Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Neurol., 22 September 2025

Sec. Neurorehabilitation

Volume 16 - 2025 | https://doi.org/10.3389/fneur.2025.1643305

The role of vagus nerve stimulation in modulating Parkinson’s disease via the microbiota-gut-brain axis: a comprehensive review

Lingqing Yang,&#x;Lingqing Yang1,2Shiyu Fan,&#x;Shiyu Fan1,2Li Sun,Li Sun1,2Jingru HanJingru Han2Meng Wang
Meng Wang1*Tao Yu,
Tao Yu1,3*
  • 1The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Medical Research Center of Acupuncture, Tianjin, China
  • 2Tianjin University of Traditional Chinese Medicine, Tianjin, China
  • 3Tianjin Xiqing District Hospital of Traditional Chinese Medicine, Tianjin, China

As the global aging trend intensifies, the incidence of neurodegenerative diseases, including Parkinson’s disease (PD), is increasing year by year. Currently, there is no effective cure for PD. Therefore, exploring safe and effective therapeutic targets is of utmost importance. Previous studies have shown that modulation of vagus nerve (VN) activity, a key communication pathway between the brain and the gut, may produce therapeutic effects in PD and influence its disease course by regulating the gut microbiota, brain plasticity, neuroimmune, and neuroendocrine systems, while the nerve itself also plays a complex role that can contribute to pathological processes like disease propagation. This review comprehensively summarizes the potential mechanisms by which vagus nerve stimulation (VNS) intervenes in PD may influence the microbiota-gut-brain axis (MGBA), including the regulation of gut microbiota composition and metabolites, inhibition of central and peripheral neuroinflammatory responses, modulation of hypothalamic–pituitary–adrenal (HPA) axis function, enhancement of brain region functional connectivity and neurotrophic factor secretion, and explores its potential value in translating into clinical therapeutic strategies. This study is the first to integrate the MGBA theory with VNS technology, revealing its cross-system regulatory network in intervening PD and providing new ideas for breaking through the limitations of traditional treatments.

1 Introduction

Parkinson’s disease (PD) is a common neurodegenerative disorder that primarily affects the middle-aged and elderly population. It is characterized by the accumulation of α-synuclein (α-Syn) in the central and peripheral autonomic nervous systems, which contributes to the gradual degeneration of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc) of the midbrain (1). The clinical manifestations of PD are diverse, encompassing a range of motor symptoms, such as bradykinesia, resting tremor, rigidity, and postural instability. In addition, non-motor symptoms, including psychiatric symptoms like anxiety and depression, as well as gastrointestinal symptoms, are also frequently observed in patients with PD (2). In recent years, the incidence of PD has been on a continuous rise. The global prevalence of PD in individuals under 50 years of age is 5.50%, while it significantly increases to 80% in those aged 80 years and above (3). This undoubtedly places a heavy burden on patients’ families and society. Currently, the treatment of PD mainly focuses on symptom control and slowing disease progression. Traditional therapies include levodopa medication and neurosurgical interventions. However, the therapeutic efficacy of levodopa frequently diminishes over time due to disease progression and pharmacokinetic changes, alongside worsening side effects. While neurosurgical interventions (e.g., lesioning, Deep Brain Stimulation) can alleviate symptoms, they involve irreversible neuronal damage (4). Therefore, the search for a safe and effective alternative therapy is imperative.

In recent years, the microbiota-gut-brain axis (MGBA) has emerged as a crucial research domain in the treatment of PD. The MGBA tightly connects the gut with the brain, forming an interactive network of bidirectional communication between the gastrointestinal system (GIS) and the central nervous system (CNS) (5). The gut microbiota, a key component of the MGBA, is closely associated with gastrointestinal dysfunction, CNS inflammation, and DA degeneration (6). The gut microbiota exerts significant regulatory effects on the brain-gut axis through immune, neuroendocrine, and direct neural mechanisms, influencing neurodevelopment, brain function, and behavior (7, 8). Research has shown that enteric nervous system (ENS) degeneration is closely linked to PD (9), with the loss of enteric neurons being implicated in the development of PD-related dysfunction. Changes in the function, connectivity, mitochondria, and/or α-Syn of enteric neurons, as well as alterations in their extrinsic innervation, may underlie the gastrointestinal dysfunction observed in PD patients (10). Sampson et al. comprehensively elucidated the critical role of the MGBA in the progression of PD, demonstrating that PD-associated alterations in the gut microbiota can lead to motor deficits, promote microglial activation, and facilitate α-Syn aggregation (11). Moreover, short-chain fatty acids (SCFAs), metabolites of the gut microbiota, exacerbate neuroinflammation and motor impairments in PD models, suggesting that SCFAs may act as potential molecular mediators in gut-brain signaling. In summary, the MGBA influences the pathogenesis and progression of PD through neuroimmune and neuroendocrine pathways.

The vagus nerve (VN), as the tenth cranial nerve, has been established as a crucial mediator of bidirectional communication between the brain and the gut (12). In recent years, the VN has been shown to be closely associated with the pathogenesis and progression of PD (13). A systematic review by Abdelnaby revealed that PD patients exhibit a degree of VN atrophy and confirmed the diagnostic value of neurosonography in PD (14). An increasing number of studies in recent years have supported Braak’s hypothesis that the VN serves as a “highway” for the propagation of pathological α-Syn from the gut to the brain, thereby elucidating the pathogenesis of PD (1, 15). Consequently, vagotomy can prevent the prion-like transmission of α-Syn, thereby reducing the risk of developing PD (16). Mondal et al. demonstrated that vagus nerve stimulation (VNS) effectively improves two-dimensional spatiotemporal gait parameters in PD patients, suggesting that VNS holds therapeutic potential for PD (17).

In summary, the VN, as a critical relay station of the MGBA, plays a significant role in the pathogenesis and progression of PD. However, the specific molecular mechanisms underlying the intervention of PD by VNS via the MGBA remain unclear, despite its emerging status as a novel therapeutic modality. Therefore, this study aims to integrate preclinical research with translational medical evidence to provide a mechanism-driven theoretical basis for the transition of VNS technology from experimental exploration to personalized precision therapy (Figure 1).

Figure 1
Illustration showing the interaction between the brain and gut via the vagus nerve. Labels include HPA axis, TNF-α, IL-1β, and IF-6 in relation to stress responses and neuronal signals. The gut microbiota composition, depicted within intestinal walls, influences these processes. A device labeled VNS is shown near the vagus nerve (VN), suggesting modulation through electrical stimulation. A legend below lists compounds and cell types: LPS, α-Syn, NE, BDNF, SCFAs, microglia, astrocyte, and dopaminergic neurons.

Figure 1. Multidimensional mechanisms of vagus nerve stimulation in Parkinson’s disease via the microbiota-gut-brain axis. α-Syn, α-synuclein; LPS, lipopolysaccharide; SCFAs, short-chain fatty acids; TNF-α, tumor necrosis factor-alpha; IL-1β, interleukin-1 beta; ROS, reactive oxygen species; BDNF, brain-derived neurotrophic factor; HPA, (hypothalamic–pituitary–adrenal) axis; VN, vagus nerve.

VNS modulates PD pathology through two synergistic pathways: (1) Peripheral (Gut) Effects: Reshapes gut microbiota composition and metabolites (↓ LPS, ↑ SCFAs), reduces intestinal inflammation and α-Syn expression, and inhibits α-Syn propagation to the CNS via the VN. (2) Central (Brain) Effects: Attenuates neuroinflammation (↓ microglia/astrocyte activation; ↓ TNF-α, IL-1β, ROS), enhances brain connectivity, ↑ BDNF secretion, and protects dopaminergic neurons. Simultaneously, VNS normalizes HPA axis function (↓ cortisol), ameliorating gut barrier dysfunction and synergistically amplifying therapeutic outcomes.

2 The relationship between the PD and the gut-microbiome-brain axis

2.1 Relationship between PD and brainstem nuclei

As a key structural component of the MGBA, the VN contains circuitry involving critical brainstem nuclei: the Nucleus Tractus Solitarius (NTS), Locus Coeruleus (LC), and dorsal motor nucleus of the vagus (DMV) (18, 19). The NTS is a vital polysynaptic hub in the human brainstem, located ventrolaterally to the dorsal motor nucleus of the VN. It serves as a major integration site for central sensory afferent neurons and is closely associated with various neural and endocrine systems (20). The NTS also plays a significant role in the pathogenesis of PD. Previous studies have suggested that PD-related respiratory failure may be associated with neuronal degeneration in the NTS. Their research identified impaired substantia nigra pars compacta (SNpc)—periaqueductal gray (PAG) - caudal nucleus of the solitary tract (cNTS) pathways in PD models, elucidating the loss of Phox2b-expressing neurons or hypoxia-activated neurons in the cNTS and subsequent respiratory dysfunction during hypoxic stimulation (21). Sun et al. demonstrated that the administration of 3,4-dihydroxyphenylacetaldehyde (DOPAL) to the VN in rats resulted in the transport of DOPAL and/or higher molecular weight α-Syn mediated by DOPAL via vagal afferent nerves to the nodose ganglion (NG) and NTS (22). This caused significant ultrastructural changes in the NG and NTS and further exacerbated PD-like autonomic dysfunction in rats.

The LC is the primary site in the brain for the production of norepinephrine (NE). NE-producing neurons in the LC widely project to many regions of the CNS to modulate functions such as attention, arousal, pain, mood, and stress responses (23, 24). Neuronal degeneration in the LC and NE signaling dysfunction are common in PD patients. The LC is also one of the earliest brain regions affected by α-Syn pathology, although the mechanisms by which α-Syn affects these neurons remain unclear (25). According to Braak’s staging of α-Syn pathology, the LC is affected early in the disease course (stage 2), before the deposition in the DA substantia nigra (SN) (stage 3), and significant loss of LC-NE neurons becomes apparent as the disease progresses (26). NE neurotransmission dysfunction is associated with non-motor symptoms of PD, including sleep disturbances, anxiety, depression, and cognitive decline. Importantly, central NE deficiency may contribute to chronic inflammation and disease progression in PD (27). A study by Butkovich et al. revealed the mechanisms by which increased α-Syn expression affects central NE transmission and related behaviors (28). The results showed that overexpression of α-Syn and the formation of oligomers have cytotoxic effects, upregulate local inflammatory markers, induce LC fiber degeneration, disrupt striatal dopamine metabolism, and exacerbate non-motor behaviors in PD. Additionally, comprehensive proteomic analysis of LC tissue from PD patients has proposed that important molecular pathogenic pathways in PD include mitochondrial dysfunction, oxidative stress, and cytoskeletal dysregulation. This study also highlighted the potential pathogenic roles of certain proteins in the LC (e.g., serine/threonine-protein kinase PAK3, mitochondrial ribosomal protein MRPS6, calcium-modulating protein regucalcin, and microtubule-associated protein KTN1) and confirmed that aminoacyl-tRNA biosynthesis pathways may also be involved in the pathogenesis of PD (29).

