An optoelectronic implantable neurostimulation platform allowing full MRI safety and optical sensing and communication

Sci Rep. 2024 May 15;14(1):11110. doi: 10.1038/s41598-024-61330-w.

ABSTRACT

A novel programmable implantable neurostimulation platform based on photonic power transfer has been developed for various clinical applications with the main focus of being safe to use with MRI scanners. The wires usually conveying electrical current from the neurostimulator to the electrodes are replaced by optical fibers. Photovoltaic cells at the tip of the fibers convert laser light to biphasic electrical impulses together with feedback signals with 54% efficiency. Furthermore, a biocompatible, implantable and ultra-flexible optical lead was developed including custom optical fibers. The neurostimulator platform incorporates advanced signal processing and optical physiological sensing capabilities thanks to a hermetically sealed transparent nonmetallic casing. Skin transparency also allowed the development of a high-speed optical transcutaneous communication channel. This implantable neurostimulation platform was first adapted to a vagus nerve stimulator for the treatment of epilepsy. This neurostimulator has been designed to fulfill the requirements of a class III long-term implantable medical device. It has been proven compliant with standard ISO/TS10974 for 1.5 T and 3 T MRI scanners. The device poses no related threat and patients can safely undergo MRI without specific or additional precautions. Especially, the RF induced heating near the implant remains below 2 °C whatever the MRI settings used. The main features of this unique advanced neurostimulator and its architecture are presented. Its functional performance is evaluated, and results are described with a focus on optoelectronics aspects and MRI safety.

PMID:38750033 | PMC:PMC11096369 | DOI:10.1038/s41598-024-61330-w

Vagus nerve stimulation in bursts can efficiently modulate gastric contractions and contraction frequency at varying gastric pressures

Neurogastroenterol Motil. 2024 May 12:e14815. doi: 10.1111/nmo.14815. Online ahead of print.

ABSTRACT

OBJECTIVE: There has been recent clinical interest in the use of vagus nerve stimulation (VNS) for treating gastrointestinal disorders as an alternative to drugs or gastric electrical stimulation. However, effectiveness of burst stimulation has not been demonstrated. We investigated the ability of bursting and continuous VNS to influence gastric and pyloric activity under a range of stimulation parameters and gastric pressures. The goals of this study were to determine which parameters could optimally excite or inhibit gastric activity.

MATERIALS AND METHODS: Data were collected from 21 Sprague-Dawley rats. Under urethane anesthesia, a rubber balloon was implanted into the stomach, connected to a pressure transducer and a saline infusion pump. A pressure catheter was inserted at the pyloric sphincter and a bipolar nerve cuff was implanted onto the left cervical vagus nerve. The balloon was filled to 15 cmH2O. Stimulation trials were conducted in a consistent order; the protocol was then repeated at 25 and 35 cmH2O. The nerve was then transected and stimulation repeated to investigate directionality of effects.

RESULTS: Bursting stimulation at the bradycardia threshold caused significant increases in gastric contraction amplitude with entrainment to the bursting frequency. Some continuous stimulation trials could also cause increased contractions but without frequency changes. Few significant changes were observed at the pylorus, except for frequency entrainment. These effects could not be uniquely attributed to afferent or efferent activity.

SIGNIFICANCE: Our findings further elucidate the effects of different VNS parameters on the stomach and pylorus and provide a basis for future studies of bursting stimulation for gastric neuromodulation.

PMID:38735698 | DOI:10.1111/nmo.14815

From the Gut to the Brain: Is Microbiota a New Paradigm in Parkinson’s Disease Treatment?

Cells. 2024 Apr 30;13(9):770. doi: 10.3390/cells13090770.

ABSTRACT

Parkinson’s disease (PD) is recognized as the second most prevalent primary chronic neurodegenerative disorder of the central nervous system. Clinically, PD is characterized as a movement disorder, exhibiting an incidence and mortality rate that is increasing faster than any other neurological condition. In recent years, there has been a growing interest concerning the role of the gut microbiota in the etiology and pathophysiology of PD. The establishment of a brain-gut microbiota axis is now real, with evidence denoting a bidirectional communication between the brain and the gut microbiota through metabolic, immune, neuronal, and endocrine mechanisms and pathways. Among these, the vagus nerve represents the most direct form of communication between the brain and the gut. Given the potential interactions between bacteria and drugs, it has been observed that the therapies for PD can have an impact on the composition of the microbiota. Therefore, in the scope of the present review, we will discuss the current understanding of gut microbiota on PD and whether this may be a new paradigm for treating this devastating disease.

PMID:38727306 | PMC:PMC11083070 | DOI:10.3390/cells13090770

Transcutaneous auricular vagus nerve stimulation as a novel therapy connecting the central and peripheral systems: a review

Int J Surg. 2024 May 9. doi: 10.1097/JS9.0000000000001592. Online ahead of print.

