1
|
Aoyagi K, Rivas E, Shababi R, Edwards R, LaValley M, Lechuga J, Napadow V, Neogi T. Safety and preliminary efficacy of transcutaneous auricular vagus nerve stimulation on chronic knee pain: A pilot trial. OSTEOARTHRITIS AND CARTILAGE OPEN 2025; 7:100545. [PMID: 39687279 PMCID: PMC11647485 DOI: 10.1016/j.ocarto.2024.100545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Objective Transcutaneous auricular vagus nerve stimulation (tVNS) may be an innovative treatment for symptoms of knee osteoarthritis (OA) due to possible shared pathological mechanisms between diminished parasympathetic function, central pain mechanisms, and knee pain. Thus, we sought to test the safety and preliminary efficacy of tVNS in people with knee OA. Design A pilot trial in which participants received a 60-min tVNS was conducted. At baseline, immediately after, and 15 min after tVNS, we assessed knee pain, pressure pain threshold (PPT), temporal summation (TS), conditioned pain modulation (CPM), and high-frequency power of heart rate variability (HF). We examined the extent to which these outcome measures changed after tVNS using linear mixed models. Results 30 participants with knee OA were included, and all completed the intervention without any major side effects. Compared to baseline, knee pain was reduced by 1.27 (95 % CI, -1.74, -0.80) immediately after and by 1.87 (-2.33, -1.40) 15 min after tVNS; CPM improved by 0.11 (0.04, 0.19) and 0.07 (-0.01, 0.15); and HF improved by 213.29 (-0.38, 426.96) and 234.17 (20.49, 447.84). PPT and TS were not changed after tVNS. Conclusions Our preliminary data demonstrated that tVNS may be a safe pain-relieving treatment for people with knee OA. Our findings suggest that improvement of knee pain might be derived from improvement of parasympathetic function and central pain mechanisms as no local therapy was applied. A large study is needed to confirm that tVNS is a novel intervention to ameliorate knee pain in people with knee OA. Clinical Trial ClinicalTrials.gov (NCT05625178).
Collapse
Affiliation(s)
- Kosaku Aoyagi
- Department of Physical Therapy and Movement Sciences, University of Texas at El Paso, United States
| | - Elias Rivas
- Department of Physical Therapy and Movement Sciences, University of Texas at El Paso, United States
| | - Roxanna Shababi
- Department of Physical Therapy and Movement Sciences, University of Texas at El Paso, United States
| | - Robert Edwards
- Brigham and Women's Hospital, Harvard Medical School, United States
| | | | - Julia Lechuga
- Department of Physical Therapy and Movement Sciences, University of Texas at El Paso, United States
| | - Vitaly Napadow
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, United States
| | - Tuhina Neogi
- Section of Rheumatology Boston University Chobanian & Avedisian School of Medicine, United States
| |
Collapse
|
2
|
Faraji N, Payami B, Ebadpour N, Gorji A. Vagus nerve stimulation and gut microbiota interactions: a novel therapeutic avenue for neuropsychiatric disorders. Neurosci Biobehav Rev 2024:105990. [PMID: 39716559 DOI: 10.1016/j.neubiorev.2024.105990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 12/19/2024] [Indexed: 12/25/2024]
Abstract
The rising prevalence of treatment-resistant neuropsychiatric disorders underscores the need for innovative and effective treatment strategies. The gut microbiota (GM) plays a pivotal role in the progression of these diseases, influencing the brain and mental health through the gut-brain axis (GBA). The vagus nerve plays a significant role in the GBA, making it a key area of focus for potential novel therapeutic interventions. Vagus nerve stimulation (VNS) was introduced and approved as a treatment for refractory forms of some neuropsychological disorders, such as depression and epilepsy. Considering its impact on several brain regions that play a vital part in mood, motivation, affection, and cognitive function, the VNS has shown significant therapeutic potential for treating a variety of neuropsychiatric disorders. Harnessing VNS to target the bidirectional communication pathways linking the GM and the VN could present an exciting and novel approach to treating neuropsychological disorders. Imbalances in the GM, such as dysbiosis, can impair the communication pathways between the gut and the brain, contributing to the development of neuropsychological disorders. VNS shows potential for modulating these interconnected systems, helping to restore balance. Interestingly, the composition of the GM may also influence the effectiveness of VNS, as it has the potential to modify the brain's response to this therapeutic approach. This study provides a comprehensive analysis of a relatively unexplored but noteworthy interaction between VNS and GM in the treatment of neuropsychiatric disorders. In addition, we discussed the mechanisms, therapeutic potential, and clinical implications of VNS on the GBA across neuropsychiatric disorders.
