical role in RA. Das’ review summarizes the current literature on the role that vagus tone plays in modulating RA and, as in so many of these inflammatory disorders, the preliminary evidence justifies further experiments.Citation83 In a recently completed study by SetPoint Medical Corporation, VNS devices were implanted in patients with RA to test the safety and efficacy of treatment. Patients were assessed after 6 weeks of treatment and showed a 20% improvement in symptoms.Citation84 Further experiments are needed to determine the role that VNS can play in the treatment of RA, in particular, the potential role of VNS in the modulation of cytokine cascade through a7 receptors.

Conclusion

VNS has been proven to be a useful treatment across a number of domains and has been used effectively to treat epilepsy and depression in adults. There is accumulating evidence to suggest that it can be used to help quell inflammation in a number of other autonomic or inflammatory disorders, which would make it useful for a wider range of pediatric patients as well. Preliminary studies have shown promise for VNS being used for stroke, autoimmune diseases, heart and lung failure, obesity, and pain management, but further studies are needed to fully elucidate the mechanistic actions that explain VNS’s potential role in treating these disorders. Many of these studies are not mechanistic in nature, and further pathway analysis and studies focused on the mechanisms by which VNS alters autonomic tone are key to further our understanding of vagus nerve modification. VNS interacts with the body’s immune system to modify inflammatory tone by altering the release of pro- and anti-inflammatory cytokines. We have summarized some of these key inflammatory markers in . There is an overwhelming evidence to suggest that vagus nerve is an important component of the immune response and manipulating vagal tone is a way to modulate the immune system. Using VNS to manipulate vagal tone provides an exciting new opportunity for minimally invasive therapeutic intervention in adult and pediatric patients.

Figure 3 Flow diagram showing the inflammatory neural circuit.

Notes: Inflammatory markers including IL-1b, IL-6, TNFa, IL-10, HMGB1, IL-11, and IL-13 alter peripheral inflammatory tone, which stimulates NFkB, resulting in increased inflammatory signaling to the CNS via the brain stem. This results in output that generates an inflammatory signal. VNS can be used to alter the input/output of this autonomic control circuitry.

Abbreviations: IL, interleukin; TNF, tumor necrosis factor; NFkB, nuclear factor kB; CNS, central nervous system; VNS, vagus nerve stimulation.

Figure 3 Flow diagram showing the inflammatory neural circuit.Notes: Inflammatory markers including IL-1b, IL-6, TNFa, IL-10, HMGB1, IL-11, and IL-13 alter peripheral inflammatory tone, which stimulates NFkB, resulting in increased inflammatory signaling to the CNS via the brain stem. This results in output that generates an inflammatory signal. VNS can be used to alter the input/output of this autonomic control circuitry.Abbreviations: IL, interleukin; TNF, tumor necrosis factor; NFkB, nuclear factor kB; CNS, central nervous system; VNS, vagus nerve stimulation.

Acknowledgments

Funding for this work was provided by intramural funding through the GRASP and Pediatric Research Initiatives (Loma Linda University) to CGW.

Disclosure

The authors report no conflicts of interest in this work.