2.2 Dysregulation of the HPA axis aggravates the progression of PD

The hypothalamic–pituitary–adrenal (HPA) axis is a critical neuroendocrine pathway within the MGBA. Dysfunction of the HPA axis is primarily characterized by alterations in peripheral cortisol levels and/or abnormal cortisol responses to stressful life events. Excess glucocorticoids (GCs) can induce neuronal damage in brain regions such as the hippocampus and SN through several mechanisms, including mitochondrial dysfunction, inhibition of inhibitory and excitatory synaptic transmission, and disruption of microglial or astrocytic functions, thereby exacerbating the progression of PD (3033). Moreover, overactivation of the HPA axis can trigger oxidative stress and modulate the diversity of gut microbiota via GCs, leading to impaired intestinal barrier function and gastrointestinal dysfunction (34). Patients with PD often exhibit GC dysregulation, which can result in abnormal gut microbial metabolism, disruption of intestinal mucosal morphology, and mediation of intestinal inflammatory responses (35). These changes can subsequently affect brain function. Research has shown that GC dysregulation can lead to persistent neuroinflammation and oxidative stress in the brain, causing damage to DA neurons and worsening PD symptoms (36). However, some researchers have proposed that GCs have a bidirectional regulatory role in PD. Specifically, GCs can induce Parkin expression via the cAMP-response element-binding protein (CREB) pathway, exerting neuroprotective effects and preventing the death of DA neurons (37). Thus, the gut microbiota and neuroendocrine system interact with each other, and their combined effects influence both motor and non-motor symptoms in PD patients.

2.3 The close relationship between intestinal microbiota and PD

In the 1980s, the first reports of Lewy body pathology in the ENS provided evidence for ENS and gut microbiota dysfunction in PD (38). Some researchers have proposed that the pathological mechanisms of PD may originate in the gut, with α-Syn pathology spreading to the brain in a bottom-up manner (39). Shannon et al. found that α-Syn had already accumulated extensively in enteric neurons in PD patients before the onset of motor symptoms through colonic biopsies (40). Additionally, increased intestinal permeability has been observed in the early stages of PD, which may be related to the increased accumulation of α-Syn in the gut, triggering intestinal inflammation (41). Statistics show that approximately 60–80% of PD patients suffer from ENS dysfunction, including chronic constipation, drooling, and dysphagia, with significantly higher prevalence than in non-PD populations (42, 43). These non-motor symptoms often precede motor symptoms (44). In recent years, Gastrointestinal Dysfunction Scale for PD (GIDS-PD) is widely used in clinical evaluation of PD. Camacho et al. showed that intestinal dysfunction may be a phenotypic feature of PD subgroups, which has implications for patient stratification and management (45). Bissacco et al. stated that the GIDS-PD score was directly related to non-motor dysfunction in PD patients (46). Nowak et al. confirmed the reliability of constipation and irritable bowel subindicators in the assessment of PD (47). Moreover, ENS dysfunction may have already accelerated PD pathology up to 10 years before the onset of clinical symptoms (48). In recent years, the role of gut microbiota and their metabolites in the progression of PD has attracted widespread attention from the scientific community. There is now substantial evidence that as PD progresses, the number of gut microbes changes, and alterations in microbial community structure are associated with disease severity (49). Huang et al. found that gut microbiota dysregulation can occur in the prodromal stage of PD, such as a reduction in short-chain fatty acid (SCFA)-producing bacteria (Lachnospira and Butyricicoccus) and butyrate-producing [Eubacterium]_ventriosum_group (50). This condition is more severe in PD patients with rapid eye movement sleep behavior disorder (RBD). This may compromise the integrity of the intestinal barrier, leading to increased intestinal permeability, activation of intestinal immune responses, and accumulation of α-Syn in the gut, ultimately triggering neuroinflammation and pathological processes in PD (51). A cross-sectional study showed that PD patients had higher abundance of three phyla (Proteobacteria, Verrucomicrobia, and Actinobacteria) and five genera (Akkermansia, Enterococcus, Hungatella, and two Ruminococcaceae) in their gut microbiome. In patients with longer disease duration, the abundance of Fournierella and DTU089 (Ruminococcaceae) increased, while that of Roseburia (Lachnospiraceae) decreased. Additionally, the composition of gut microbiota also varied among patients with different motor subtypes (52).

In addition, metabolites of the gut microbiota, such as SCFAs and lipopolysaccharides (LPS), have been shown to play important roles in the pathogenesis and progression of PD. A healthy gut microbiota often promotes the integrity of the blood–brain barrier (BBB) by regulating tight junction protein expression mediated by SCFAs (e.g., occludin and claudin-5) (53). SCFAs also play a crucial role in maintaining the integrity of the intestinal barrier by preventing microbial translocation, thereby alleviating local intestinal inflammation, systemic inflammation, and neuroinflammation (54). LPS, an endotoxin produced by Gram-negative bacteria, interacts with immune cells in the bloodstream, upregulating the expression of pro-inflammatory cytokines (e.g., tumor necrosis factor-α (TNF-α) and interleukins) and systemic inflammation (55). In individuals with PD, certain microbial compositions (e.g., dysbiosis) have been found to stimulate the production of inflammatory cytokines and LPS, leading to intestinal epithelial damage and compromised barrier integrity (56). Studies have also shown that LPS can induce a structurally unique self-replicating α-Syn fibril strain in mice, which triggers a signature pattern of synucleinopathy similar to that induced by wild-type α-Syn commonly observed in PD (57). In addition, gut microbiota metabolites derived from dietary polyphenols (e.g., phenolic sulfates) can cross the BBB and directly modulate microglia activity and neuroinflammation (58, 59). For instance, pyrosulfate (Pyr-sulf) enhances BBB integrity while suppressing nuclear factor-κB (NF-κB) signaling pathways (60, 61). Clinical studies have further confirmed these metabolites’ presence in cerebrospinal fluid (62). Therefore, maintaining gut homeostasis, ensuring normal ENS function, and regulating the quantity and community structure of gut microbiota are of great importance for the prevention and treatment of PD.

2.4 Neuroinflammation in PD

PD is characterized by the slow and progressive degeneration of DA neurons in the SNc. Neuroinflammatory mechanisms are likely to be one of the causes of neuronal degeneration. Multiple studies have demonstrated that alterations in the peripheral immune system precede the emergence of classic motor symptoms in PD patients. PET imaging reveals microglia activation in the SN before motor symptoms appear, while peripheral immune cells (e.g., CD8 + T cells and monocytes) can infiltrate the central nervous system through compromised BBB (63). The pathological process of PD may originate from inflammation in the peripheral nervous system surrounding visceral organs and progress to the brainstem and SNc via the VN (64). Postmortem analyses of PD patients have shown significant increases in activated microglia, HLA-positive microglia, and astrocyte density in the SNc (6567). These proliferating and activated glial cells induce inflammatory mediators, which subsequently cause oxidative damage and accelerate the degeneration of DA neurons in the SNc. It has been reported that pro-inflammatory changes are present in the blood and cerebrospinal fluid (CSF) of PD patients. Levels of TNF-α, interleukin 1β (IL-1β), and interleukin 6 (IL-6) are elevated in the CSF samples of PD patients (68, 69). Antibodies that recognize various components of DA neurons, including the products of dopamine oxidation, have been identified in the serum of PD patients (70). Studies have also shown that the number of CD4 + and CD45RA + T cells (representing naive lymphocytes) is reduced in the serum of PD patients, while the number of CD4 + and CD45RO + T cells (representing activated T cells) is increased, indicating that peripheral lymphocytes are activated and the inflammatory response is exacerbated (71). Moreover, gut microbiota dysbiosis mediates CNS inflammatory responses by damaging the intestinal epithelial barrier (IEB), disrupting intercellular junctions, increasing intestinal permeability, and promoting inflammatory mediator translocation (72). In pre-symptomatic PD patients (e.g., those with RBD), this dysbiosis reduces short-chain fatty acid-producing bacteria, further elevating permeability and facilitating CNS infiltration of inflammatory mediators like LPS and α-Syn (73). Pathological α-Syn aggregation then accelerates DA neuron degeneration and PD progression. Reducing inflammatory responses is therefore crucial to slowing PD advancement.

3 The relationship between the VN and the gut-microbiome-brain axis

The VN, as a component of the parasympathetic nervous system, has multiple physiological functions, including regulating immune responses, digestion, heart rate, and emotional control (74). It is also considered a vital connection between the human brain and the gut, and the pathological biomarker associated with PD, α-Syn, may propagate bidirectionally between the gut and brain via the VN (75). The VN may exert therapeutic effects on PD through the MGBA, primarily by regulating brain plasticity, maintaining gut homeostasis, modulating the HPA axis, and exerting anti-neuroinflammatory effects (Table 1).

Table 1
www.frontiersin.org

Table 1. Vagus nerve stimulation mechanisms targeting Parkinson’s disease hallmarks via the microbiota-gut-brain axis.