Collapse
Affiliation(s)
- Navid Faraji
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahareh Payami
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Ebadpour
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Epilepsy Research Center, Department of Neurosurgery, Münster University, Germany; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
| |
Collapse
|
3
|
Atalar K, Alim E, Yigman Z, Belen HB, Erten F, Sahin K, Soylu A, Dizakar SOA, Bahcelioglu M. Transauricular vagal nerve stimulation suppresses inflammatory responses in the gut and brain in an inflammatory bowel disease model. J Anat 2024. [PMID: 39707162 DOI: 10.1111/joa.14178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 12/23/2024] Open
Abstract
Inflammatory bowel disease (IBD) encompasses Crohn's disease (CD) and ulcerative colitis (UC), is a major health problem on a global scale and its treatment is unsatisfactory. We aimed to investigate the effects of transauricular vagal nerve stimulation (tVNS) on inflammation in rats with IBD induced by trinitrobenzene sulfonic acid (TNBS). A total of 36 adult female Sprague-Dawley rats were given TNBS, or vehicle, and tVNS, or sham, every other day for 30 min for 10 days. Postmortem macroscopic and microscopic colon morphology were evaluated by histological staining. Additionally, IL-1β, IL-6, IL-10, and TNF-α cytokine levels in the colon and the brain were evaluated by immunohistochemistry and western blotting analysis. TNBS induced epithelial damage, inflammation, ulceration, and thickened mucosal layer in the colonic tissues. Administration of tVNS significantly ameliorated the severity of TNBS-induced tissue damage and inflammatory response. TNBS also alters pro-inflammatory and anti-inflammatory balance in the brain tissue. TVNS application significantly suppressed the protein levels of pro-inflammatory cytokines, namely IL-1β, IL-6, and TNF- α while augmenting the level of anti-inflammatory cytokine IL-10 in the colonic and the brain tissue. We have shown that TNBS-mediated colonic inflammation and tissue damage are associated with neuroinflammatory responses in the brain tissue. Also demonstrated for the first time that neuroinflammatory response in the gut-brain axis is suppressed by tVNS in the IBD model. Non-invasive tVNS stands out as a new potential treatment option for types of IBD.
Collapse
Affiliation(s)
- Kerem Atalar
- Department of Anatomy, Faculty of Medicine, Neuroscience and Neurotechnology Center of Excellence (NÖROM) and Neuropsychiatry Center, Gazi University, Ankara, Türkiye
| | - Ece Alim
- Department of Anatomy, Faculty of Medicine, Neuroscience and Neurotechnology Center of Excellence (NÖROM), Gazi University, Ankara, Türkiye
| | - Zeynep Yigman
- Department of Histology and Embryology, Faculty of Medicine, Neuroscience and Neurotechnology Center of Excellence (NÖROM), Gazi University, Türkiye
| | - Hayrunnisa Bolay Belen
- Department of Neurology and Algology, Faculty of Medicine, Neuroscience and Neurotechnology Center of Excellence (NÖROM), Neuropsychiatry Center, Gazi University, Ankara, Türkiye
| | - Fusun Erten
- Department of Veterinary Medicine, Pertek Sakine Genc Vocational School, Munzur University, Tunceli, Türkiye
| | - Kazım Sahin
- Department of Animal Nutrition and Nutritional Diseases, Faculty of Veterinary Medicine, Fırat University, Elazığ, Türkiye
| | - Ayse Soylu
- Department of Anatomy Faculty of Medicine, Gazi University, Ankara, Türkiye
| | | | - Meltem Bahcelioglu
- Department of Anatomy, Faculty of Medicine, Neuroscience and Neurotechnology Center of Excellence (NÖROM) and Neuropsychiatry Center, Gazi University, Ankara, Türkiye
| |
Collapse
|
4
|
Brem S. Vagus nerve stimulation: Novel concept for the treatment of glioblastoma and solid cancers by cytokine (interleukin-6) reduction, attenuating the SASP, enhancing tumor immunity. Brain Behav Immun Health 2024; 42:100859. [PMID: 39512605 PMCID: PMC11541944 DOI: 10.1016/j.bbih.2024.100859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/31/2024] [Accepted: 09/07/2024] [Indexed: 11/15/2024] Open
Abstract
