Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.136
Filter
1.
BMJ Open ; 14(5): e082906, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38772894

ABSTRACT

INTRODUCTION: With an increasing incidence and significant effects on patients, tinnitus has become a major disease burden. There is a dearth of therapies with established efficacy for tinnitus. Transcutaneous auricular vagus nerve stimulation (ta-VNS) is being investigated as a potential therapy for tinnitus, but the current body of evidence remains inconclusive due to conflicting results across different studies. As a result, this protocol aims to synthesise and update the evidence to clarify whether ta-VNS is effective and safe for alleviating tinnitus. METHODS AND ANALYSIS: To identify relevant randomised controlled trials (RCTs), seven representative bibliographical databases will be searched from their inception to December 2023: PubMed, Embase (via OVID), Cochrane Library, Chinese National Knowledge Infrastructure, Wangfang Database, Chinese BioMedical Literature Database, and Chongqing VIP Chinese Science and Technology Periodical Database. Publications in English or Chinese will be considered for inclusion. RCTs comparing ta-VNS with active treatments, no intervention, waitlist control or sham ta-VNS in adult patients with subjective tinnitus will be included. Studies on objective tinnitus will be excluded. Primary outcome is tinnitus symptom severity measured by validated scales. With all eligible trials included, when applicable, quantitative analysis via meta-analyses will be performed using RevMan V.5.4.1 software. Otherwise, a qualitative analysis will be conducted. The methodological quality of the included RCTs will be assessed using the Risk of Bias 2.0 tool. Sensitivity analyses, subgroup analysis and publication bias evaluation will also be performed. The Grading of Recommendations, Assessment, Development, and Evaluation approach will be used to grade the certainty of the evidence. ETHICS AND DISSEMINATION: Ethical approval is not required for this systematic review, as no primary data will be collected. The results will be reported and disseminated through publication in a peer-reviewed journal. PROSPERO REGISTRATION NUMBER: CRD42022351917.


Subject(s)
Meta-Analysis as Topic , Systematic Reviews as Topic , Tinnitus , Transcutaneous Electric Nerve Stimulation , Vagus Nerve Stimulation , Humans , Tinnitus/therapy , Vagus Nerve Stimulation/methods , Transcutaneous Electric Nerve Stimulation/methods , Randomized Controlled Trials as Topic , Research Design , Treatment Outcome
2.
CNS Neurosci Ther ; 30(5): e14757, 2024 May.
Article in English | MEDLINE | ID: mdl-38747078

ABSTRACT

BACKGROUND: With the improvement of emergency techniques, the survival rate of patients with severe brain injury has increased. However, this has also led to an annual increase in the number of patients with prolonged disorders of consciousness (pDoC). Hence, recovery of consciousness is an important part of treatment. With advancing techniques, noninvasive neuromodulation seems a promising intervention. The objective of this review was to summarize the latest techniques and provide the basis for protocols of noninvasive neuromodulations in pDoC. METHODS: This review summarized the advances in noninvasive neuromodulation in the treatment of pDoC in the last 5 years. RESULTS: Variable techniques of neuromodulation are used in pDoC. Transcranial ultrasonic stimulation (TUS) and transcutaneous auricular vagus nerve stimulation (taVNS) are very new techniques, while transcranial direct current stimulation (tDCS) and transcranial magnetic stimulation (TMS) are still the hotspots in pDoC. Median nerve electrical stimulation (MNS) has received little attention in the last 5 years. CONCLUSIONS: Noninvasive neuromodulation is a valuable and promising technique to treat pDoC. Further studies are needed to determine a unified stimulus protocol to achieve optimal effects as well as safety.


Subject(s)
Consciousness Disorders , Transcranial Direct Current Stimulation , Transcranial Magnetic Stimulation , Vagus Nerve Stimulation , Humans , Consciousness Disorders/therapy , Transcranial Magnetic Stimulation/methods , Transcranial Magnetic Stimulation/trends , Transcranial Direct Current Stimulation/methods , Vagus Nerve Stimulation/methods , Vagus Nerve Stimulation/trends , Transcutaneous Electric Nerve Stimulation/methods , Transcutaneous Electric Nerve Stimulation/trends
3.
CNS Neurosci Ther ; 30(5): e14755, 2024 May.
Article in English | MEDLINE | ID: mdl-38752512