ABSTRACT

Currently, clinical practice and scientific research mostly revolve around a single disease or system, but the single disease-oriented diagnostic and therapeutic paradigm needs to be revised. This review describes how transcutaneous auricular vagus nerve stimulation (taVNS), a novel noninvasive neuromodulation approach, connects the central and peripheral systems of the body. Through stimulation of the widely distributed vagus nerve from the head to the abdominal cavity, this therapy can improve and treat central system disorders, peripheral system disorders, and central-peripheral comorbidities caused by autonomic dysfunction. In the past, research on taVNS has focused on the treatment of central system disorders by modulating this brain nerve. As the vagus nerve innervates the heart, lungs, liver, pancreas, gastrointestinal tract, spleen and other peripheral organs, taVNS could have an overall modulatory effect on the region of the body where the vagus nerve is widespread. Based on this physiological basis, we summarize the existing evidence of the taVNS ability to regulate cardiac function, adiposity, glucose levels, gastrointestinal function, and immune function, among others, to treat peripheral system diseases, and complex diseases with central and peripheral comorbidities. This review shows the successful examples and research progress of taVNS using peripheral neuromodulation mechanisms from more perspectives, demonstrating the expanded scope and value of taVNS to provide new ideas and approaches for holistic therapy from both central and peripheral perspectives.

PMID:38729100 | DOI:10.1097/JS9.0000000000001592

Genetic changes in the FH gene cause vagal paraganglioma

Front Endocrinol (Lausanne). 2024 Apr 24;15:1381093. doi: 10.3389/fendo.2024.1381093. eCollection 2024.

ABSTRACT

Vagal paraganglioma (VPGL) is a rare neuroendocrine tumor that originates from the paraganglion associated with the vagus nerve. VPGLs present challenges in terms of diagnostics and treatment. VPGL can occur as a hereditary tumor and, like other head and neck paragangliomas, is most frequently associated with mutations in the SDHx genes. However, data regarding the genetics of VPGL are limited. Herein, we report a rare case of a 41-year-old woman with VPGL carrying a germline variant in the FH gene. Using whole-exome sequencing, a variant, FH p.S249R, was identified; no variants were found in other PPGL susceptibility and candidate genes. Loss of heterozygosity analysis revealed the loss of the wild-type allele of the FH gene in the tumor. The pathogenic effect of the p.S249R variant on FH activity was confirmed by immunohistochemistry for S-(2-succino)cysteine (2SC). Potentially deleterious somatic variants were found in three genes, SLC7A7, ZNF225, and MED23. The latter two encode transcriptional regulators that can impact gene expression deregulation and are involved in tumor development and progression. Moreover, FH-mutated VPGL was characterized by a molecular phenotype different from SDHx-mutated PPGLs. In conclusion, the association of genetic changes in the FH gene with the development of VPGL was demonstrated. The germline variant FH: p.S249R and somatic deletion of the second allele can lead to biallelic gene damage that promotes tumor initiation. These results expand the clinical and mutation spectra of FH-related disorders and improve our understanding of the molecular genetic mechanisms underlying the pathogenesis of VPGL.

PMID:38721148 | PMC:PMC11076847 | DOI:10.3389/fendo.2024.1381093

Compatibility of transvenous implantable cardioverter defibrillator and vagus nerve stimulation device: a case report

Eur Heart J Case Rep. 2024 Apr 23;8(5):ytae214. doi: 10.1093/ehjcr/ytae214. eCollection 2024 May.

ABSTRACT

BACKGROUND: Vagus nerve stimulation (VNS) is an established therapy for drug-resistant epilepsy and depression. While VNS co-existence with cardiac pacemakers is considered safe, its interaction with implantable cardioverter defibrillators (ICDs) remains poorly understood. The concern revolves around the potential for VNS stimulation to interfere with ICD function, potentially resulting in inappropriate therapy or changes in cardiac pacing.

CASE SUMMARY: We present the case of a 50-year-old woman with drug-resistant epilepsy who underwent VNS device implantation and subsequent transvenous ICD placement for primary prevention post-myocardial infarction. These devices were thoughtfully situated contralaterally, with a minimum 10 cm separation. Comprehensive testing and follow-up demonstrated no interactions during device programming or serial assessments. Simultaneous interrogation of both devices with their respective telemetry wands caused chaotic artefacts in all channels on the ICD, likely due to electromagnetic interference. Importantly, this interference did not affect ICD sensing.

DISCUSSION: The co-existence of VNS and ICD in a patient is an emerging scenario with limited previous reports, yet our findings align with prior cases involving VNS and pacemakers. Emphasizing the need for optimal device separation and meticulous evaluation, particularly at maximum VNS output and ICD sensitivity settings, ensures their safe and feasible co-existence. As the use of VNS alongside cardiac implantable electronic devices becomes more common, a diligent evaluation for potential interactions is imperative. Our case highlights the successful co-existence of VNS and ICD, underscoring the importance of careful monitoring and evaluation to guarantee the safe utilization of these two devices.