3.1 VNS regulates brain plasticity

VNS can optimize brain plasticity, mainly by improving functional connectivity between brain regions, regulating neurotransmitters, and modulating the secretion of neurotrophic factors. A study by Yang et al. found that transcutaneous VNS (tVNS) can protect the integrity of the BBB in ischemic injury rat models and significantly reduce the infarct size caused by ischemic stroke (76). Immediate studies have shown that tVNS can alter the functional connectivity between the parietal and temporal lobes in patients with mild cognitive impairment (MCI) and enhance the activity of multiple brain regions, thereby improving brain function in MCI patients, with the cingulate gyrus possibly being one of the targets regulated by tVNS in MCI (77). Another study showed that transcutaneous auricular VNS (taVNS) can significantly activate the left triangular part of the inferior frontal gyrus (IFG) in PD patients with anxiety, increase the concentration of oxyhemoglobin in this brain region, and thereby improve the patients’ anxiety (78). Dysfunction of the thalamocortical connectivity network is considered the basis of migraine pathophysiology. Clinical research results by Zhang et al. confirmed that taVNS may relieve migraines by increasing connectivity between motor-related thalamic subregions and the anterior cingulate cortex/medial prefrontal cortex, while reducing connectivity between occipital cortex-related thalamic subregions and the postcentral gyrus/precuneus (79). Wang et al. found that right vagus nerve stimulation (RVNS) significantly upregulated the levels of tyrosine hydroxylase (TH) and vesicular monoamine transporter 2 (VMAT2) in the midbrain and reduced α-Syn expression in the SN, exerting neuroprotective effects on brain DA neurons (80). Motor behavior in PD rats was also improved after intervention. Thus, RVNS may be a potential therapeutic option for PD. Some researchers believe that PD-related motor symptoms (such as bradykinesia and rigidity) may be associated with the overexpression of pathological brain rhythms in the β band within the subthalamic nucleus (STN) (81). A study by Marano et al. showed that left taVNS can interact with the right STN circuit, reduce the total β power in the right STN, and effectively improve subclinical gait parameters (Timed Up and Go time, velocity, and variability) in PD patients (82). Other studies have proposed that VNS can exert neuroprotective effects on PD models and improve PD-related behavioral deficits by increasing brain-derived neurotrophic factor (BDNF) and enhancing the survival-promoting mechanisms of its receptor, tropomyosin receptor kinase-B (TrkB) (83).

3.2 VNS modulates gut microbiota and repairs the intestinal barrier

The ENS and the VN together constitute the main neural pathways of the MGBA. The ENS, a division of the autonomic nervous system (ANS), communicates with the CNS via sensory and motor neurons and neurotransmitters. This communication is primarily mediated by the VN’s afferent and efferent fibers (84). Previous studies have found that VNS can effectively alleviate intestinal barrier damage and restore intestinal permeability (85). Wang et al. demonstrated that VNS treatment for post-stroke hemiplegia is somewhat effective and can improve BBB and intestinal barrier damage following ischemic stroke (86). VNS can also alter the gut microenvironment and microbiota. Preclinical research has shown that subdiaphragmatic vagotomy (SDV) can lower blood pressure in spontaneously hypertensive rats (SHR) while reducing the abundance of Defluviitaleaceae bacteria in feces (87). Another study found that SDV can prevent depressive-like behaviors and gut microbiota dysbiosis in mice treated with LPS, further supporting the role of the VN in the MGBA (88). Liu et al. showed that taVNS can alter the natural course of constipation-predominant irritable bowel syndrome (IBS-C), with 16S rRNA sequencing analysis results indicating that taVNS restored the abundance of Lactobacillus and increased the abundance of Bifidobacterium probiotics at the genus level (89). Other studies have shown that electroacupuncture stimulation of the VN can reverse the decrease in Firmicutes abundance and increase in Bacteroidetes abundance caused by ischemic stroke, thereby improving gut microbiota dysbiosis (86). Additionally, a study on adolescent irritable bowel syndrome showed that subjects with higher abundance of Blautia after tVNS had better outcomes. This suggests that patients with specific microbial profiles may be more responsive to tVNS (90). In summary, VNS can influence the ENS to some extent, improve intestinal barrier damage, and maintain the homeostasis of gut microbiota, thereby alleviating brain and gut-related diseases.

3.3 VNS modulates the HPA Axis

The VN is a key component of the bidirectional communication between the gut and the brain and is a major regulatory element for sensing the “internal environment” and promoting neuroendocrine responses to maintain gut health. As one of the important pathways for emotional and cognitive regulation, dysregulation of the HPA axis is associated with the induction, exacerbation, or progression of PD (91). Therefore, maintaining normal HPA axis function is of great significance for the prevention and treatment of PD in clinical practice. Previous studies have found that modulating the HPA axis can effectively alleviate PD-related depressive symptoms. van Wamelen et al. proposed that HPA axis hyperactivation can occur in the prodromal stage of PD, manifesting as elevated cortisol levels (92). This is closely related to the occurrence of non-motor symptoms in PD, such as depression, anxiety, sleep, and cognition. They further speculated that modulating cortisol levels might be one of the therapeutic targets for PD-related neuropsychiatric symptoms. VNS is believed to play an important role in maintaining neuroendocrine homeostasis by restoring stress-induced HPA axis responses (93). Keller et al. (94) found that VNS may modulate HPA axis function by increasing corticotropin-releasing factor (CRF) mRNA in the paraventricular nucleus of the hypothalamus (PVN) and the firing of specific PVN CRF neurons, thereby upregulating corticosterone levels. VNS has previously been shown to alleviate chronic depression by modulating the HPA axis and reducing the CRH (corticotropin-releasing hormone)/ACTH (adrenocorticotropic hormone) response (95). VNS can also activate the anti-inflammatory effects of the HPA axis via vagal afferent fibers. In rats treated with vagotomy, inflammatory markers (myeloperoxidase activity, serum amyloid P levels, and interleukin [IL]-1β, IL-6, and TNF-α) were significantly elevated (96). Eugenijus Kaniusas et al. confirmed that taVNS can reduce pro-inflammatory cytokines and regulate lung injury by activating the HPA anti-inflammatory pathway, thereby alleviating acute respiratory distress syndrome caused by Covid-19 (97). In summary, the HPA axis is a key component of the MGBA, and a possible mechanism is that VNS modulates PD-related neuropsychiatric symptoms by regulating HPA axis function via the vagus nerve.

3.4 Anti-inflammatory effect of VNS

Recent studies have shown that the VN has anti-inflammatory effects, mediated through multiple pathways, including the cholinergic anti-inflammatory pathway (CAP), the HPA axis anti-inflammatory pathway, and the splenic sympathetic nerve anti-inflammatory pathway (98). The cholinergic system is one of the important pathways through which VNS exerts its anti-inflammatory effects. When external stimuli excite the vagus nerve, the nerve terminals produce the anti-inflammatory parasympathetic neurotransmitter acetylcholine (ACh), which activates the α7 nicotinic acetylcholine receptors (α7nAChR) on monocytes and macrophages. This further activates the intracellular NF-κB signaling pathway and the Janus kinase 2/signal transducers and activators of transcription 3 (JAK2/STAT3) pathway, thereby inhibiting the production of pro-inflammatory cytokines such as IL-1β and TNF-α, ultimately exerting anti-inflammatory effects (99). In recent years, the CAP has been considered one of the therapeutic targets for PD. On the one hand, it has anti-neuroinflammatory properties; on the other hand, it can regulate DA release, prevent the degeneration of DA neurons, and rebalance the direct and indirect signaling pathways in the striatum (100). Previous studies have explored the therapeutic potential of VNS in PD through the CAP pathway in PD rat models. Farrand et al. found that after VNS intervention, PD rats showed significant improvements in motor symptoms and increased expression of TH (101). Additionally, reduced expression of glial fibrillary acidic protein (GFAP) and lonized calcium-binding adapter molecule 1 (Iba-1) in glial cells was observed, indicating decreased inflammatory responses. Similar results were observed in another study, where researchers first injected 6-hydroxydopamine into the medial forebrain bundle of Wistar rats and then performed aVNS treatment. The treatment significantly improved motor deficits in PD rats, increased TH and α7 nAChR expression, and reduced levels of pro-inflammatory cytokines (TNF-α and interleukin-1β [IL-1β]). Moreover, it increased the number of regulatory T cells (Treg) while reducing the number of T helper cell 17 (Th17) (102). These studies suggest that VNS may exert neuroprotective effects against DA neuron damage by inhibiting the inflammatory process and modulating innate immune responses.

The VN is also believed to activate splenic sympathetic nerves via a vagal-sympathetic reflex, releasing NE that binds to splenic lymphocytes, promoting the release of ACh, which in turn inhibits the secretion of pro-inflammatory cytokines from splenic macrophages through a negative feedback loop, thereby exerting anti-inflammatory effects via the splenic sympathetic nerve pathway (103, 104). It has been reported that taVNS may inhibit peripheral inflammatory responses by modulating the α7nAChR/JAK2/STAT3 signaling pathway in the spleen, reducing the release of chemokine C-X-C Motif Chemokine Ligand 1 (CXCL1) and exerting anti-inflammatory effects, thereby improving LPS-induced depressive-like behaviors in rats (105). However, this theory may be controversial. Bratton et al. found in their experiments that the vagal efferent nerves in rats do not synapse with splenic sympathetic neurons nor drive their sustained activity (106). Based on this, Martelli et al. proposed a non-neural connection model between the VN and the spleen (107). They suggested that α7nAChRs are located on the peripheral terminals of splenic sympathetic nerves. When afferent T cells are stimulated by ACh, these terminals release NE, which then acts on β-adrenergic receptors on splenic macrophages, inhibiting their release of TNF-α. Other studies have suggested that VNS may induce the release of ACh in the celiac mesenteric ganglia, which binds to post-synaptic α7nAChRs in the splenic nerve, releasing NE in the spleen to exert anti-inflammatory effects (108).

Moreover, significant degeneration of NE neurons in the LC can also exacerbate the neuroinflammatory process in PD (109). Ittetsu Kin et al. demonstrated in a PD rat model that VNS increased NE in the LC and significantly inhibited the activation of microglia and astrocytes induced by 6-Hydroxydopamine (6-OHDA), exerting a protective effect on DA neurons in the SN (110). This alleviated PD-like motor symptoms and DA neuronal degeneration in rats. Their study found that mild stimulation (0.25–0.5 mA) provided optimal anti-inflammatory and neuroprotective effects in PD rats. Other studies confirmed that continuous afferent vagal stimulation reduces inflammatory neuroglia in the SN, upregulates rate-limiting enzyme density in the LC, and alleviates motor deficits in PD rat models (111).