ABSTRACT

BACKGROUND: Depression is a common psychiatric disorder in diabetic patients. Depressive mood associated with obesity/metabolic disorders is related to the inflammatory response caused by long-term consumption of high-fat diets, but its molecular mechanism is unclear. In this study, we investigated whether the antidepressant effect of transcutaneous auricular vagus nerve stimulation (taVNS) in high-fat diet rats works through the P2X7R/NLRP3/IL-1ß pathway. METHODS: We first used 16S rRNA gene sequencing analysis and LC-MS metabolomics assays in Zucker diabetic fatty (ZDF) rats with long-term high-fat diet (Purina #5008) induced significant depression-like behaviors. Next, the forced swimming test (FST) and open field test (OFT) were measured to evaluate the antidepressive effect of taVNS. Immunofluorescence and western blotting (WB) were used to measure the microglia state and the expression of P2X7R, NLRP3, and IL-1ß in PFC. RESULTS: Purina#5008 diet induced significant depression-like behaviors in ZDF rats and was closely related to purine and inflammatory metabolites. Consecutive taVNS increased plasma insulin concentration, reduced glycated hemoglobin and glucagon content in ZDF rats, significantly improved the depressive-like phenotype in ZDF rats through reducing the microglia activity, and increased the expression of P2X7R, NLRP3, and IL-1ß in the prefrontal cortex (PFC). CONCLUSION: The P2X7R/NLRP3/IL-1ß signaling pathway may play an important role in the antidepressant-like behavior of taVNS, which provides a promising mechanism for taVNS clinical treatment of diabetes combined with depression.


Subject(s)
Depression , Diet, High-Fat , Interleukin-1beta , NLR Family, Pyrin Domain-Containing 3 Protein , Prefrontal Cortex , Rats, Zucker , Receptors, Purinergic P2X7 , Vagus Nerve Stimulation , Animals , Prefrontal Cortex/metabolism , Diet, High-Fat/adverse effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Depression/metabolism , Depression/therapy , Depression/etiology , Male , Rats , Interleukin-1beta/metabolism , Vagus Nerve Stimulation/methods , Receptors, Purinergic P2X7/metabolism , Phenotype
4.
Sci Rep ; 14(1): 11224, 2024 05 16.
Article in English | MEDLINE | ID: mdl-38755234

ABSTRACT

The present study examined the effects of transcutaneous auricular vagus nerve stimulation (taVNS) on short-latency afferent inhibition (SAI), as indirect biomarker of cholinergic system activation. 24 healthy adults underwent intermittent taVNS (30 s on/30 s off, 30 min) or continuous taVNS at a frequency of 25 Hz (15 min) along with earlobe temporary stimulation (15 min or 30 min) were performed in random order. The efficiency with which the motor evoked potential from the abductor pollicis brevis muscle by transcranial magnetic stimulation was attenuated by the preceding median nerve conditioning stimulus was compared before taVNS, immediately after taVNS, and 15 min after taVNS. Continuous taVNS significantly increased SAI at 15 min post-stimulation compared to baseline. A positive correlation (Pearson coefficient = 0.563, p = 0.004) was observed between baseline SAI and changes after continuous taVNS. These results suggest that 15 min of continuous taVNS increases the activity of the cholinergic nervous system, as evidenced by the increase in SAI. In particular, the increase after taVNS was more pronounced in those with lower initial SAI. This study provides fundamental insight into the clinical potential of taVNS for cholinergic dysfunction.


Subject(s)
Transcutaneous Electric Nerve Stimulation , Vagus Nerve Stimulation , Humans , Male , Female , Adult , Vagus Nerve Stimulation/methods , Transcutaneous Electric Nerve Stimulation/methods , Young Adult , Evoked Potentials, Motor/physiology , Transcranial Magnetic Stimulation/methods , Vagus Nerve/physiology
5.
Sensors (Basel) ; 24(10)2024 May 16.
Article in English | MEDLINE | ID: mdl-38794024

ABSTRACT

An electroceutical is a medical device that uses electrical signals to control biological functions. It can be inserted into the human body as an implant and has several crucial advantages over conventional medicines for certain diseases. This research develops a new vagus nerve simulation (VNS) electroceutical through an innovative approach to overcome the communication limitations of existing devices. A phased array antenna with a better communication performance was developed and applied to the electroceutical prototype. In order to effectively respond to changes in communication signals, we developed the steering algorithm and firmware, and designed the smart communication protocol that operates at a low power that is safe for the patients. This protocol is intended to improve a communication sensitivity related to the transmission and reception distance. Based on this technical approach, the heightened effectiveness and safety of the prototype have been ascertained, with the actual clinical tests using live animals. We confirmed the signal attenuation performance to be excellent, and a smooth communication was achieved even at a distance of 7 m. The prototype showed a much wider communication range than any other existing products. Through this, it is conceivable that various problems due to space constraints can be resolved, hence presenting many benefits to the patients whose last resort to the disease is the VNS electroceutical.


Subject(s)
Algorithms , Vagus Nerve , Vagus Nerve/physiology , Animals , Humans , Prostheses and Implants , Vagus Nerve Stimulation/methods , Vagus Nerve Stimulation/instrumentation , Signal Processing, Computer-Assisted
6.
Medicina (Kaunas) ; 60(5)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38792911