References

  • BonazBPicqCSinnigerVMayolJFClarençonDVagus nerve stimulation: from epilepsy to the cholinergic anti-inflammatory pathwayNeurogastroenterol Motil201325320822123360102
  • TraceyKJThe inflammatory reflexNature2002420691785385912490958
  • BorovikovaLVIvanovaSZhangMVagus nerve stimulation attenuates the systemic inflammatory response to endotoxinNature2000405678545846210839541
  • MatteoliGGomez-PinillaPJNemethovaAA distinct vagal anti-inflammatory pathway modulates intestinal muscularis resident macrophages independent of the spleenGut201463693894823929694
  • Rosas-BallinaMOlofssonPSOchaniMAcetylcholine- synthesizing T cells relay neural signals in a vagus nerve circuitScience201133460529810121921156
  • Rosas-BallinaMOchaniMParrishWRSplenic nerve is required for cholinergic antiinflammatory pathway control of TNF in endotoxemiaProc Natl Acad Sci U S A200810531110081101318669662
  • BerthoudHRNeuhuberWLFunctional and chemical anatomy of the afferent vagal systemAuton Neurosci2000851–311711189015
  • YuZJWellerRASandidgeKWellerEBVagus nerve stimulation: can it be used in adolescents or children with treatment-resistant depression?Curr Psychiatry Rep200810211612218474201
  • BrowningKNTravagliRACentral nervous system control of gastrointestinal motility and secretion and modulation of gastrointestinal functionsCompr Physiol2014441339136825428846
  • VivasACBaajAABenbadisSRValeFLThe health care burden of patients with epilepsy in the United States: an analysis of a nationwide database over 15 yearsNeurosurg Focus2012323E1
  • LanskaDJJ.L. Corning and vagal nerve stimulation for seizures in the 1880sNeurology200258345245911839848
  • CouchJDGilmanAMDoyleWKLong-term expectations of vagus nerve stimulation: a look at battery replacement and revision surgeryNeurosurgery2016781424626678088
  • SpuckSNowakGRennebergATronnierVSpernerJRight-sided vagus nerve stimulation in humans: an effective therapy?Epilepsy Res2008822–323223418801642
  • NavasMNavarreteEGPascualJMTreatment of refractory epilepsy in adult patients with right-sided vagus nerve stimulationEpilepsy Res2010901–21720488666
  • McGregorAWhelessJBaumgartnerJBettisDRight-sided vagus nerve stimulation as a treatment for refractory epilepsy in humansEpilepsia20054619196
  • GrovesDABrownVJVagal nerve stimulation: a review of its applications and potential mechanisms that mediate its clinical effectsNeurosci Biobehav Rev200529349350015820552
  • Manuals – Product Information / Manuals – Healthcare Professionals, on VNS Therapy for Epilepsy | Cyberonics US [webpage on the Internet] Available from: http://us.livanova.cyberonics.com/en/vns-therapy-for-epilepsy/healthcare-professionals/vns-therapy/manuals-page/Accessed December 15, 2015
  • ChakravarthyKChaudhryHWilliamsKChristoPJReview of the uses of vagal nerve stimulation in chronic pain managementCurr Pain Headache Rep201519125426493698
  • ElliottRERodgersSDBassaniLVagus nerve stimulation for children with treatment-resistant epilepsy: a consecutive series of 141 casesJ Neurosurg Pediatr20117549150021529189
  • MorrisGLMuellerWMLong-term treatment with vagus nerve stimulation in patients with refractory epilepsy. The Vagus Nerve Stimulation Study Group E01-E05Neurology19995381731173510563620
  • KrahlSEClarkKBVagus nerve stimulation for epilepsy: a review of central mechanismsSurg Neurol Int20123Suppl 4S255S25923230530
  • MacDonaldSCBatemanBTMcElrathTFHernández-DíazSMortality and morbidity during delivery hospitalization among pregnant women with epilepsy in the United StatesJAMA Neurol201572998198826147878
  • PatelSIPennellPBManagement of epilepsy during pregnancy: an updateTher Adv Neurol Disord20169211812927006699
  • HouserMVHennessyMDHowardBCVagal nerve stimulator use during pregnancy for treatment of refractory seizure disorderObstet Gynecol20101152 Pt 241741920093864
  • SalernoGPassamontiCCecchiAZamponiNVagus nerve stimulation during pregnancy: an instructive caseChilds Nerv Syst201632120921126350804
  • ZackMMNational and state estimates of the numbers of adults and children with active epilepsy – United States, 2015MMWR Morb Mortal Wkly Rep20176682182528796763
  • CianchettiCMessinaPPupilloEThe perceived burden of epilepsy: Impact on the quality of life of children and adolescents and their familiesSeizure2015249310125264356
  • TerraVCde PaolaLSilvadoCEAre children affected by epileptic neuropsychiatric comorbidities?Epilepsy Behav20143881224239433
  • HeWWangX-YZhouLTranscutaneous auricular vagus nerve stimulation for pediatric epilepsy: study protocol for a randomized controlled trialTrials20151637126292720
  • HallböökTLundgrenJStjernqvistKBlennowGStrömbladL-GRosénIVagus nerve stimulation in 15 children with therapy resistant epilepsy; its impact on cognition, quality of life, behaviour and moodSeizure200514750451316176878