Immuno-oncology, specifically immune checkpoint inhibitors (ICIs), has revolutionized cancer care with dramatic, long-term responses and increased survival, including patients with metastatic cancer to the brain. Glioblastomas, and other primary brain tumors, are refractory to ICIs as monotherapy or in combination with standard therapy. The tumor microenvironment (TME) poses multiple biological hurdles: blood-brain barrier, immune suppression, heterogeneity, and tumor infiltration. Genomic analysis of the senescence-associated secretory phenotype (SASP) and preclinical models of glioma suggest that an exciting approach would entail reprogramming of the glioma microenvironment, attenuating the pro-inflammatory, pro-tumorigenic cytokines of the SASP, especially interleukin-6 (IL-6). A testable hypothesis now proposed is to modulate the immune system by harnessing the body's 'inflammatory reflex' to reduce cytokines. Vagus nerve stimulation can activate T cell immunity by the cholinergic, α7nicotinic acetylcholine receptor agonist (α7nAchR), and suppress IL-6 systemically, as well as other pro-inflammatory cytokines of the SASP, interleukin -1β (IL-1β) and tumor necrosis factor-alpha (TNF-α). The hypothesis predicts that electrical activation of the vagus nerve, with cytokine reduction, in combination with ICIs, would convert an immune resistant ("cold") tumor to an immune responsive ("hot") tumor, and halt glioma progression. The hypothesis also envisions cancer as an immune "dysautonomia" whereby a therapeutic intervention, vagus nerve stimulation (VNS), resets the systemic and local cytokine levels. A prospective, randomized, phase II clinical trial, to confirm the hypothesis, is a logical, exigent, next step. Cytokine reduction by VNS could also be useful for other forms of human cancer, e.g., breast, colorectal, head and neck, lung, melanoma, ovarian, pancreatic, and prostate cancer, as the emerging field of "cancer neuroscience" shows a role for neural regulation of multiple tumor types. Because IL-6, and companion pro-inflammatory cytokines, participate in the initiation, progression, spread and recurrence of cancer, minimally invasive VNS could be employed to suppress glioma or cancer progression, while also mitigating depression and/or seizures, thereby enhancing quality of life. The current hypothesis reimagines glioma pathophysiology as a dysautonomia with the therapeutic objective to reset the autonomic nervous system and form an immune responsive state to halt tumor progression and prevent recurrence. VNS, as a novel method to control cancer, can be administered with ICIs, standard therapy, or in clinical trials, combined with emerging immunotherapy: dendritic cell, mRNA, or chimeric antigen receptor (CAR) T cell vaccines.
Collapse
Affiliation(s)
- Steven Brem
- University of Pennsylvania, Department of Neurosurgery, Perelman Center for Advanced Medicine, 15-141, 3400 Civic Center Blvd., Philadelphia, PA, 19104, United States
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, United States
| |
Collapse
|
5
|
Duff IT, Likar R, Perruchoud C, Kampusch S, Köstenberger M, Sator S, Stremnitzer C, Wolf A, Neuwersch-Sommeregger S, Abd-Elsayed A. Clinical Efficacy of Auricular Vagus Nerve Stimulation in the Treatment of Chronic and Acute Pain: A Systematic Review and Meta-analysis. Pain Ther 2024; 13:1407-1427. [PMID: 39382792 PMCID: PMC11543973 DOI: 10.1007/s40122-024-00657-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/29/2024] [Indexed: 10/10/2024] Open
Abstract
INTRODUCTION Current guidelines for pain treatment recommend a personalized, multimodal and interdisciplinary approach as well as the use of a combination of drug and non-drug therapies. Risk factors for chronification should already be reduced in patients with acute pain, e.g., after surgery or trauma. Auricular vagus nerve stimulation (aVNS) could be an effective non-drug therapy in the multimodal treatment of chronic and acute pain. The aim of this systematic review and meta-analysis is to evaluate the clinical efficacy and safety of aVNS in treating chronic and acute pain conditions. METHODS A systematic literature search was performed regarding the application of auricular electrical stimulation in chronic and acute pain. Studies were classified according to their level of evidence (Jadad scale), scientific validity and risk of bias (RoB 2 tool) and analyzed regarding indication, method, stimulation parameters, duration of treatment and efficacy and safety. A meta-analysis on (randomized) controlled trials (using different comparators) was performed for chronic and acute pain conditions, respectively, including subgroup analysis for percutaneous (pVNS-needle electrodes) and transcutaneous (tVNS-surface electrodes) aVNS. The visual analog pain scale (VAS) was defined as primary efficacy endpoint. RESULTS A total of n = 1496 patients