ABSTRACT

Background and Objectives: This review systematically evaluates the potential of electrical neuromodulation techniques-vagus nerve stimulation (VNS), sacral nerve stimulation (SNS), and tibial nerve stimulation (TNS)-as alternative treatments for inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's Disease (CD). It aims to synthesize current evidence on the efficacy and safety of these modalities, addressing the significant burden of IBD on patient quality of life and the limitations of existing pharmacological therapies. Materials and Methods: We conducted a comprehensive analysis of studies from PubMed, focusing on research published between 1978 and 2024. The review included animal models and clinical trials investigating the mechanisms, effectiveness, and safety of VNS, SNS, and TNS in IBD management. Special attention was given to the modulation of inflammatory responses and its impact on gastrointestinal motility and functional gastrointestinal disorders associated with IBD. Results: Preliminary findings suggest that VNS, SNS, and TNS can significantly reduce inflammatory markers and improve symptoms in IBD patients. These techniques also show potential in treating related gastrointestinal disorders during IBD remission phases. However, the specific mechanisms underlying these benefits remain to be fully elucidated, and there is considerable variability in treatment parameters. Conclusions: Electrical neuromodulation holds promise as a novel therapeutic avenue for IBD, offering an alternative to patients who do not respond to traditional treatments or experience adverse effects. The review highlights the need for further rigorous studies to optimize stimulation parameters, understand long-term outcomes, and integrate neuromodulation effectively into IBD treatment protocols.


Subject(s)
Electric Stimulation Therapy , Inflammatory Bowel Diseases , Humans , Inflammatory Bowel Diseases/therapy , Inflammatory Bowel Diseases/physiopathology , Electric Stimulation Therapy/methods , Animals , Vagus Nerve Stimulation/methods , Tibial Nerve/physiology , Quality of Life
7.
Int J Mol Sci ; 25(8)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38673781

ABSTRACT

Vagus nerve stimulation (VNS) represents a long-term adjunctive treatment option in patients with difficult-to-treat depression (DTD). Anti-inflammatory effects have been discussed as a key mechanism of action of VNS. However, long-term investigations in real-world patients are sparse. In this naturalistic observational study, we collected data on cytokines in peripheral blood in n = 6 patients (mean age 47.8) with DTD and VNS treatment at baseline and at 6 months follow-up. We have identified clusters of peripheral cytokines with a similar dynamic over the course of these 6 months using hierarchical clustering. We have investigated cytokine changes from baseline to 6 months as well as the relationship between the cytokine profile at 6 months and long-term response at 12 months. After 6 months of VNS, we observed significant correlations between cytokines (p < 0.05) within the identified three cytokine-pairs which were not present at baseline: IL(interleukin)-6 and IL-8; IL-1ß and TNF-α; IFN-α2 and IL-33. At 6 months, the levels of all the cytokines of interest had decreased (increased in non-responders) and were lower (5-534 fold) in responders to VNS than in non-responders: however, these results were not statistically significant. VNS-associated immunomodulation might play a role in long-term clinical response to VNS.


Subject(s)
Cytokines , Vagus Nerve Stimulation , Humans , Cytokines/blood , Cytokines/metabolism , Male , Female , Middle Aged , Vagus Nerve Stimulation/methods , Adult , Depression/therapy , Depression/immunology , Treatment Outcome , Immunomodulation
8.
Brain Stimul ; 17(2): 460-468, 2024.
Article in English | MEDLINE | ID: mdl-38593972

ABSTRACT

BACKGROUND: Working memory is essential to a wide range of cognitive functions and activities. Transcutaneous auricular vagus nerve stimulation (taVNS) is a promising method to improve working memory performance. However, the feasibility and scalability of electrical stimulation are constrained by several limitations, such as auricular discomfort and inconsistent electrical contact. OBJECTIVE: We aimed to develop a novel and practical method, vibrotactile taVNS, to improve working memory. Further, we investigated its effects on arousal, measured by skin conductance and pupil diameter. METHOD: This study included 20 healthy participants. Behavioral response, skin conductance, and eye tracking data were concurrently recorded while the participants performed N-back tasks under three conditions: vibrotactile taVNS delivered to the cymba concha, earlobe (sham control), and no stimulation (baseline control). RESULTS: In 4-back tasks, which demand maximal working memory capacity, active vibrotactile taVNS significantly improved the performance metric d' compared to the baseline but not to the sham. Moreover, we found that the reduction rate of d' with increasing task difficulty was significantly smaller during vibrotactile taVNS sessions than in both baseline and sham conditions. Arousal, measured as skin conductance and pupil diameter, declined over the course of the tasks. Vibrotactile taVNS rescued this arousal decline, leading to arousal levels corresponding to optimal working memory levels. Moreover, pupil diameter and skin conductance level were higher during high-cognitive-load tasks when vibrotactile taVNS was delivered to the concha compared to baseline and sham. CONCLUSION: Our findings suggest that vibrotactile taVNS modulates the arousal pathway and could be a potential intervention for enhancing working memory.