  • SmythMDTubbsRSBebinEMGrabbPABlountJPComplications of chronic vagus nerve stimulation for epilepsy in childrenJ Neurosurg200399350050312959437
  • FarwellJRLeeYJHirtzDGSulzbacherSIEllenbergJHNelsonKBPhenobarbital for febrile seizures – effects on intelligence and on seizure recurrenceN Engl J Med199032263643692242106
  • LagaeLCognitive side effects of anti-epileptic drugs. The relevance in childhood epilepsySeizure200615423524116563808
  • FernandezLGedelaSTamberMSogawaYVagus nerve stimulation in children less than 3 years with medically intractable epilepsyEpilepsy Res2015112374225847337
  • LevyMLLevyKMHoffDVagus nerve stimulation therapy in patients with autism spectrum disorder and intractable epilepsy: results from the vagus nerve stimulation therapy patient outcome registryJ Neurosurg Pediatr20105659560220515333
  • GreenbergPEFournierA-ASisitskyTPikeCTKesslerRCThe economic burden of adults with major depressive disorder in the United States (2005 and 2010)J Clin Psychiatry201576215516225742202
  • BajboujMMerklASchlaepferTETwo-year outcome of vagus nerve stimulation in treatment-resistant depressionJ Clin Psychopharmacol201030327328120473062
  • American Psychiatric AssociationDiagnostic and Statistical Manual of Mental DisordersWashington, DCAmerican Psychiatric Association1980
  • AndrewsGBrughaTThaseMEDuffyFFRucciPSladeTDimensionality and the category of major depressive episodeInt J Methods Psychiatr Res200716Suppl 1S41S5117623394
  • ZimmermanMChelminskiIPosternakMA review of studies of the Hamilton depression rating scale in healthy controls: implications for the definition of remission in treatment studies of depressionJ Nerv Ment Dis2004192959560115348975
  • NahasZMarangellLBHusainMMTwo-year outcome of vagus nerve stimulation (VNS) for treatment of major depressive episodesJ Clin Psychiatry20056691097110416187765
  • RushAJKraemerHCSackeimHAReport by the ACNP Task Force on response and remission in major depressive disorderNeuropsychopharmacology20063191841185316794566
  • WeisskopfEFumeauxCJFBickle GrazMBon usage des antidépresseurs ISRS durant la grossesse - le défi de l’évaluation de la balance bénéfice-risque. [SSRI antidepressant use during pregnancy and the assessment of the risk-benefit ratio]Rev Médicale Suisse201612510561566 French
  • PretiACardasciaLZenTObstetric complications in patients with depression – a population-based case-control studyJ Affect Disord2000611–210110611099747
  • HusainMMStegmanDTrevinoKPregnancy and delivery while receiving vagus nerve stimulation for the treatment of major depression: a case reportAnn Gen Psychiatry200541616168055
  • JudkinsAJohnsonRLMurraySTYellonSMWilsonCGVagus nerve stimulation in pregnant rats and effects on inflammatory markers in the brain stem of neonatesPediatr Res201883251451929053705
  • BotteronKNGellerBRefractory depression in children and adolescentsDepress Anxiety1997542122239338113
  • Antidepressant Medications for Children and Adolescents: Information for Parents and Caregivers Available from: http://www.nimh.nih.gov/health/topics/child-and-adolescent-mental-health/antidepressant-medications-for-children-and-adolescents-information-for-parents-and-caregivers.shtmlAccessed January 6, 2016
  • SladeETamberPSVincentJ-LThe Surviving Sepsis Campaign: raising awareness to reduce mortalityCrit Care Lond Engl20037112
  • KesslerWDiedrichSMengesPThe role of the vagus nerve: modulation of the inflammatory reaction in murine polymicrobial sepsisMediators Inflamm2012201246762022547905
  • HuangJWangYJiangDZhouJHuangXThe sympathetic-vagal balance against endotoxemiaJ Neural Transm (Vienna)2010117672973520458507
  • JohnsonRLMurraySTCamachoDKWilsonCGVagal nerve stimulation attenuates IL-6 and TNFa expression in respiratory regions of the developing rat brain stemRespir Physiol Neurobiol20162291427049312
  • BarbantiPGrazziLEgeoGPadovanAMLieblerEBussoneGNoninvasive vagus nerve stimulation for acute treatment of high-frequency and chronic migraine: an open-label studyJ Headache Pain2015166126123825
  • PatelYASaxenaTBellamkondaRVButeraRJKilohertz frequency nerve block enhances anti-inflammatory effects of vagus nerve stimulationSci Rep201773981028054557
  • PatelYASaxenaTBellamkondaRVButeraRJCorrigendum: kilohertz frequency nerve block enhances anti-inflammatory effects of vagus nerve stimulationSci Rep201774684828589957
  • LangeGJanalMNManikerASafety and efficacy of vagus nerve stimulation in fibromyalgia: a phase I/II proof of concept trialPain Med20111291406141321812908
  • RandichAGebhartGFVagal afferent modulation of nociceptionBrain Res Brain Res Rev199217277991327371
  • SilbersteinSDMechtlerLLKudrowDBNon-invasive vagus nerve stimulation for the acute treatment of cluster headache: findings from the randomized, double-blind, sham-controlled ACT1 studyHeadache20165681317133227593728