4 Innovative strategies for treating PD with VNS

4.1 Stimulation modalities

At the end of the 19th century, American neurologist James Corning first used VNS to treat epilepsy. Although the therapeutic effect was not satisfactory, the concept of VNS was introduced to the world (112). With the continuous development of technology, the clinical efficacy and safety of VNS have been greatly improved. In 1997, the US Food and Drug Administration (FDA) approved the implantable left cervical VNS device for the treatment of refractory epilepsy (TRE) (113, 114). In 2005, VNS was approved for the treatment of treatment-resistant depression (TRD) (115). Cheng et al. proposed that VNS might become a novel therapeutic approach for PD, with neuroprotective effects, and indicated that activation of noradrenergic neurons in the LC may play an important role in VNS treatment for PD (116). The stimulation modalities of the VN have gradually evolved into various forms of non-invasive stimulation. Compared with invasive VNS, non-invasive VNS often has the advantages of high safety, portability, and long-lasting therapeutic effects, although it may be slightly inferior to invasive stimulation in terms of stimulation intensity. Currently, the commonly used tVNS in clinical practice includes taVNS, transcutaneous cervical VNS (tcVNS), and closed-loop taVNS (CL-taVNS). TaVNS is a promising neuromodulatory approach developed based on VNS technology and traditional Chinese medicine auricular acupuncture. This technique can stimulate the cutaneous receptive areas of the auricular branches of the VN to regulate bodily functions and achieve therapeutic effects. In 2010, taVNS was approved in Europe for the treatment of epilepsy and depression, and in 2012, it was approved for the treatment of pain (117, 118). In recent years, taVNS has been considered to have an intervention effect on the occurrence and development of PD. Marano et al. showed that taVNS can effectively improve gait parameters such as stride length, swing amplitude, gait velocity, and gait time in PD patients (119). Other studies have shown that taVNS may exert neuroprotective effects on DA damage in PD models by inhibiting inflammatory evolution and modulating innate immune responses (102). In addition, tcVNS works by stimulating the cervical branches of the VN and is also recognized for the prevention and treatment of epilepsy, primary headache, anxiety, depression, and other diseases (120, 121). Morris et al. conducted a pilot study and found that tcVNS can effectively improve gait abnormalities in PD, especially in terms of gait time and stride length (122). CL-taVNS is an automatically controlled taVNS system, the process of which is regulated by biofeedback signals (such as behavioral changes, respiratory changes, brain activity, etc.) (123). This device can more sensitively adapt to dynamically detectable changes in the clinic, thereby providing personalized taVNS protocols and improving therapeutic efficiency. A recent study showed that electroencephalogram (EEG)-gated taVNS can significantly downregulate delta wave power to treat delirium, indicating that EEG-gated taVNS may be a promising option for intervening in clinical disorders of consciousness (124).

4.2 Stimulation parameters

Given the good efficacy and high safety of non-invasive VNS, research on tVNS has gradually increased in recent years. However, there is currently no unified standard for stimulation parameters in clinical practice. Reviewing previous studies on tVNS intervention in PD, we found that in improving PD-related motor disorders, the tVNS stimulation frequency is often set at 20 Hz, the stimulation intensity is usually adjusted according to the sensitivity threshold, and the pulse width is mainly 0.2 ms and 0.5 ms (125). van Midden et al. compared the effects of 25 Hz and 100 Hz taVNS on PD gait and found that a stimulation frequency of 25 Hz was more effective in improving gait disorders, while a frequency of 100 Hz was more significant in increasing arm swing velocity (126). Currently, there are few studies using VNS to treat PD-related non-motor symptoms. In a recent study on taVNS intervention for PD-related anxiety symptoms, an intermittent alternating stimulation pattern of 20 Hz and 4 Hz was used. The results showed that taVNS not only improved anxiety symptoms in PD with anxiety (PD-A) patients but also modulated the function of the IFG (78). In summary, research on different stimulation parameters for improving PD symptoms is relatively lacking. Future studies should explore the optimal stimulation parameters for tVNS targeting different PD symptoms on the basis of proving the efficacy of tVNS, in order to provide more precise and efficient treatment options for PD patients.

4.3 Controversies, open questions, and future directions

Despite the promising role of VNS in modulating PD via the MGBA, several controversies and unresolved issues warrant attention. Key controversies include the dual role of GCs, which may promote neuroinflammation in some contexts (e.g., through HPA axis dysregulation exacerbating DA neuronal damage) yet offer neuroprotective effects in others (e.g., by inducing Parkin expression via CREB pathways to prevent neuronal death) (37, 91). Similarly, vagotomy has been proposed as a preventive measure against α-synuclein propagation (16), but it may compromise anti-inflammatory benefits associated with intact vagal function, as vagal ablation can exacerbate inflammatory responses in models like LPS-induced depression (96). Additionally, the mechanisms underlying VNS-mediated anti-inflammatory effects—particularly through splenic sympathetic pathways—remain debated, with conflicting evidence on neural versus non-neural connections (107, 108).

Open questions persist regarding the specificity of VNS effects, such as how it precisely regulates gut microbiota homeostasis and whether these changes directly influence PD progression in humans. The optimal stimulation parameters (e.g., frequency, intensity) for targeting different PD symptoms (motor vs. non-motor) are also underexplored, as current studies show variable efficacy (125, 126). Future research should prioritize personalized VNS therapies, leveraging closed-loop systems integrated with multimodal biomarkers (e.g., EEG-gated taVNS) to dynamically optimize stimulation in real-time (123). Large-scale clinical trials are needed to validate VNS efficacy across diverse PD subtypes and address non-motor symptoms like cognitive impairment and gastrointestinal dysfunction, which have been neglected in prior studies.

5 Conclusion

This review synthesizes evidence supporting VNS as a promising intervention for PD through modulation of the MGBA. By targeting key pathways—including gut microbiota homeostasis, HPA axis function, neuroinflammation, and brain plasticity—VNS demonstrates potential to mitigate PD progression across multiple systems.

While preclinical and clinical studies highlight its therapeutic value, translating VNS into clinically actionable strategies requires resolving some controversies. Future efforts should prioritize large-scale trials validating efficacy for both motor and non-motor symptoms, alongside development of personalized closed-loop systems to enhance precision.

In summary, VNS technology holds promise to break through the limitations of traditional PD treatments through mechanism-driven targeted interventions and clinically adaptable technological empowerment, ultimately achieving a paradigm shift from “symptom control” to “disease modification.”

Author contributions

LY: Writing – review & editing, Writing – original draft. SF: Writing – review & editing, Writing – original draft. LS: Writing – original draft, Visualization. JH: Writing – review & editing. MW: Resources, Writing – review & editing. TY: Writing – review & editing.

Funding

The author(s) declare that financial support was received for the research and/or publication of this article. This research was supported by Tianjin Scientific Research Projects in Key Areas of Traditional Chinese Medicine (2025017).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The authors declare that no Gen AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Kalia, LV, and Lang, AE. Parkinson's disease. Lancet (London, England). (2015) 386:896–912. doi: 10.1016/S0140-6736(14)61393-3

PubMed Abstract | Crossref Full Text | Google Scholar

2. Tolosa, E, Garrido, A, Scholz, SW, and Poewe, W. Challenges in the diagnosis of Parkinson's disease. Lancet Neurol. (2021) 20:385–97. doi: 10.1016/S1474-4422(21)00030-2

PubMed Abstract | Crossref Full Text | Google Scholar

3. Zhu, J, Cui, Y, Zhang, J, Yan, R, Su, D, Zhao, D, et al. Temporal trends in the prevalence of Parkinson's disease from 1980 to 2023: a systematic review and meta-analysis. Lancet Healthy Longev. (2024) 5:e464–79. doi: 10.1016/S2666-7568(24)00094-1

PubMed Abstract | Crossref Full Text | Google Scholar

4. Przytuła, F, Dulski, J, Sobstyl, M, and Sławek, J. Battery for deep brain stimulation depletion in Parkinson's disease and dystonia patients - a systematic review. Neurol Neurochir Pol. (2021) 55:346–50. doi: 10.5603/PJNNS.a2021.0041

PubMed Abstract | Crossref Full Text | Google Scholar

5. Rhee, SH, Pothoulakis, C, and Mayer, EA. Principles and clinical implications of the brain-gut-enteric microbiota axis. Nat Rev Gastroenterol Hepatol. (2009) 6:306–14. doi: 10.1038/nrgastro.2009.35

PubMed Abstract | Crossref Full Text | Google Scholar

6. Borre, YE, Moloney, RD, Clarke, G, Dinan, TG, and Cryan, JF. The impact of microbiota on brain and behavior: mechanisms & therapeutic potential. Adv Exp Med Biol. (2014) 817:373–403. doi: 10.1007/978-1-4939-0897-4_17

PubMed Abstract | Crossref Full Text | Google Scholar

7. Mayer, EA, Tillisch, K, and Gupta, A. Gut/brain axis and the microbiota. J Clin Invest. (2015) 125:926–38. doi: 10.1172/JCI76304

PubMed Abstract | Crossref Full Text | Google Scholar

8. Yang, D, Zhao, D, Ali Shah, SZ, Wu, W, Lai, M, Zhang, X, et al. The role of the gut microbiota in the pathogenesis of Parkinson's disease. Front Neurol. (2019) 10:1155. doi: 10.3389/fneur.2019.01155

PubMed Abstract | Crossref Full Text | Google Scholar

9. McQuade, RM, Singleton, LM, Wu, H, Lee, S, Constable, R, Di Natale, M, et al. The association of enteric neuropathy with gut phenotypes in acute and progressive models of Parkinson's disease. Sci Rep. (2021) 11:7934. doi: 10.1038/s41598-021-86917-5

PubMed Abstract | Crossref Full Text | Google Scholar

10. O'Day, C, Finkelstein, DI, Diwakarla, S, and McQuade, RM. A critical analysis of intestinal enteric neuron loss and constipation in Parkinson's disease. J Parkinsons Dis. (2022) 12:1841–61. doi: 10.3233/JPD-223262

PubMed Abstract | Crossref Full Text | Google Scholar

11. Sampson, TR, Debelius, JW, Thron, T, Janssen, S, Shastri, GG, Ilhan, ZE, et al. Gut microbiota regulate motor deficits and Neuroinflammation in a model of Parkinson's disease. Cell. (2016) 167:1469–80.e12. doi: 10.1016/j.cell.2016.11.018

PubMed Abstract | Crossref Full Text | Google Scholar

12. Bonaz, B, Bazin, T, and Pellissier, S. The Vagus nerve at the Interface of the microbiota-gut-brain Axis. Front Neurosci. (2018) 12:49. doi: 10.3389/fnins.2018.00049

PubMed Abstract | Crossref Full Text | Google Scholar

13. Ma, YY, Li, X, Yu, JT, and Wang, YJ. Therapeutics for neurodegenerative diseases by targeting the gut microbiome: from bench to bedside. Transl Neurodegen. (2024) 13:12. doi: 10.1186/s40035-024-00404-1

PubMed Abstract | Crossref Full Text | Google Scholar

14. Abdelnaby, R, Elsayed, M, Mohamed, KA, Dardeer, KT, Sonbol, YT, A, EL, et al. Vagus nerve ultrasonography in Parkinson's disease: a systematic review and meta-analysis. Auton Neurosci. (2021) 234:102835. doi: 10.1016/j.autneu.2021.102835