Subject(s)
Memory, Short-Term , Humans , Memory, Short-Term/physiology , Male , Female , Adult , Young Adult , Vagus Nerve Stimulation/methods , Vibration , Pupil/physiology , Galvanic Skin Response/physiology , Vagus Nerve/physiology
9.
Int Immunopharmacol ; 132: 112030, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38603861

ABSTRACT

Mast cells (MCs) play a significant role in various diseases, and their activation and degranulation can trigger inflammatory responses and barrier damage. Several studies have indicated that vagus nerve stimulation (VNS) exerts ameliorates neurological injury, and regulates gut MC degranulation. However, there is limited research on the modulatory effect of VNS on MCs in both the gut and brain in brain ischemia-reperfusion (I/R) injury in this process. We aim to develop a minimally invasive, targeted and convenient VNS approach to assess the impact of VNS and to clarify the relationship between VNS and MCs on the prognosis of acute ischemic stroke. We utilized middle cerebral artery occlusion/reperfusion (MCAO/r) to induce brain I/R injury. After the experiment, the motor function and neurofunctional impairments of the rats were detected, and the gastrointestinal function, blood-brain barrier (BBB) and intestinal barrier damage, and systemic and local inflammation were evaluated by Nissl, TTC staining, Evans blue, immunofluorescence staining, transmission electron microscopy, western blot assays, ELISA, and fecal 16S rRNA sequencing methods. Our research confirmed that our minimally invasive VNS method is a novel approach for stimulating the vagus nerve. VNS alleviated motor deficits and gastrointestinal dysfunction while also suppressing intestinal and neuroinflammation. Additionally, VNS ameliorated gut microbiota dysbiosis in rats. Furthermore, our analysis indicated that VNS reduces chymase secretion by modulating MCs degranulation and improves intestinal and BBB damage. Our results showed that VNS treatment can alleviate the damage of BBB and colonic barrier after cerebral I/R by modulating mast cell degranulation, and alleviates systemic inflammatory responses.


Subject(s)
Blood-Brain Barrier , Brain-Gut Axis , Cell Degranulation , Gastrointestinal Microbiome , Ischemic Stroke , Mast Cells , Rats, Sprague-Dawley , Reperfusion Injury , Vagus Nerve Stimulation , Animals , Mast Cells/immunology , Vagus Nerve Stimulation/methods , Male , Rats , Reperfusion Injury/therapy , Reperfusion Injury/immunology , Ischemic Stroke/therapy , Brain-Gut Axis/physiology , Infarction, Middle Cerebral Artery/therapy , Disease Models, Animal , Brain Ischemia/therapy , Brain Ischemia/immunology
10.
Seizure ; 117: 298-304, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38615369

ABSTRACT

BACKGROUND: Right-sided vagus nerve stimulation (RS-VNS) is indicated when the procedure was deemed not technically feasible or too risky on the indicated left side. OBJECTIVE: The present study aims to systematically review the literature on RS-VNS, assessing its effectiveness and safety. METHODS: A systematic review following PRISMA guidelines was conducted: Pubmed/MEDLINE, The Cochrane Library, Scopus, Embase and Web of science databases were searched from inception to August 13th,2023. Gray literature was searched in two libraries. Eligible studies included all studies reporting, at least, one single case of RS-VNS in patients for the treatment of drug-resistant epilepsy. RESULTS: Out of 2333 initial results, 415 studies were screened by abstract. Only four were included in the final analysis comprising seven patients with RS-VNS for a drug-resistant epilepsy. One patient experienced nocturnal asymptomatic bradycardia whereas the other six patients did not display any cardiac symptom. RS-VNS was discontinued in one case due to exercise-induced airway disease exacerbation. Decrease of epileptic seizure frequency after RS-VNS ranged from 25 % to 100 % in six cases. In the remaining case, VNS effectiveness was unclear. In one case, RS-VNS was more efficient than left-sided VNS (69 % vs 50 %, respectively) whereas in another case, RS-VNS was less efficient (50 % vs 95 %, respectively). CONCLUSION: Literature on the present topic is limited. In six out of seven patients, RS-VNS for drug-resistant epilepsy displayed reasonable effectiveness with a low complication rate. Further research, including prospective studies, is necessary to assess safety and effectiveness of RS-VNS for drug-resistant epilepsy patients.


Subject(s)
Drug Resistant Epilepsy , Vagus Nerve Stimulation , Humans , Vagus Nerve Stimulation/methods , Drug Resistant Epilepsy/therapy
11.
Sci Adv ; 10(17): eadn3760, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38669336

ABSTRACT

Acetylcholine is produced in the spleen in response to vagus nerve activation; however, the effects on antibody production have been largely unexplored. Here, we use a chronic vagus nerve stimulation (VNS) mouse model to study the effect of VNS on T-dependent B cell responses. We observed lower titers of high-affinity IgG and fewer antigen-specific germinal center (GC) B cells. GC B cells from chronic VNS mice exhibited altered mRNA and protein expression suggesting increased apoptosis and impaired plasma cell differentiation. Follicular dendritic cell (FDC) cluster dispersal and altered gene expression suggested poor function. The absence of acetylcholine-producing CD4+ T cells diminished these alterations. In vitro studies revealed that α7 and α9 nicotinic acetylcholine receptors (nAChRs) directly regulated B cell production of TNF, a cytokine crucial to FDC clustering. α4 nAChR inhibited coligation of CD19 to the B cell receptor, presumably decreasing B cell survival. Thus, VNS-induced GC impairment can be attributed to distinct effects of nAChRs on B cells.