PMID:38721251 | PMC:PMC11078306 | DOI:10.1093/ehjcr/ytae214

High-Frequency Heart Rate Variability Is Prospectively Associated With Sleep Complaints in a Healthy Working Cohort

Psychosom Med. 2024 May 1;86(4):342-348. doi: 10.1097/PSY.0000000000001302. Epub 2024 Mar 4.

ABSTRACT

OBJECTIVE: Vagus nerve functioning, as indexed by high-frequency heart rate variability (HF-HRV), has been implicated in a wide range of mental and physical health conditions, including sleep complaints. This study aimed to test associations between HF-HRV measured during sleep (sleep HF-HRV) and subjective sleep complaints 4 years later.

METHODS: One hundred forty-three healthy employees (91% male; MAge = 47.8 years [time 2], SD = 8.3 years) of an industrial company in Southern Germany completed the Jenkins Sleep Problems Scale, participated in a voluntary health assessment, and were given a 24-hour ambulatory heart rate recording device in 2007. Employees returned for a health assessment and completed the Jenkins Sleep Problems Scale 4 years later.

RESULTS: Hierarchical regression analyses showed that lower sleep HF-HRV measured in 2007 was associated with higher self-reported sleep complaints 4 years later after controlling for covariates (rab,c = -0.096, b = -0.108, 95% CI, -0.298 to 0.081, ΔR2 = 0.009, p = .050).

CONCLUSIONS: These data are the first to show that lower sleep HF-HRV predicted worse sleep 4 years later, highlighting the importance of vagus nerve functioning in adaptability and health.

PMID:38724040 | DOI:10.1097/PSY.0000000000001302

A survey of preferences and expectations for surgical interventions targeting atonic seizures in Lennox-Gastaut syndrome

Childs Nerv Syst. 2024 May 8. doi: 10.1007/s00381-024-06397-6. Online ahead of print.

ABSTRACT

PURPOSE: To assess preferences and outcome expectations for vagus nerve stimulation (VNS) and corpus callosotomy (CC) surgeries in the treatment of atonic seizure in Lennox-Gastaut syndrome (LGS).

METHODS: A total of 260 surveys were collected from patients are caregivers of LGS patients via Research Electronic Data Capture (REDCap).

RESULTS: Respondents reported an average acceptable atonic seizure reduction rate of 55.9% following VNS and 74.7% following CC. 21.3% (n = 50) were willing to be randomized. Respondents reported low willingness for randomization and a higher seizure reduction expectation with CC.

CONCLUSION: Our findings guide surgical approaches for clinicians to consider patient preference in order to design future studies comparing effectiveness between these two procedures.

PMID:38717604 | DOI:10.1007/s00381-024-06397-6

Reinforcement Learning for Closed-loop Regulation of Cardiovascular System with Vagus Nerve Stimulation: A Computational Study

J Neural Eng. 2024 May 8. doi: 10.1088/1741-2552/ad48bb. Online ahead of print.

ABSTRACT

(Objective). Vagus nerve stimulation (VNS) is being investigated as a potential therapy for cardiovascular diseases including heart failure, cardiac arrhythmia, and hypertension. The lack of a systematic approach for controlling and tuning the VNS parameters poses a significant challenge. Closed-loop VNS strategies combined with artificial intelligence (AI) approaches offer a framework for systematically learning and adapting the optimal stimulation parameters. In this study, we presented an interactive AI framework using reinforcement learning (RL) for automated data-driven design of closed-loop VNS control systems in a computational study. (Approach). Multiple simulation environments with a standard application programming interface were developed to facilitate the design and evaluation of the automated data-driven closed-loop VNS control systems. These environments simulate the hemodynamic response to multi-location VNS using biophysics-based computational models of healthy and hypertensive rat cardiovascular systems in resting and exercise states. We designed and implemented the RL-based closed-loop VNS control frameworks in the context of controlling the heart rate (HR) and the mean arterial pressure (MAP) for a set point tracking task. Our experimental design included two approaches; a general policy using deep RL algorithms and a sample-efficient adaptive policy using probabilistic inference for learning and control (PILCO). (Main results). Our simulation results demonstrated the capabilities of the closed-loop RL-based approaches to learn optimal VNS control policies and to adapt to variations in the target set points and the underlying dynamics of the cardiovascular system. Our findings highlighted the trade-off between sample-efficiency and generalizability, providing insights for proper algorithm selection. Finally, we demonstrated that transfer learning improves the sample efficiency of Deep RL algorithms allowing the development of more efficient and personalized closed-loop VNS systems. (Significance). We demonstrated the capability of RL-based closed-loop VNS systems. Our approach provided a systematic adaptable framework for learning control strategies without requiring prior knowledge about the underlying dynamics.

PMID:38718787 | DOI:10.1088/1741-2552/ad48bb