Crossref Full Text | Google Scholar

15. Uemura, N, Yagi, H, Uemura, MT, Hatanaka, Y, Yamakado, H, and Takahashi, R. Inoculation of α-synuclein preformed fibrils into the mouse gastrointestinal tract induces Lewy body-like aggregates in the brainstem via the vagus nerve. Mol Neurodegener. (2018) 13:21. doi: 10.1186/s13024-018-0257-5

PubMed Abstract | Crossref Full Text | Google Scholar

16. Kaufmann, H. Cutting the vagal highway blocks one point of entry for prion-like alpha-synuclein. Ann Neurol. (2015) 78:520–1. doi: 10.1002/ana.24492

PubMed Abstract | Crossref Full Text | Google Scholar

17. Mondal, B, Choudhury, S, Simon, B, Baker, MR, and Kumar, H. Noninvasive vagus nerve stimulation improves gait and reduces freezing of gait in Parkinson's disease. Move Disord. (2019) 34:917–8. doi: 10.1002/mds.27662

PubMed Abstract | Crossref Full Text | Google Scholar

18. Frangos, E, Ellrich, J, and Komisaruk, BR. Non-invasive access to the Vagus nerve central projections via electrical stimulation of the external ear: fMRI evidence in humans. Brain Stimul. (2015) 8:624–36. doi: 10.1016/j.brs.2014.11.018

PubMed Abstract | Crossref Full Text | Google Scholar

19. Travagli, RA, and Anselmi, L. Vagal neurocircuitry and its influence on gastric motility. Nat Rev Gastroenterol Hepatol. (2016) 13:389–401. doi: 10.1038/nrgastro.2016.76

PubMed Abstract | Crossref Full Text | Google Scholar

20. Andresen, MC, and Kunze, DL. Nucleus tractus solitarius--gateway to neural circulatory control. Annu Rev Physiol. (1994) 56:93–116. doi: 10.1146/annurev.ph.56.030194.000521

PubMed Abstract | Crossref Full Text | Google Scholar

21. Naccarato, MC, Oliveira, LM, Ferreira, CB, Moreira, TS, and Takakura, AC. Nucleus of the solitary tract neuronal degeneration and impaired hypoxia response in a model of Parkinson's disease. Exp Neurol. (2024) 380:114924. doi: 10.1016/j.expneurol.2024.114924

PubMed Abstract | Crossref Full Text | Google Scholar

22. Sun, J, He, C, Yan, QX, Wang, HD, Li, KX, Sun, X, et al. Parkinson-like early autonomic dysfunction induced by vagal application of DOPAL in rats. CNS Neurosci Ther. (2021) 27:540–51. doi: 10.1111/cns.13589

PubMed Abstract | Crossref Full Text | Google Scholar

23. Poe, GR, Foote, S, Eschenko, O, Johansen, JP, Bouret, S, Aston-Jones, G, et al. Locus coeruleus: a new look at the blue spot. Nat Rev Neurosci. (2020) 21:644–59. doi: 10.1038/s41583-020-0360-9

PubMed Abstract | Crossref Full Text | Google Scholar

24. Suárez-Pereira, I, Llorca-Torralba, M, Bravo, L, Camarena-Delgado, C, Soriano-Mas, C, and Berrocoso, E. The role of the locus Coeruleus in pain and associated stress-related disorders. Biol Psychiatry. (2022) 91:786–97. doi: 10.1016/j.biopsych.2021.11.023

PubMed Abstract | Crossref Full Text | Google Scholar

25. Weinshenker, D. Long road to ruin: noradrenergic dysfunction in neurodegenerative disease. Trends Neurosci. (2018) 41:211–23. doi: 10.1016/j.tins.2018.01.010

PubMed Abstract | Crossref Full Text | Google Scholar

26. Braak, H, Del Tredici, K, Rüb, U, de Vos, RA, Jansen Steur, EN, and Braak, E. Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol Aging. (2003) 24:197–211. doi: 10.1016/s0197-4580(02)00065-9

PubMed Abstract | Crossref Full Text | Google Scholar

27. Song, S, Jiang, L, Oyarzabal, EA, Wilson, B, Li, Z, Shih, YI, et al. Loss of brain norepinephrine elicits neuroinflammation-mediated oxidative injury and selective caudo-rostral neurodegeneration. Mol Neurobiol. (2019) 56:2653–69. doi: 10.1007/s12035-018-1235-1

PubMed Abstract | Crossref Full Text | Google Scholar

28. Butkovich, LM, Houser, MC, Chalermpalanupap, T, Porter-Stransky, KA, Iannitelli, AF, Boles, JS, et al. Transgenic mice expressing human α-Synuclein in noradrenergic neurons develop locus Ceruleus pathology and nonmotor features of Parkinson's disease. J Neurosci. (2020) 40:7559–76. doi: 10.1523/JNEUROSCI.1468-19.2020

PubMed Abstract | Crossref Full Text | Google Scholar

29. van Dijk, KD, Berendse, HW, Drukarch, B, Fratantoni, SA, Pham, TV, Piersma, SR, et al. The proteome of the locus ceruleus in Parkinson's disease: relevance to pathogenesis. Brain Pathol (Zurich, Switzerland). (2012) 22:485–98. doi: 10.1111/j.1750-3639.2011.00540.x

PubMed Abstract | Crossref Full Text | Google Scholar

30. Choi, GE, Lee, HJ, Chae, CW, Cho, JH, Jung, YH, Kim, JS, et al. BNIP3L/NIX-mediated mitophagy protects against glucocorticoid-induced synapse defects. Nat Commun. (2021) 12:487. doi: 10.1038/s41467-020-20679-y

PubMed Abstract | Crossref Full Text | Google Scholar

31. Zhang, S, Cheon, M, Park, H, Kim, T, and Chung, C. Interaction between glucocorticoid receptors and FKBP5 in regulating neurotransmission of the Hippocampus. Neuroscience. (2022) 483:95–103. doi: 10.1016/j.neuroscience.2021.12.020

PubMed Abstract | Crossref Full Text | Google Scholar

32. Maatouk, L, Compagnion, AC, Sauvage, MC, Bemelmans, AP, Leclere-Turbant, S, Cirotteau, V, et al. TLR9 activation via microglial glucocorticoid receptors contributes to degeneration of midbrain dopamine neurons. Nat Commun. (2018) 9:2450. doi: 10.1038/s41467-018-04569-y

Crossref Full Text | Google Scholar

33. Maatouk, L, Yi, C, Carrillo-de Sauvage, MA, Compagnion, AC, Hunot, S, Ezan, P, et al. Glucocorticoid receptor in astrocytes regulates midbrain dopamine neurodegeneration through connexin hemichannel activity. Cell Death Differ. (2019) 26:580–96. doi: 10.1038/s41418-018-0150-3

PubMed Abstract | Crossref Full Text | Google Scholar

34. Barnett, JA, Bandy, ML, and Gibson, DL. Is the use of glyphosate in modern agriculture resulting in increased neuropsychiatric conditions through modulation of the gut-brain-microbiome Axis? Front Nutr. (2022) 9:827384. doi: 10.3389/fnut.2022.827384

PubMed Abstract | Crossref Full Text | Google Scholar

35. Niesler, B, Kuerten, S, Demir, IE, and Schäfer, KH. Disorders of the enteric nervous system - a holistic view. Nat Rev Gastroenterol Hepatol. (2021) 18:393–410. doi: 10.1038/s41575-020-00385-2

Crossref Full Text | Google Scholar

36. Claros, S, Cabrera, P, Valverde, N, Romero-Zerbo, SY, López-González, MV, Shumilov, K, et al. Insulin-like growth factor II prevents MPP+ and glucocorticoid mitochondrial-oxidative and neuronal damage in dopaminergic neurons. Antioxidants (Basel, Switzerland). (2021) 11:41. doi: 10.3390/antiox11010041

Crossref Full Text | Google Scholar

37. Ham, S, Lee, YI, Jo, M, Kim, H, Kang, H, Jo, A, et al. Hydrocortisone-induced parkin prevents dopaminergic cell death via CREB pathway in Parkinson's disease model. Sci Rep. (2017) 7:525. doi: 10.1038/s41598-017-00614-w

PubMed Abstract | Crossref Full Text | Google Scholar

38. Wakabayashi, K, Takahashi, H, Ohama, E, and Ikuta, F. Parkinson's disease: an immunohistochemical study of Lewy body-containing neurons in the enteric nervous system. Acta Neuropathol. (1990) 79:581–3. doi: 10.1007/BF00294234

PubMed Abstract | Crossref Full Text | Google Scholar

39. Rietdijk, CD, Perez-Pardo, P, Garssen, J, van Wezel, RJ, and Kraneveld, AD. Exploring Braak's hypothesis of Parkinson's disease. Front Neurol. (2017) 8:37. doi: 10.3389/fneur.2017.00037

PubMed Abstract | Crossref Full Text | Google Scholar

40. Shannon, KM, Keshavarzian, A, Dodiya, HB, Jakate, S, and Kordower, JH. Is alpha-synuclein in the colon a biomarker for premotor Parkinson's disease? Evidence from 3 cases. Move Disord. (2012) 27:716–9. doi: 10.1002/mds.25020

PubMed Abstract | Crossref Full Text | Google Scholar

41. Forsyth, CB, Shannon, KM, Kordower, JH, Voigt, RM, Shaikh, M, Jaglin, JA, et al. Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson's disease. PLoS One. (2011) 6:e28032. doi: 10.1371/journal.pone.0028032

PubMed Abstract | Crossref Full Text | Google Scholar

42. Ueki, A, and Otsuka, M. Life style risks of Parkinson's disease: association between decreased water intake and constipation. J Neurol. (2004) 251:vII18–23. doi: 10.1007/s00415-004-1706-3

Crossref Full Text | Google Scholar

43. Siddiqui, MF, Rast, S, Lynn, MJ, Auchus, AP, and Pfeiffer, RF. Autonomic dysfunction in Parkinson's disease: a comprehensive symptom survey. Parkinsonism Relat Disord. (2002) 8:277–84. doi: 10.1016/S1353-8020(01)00052-9

PubMed Abstract | Crossref Full Text | Google Scholar

44. Adams-Carr, KL, Bestwick, JP, Shribman, S, Lees, A, Schrag, A, and Noyce, AJ. Constipation preceding Parkinson's disease: a systematic review and meta-analysis. J Neurol Neurosurg Psychiatry. (2016) 87:710–6. doi: 10.1136/jnnp-2015-311680