Subject(s)
B-Lymphocytes , Germinal Center , Receptors, Nicotinic , Vagus Nerve Stimulation , alpha7 Nicotinic Acetylcholine Receptor , Animals , Germinal Center/metabolism , Germinal Center/immunology , Vagus Nerve Stimulation/methods , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Mice , Receptors, Nicotinic/metabolism , Receptors, Nicotinic/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics , Dendritic Cells, Follicular/metabolism , Dendritic Cells, Follicular/immunology , Receptors, Cholinergic/metabolism , Receptors, Cholinergic/immunology , Receptors, Antigen, B-Cell/metabolism , Cell Differentiation , Mice, Inbred C57BL , Immunoglobulin G/immunology , Vagus Nerve/metabolism , Vagus Nerve/physiology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology
12.
Nat Rev Neurol ; 20(5): 259-268, 2024 05.
Article in English | MEDLINE | ID: mdl-38570705

ABSTRACT

Neurostimulation, the use of electrical stimulation to modulate the activity of the nervous system, is now commonly used for the treatment of chronic pain, movement disorders and epilepsy. Many neurostimulation techniques have now shown promise for the treatment of physical impairments in people with stroke. In 2021, vagus nerve stimulation was approved by the FDA as an adjunct to intensive rehabilitation therapy for the treatment of chronic upper extremity deficits after ischaemic stroke. In 2024, pharyngeal electrical stimulation was conditionally approved by the UK National Institute for Health and Care Excellence for neurogenic dysphagia in people with stroke who have a tracheostomy. Many other approaches have also been tested in pivotal device trials and a number of approaches are in early-phase study. Typically, neurostimulation techniques aim to increase neuroplasticity in response to training and rehabilitation, although the putative mechanisms of action differ and are not fully understood. Neurostimulation techniques offer a number of practical advantages for use after stroke, such as precise dosing and timing, but can be invasive and costly to implement. This Review focuses on neurostimulation techniques that are now in clinical use or that have reached the stage of pivotal trials and show considerable promise for the treatment of post-stroke impairments.


Subject(s)
Electric Stimulation Therapy , Stroke Rehabilitation , Stroke , Humans , Stroke/complications , Stroke/therapy , Electric Stimulation Therapy/methods , Stroke Rehabilitation/methods , Vagus Nerve Stimulation/methods
13.
Compr Psychiatry ; 132: 152488, 2024 07.
Article in English | MEDLINE | ID: mdl-38657358

ABSTRACT

BACKGROUND: Major depressive disorder (MDD) is often marked by impaired motivation and reward processing, known as anhedonia. Many patients do not respond to first-line treatments, and improvements in motivation can be slow, creating an urgent need for rapid interventions. Recently, we demonstrated that transcutaneous auricular vagus nerve stimulation (taVNS) acutely boosts effort invigoration in healthy participants, but its effects on depression remain unclear. OBJECTIVE: To assess the impact of taVNS on effort invigoration and maintenance in a sample that includes patients with MDD, evaluating the generalizability of our findings. METHODS: We used a single-blind, randomized crossover design in 30 patients with MDD and 29 matched (age, sex, and BMI) healthy control participants (HCP). RESULTS: Consistent with prior findings, taVNS increased effort invigoration for rewards in both groups during Session 1 (p = .040), particularly for less wanted rewards in HCP (pboot < 0.001). However, invigoration remained elevated in all participants, and no acute changes were observed in Session 2 (Δinvigoration = 3.3, p = .12). Crucially, throughout Session 1, we found taVNS-induced increases in effort invigoration (pboot = 0.008) and wanting (pboot = 0.010) in patients with MDD, with gains in wanting maintained across sessions (Δwanting = 0.06, p = .97). CONCLUSIONS: Our study replicates the invigorating effects of taVNS in Session 1 and reveals its generalizability to depression. Furthermore, we expand upon previous research by showing taVNS-induced conditioning effects on invigoration and wanting within Session 1 in patients that were largely sustained. While enduring motivational improvements present challenges for crossover designs, they are highly desirable in interventions and warrant further follow-up research.


Subject(s)
Cross-Over Studies , Depressive Disorder, Major , Motivation , Reward , Vagus Nerve Stimulation , Humans , Female , Male , Vagus Nerve Stimulation/methods , Depressive Disorder, Major/therapy , Depressive Disorder, Major/psychology , Adult , Single-Blind Method , Middle Aged , Anhedonia
14.
Acta Neurochir (Wien) ; 166(1): 193, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38662025

ABSTRACT

Vagal neuropathy causing vocal fold palsy is an uncommon complication of vagal nerve stimulator (VNS) placement. It may be associated with intraoperative nerve injury or with device stimulation. Here we present the first case of delayed, compressive vagal neuropathy associated with VNS coil placement which presented with progressive hoarseness and vocal cord paralysis. Coil removal and vagal neurolysis was performed to relieve the compression. Larger 3 mm VNS coils were placed for continuation of therapy. Coils with a larger inner diameter should be employed where possible to prevent this complication. The frequency of VNS-associated vagal nerve compression may warrant further investigation.