were treated with aVNS in 23 identified and analyzed studies in chronic pain, 12 studies in acute postoperative pain and 7 studies in experimental acute pain. Of these, seven studies for chronic pain and six studies for acute postoperative pain were included in the meta-analysis. In chronic pain conditions, including back pain, migraine and abdominal pain, a statistically significant reduction in VAS pain intensity for active compared to sham aVNS or control treatment with an effect size Hedges' g/mean difference of - 1.95 (95% confidence interval [CI]: - 3.94 to 0.04, p = 0.008) could be shown and a more favorable effect in pVNS compared to tVNS (- 5.40 [- 8.94; - 1.85] vs. - 1.00 [- 1.55; - 0.44]; p = 0.015). In acute pain conditions, single studies showed significant improvements with aVNS, e.g., in kidney donor surgery or tonsillectomy but, overall, a non-statistically significant reduction in VAS pain intensity for active compared to sham aVNS or control with - 0.70 [- 2.34; 0.93] (p = 0.15) could be observed in the meta-analysis. In acute pain results vary greatly between studies depending especially on co-medication and timepoints of assessment after surgery. A significant reduction in analgesics or opiate intake was documented in most studies evaluating this effect in chronic and acute pain. In 3 of the 12 randomized controlled trials in patients with chronic pain, a sustainable pain reduction over a period of up to 12 months was shown. Overall, aVNS was very well tolerated. CONCLUSION This systematic review and meta-analysis indicate that aVNS can be an effective and safe non-drug treatment in patients with specific chronic and acute postoperative pain conditions. Further research is needed to identify the influence of simulation parameters and find optimal and standardized treatment protocols while considering quality-of-life outcome parameters and prolonged follow-up periods. A more standardized approach and harmonization in study designs would improve comparability and robustness of outcomes.
Collapse
Affiliation(s)
- Irina T Duff
- Neurosurgery Department, Johns Hopkins University, Baltimore, MD, USA
| | - Rudolf Likar
- Department for Anesthesia and Critical Care, Klinikum Klagenfurt am Wörthersee, Klagenfurt, Austria
- Sigmund Freud University, Vienna, Austria
| | | | | | - Markus Köstenberger
- Department for Anesthesia and Critical Care, Klinikum Klagenfurt am Wörthersee, Klagenfurt, Austria
- Medical University of Graz, Graz, Austria
| | - Sabine Sator
- Department for Anesthesia, Critical Care and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Andreas Wolf
- Department of Anesthesia, Krankenhaus St. Vinzenz, Zams, Austria
| | - Stefan Neuwersch-Sommeregger
- Medical University of Graz, Graz, Austria
- Department for Anesthesiology and Intensive Care, Krankenhaus der Barmherzigen Brüder, St. Veit/Glan, Austria
| | - Alaa Abd-Elsayed
- Department of Anesthesiology, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
6
|
Meerschaert KA, Chiu IM. The gut-brain axis and pain signalling mechanisms in the gastrointestinal tract. Nat Rev Gastroenterol Hepatol 2024:10.1038/s41575-024-01017-9. [PMID: 39578592 DOI: 10.1038/s41575-024-01017-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 11/24/2024]
Abstract
Visceral pain is a major clinical problem and one of the most common reasons patients with gastrointestinal disorders seek medical help. Peripheral sensory neurons that innervate the gut can detect noxious stimuli and send signals to the central nervous system that are perceived as pain. There is a bidirectional communication network between the gastrointestinal tract and the nervous system that mediates pain through the gut-brain axis. Sensory neurons detect mechanical and chemical stimuli within the intestinal tissues, and receive signals from immune cells, epithelial cells and the gut microbiota, which results in peripheral sensitization and visceral pain. This Review focuses on molecular communication between these non-neuronal cell types and neurons in visceral pain. These bidirectional interactions can be dysregulated during gastrointestinal diseases to exacerbate visceral pain. We outline the anatomical pathways involved in pain processing in the gut and how cell-cell communication is integrated into this gut-brain axis. Understanding how bidirectional communication between the gut and nervous system is altered during disease could provide new therapeutic targets for treating visceral pain.