PubMed Abstract | Crossref Full Text | Google Scholar

45. Camacho, M, Greenland, JC, Daruwalla, C, Scott, KM, Patel, B, Apostolopoulos, D, et al. The profile of gastrointestinal dysfunction in prodromal to late-stage Parkinson's disease. NPJ Parkinson’s Dis. (2025) 11:123. doi: 10.1038/s41531-025-00900-9

PubMed Abstract | Crossref Full Text | Google Scholar

46. Bissacco, J, Bovenzi, R, Conti, M, Simonetta, C, Mascioli, D, Cerroni, R, et al. Gastrointestinal dysfunction bears on the clinical-biological profile of Parkinson's disease. Move Disord Clin Pract. (2025) 12:497–503. doi: 10.1002/mdc3.14319

PubMed Abstract | Crossref Full Text | Google Scholar

47. Nowak, JM, Antoniak, A, Kopczyński, M, Zając, W, Sadowski, K, Milanowski, Ł, et al. Validation of polish version of gastrointestinal dysfunction scale for Parkinson's disease. Neurol Neurochir Pol. (2024) 58:338–46. doi: 10.5603/pjnns.98275

PubMed Abstract | Crossref Full Text | Google Scholar

48. Lesser, GT. Frequency of bowel movements and future risk of Parkinson's disease. Neurology. (2002) 58:838; author reply -9–9. doi: 10.1212/WNL.58.5.838-a

PubMed Abstract | Crossref Full Text | Google Scholar

49. Shi, J, Wang, Y, Chen, D, Xu, X, Li, W, Li, K, et al. The alteration of intestinal mucosal α-synuclein expression and mucosal microbiota in Parkinson's disease. Appl Microbiol Biotechnol. (2023) 107:1917–29. doi: 10.1007/s00253-023-12410-w

PubMed Abstract | Crossref Full Text | Google Scholar

50. Huang, B, Chau, SWH, Liu, Y, Chan, JWY, Wang, J, Ma, SL, et al. Gut microbiome dysbiosis across early Parkinson's disease, REM sleep behavior disorder and their first-degree relatives. Nat Commun. (2023) 14:2501. doi: 10.1038/s41467-023-38248-4

PubMed Abstract | Crossref Full Text | Google Scholar

51. Kelly, LP, Carvey, PM, Keshavarzian, A, Shannon, KM, Shaikh, M, Bakay, RA, et al. Progression of intestinal permeability changes and alpha-synuclein expression in a mouse model of Parkinson's disease. Mov Disord. (2014) 29:999–1009. doi: 10.1002/mds.25736

PubMed Abstract | Crossref Full Text | Google Scholar

52. Zhang, K, Paul, KC, Jacobs, JP, Chou, HL, Duarte Folle, A, Del Rosario, I, et al. Parkinson's disease and the gut microbiome in rural California. J Parkinsons Dis. (2022) 12:2441–52. doi: 10.3233/JPD-223500

PubMed Abstract | Crossref Full Text | Google Scholar

53. Tran, SM, and Mohajeri, MH. The role of gut bacterial metabolites in brain development, aging and disease. Nutrients. (2021) 13:732. doi: 10.3390/nu13030732

PubMed Abstract | Crossref Full Text | Google Scholar

54. Wang, RX, Lee, JS, Campbell, EL, and Colgan, SP. Microbiota-derived butyrate dynamically regulates intestinal homeostasis through regulation of actin-associated protein synaptopodin. Proc Natl Acad Sci USA. (2020) 117:11648–57. doi: 10.1073/pnas.1917597117

PubMed Abstract | Crossref Full Text | Google Scholar

55. Ghosh, SS, Wang, J, Yannie, PJ, and Ghosh, S. Intestinal barrier dysfunction, LPS translocation, and disease development. J Endocr Soc. (2020) 4:bvz039. doi: 10.1210/jendso/bvz039

PubMed Abstract | Crossref Full Text | Google Scholar

56. ISCD, v, and P, D. The intestinal barrier in Parkinson's disease: current state of knowledge. J Parkinsons Dis. (2019) 9:S323–9. doi: 10.3233/JPD-191707

Crossref Full Text | Google Scholar

57. Kim, C, Lv, G, Lee, JS, Jung, BC, Masuda-Suzukake, M, Hong, CS, et al. Exposure to bacterial endotoxin generates a distinct strain of α-synuclein fibril. Sci Rep. (2016) 6:30891. doi: 10.1038/srep30891

PubMed Abstract | Crossref Full Text | Google Scholar

58. Carregosa, D, Carecho, R, Figueira, I, and Santos, CN. Low-molecular weight metabolites from polyphenols as effectors for attenuating neuroinflammation. J Agric Food Chem. (2020) 68:1790–807. doi: 10.1021/acs.jafc.9b02155

Crossref Full Text | Google Scholar

59. Carregosa, D, Pinto, C, Ávila-Gálvez, M, Bastos, P, Berry, D, and Santos, CN. A look beyond dietary (poly)phenols: the low molecular weight phenolic metabolites and their concentrations in human circulation. Compr Rev Food Sci Food Saf. (2022) 21:3931–62. doi: 10.1111/1541-4337.13006

PubMed Abstract | Crossref Full Text | Google Scholar

60. Figueira, I, Garcia, G, Pimpão, RC, Terrasso, AP, Costa, I, Almeida, AF, et al. Polyphenols journey through blood-brain barrier towards neuronal protection. Sci Rep. (2017) 7:11456. doi: 10.1038/s41598-017-11512-6

PubMed Abstract | Crossref Full Text | Google Scholar

61. Carecho, R, Marques, D, Carregosa, D, Masuero, D, Garcia-Aloy, M, Tramer, F, et al. Circulating low-molecular-weight (poly)phenol metabolites in the brain: unveiling in vitro and in vivo blood-brain barrier transport. Food Funct. (2024) 15:7812–27. doi: 10.1039/D4FO01396D

PubMed Abstract | Crossref Full Text | Google Scholar

62. Le Sayec, M, Carregosa, D, Khalifa, K, de Lucia, C, Aarsland, D, Santos, CN, et al. Identification and quantification of (poly)phenol and methylxanthine metabolites in human cerebrospinal fluid: evidence of their ability to cross the BBB. Food Funct. (2023) 14:8893–902. doi: 10.1039/D3FO01913F

PubMed Abstract | Crossref Full Text | Google Scholar

63. Roodveldt, C, Bernardino, L, Oztop-Cakmak, O, Dragic, M, Fladmark, KE, Ertan, S, et al. The immune system in Parkinson's disease: what we know so far. Brain. (2024) 147:3306–24. doi: 10.1093/brain/awae177

PubMed Abstract | Crossref Full Text | Google Scholar

64. Braak, H, Rüb, U, Gai, WP, and Del Tredici, K. Idiopathic Parkinson's disease: possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J Neural Transm (Vienna, Austria: 1996). (2003) 110:517–36. doi: 10.1007/s00702-002-0808-2

PubMed Abstract | Crossref Full Text | Google Scholar

65. McGeer, PL, Itagaki, S, Boyes, BE, and McGeer, EG. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson's and Alzheimer's disease brains. Neurology. (1988) 38:1285–91. doi: 10.1212/wnl.38.8.1285

PubMed Abstract | Crossref Full Text | Google Scholar

66. Damier, P, Hirsch, EC, Zhang, P, Agid, Y, and Javoy-Agid, F. Glutathione peroxidase, glial cells and Parkinson's disease. Neuroscience. (1993) 52:1–6. doi: 10.1016/0306-4522(93)90175-f

PubMed Abstract | Crossref Full Text | Google Scholar

67. Ricardo, C, A, MF, T, D, E-B, RS, M, L, G, J, et al. Astrocytes role in Parkinson: A double-edged sword In: U K, editor. Neurodegenerative diseases. Rijeka: IntechOpen (2013). Ch. 20.

Google Scholar

68. Mogi, M, Harada, M, Riederer, P, Narabayashi, H, Fujita, K, and Nagatsu, T. Tumor necrosis factor-alpha (TNF-alpha) increases both in the brain and in the cerebrospinal fluid from parkinsonian patients. Neurosci Lett. (1994) 165:208–10. doi: 10.1016/0304-3940(94)90746-3

PubMed Abstract | Crossref Full Text | Google Scholar

69. Blum-Degen, D, Müller, T, Kuhn, W, Gerlach, M, Przuntek, H, and Riederer, P. Interleukin-1 beta and interleukin-6 are elevated in the cerebrospinal fluid of Alzheimer's and de novo Parkinson's disease patients. Neurosci Lett. (1995) 202:17–20. doi: 10.1016/0304-3940(95)12192-7

PubMed Abstract | Crossref Full Text | Google Scholar

70. Rowe, DB, Le, W, Smith, RG, and Appel, SH. Antibodies from patients with Parkinson's disease react with protein modified by dopamine oxidation. J Neurosci Res. (1998) 53:551–8. doi: 10.1002/(SICI)1097-4547(19980901)53:5<551::AID-JNR5>3.0.CO;2-8

PubMed Abstract | Crossref Full Text | Google Scholar

71. Fiszer, U, Mix, E, Fredrikson, S, Kostulas, V, Olsson, T, and Link, H. Gamma delta+ T cells are increased in patients with Parkinson's disease. J Neurol Sci. (1994) 121:39–45. doi: 10.1016/0022-510x(94)90154-6

PubMed Abstract | Crossref Full Text | Google Scholar

72. Ghosh, S, Whitley, CS, Haribabu, B, and Jala, VR. Regulation of intestinal barrier function by microbial metabolites. Cell Mol Gastroenterol Hepatol. (2021) 11:1463–82. doi: 10.1016/j.jcmgh.2021.02.007

PubMed Abstract | Crossref Full Text | Google Scholar

73. Greenland, JC, Holbrook, J, Kahanawita, L, Camacho, M, Fryer, TD, Hong, YT, et al. Peripheral-central immune crosstalk in Parkinson's disease and its association with clinical severity. Brain Behav Immun. (2025) 128:558–70. doi: 10.1016/j.bbi.2025.04.028

PubMed Abstract | Crossref Full Text | Google Scholar

74. Breit, S, Kupferberg, A, Rogler, G, and Hasler, G. Vagus nerve as modulator of the brain-gut Axis in psychiatric and inflammatory disorders. Front Psych. (2018) 9:44. doi: 10.3389/fpsyt.2018.00044

PubMed Abstract | Crossref Full Text | Google Scholar

75. Kim, S, Kwon, SH, Kam, TI, Panicker, N, Karuppagounder, SS, Lee, S, et al. Transneuronal propagation of pathologic α-Synuclein from the gut to the brain models Parkinson's disease. Neuron. (2019) 103:627–41.e7. doi: 10.1016/j.neuron.2019.05.035

PubMed Abstract | Crossref Full Text | Google Scholar

76. Yang, Y, Yang, LY, Orban, L, Cuylear, D, Thompson, J, Simon, B, et al. Non-invasive vagus nerve stimulation reduces blood-brain barrier disruption in a rat model of ischemic stroke. Brain Stimul. (2018) 11:689–98. doi: 10.1016/j.brs.2018.01.034

PubMed Abstract | Crossref Full Text | Google Scholar

77. Guo, Chunlei (2023). Efficacy and brain mechanism of percutaneous auricular vagus nerve stimulation for mild cognitive impairment: an fMRI study. [Master's Thesis].