Subject(s)
Vagus Nerve Stimulation , Vocal Cord Paralysis , Humans , Male , Nerve Compression Syndromes/etiology , Nerve Compression Syndromes/surgery , Vagus Nerve , Vagus Nerve Diseases/etiology , Vagus Nerve Diseases/surgery , Vagus Nerve Stimulation/adverse effects , Vagus Nerve Stimulation/instrumentation , Vagus Nerve Stimulation/methods , Vocal Cord Paralysis/etiology , Aged
15.
Seizure ; 118: 65-70, 2024 May.
Article in English | MEDLINE | ID: mdl-38642446

ABSTRACT

INTRODUCTION: For patients with drug-resistant epilepsy (DRE) who are not suitable for surgical resection, neuromodulation with vagus nerve stimulation (VNS) is an established approach. However, there is limited evidence of seizure reduction when replacing traditional VNS (tVNS) device with a cardiac-based one (cbVNS). This meta-analysis compares the seizure reduction achieved by replacing tVNS with cbVNS in a population with DRE. METHODS: We systematically searched PubMed, Embase, and Cochrane Central following PRISMA guidelines. The main outcomes were number of patients experiencing a ≥ 50 % and ≥80 % reduction in seizures, as defined by the McHugh scale. Additionally, we assessed the number of patients achieving freedom from seizures. RESULTS: We included 178 patients with DRE from 7 studies who were initially treated with tVNS and subsequently had it replaced by cbVNS. The follow-up for cbVNS ranged from 6 to 37.5 months. There was a statistically significant reduction in seizure frequency with the replacement of tVNS by cbVNS, using a ≥ 50 % (OR 1.79; 95 % CI 1.07 to 2.97; I²=0 %; p = 0.03) and a ≥ 80 % (OR 2.06; 95 % CI 1.17 to 3.62; I²=0 %; p = 0.01) reduction threshold. Nineteen (13 %) participants achieved freedom from seizures after switching to cbVNS. There was no difference in the rate of freedom from seizures between groups (OR 1.85; 95 % CI 0.81 to 4.21; I²=0 %; p = 0.14). CONCLUSION: In patients with DRE undergoing battery replacement, cbVNS might be associated with seizure reduction (≥50 % and ≥80 % threshold) after switching from tVNS. Randomised controlled trials are necessary to validate these findings.


Subject(s)
Seizures , Vagus Nerve Stimulation , Humans , Vagus Nerve Stimulation/methods , Vagus Nerve Stimulation/instrumentation , Seizures/therapy , Drug Resistant Epilepsy/therapy
16.
BMJ Open ; 14(4): e082764, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38604630

ABSTRACT

INTRODUCTION: Poststroke cognitive impairment is a common complication in stroke survivors, seriously affecting their quality of life. Therefore, it is crucial to improve cognitive function of patients who had a stroke. Transcranial direct current stimulation (tDCS) and transcutaneous auricular vagus nerve stimulation (taVNS) are non-invasive, safe treatments with great potential to improve cognitive function in poststroke patients. However, further improvements are needed in the effectiveness of a single non-invasive brain stimulation technique for cognitive rehabilitation. This study protocol aims to investigate the effect and neural mechanism of the combination of tDCS and taVNS on cognitive function in patients who had a stroke. METHODS AND ANALYSIS: In this single-centre, prospective, parallel, randomised controlled trial, a total of 66 patients with poststroke cognitive impairment will be recruited and randomly assigned (1:1:1) to the tDCS group, the taVNS group and the combination of tDCS and taVNS group. Each group will receive 30 min of treatment daily, five times weekly for 3 weeks. Primary clinical outcome is the Montreal Cognitive Assessment. Secondary clinical outcomes include the Mini-Mental State Examination, Stroop Colour Word Test, Trail Marking Test, Symbol Digit Modalities Test and Modified Barthel Index. All clinical outcomes, functional MRI and diffusion tensor imaging will be measured at preintervention and postintervention. ETHICS AND DISSEMINATION: The trial has been approved by the Ethics Committee of the First Affiliated Hospital of Yangtze University (approval no: KY202390). The results will be submitted for publication in peer-reviewed journals or at scientific conferences. TRIAL REGISTRATION NUMBER: ChiCTR2300076632.