Collapse
Affiliation(s)
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Liu FJ, Wu J, Gong LJ, Yang HS, Chen H. Non-invasive vagus nerve stimulation in anti-inflammatory therapy: mechanistic insights and future perspectives. Front Neurosci 2024; 18:1490300. [PMID: 39605787 PMCID: PMC11599236 DOI: 10.3389/fnins.2024.1490300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Non-invasive vagus nerve stimulation (VNS) represents a transformative approach for managing a broad spectrum of inflammatory and autoimmune conditions, including rheumatoid arthritis and inflammatory bowel disease. This comprehensive review delineates the mechanisms underlying VNS, emphasizing the cholinergic anti-inflammatory pathway, and explores interactions within the neuro-immune and vagus-gut axes based on both clinical outcomes and pre-clinical models. Clinical applications have confirmed the efficacy of VNS in managing specific autoimmune diseases, such as rheumatoid arthritis, and chronic inflammatory conditions like inflammatory bowel disease, showcasing the variability in stimulation parameters and patient responses. Concurrently, pre-clinical studies have provided insights into the potential of VNS in modulating cardiovascular and broader inflammatory responses, paving the way for its translational application in clinical settings. Innovations in non-invasive VNS technology and precision neuromodulation are enhancing its therapeutic potential, making it a viable option for patients who are unresponsive to conventional treatments. Nonetheless, the widespread adoption of this promising therapy is impeded by regulatory challenges, patient compliance issues, and the need for extensive studies on long-term efficacy and safety. Future research directions will focus on refining VNS technology, optimizing treatment parameters, and exploring synergistic effects with other therapeutic modalities, which could revolutionize the management of chronic inflammatory and autoimmune disorders.
Collapse
Affiliation(s)
- Fu-Jun Liu
- Neurology Medical Center II, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, China
| | - Jing Wu
- Department of Medical Imaging, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, China
| | - Li-Jun Gong
- Center of Surgical Anesthesia, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hong-Shuai Yang
- Central Operating Room, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, China
| | - Huan Chen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Shin JH, Lee CM, Song JJ. Transcutaneous auricular vagus nerve stimulation mitigates gouty inflammation by reducing neutrophil infiltration in BALB/c mice. Sci Rep 2024; 14:25630. [PMID: 39463429 PMCID: PMC11514149 DOI: 10.1038/s41598-024-77272-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
Gouty inflammation, caused by uric acid crystal deposition, primarily affects tissues around the toe joints and triggers potent inflammatory responses. Current treatments focus on alleviating inflammation and pain using pharmaceutical agents, which can lead to side effects and complications. This has generated interest in non-pharmacological interventions, such as non-invasive vagus nerve stimulation (VNS). In this study, we explored the anti-inflammatory mechanisms of transcutaneous auricular vagus nerve stimulation (taVNS) in a mouse model of acute gout. Gouty inflammation was induced by injecting monosodium urate (MSU) crystals into the ankle joints of BALB/c mice. The effects of taVNS on the expression of inflammatory cytokines and chemokines in the ankle joint tissue were assessed using real-time quantitative PCR (qPCR), western blotting, histological assessments (H&E staining), and immunohistochemistry (IHC). The role of α7 nicotinic acetylcholine receptors (α7nAChR) was also evaluated by signal blocking. Our findings revealed that MSU significantly elevated gout-associated inflammatory cascades and mediators in the ankle joint. Notably, taVNS at 200 µA and 25 Hz effectively reduced these inflammatory responses, decreasing neutrophil infiltration and chemoattraction within the tissue. taVNS showed significant anti-inflammatory properties by suppressing neutrophil activity, offering a novel therapeutic approach for gout beyond conventional pharmacological methods. Additionally, taVNS holds potential for managing various chronic joint diseases. These results highlight taVNS as a promising non-pharmacological therapy for chronic inflammation.