Google Scholar

78. Zhang, H, Shan, AD, Wan, CH, Cao, XY, Yuan, YS, Ye, SY, et al. Transcutaneous auricular vagus nerve stimulation improves anxiety symptoms and cortical activity during verbal fluency task in Parkinson's disease with anxiety. J Affect Disord. (2024) 361:556–63. doi: 10.1016/j.jad.2024.06.083

PubMed Abstract | Crossref Full Text | Google Scholar

79. Zhang, Y, Huang, Y, Li, H, Yan, Z, Zhang, Y, Liu, X, et al. Transcutaneous auricular vagus nerve stimulation (taVNS) for migraine: an fMRI study. Reg Anesth Pain Med. (2021) 46:145–50. doi: 10.1136/rapm-2020-102088

PubMed Abstract | Crossref Full Text | Google Scholar

80. Wang, C, Su, T, Xiao, L, Wang, Y, Huo, X, Li, W, et al. Right vagus nerve stimulation improves motor behavior by exerting neuroprotective effects in Parkinson's disease rats. Ann Transl Med. (2022) 10:1314. doi: 10.21037/atm-22-5366

PubMed Abstract | Crossref Full Text | Google Scholar

81. Brown, P, Oliviero, A, Mazzone, P, Insola, A, Tonali, P, and Di Lazzaro, V. Dopamine dependency of oscillations between subthalamic nucleus and pallidum in Parkinson's disease. J Neurosci. (2001) 21:1033–8. doi: 10.1523/JNEUROSCI.21-03-01033.2001

PubMed Abstract | Crossref Full Text | Google Scholar

82. Marano, M, Anzini, G, Saltarocchi, L, Ricciuti, R, Capone, F, Tan, H, et al. Left Vagus stimulation modulates contralateral subthalamic β power improving the gait in Parkinson's disease. Move Disord. (2024) 39:424–8. doi: 10.1002/mds.29690

PubMed Abstract | Crossref Full Text | Google Scholar

83. Farrand, AQ, Helke, KL, Aponte-Cofresí, L, Gooz, MB, Gregory, RA, Hinson, VK, et al. Effects of vagus nerve stimulation are mediated in part by TrkB in a parkinson's disease model. Behav Brain Res. (2019) 373:112080. doi: 10.1016/j.bbr.2019.112080

PubMed Abstract | Crossref Full Text | Google Scholar

84. Forsythe, P, Bienenstock, J, and Kunze, WA. Vagal pathways for microbiome-brain-gut axis communication. Adv Exp Med Biol. (2014) 817:115–33. doi: 10.1007/978-1-4939-0897-4_5

PubMed Abstract | Crossref Full Text | Google Scholar

85. Levy, G, Fishman, JE, Xu, DZ, Dong, W, Palange, D, Vida, G, et al. Vagal nerve stimulation modulates gut injury and lung permeability in trauma-hemorrhagic shock. J Trauma Acute Care Surg (2012); 73: 338–42; discussion 342. doi: 10.1097/TA.0b013e31825debd3

Crossref Full Text | Google Scholar

86. Wang, Y, Tan, Q, Pan, M, Yu, J, Wu, S, Tu, W, et al. Minimally invasive vagus nerve stimulation modulates mast cell degranulation via the microbiota-gut-brain axis to ameliorate blood-brain barrier and intestinal barrier damage following ischemic stroke. Int Immunopharmacol. (2024) 132:112030. doi: 10.1016/j.intimp.2024.112030

PubMed Abstract | Crossref Full Text | Google Scholar

87. Dirr, EW, Jiracek, LG, Baekey, DM, C, JM, Otto, KJ, and Zubcevic, J. Subdiaphragmatic vagal nerve stimulation attenuates the development of hypertension and alters nucleus of the solitary tract transcriptional networks in the spontaneously hypertensive rat. Physiol Genomics. (2023) 55:606–17. doi: 10.1152/physiolgenomics.00016.2023

Crossref Full Text | Google Scholar

88. Zhang, J, Ma, L, Chang, L, Pu, Y, Qu, Y, and Hashimoto, K. A key role of the subdiaphragmatic vagus nerve in the depression-like phenotype and abnormal composition of gut microbiota in mice after lipopolysaccharide administration. Transl Psychiatry. (2020) 10:186. doi: 10.1038/s41398-020-00878-3

PubMed Abstract | Crossref Full Text | Google Scholar

89. Liu, J, Dai, Q, Qu, T, Ma, J, Lv, C, Wang, H, et al. Ameliorating effects of transcutaneous auricular vagus nerve stimulation on a mouse model of constipation-predominant irritable bowel syndrome. Neurobiol Dis. (2024) 193:106440. doi: 10.1016/j.nbd.2024.106440

PubMed Abstract | Crossref Full Text | Google Scholar

90. Bora, G, Atkinson, SN, Pan, A, Sood, M, Salzman, N, and Karrento, K. Impact of auricular percutaneous electrical nerve field stimulation on gut microbiome in adolescents with irritable bowel syndrome: A pilot study. J Dig Dis. (2023) 24:348–58. doi: 10.1111/1751-2980.13203

PubMed Abstract | Crossref Full Text | Google Scholar

91. Soares, NM, Pereira, GM, Altmann, V, de Almeida, RMM, and Rieder, CRM. Cortisol levels, motor, cognitive and behavioral symptoms in Parkinson's disease: a systematic review. J Neural Transm (Vienna, Austria: 1996). (2019) 126:219–32. doi: 10.1007/s00702-018-1947-4

PubMed Abstract | Crossref Full Text | Google Scholar

92. van Wamelen, DJ, Wan, YM, Ray Chaudhuri, K, and Jenner, P. Stress and cortisol in Parkinson's disease. Int Rev Neurobiol. (2020) 152:131–56. doi: 10.1016/bs.irn.2020.01.005

PubMed Abstract | Crossref Full Text | Google Scholar

93. Thrivikraman, KV, Zejnelovic, F, Bonsall, RW, and Owens, MJ. Neuroendocrine homeostasis after vagus nerve stimulation in rats. Psychoneuroendocrinology. (2013) 38:1067–77. doi: 10.1016/j.psyneuen.2012.10.015

PubMed Abstract | Crossref Full Text | Google Scholar

94. Keller, BN, Snyder, AE, Coker, CR, Aguilar, EA, O'Brien, MK, Lookfong, NA, et al. Vagus nerve damage increases alcohol intake and preference in a nonpreferring rat line: relationship to vagal regulation of the hypothalamic-pituitary-adrenal axis. Alcohol Clin Exp Res. (2024) 48:488–98. doi: 10.1111/acer.15264

PubMed Abstract | Crossref Full Text | Google Scholar

95. O'Keane, V, Dinan, TG, Scott, L, and Corcoran, C. Changes in hypothalamic-pituitary-adrenal axis measures after vagus nerve stimulation therapy in chronic depression. Biol Psychiatry. (2005) 58:963–8. doi: 10.1016/j.biopsych.2005.04.049

PubMed Abstract | Crossref Full Text | Google Scholar

96. Ghia, JE, Blennerhassett, P, Kumar-Ondiveeran, H, Verdu, EF, and Collins, SM. The vagus nerve: a tonic inhibitory influence associated with inflammatory bowel disease in a murine model. Gastroenterology. (2006) 131:1122–30. doi: 10.1053/j.gastro.2006.08.016

PubMed Abstract | Crossref Full Text | Google Scholar

97. Kaniusas, E, Szeles, JC, Kampusch, S, Alfageme-Lopez, N, Yucuma-Conde, D, Li, X, et al. Non-invasive auricular Vagus nerve stimulation as a potential treatment for Covid19-originated acute respiratory distress syndrome. Front Physiol. (2020) 11:890. doi: 10.3389/fphys.2020.00890

PubMed Abstract | Crossref Full Text | Google Scholar

98. Bonaz, B, Sinniger, V, and Pellissier, S. Vagus nerve stimulation: a new promising therapeutic tool in inflammatory bowel disease. J Intern Med. (2017) 282:46–63. doi: 10.1111/joim.12611

PubMed Abstract | Crossref Full Text | Google Scholar

99. Wang, H, Yu, M, Ochani, M, Amella, CA, Tanovic, M, Susarla, S, et al. Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature. (2003) 421:384–8. doi: 10.1038/nature01339

PubMed Abstract | Crossref Full Text | Google Scholar

100. Liu, C. Targeting the cholinergic system in Parkinson's disease. Acta Pharmacol Sin. (2020) 41:453–63. doi: 10.1038/s41401-020-0380-z

PubMed Abstract | Crossref Full Text | Google Scholar

101. Farrand, AQ, Helke, KL, Gregory, RA, Gooz, M, Hinson, VK, and Boger, HA. Vagus nerve stimulation improves locomotion and neuronal populations in a model of Parkinson's disease. Brain Stimul. (2017) 10:1045–54. doi: 10.1016/j.brs.2017.08.008

PubMed Abstract | Crossref Full Text | Google Scholar

102. Jiang, Y, Cao, Z, Ma, H, Wang, G, Wang, X, Wang, Z, et al. Auricular Vagus nerve stimulation exerts Antiinflammatory effects and immune regulatory function in a 6-OHDA model of Parkinson's disease. Neurochem Res. (2018) 43:2155–64. doi: 10.1007/s11064-018-2639-z

PubMed Abstract | Crossref Full Text | Google Scholar

103. Kaniusas, E, Kampusch, S, Tittgemeyer, M, Panetsos, F, Gines, RF, Papa, M, et al. Current directions in the auricular Vagus nerve stimulation I - A physiological perspective. Front Neurosci. (2019) 13:854. doi: 10.3389/fnins.2019.00854