Subject(s)
Cognitive Dysfunction , Stroke Rehabilitation , Stroke , Transcranial Direct Current Stimulation , Vagus Nerve Stimulation , Humans , Transcranial Direct Current Stimulation/methods , Diffusion Tensor Imaging , Prospective Studies , Vagus Nerve Stimulation/methods , Quality of Life , Cognitive Dysfunction/etiology , Cognitive Dysfunction/therapy , Stroke/complications , Stroke/therapy , Randomized Controlled Trials as Topic
17.
Diabetologia ; 67(6): 1122-1137, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38546822

ABSTRACT

AIMS/HYPOTHESIS: Diabetic gastroenteropathy frequently causes debilitating gastrointestinal symptoms. Previous uncontrolled studies have shown that transcutaneous vagal nerve stimulation (tVNS) may improve gastrointestinal symptoms. To investigate the effect of cervical tVNS in individuals with diabetes suffering from autonomic neuropathy and gastrointestinal symptoms, we conducted a randomised, sham-controlled, double-blind (participants and investigators were blinded to the allocated treatment) study. METHODS: This study included adults (aged 20-86) with type 1 or 2 diabetes, gastrointestinal symptoms and autonomic neuropathy recruited from three Steno Diabetes Centres in Denmark. Participants were randomly allocated 1:1 to receive active or sham stimulation. Active cervical tVNS or sham stimulation was self-administered over two successive study periods: 1 week of four daily stimulations and 8 weeks of two daily stimulations. The primary outcome measures were gastrointestinal symptom changes as measured using the gastroparesis cardinal symptom index (GCSI) and the gastrointestinal symptom rating scale (GSRS). Secondary outcomes included gastrointestinal transit times and cardiovascular autonomic function. RESULTS: Sixty-eight participants were randomised to the active group, while 77 were randomised to the sham group. Sixty-three in the active and 68 in the sham group remained for analysis in study period 1, while 62 in each group were analysed in study period 2. In study period 1, active and sham tVNS resulted in similar symptom reductions (GCSI: -0.26 ± 0.64 vs -0.17 ± 0.62, p=0.44; GSRS: -0.35 ± 0.62 vs -0.32 ± 0.59, p=0.77; mean ± SD). In study period 2, active stimulation also caused a mean symptom decrease that was comparable to that observed after sham stimulation (GCSI: -0.47 ± 0.78 vs -0.33 ± 0.75, p=0.34; GSRS: -0.46 ± 0.90 vs -0.35 ± 0.79, p=0.50). Gastric emptying time was increased in the active group compared with sham (23 min vs -19 min, p=0.04). Segmental intestinal transit times and cardiovascular autonomic measurements did not differ between treatment groups (all p>0.05). The tVNS was well-tolerated. CONCLUSIONS/INTERPRETATION: Cervical tVNS, compared with sham stimulation, does not improve gastrointestinal symptoms among individuals with diabetes and autonomic neuropathy. TRIAL REGISTRATION: ClinicalTrials.gov NCT04143269 FUNDING: The study was funded by the Novo Nordisk Foundation (grant number NNF180C0052045).


Subject(s)
Transcutaneous Electric Nerve Stimulation , Vagus Nerve Stimulation , Humans , Female , Male , Middle Aged , Double-Blind Method , Vagus Nerve Stimulation/methods , Adult , Aged , Transcutaneous Electric Nerve Stimulation/methods , Diabetic Neuropathies/therapy , Diabetic Neuropathies/physiopathology , Gastrointestinal Diseases/therapy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/therapy , Aged, 80 and over , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/therapy , Treatment Outcome , Young Adult
18.
Brain Stimul ; 17(2): 382-391, 2024.
Article in English | MEDLINE | ID: mdl-38499287

ABSTRACT

BACKGROUND: Vagus nerve stimulation (VNS) at low frequencies (≤30 Hz) has been an established treatment for drug-resistant epilepsy (DRE) for over 25 years. OBJECTIVE: To examine the initial safety and efficacy performance of an investigational, high-frequency (≥250 Hz) VNS paradigm herein called "Microburst VNS" (µVNS). µVNS consists of short, high-frequency bursts of electrical pulses believed to preferentially modulate certain brain regions. METHODS: Thirty-three (33) participants were enrolled into an exploratory feasibility study, 21 with focal-onset seizures and 12 with generalized-onset seizures. Participants were titrated to a personalized target dose of µVNS using an investigational fMRI protocol. Participants were then followed for up to 12 months, with visits every 3 months, and monitored for side-effects at all time points. This study was registered as NCT03446664 on February 27th, 2018. RESULTS: The device was well-tolerated. Reported adverse events were consistent with typical low frequency VNS outcomes and tended to diminish in severity over time, including dysphonia, cough, dyspnea, and implant site pain. After 12 months of µVNS, the mean seizure frequency reduction for all seizures was 61.3% (median reduction: 70.4%; 90% CI of median: 48.9%-83.3%). The 12-month responder rate (≥50% reduction) was 63.3% (90% CI: 46.7%-77.9%) and the super-responder rate (≥80% reduction) was 40% (90% CI: 25.0%-56.6%). Participants with focal-onset seizures appeared to benefit similarly to participants with generalized-onset seizures (mean reduction in seizures at 12 months: 62.6% focal [n = 19], versus 59.0% generalized [n = 11]). CONCLUSION: Overall, µVNS appears to be safe and potentially a promising therapeutic alternative to traditional VNS. It merits further investigation in randomized controlled trials which will help determine the impact of investigational variables and which patients are most suitable for this novel therapy.