Collapse
Affiliation(s)
- Jae Hee Shin
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-dong, Guro-gu, Seoul, 08308, Korea
- Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul, 08308, Korea
| | - Chan Mi Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-dong, Guro-gu, Seoul, 08308, Korea
| | - Jae-Jun Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-dong, Guro-gu, Seoul, 08308, Korea.
- Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul, 08308, Korea.
- Neurive Institute, Neurive Co., Ltd., Seoul, 08308, Korea.
| |
Collapse
|
9
|
Pérez-Alcalde AI, Galán-Del-Río F, Fernández-Rodríguez FJ, de la Plaza San Frutos M, García-Arrabé M, Giménez MJ, Ruiz-Ruiz B. The Effects of a Single Vagus Nerve's Neurodynamics on Heart Rate Variability in Chronic Stress: A Randomized Controlled Trial. SENSORS (BASEL, SWITZERLAND) 2024; 24:6874. [PMID: 39517768 PMCID: PMC11548125 DOI: 10.3390/s24216874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The modulation of the autonomic nervous system's activity, particularly increasing its parasympathetic tone, is of significant interest in clinical physiotherapy due to its potential benefits for stress-related conditions and recovery processes. This study evaluated the effectiveness of the addition of neurodynamics in enhancing parasympathetic activation in subjects with chronic stress. METHODS A clinical trial randomly assigned participants to a group with neurodynamics (6 bpm breathing protocol + manual therapy + neurodynamic technique) or a group without neurodynamics (6 bpm breathing protocol + manual therapy only). Metrics of heart rate variability (HRV), including the Mean Heart Rate (Mean HR), standard deviation of intervals between consecutive heartbeats (SDNN), Heart Rate Difference (Diff. HR), Root Mean Square of Successive Differences (RMSSD), number of intervals differing by more than 50 ms (NN50), percentage of consecutive NN intervals that differed by more than 50 ms (pNN50), and the high-frequency component measured in standardized units (HF), were assessed before, during, and after the intervention. RESULTS During the intervention, the group with neurodynamics showed significant changes in all variables except in the pNN50 and HF while the group without neurodynamics only showed improvements in the Mean HR, SDNN, and RMSSD. In the post-intervention phase, the group with neurodynamics maintained an increase in HRV while the group without neurodynamics experienced a decrease, suggesting an increase in sympathetic activity. CONCLUSIONS Vagal nerve neurodynamics appear to represent an effective method for enhancing parasympathetic activation in patients with chronic stress. The results highlight the importance of a more comprehensive analysis of HRV variables in order to obtain a correct picture of the impact of interventions on the complex and multifaceted functioning of the autonomic nervous system.
Collapse
Affiliation(s)
- Ana Isabel Pérez-Alcalde
- Department of Physiotherapy, Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain
- International Doctoral School, Rey Juan Carlos University, 28008 Madrid, Spain
| | - Fernando Galán-Del-Río
- Department of Physical Therapy, Occupational Therapy, Rehabilitation and Physical Medicine, Faculty of Health Sciences, Rey Juan Carlos University, 28922 Madrid, Spain
| | | | | | - María García-Arrabé
- Department of Physiotherapy, Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain
| | - María-José Giménez
- Department of Physiotherapy, Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Beatriz Ruiz-Ruiz
- Department of Physiotherapy, Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain
| |
Collapse
|
10
|
Goyal M, Goyal R, Sanguinetti JL. Ultrasound stimulation of the vagus nerve as a treatment modality for anxiety. Front Psychiatry 2024; 15:1376140. [PMID: 39415887 PMCID: PMC11480057 DOI: 10.3389/fpsyt.2024.1376140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 08/19/2024] [Indexed: 10/19/2024] Open
Abstract
Anxiety is an increasingly prevalent mental disorder, causing widespread hardship and interfering with society's economic progression. Standard treatments include various talk therapies with poor prognoses or drug interventions with complex side effects, both introducing unnecessary burdens to patients. To remedy this, non-invasive ultrasound stimulation to the vagus nerve is a novel, low-cost treatment that is showing promise. Although vagus nerve stimulation is already approved for epilepsy and other conditions, it requires regular maintenance. In contrast, studies using non-invasive ultrasound stimulation have shown preliminary positive results in affecting vagal activity with minimal drawbacks. This review covers a variety of studies investigating the effects of ultrasound stimulation on the vagus nerve. With rising levels of anxiety with each generation, there is a pressing need for more innovative and diverse treatments with fewer costs and more benefits.