  • FastStats [webpage on the Internet]Obesity and overweight Available from: http://www.cdc.gov/nchs/fastats/obesity-overweight.htmAccessed January 7, 2016
  • BurneoJGFaughtEKnowltonRMorawetzRKuznieckyRWeight loss associated with vagus nerve stimulationNeurology200259346346412177391
  • BodenlosJSKoseSBorckardtJJVagus nerve stimulation acutely alters food craving in adults with depressionAppetite200748214515317081655
  • Val-LailletDBirabenARanduineauGMalbertCHChronic vagus nerve stimulation decreased weight gain, food consumption and sweet craving in adult obese minipigsAppetite201055224525220600417
  • RaybouldHETacheYCholecystokinin inhibits gastric motility and emptying via a capsaicin-sensitive vagal pathway in ratsAm J Physiol19882552G242G2463136661
  • IkramuddinSBlackstoneRPBrancatisanoAEffect of reversible intermittent intra-abdominal vagal nerve blockade on morbid obesity: the ReCharge randomized clinical trialJAMA2014312991592225182100
  • BoronWFBoulpaepELMedical Physiology, 2e Updated Edition: With STUDENT CONSULT Online AccessAtlanta, GAElsevier Health Sciences2012
  • DasUNVagal nerve stimulation in prevention and management of coronary heart diseaseWorld J Cardiol20113410511021526047
  • SloanRPMcCreathHTraceyKJSidneySLiuKSeemanTRR interval variability is inversely related to inflammatory markers: the CARDIA studyMol Med Camb Mass2007133–417818417592552
  • ZhangYPopovicZBBibevskiSChronic vagus nerve stimulation improves autonomic control and attenuates systemic inflammation and heart failure progression in a canine high-rate pacing modelCirc Heart Fail20092669269919919995
  • ZhaoMHeXBiX-YYuX-JGil WierWZangW-JVagal stimulation triggers peripheral vascular protection through the cholinergic anti-inflammatory pathway in a rat model of myocardial ischemia/reperfusionBasic Res Cardiol2013108334523519622
  • ChapleauMWRotellaDLRehoJJRahmouniKStaussHMChronic vagal nerve stimulation prevents high-salt diet-induced endothelial dysfunction and aortic stiffening in stroke-prone spontaneously hypertensive ratsAm J Physiol Heart Circ Physiol20163111H276H28527208157
  • dos SantosCCShanYAkramASlutskyASHaitsmaJJNeuroimmune regulation of ventilator-induced lung injuryAm J Respir Crit Care Med2011183447148220870758
  • LevyGFishmanJEXuDVagal nerve stimulation modulates gut injury and lung permeability in trauma-hemorrhagic shockJ Trauma Acute Care Surg2012732338342 discussion 34222846937
  • TarrasSLDiebelLNLiberatiDMGinnebaughKPharmacologic stimulation of the nicotinic anti-inflammatory pathway modulates gut and lung injury after hypoxia-reoxygenation injurySurgery20131544841847 discussion 847–84824074423
  • ReysLGOrtiz-PomalesYTLopezNUncovering the neuroenteric-pulmonary axis: vagal nerve stimulation prevents acute lung injury following hemorrhagic shockLife Sci2013921378379223439327
  • BansalVRyuSYLopezNVagal stimulation modulates inflammation through a ghrelin mediated mechanism in traumatic brain injuryInflammation201235121422021360048
  • PalGKAdithanCAnanthanarayananPHSympathovagal imbalance contributes to prehypertension status and cardiovascular risks attributed by insulin resistance, inflammation, dyslipidemia and oxidative stress in first degree relatives of type 2 diabeticsPLoS One2013811e7807224265679
  • WoieKReedRKAlloxan diabetes abolishes the increased negativity of interstitial fluid pressure in rat trachea induced by vagal nerve stimulationActa Physiol Scand199716111131199381943
  • MeyersEEKronembergerALiraVRahmouniKStaussHMContrasting effects of afferent and efferent vagal nerve stimulation on insulin secretion and blood glucose regulationPhysiol Rep201644e1271826884478
  • PatelYAButeraRJDifferential fiber-specific block of nerve conduction in mammalian peripheral nerves using kilohertz electrical stimulationJ Neurophysiol2015113103923392925878155
  • FeldmannMBrennanFMMainiRNRole of cytokines in rheumatoid arthritisAnnu Rev Immunol1996143974408717520
  • TettaCCamussiGModenaVDi VittorioCBaglioniCTumour necrosis factor in serum and synovial fluid of patients with active and severe rheumatoid arthritisAnn Rheum Dis19904996656671700672
  • WestmanMEngströmMCatrinaAILampaJCell specific synovial expression of nicotinic alpha 7 acetylcholine receptor in rheumatoid arthritis and psoriatic arthritisScand J Immunol200970213614019630919
  • DasUNCan vagus nerve stimulation halt or ameliorate rheumatoid arthritis and lupus?Lipids Health Dis2011101921261967
  • ClinicalTrials.gov [webpage on the Internet]Safety and Efficacy Vagal Nerve Stimulation in Patients with Rheumatoid Arthritis – Tabular View2012 Available from: https://clinicaltrials.gov/ct2/show/NCT01552941?term=NCT01552941&rank=1Accessed January 7, 2016
  • KirchbergerKHummelCStefanHPostoperative multichannel magnetoencephalography in patients with recurrent seizures after epilepsy surgeryActa Neurol Scand1998981179696520