Subject(s)
Drug Resistant Epilepsy , Feasibility Studies , Vagus Nerve Stimulation , Humans , Male , Female , Vagus Nerve Stimulation/methods , Vagus Nerve Stimulation/instrumentation , Vagus Nerve Stimulation/adverse effects , Adult , Drug Resistant Epilepsy/therapy , Middle Aged , Young Adult , Epilepsy, Generalized/therapy , Epilepsy, Generalized/physiopathology , Treatment Outcome , Epilepsies, Partial/therapy , Epilepsies, Partial/physiopathology , Adolescent , Magnetic Resonance Imaging
19.
Neurotherapeutics ; 21(3): e00339, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38430811

ABSTRACT

Prader-Willi syndrome (PWS) is a complex, genetic disorder characterized by multisystem involvement, including hyperphagia, maladaptive behaviors and endocrinological derangements. Recent developments in advanced neuroimaging have led to a growing understanding of PWS as a neural circuit disorder, as well as subsequent interests in the application of neuromodulatory therapies. Various non-invasive and invasive device-based neuromodulation methods, including vagus nerve stimulation (VNS), transcranial direct current stimulation (tDCS), repetitive transcranial magnetic stimulation (rTMS), and deep brain stimulation (DBS) have all been reported to be potentially promising treatments for addressing the major symptoms of PWS. In this systematic literature review, we summarize the recent literature that investigated these therapies, discuss the underlying circuits which may underpin symptom manifestations, and cover future directions of the field. Through our comprehensive search, there were a total of 47 patients who had undergone device-based neuromodulation therapy for PWS. Two articles described VNS, 4 tDCS, 1 rTMS and 2 DBS, targeting different symptoms of PWS, including aberrant behavior, hyperphagia and weight. Multi-center and multi-country efforts will be required to advance the field given the low prevalence of PWS. Finally, given the potentially vulnerable population, neuroethical considerations and dialogue should guide the field.


Subject(s)
Deep Brain Stimulation , Prader-Willi Syndrome , Transcranial Direct Current Stimulation , Transcranial Magnetic Stimulation , Vagus Nerve Stimulation , Humans , Prader-Willi Syndrome/therapy , Vagus Nerve Stimulation/methods , Vagus Nerve Stimulation/instrumentation , Transcranial Magnetic Stimulation/methods , Deep Brain Stimulation/methods , Deep Brain Stimulation/instrumentation , Transcranial Direct Current Stimulation/methods
20.
Ann Clin Transl Neurol ; 11(5): 1135-1147, 2024 May.
Article in English | MEDLINE | ID: mdl-38532258

ABSTRACT

OBJECTIVE: In parallel to standard vagus nerve stimulation (VNS), microburst stimulation delivery has been developed. We evaluated the fMRI-related signal changes associated with standard and optimized microburst stimulation in a proof-of-concept study (NCT03446664). METHODS: Twenty-nine drug-resistant epilepsy patients were prospectively implanted with VNS. Three 3T fMRI scans were collected 2 weeks postimplantation. The maximum tolerated VNS intensity was determined prior to each scan starting at 0.125 mA with 0.125 mA increments. FMRI scans were block-design with alternating 30 sec stimulation [ON] and 30 sec no stimulation [OFF]: Scan 1 utilized standard VNS and Scan 3 optimized microburst parameters to determine target settings. Semi-automated on-site fMRI data processing utilized ON-OFF block modeling to determine VNS-related fMRI activation per stimulation setting. Anatomical thalamic mask was used to derive highest mean thalamic t-value for determination of microburst stimulation parameters. Paired t-tests corrected at P < 0.05 examined differences in fMRI responses to each stimulation type. RESULTS: Standard and microburst stimulation intensities at Scans 1 and 3 were similar (P = 0.16). Thalamic fMRI responses were obtained in 28 participants (19 with focal; 9 with generalized seizures). Group activation maps showed standard VNS elicited thalamic activation while optimized microburst VNS showed widespread activation patterns including thalamus. Comparison of stimulation types revealed significantly greater cerebellar, midbrain, and parietal fMRI signal changes in microburst compared to standard VNS. These differences were not associated with seizure responses. INTERPRETATION: While standard and optimized microburst VNS elicited thalamic activation, microburst also engaged other brain regions. Relationship between these fMRI activation patterns and clinical response warrants further investigation. CLINICAL TRIAL REGISTRATION: The study was registered with clinicaltrials.gov (NCT03446664).


Subject(s)
Drug Resistant Epilepsy , Magnetic Resonance Imaging , Thalamus , Vagus Nerve Stimulation , Adolescent , Adult , Female , Humans , Male , Middle Aged , Young Adult , Drug Resistant Epilepsy/therapy , Drug Resistant Epilepsy/diagnostic imaging , Drug Resistant Epilepsy/physiopathology , Functional Neuroimaging/standards , Functional Neuroimaging/methods , Proof of Concept Study , Thalamus/diagnostic imaging , Vagus Nerve Stimulation/methods , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...