PubMed Abstract | Crossref Full Text | Google Scholar

104. Rosas-Ballina, M, Ochani, M, Parrish, WR, Ochani, K, Harris, YT, Huston, JM, et al. Splenic nerve is required for cholinergic antiinflammatory pathway control of TNF in endotoxemia. Proc Natl Acad Sci USA. (2008) 105:11008–13. doi: 10.1073/pnas.0803237105

PubMed Abstract | Crossref Full Text | Google Scholar

105. Chen, Y, Wang, J, Zhang, Y, Zhang, Z, Wang, Y, and Rong, P. Effects of percutaneous auricular vagus nerve stimulation on the α7-nAchR/JAK2/STAT3 signaling pathway in lipopolysaccharide-induced depression-like behavior in rat spleen. J Acupunct Moxibust. (2023) 48:933–8. doi: 10.13702/j.1000-0607.20220402

Crossref Full Text | Google Scholar

106. Bratton, BO, Martelli, D, McKinley, MJ, Trevaks, D, Anderson, CR, and McAllen, RM. Neural regulation of inflammation: no neural connection from the vagus to splenic sympathetic neurons. Exp Physiol. (2012) 97:1180–5. doi: 10.1113/expphysiol.2011.061531

PubMed Abstract | Crossref Full Text | Google Scholar

107. Martelli, D, McKinley, MJ, and McAllen, RM. The cholinergic anti-inflammatory pathway: a critical review. Auton Neurosci. (2014) 182:65–9. doi: 10.1016/j.autneu.2013.12.007

PubMed Abstract | Crossref Full Text | Google Scholar

108. Rosas-Ballina, M, Olofsson, PS, Ochani, M, Valdés-Ferrer, SI, Levine, YA, Reardon, C, et al. Acetylcholine-synthesizing T cells relay neural signals in a vagus nerve circuit. Science (New York, NY). (2011) 334:98–101. doi: 10.1126/science.1209985

PubMed Abstract | Crossref Full Text | Google Scholar

109. Hirsch, EC, and Hunot, S. Neuroinflammation in Parkinson's disease: a target for neuroprotection? Lancet Neurol. (2009) 8:382–97. doi: 10.1016/S1474-4422(09)70062-6

PubMed Abstract | Crossref Full Text | Google Scholar

110. Kin, I, Sasaki, T, Yasuhara, T, Kameda, M, Agari, T, Okazaki, M, et al. Vagus nerve stimulation with mild stimulation intensity exerts anti-inflammatory and neuroprotective effects in Parkinson's disease model rats. Biomedicine. (2021) 9:789. doi: 10.3390/biomedicines9070789

PubMed Abstract | Crossref Full Text | Google Scholar

111. Hosomoto, K, Sasaki, T, Yasuhara, T, Kameda, M, Sasada, S, Kin, I, et al. Continuous vagus nerve stimulation exerts beneficial effects on rats with experimentally induced Parkinson's disease: evidence suggesting involvement of a vagal afferent pathway. Brain Stimul. (2023) 16:594–603. doi: 10.1016/j.brs.2023.03.003

PubMed Abstract | Crossref Full Text | Google Scholar

112. Lanska, DJ. J.L. corning and vagal nerve stimulation for seizures in the 1880s. Neurology. (2002) 58:452–9. doi: 10.1212/wnl.58.3.452

PubMed Abstract | Crossref Full Text | Google Scholar

113. Morris, GL 3rd, Gloss, D, Buchhalter, J, Mack, KJ, Nickels, K, and Harden, C. Evidence-based guideline update: vagus nerve stimulation for the treatment of epilepsy: report of the guideline development subcommittee of the American Academy of Neurology. Neurology. (2013) 81:1453–9. doi: 10.1212/WNL.0b013e3182a393d1

PubMed Abstract | Crossref Full Text | Google Scholar

114. DeGiorgio, CM, Schachter, SC, Handforth, A, Salinsky, M, Thompson, J, Uthman, B, et al. Prospective long-term study of vagus nerve stimulation for the treatment of refractory seizures. Epilepsia. (2000) 41:1195–200. doi: 10.1111/j.1528-1157.2000.tb00325.x

Crossref Full Text | Google Scholar

115. Howland, RH. Vagus nerve stimulation. Curr Behav Neurosci Rep. (2014) 1:64–73. doi: 10.1007/s40473-014-0010-5

PubMed Abstract | Crossref Full Text | Google Scholar

116. Cheng, W, Fang, K, Ouyang, X, Jin, L, Song, Y, and Yu, B. Vagus nerve stimulation with a small total charge transfer improves motor behavior and reduces neuroinflammation in a mouse model of Parkinson's disease. Neurochem Int. (2024) 180:105871. doi: 10.1016/j.neuint.2024.105871

PubMed Abstract | Crossref Full Text | Google Scholar

117. Wang, Y, Li, SY, Wang, D, Wu, MZ, He, JK, Zhang, JL, et al. Transcutaneous auricular Vagus nerve stimulation: from concept to application. Neurosci Bull. (2021) 37:853–62. doi: 10.1007/s12264-020-00619-y

PubMed Abstract | Crossref Full Text | Google Scholar

118. Howland, RH. New developments with vagus nerve stimulation therapy. J Psychosoc Nurs Ment Health Serv. (2014) 52:11–4. doi: 10.3928/02793695-20140218-01

PubMed Abstract | Crossref Full Text | Google Scholar

119. Marano, M, Anzini, G, Musumeci, G, Magliozzi, A, Pozzilli, V, Capone, F, et al. Transcutaneous auricular Vagus stimulation improves gait and reaction time in Parkinson's disease. Move Disord. (2022) 37:2163–4. doi: 10.1002/mds.29166

PubMed Abstract | Crossref Full Text | Google Scholar

120. Grazzi, L, Usai, S, and Bussone, G. EHMTI-0036. Gammacore device for treatment of migraine attack: preliminary report. J Headache Pain. (2014) 15:G12. doi: 10.1186/1129-2377-15-S1-G12

Crossref Full Text | Google Scholar

121. Gaul, C, Diener, H, Solbach, K, Silver, N, Straube, A, Magis, D, et al. EHMTI-0364. Non-invasive vagus nerve stimulation using gammacore® for prevention and acute treatment of chronic cluster headache: report from the randomized phase of the preva study. J Headache Pain. (2014) 15:I7. doi: 10.1186/1129-2377-15-S1-I7

PubMed Abstract | Crossref Full Text | Google Scholar

122. Morris, R, Yarnall, AJ, Hunter, H, Taylor, JP, Baker, MR, and Rochester, L. Noninvasive vagus nerve stimulation to target gait impairment in Parkinson's disease. Move Disord. (2019) 34:918–9. doi: 10.1002/mds.27664

PubMed Abstract | Crossref Full Text | Google Scholar

123. Cook, DN, Thompson, S, Stomberg-Firestein, S, Bikson, M, George, MS, Jenkins, DD, et al. Design and validation of a closed-loop, motor-activated auricular vagus nerve stimulation (MAAVNS) system for neurorehabilitation. Brain Stimul. (2020) 13:800–3. doi: 10.1016/j.brs.2020.02.028

PubMed Abstract | Crossref Full Text | Google Scholar

124. Anzolin, A, Das, P, Garcia, RG, Chen, A, Grahl, A, Ellis, S, et al. Delta power during sleep is modulated by EEG-gated auricular vagal afferent nerve stimulation (EAVANS). Annual International Conference of the IEEE Engineering in Medicine and Biology Society IEEE Engineering in Medicine and Biology Society Annual International Conference. (2023); 2023: 1–4.

Google Scholar

125. Yap, JYY, Keatch, C, Lambert, E, Woods, W, Stoddart, PR, and Kameneva, T. Critical review of transcutaneous Vagus nerve stimulation: challenges for translation to clinical practice. Front Neurosci. (2020) 14:284. doi: 10.3389/fnins.2020.00284

PubMed Abstract | Crossref Full Text | Google Scholar

126. van Midden, V, Simončič, U, Pirtošek, Z, and Kojović, M. The effect of ta VNS at 25 Hz and 100 Hz on Parkinson's disease gait-a randomized motion sensor study. Mov Disord. (2024) 39:1375–85. doi: 10.1002/mds.29826

PubMed Abstract | Crossref Full Text | Google Scholar

127. Zhang, H, Cao, XY, Wang, LN, Tong, Q, Sun, HM, Gan, CT, et al. Transcutaneous auricular vagus nerve stimulation improves gait and cortical activity in Parkinson's disease: A pilot randomized study. CNS Neurosci Ther. (2023) 29:3889–900. doi: 10.1111/cns.14309

PubMed Abstract | Crossref Full Text | Google Scholar

128. Faraji, N, Payami, B, Ebadpour, N, and Gorji, A. Vagus nerve stimulation and gut microbiota interactions: a novel therapeutic avenue for neuropsychiatric disorders. Neurosci Biobehav Rev. (2025) 169:105990. doi: 10.1016/j.neubiorev.2024.105990

PubMed Abstract | Crossref Full Text | Google Scholar

129. Liu, L, Lou, S, Fu, D, Ji, P, Xia, P, Shuang, S, et al. Neuro-immune interactions: exploring the anti-inflammatory role of the vagus nerve. Int Immunopharmacol. (2025) 159:114941. doi: 10.1016/j.intimp.2025.114941

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: vagus nerve stimulation, Parkinson’s disease, microbiota-gut-brain axis, mechanism, intestinal flora

Citation: Yang L, Fan S, Sun L, Han J, Wang M and Yu T (2025) The role of vagus nerve stimulation in modulating Parkinson’s disease via the microbiota-gut-brain axis: a comprehensive review. Front. Neurol. 16:1643305. doi: 10.3389/fneur.2025.1643305

Received: 08 June 2025; Accepted: 20 August 2025;
Published: 22 September 2025.

Edited by:

Marcelo Mendonça, Champalimaud Foundation, Portugal

Reviewed by:

Inês Figueira, NOVA University of Lisbon, Portugal
Alina Sacarescu, Grigore T. Popa University of Medicine and Pharmacy, Romania

Copyright © 2025 Yang, Fan, Sun, Han, Wang and Yu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Meng Wang, dGp3YW5nbWVuZzE5ODhAMTYzLmNvbQ==; Tao Yu, ZG9jdG9yeXV0YW9AMTYzLmNvbQ==

These authors have contributed equally to this work and share first authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.