Collapse
Affiliation(s)
- Michell Goyal
- Department of Physiology, University of Arizona, Tucson, AZ, United States
| | - Ravi Goyal
- Department of Obstetrics and Gynecology, University of Arizona, Tucson, AZ, United States
| | | |
Collapse
|
11
|
Agnesi F, Zinno C, Strauss I, Dushpanova A, Casieri V, Bernini F, Terlizzi D, Gabisonia K, Paggi V, Lacour SP, Lionetti V, Micera S. Cardiovascular Response to Intraneural Right Vagus Nerve Stimulation in Adult Minipig. Neuromodulation 2024; 27:1187-1195. [PMID: 36997453 DOI: 10.1016/j.neurom.2023.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/27/2023] [Accepted: 03/06/2023] [Indexed: 03/31/2023]
Abstract
OBJECTIVE This study explored intraneural stimulation of the right thoracic vagus nerve (VN) in sexually mature male minipigs to modulate safe heart rate and blood pressure response. MATERIAL AND METHODS We employed an intraneural electrode designed for the VN of pigs to perform VN stimulation (VNS). This was delivered using different numbers of contacts on the electrode and different stimulation parameters (amplitude, frequency, and pulse width), identifying the most suitable stimulation configuration. All the parameter ranges had been selected from a computational cardiovascular system model. RESULTS Clinically relevant responses were observed when stimulating with low current intensities and relatively low frequencies delivered with a single contact. Selecting a biphasic, charge-balanced square wave for VNS with a current amplitude of 500 μA, frequency of 10 Hz, and pulse width of 200 μs, we obtained heart rate reduction of 7.67 ± 5.19 beats per minute, systolic pressure reduction of 5.75 ± 2.59 mmHg, and diastolic pressure reduction of 3.39 ± 1.44 mmHg. CONCLUSION Heart rate modulation was obtained without inducing any observable adverse effects, underlining the high selectivity of the intraneural approach.
Collapse
Affiliation(s)
- Filippo Agnesi
- BioRobotics Institute, Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Ciro Zinno
- BioRobotics Institute, Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Ivo Strauss
- Institut für Mikrosystemtechnik, University of Frieberg, IMTEK, Freiburg, Germany
| | - Anar Dushpanova
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, Interdisciplinary Research Center "Health Science," Scuola Superiore Sant'Anna, Pisa, Italy; Health Research Institute, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Valentina Casieri
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, Interdisciplinary Research Center "Health Science," Scuola Superiore Sant'Anna, Pisa, Italy; BioMedLab, Scuola Superiore Sant'Anna, Pisa, Italy
| | | | | | | | - Valentina Paggi
- Bertarelli Foundation Chair in Neuroprosthetic Technology, Laboratory for Soft Bioelectric Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Stéphanie P Lacour
- Bertarelli Foundation Chair in Neuroprosthetic Technology, Laboratory for Soft Bioelectric Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Vincenzo Lionetti
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, Interdisciplinary Research Center "Health Science," Scuola Superiore Sant'Anna, Pisa, Italy; BioMedLab, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Silvestro Micera
- BioRobotics Institute, Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, Pisa, Italy; Bertarelli Foundation Chair in Translational NeuroEngineering, Centre for Neuroprosthetics and Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| |
Collapse
|
12
|
Habibollahi M, Jiang D, Lancashire HT, Demosthenous A. Active Neural Interface Circuits and Systems for Selective Control of Peripheral Nerves: A Review. IEEE TRANSACTIONS ON BIOMEDICAL CIRCUITS AND SYSTEMS 2024; 18:954-975. [PMID: 39018210 DOI: 10.1109/tbcas.2024.3430038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
Interfaces with peripheral nerves have been widely developed to enable bioelectronic control of neural activity. Peripheral nerve neuromodulation shows great potential in addressing motor dysfunctions, neurological disorders, and psychiatric conditions. The integration of high-density neural electrodes with stimulation and recording circuits poses a challenge in the design of neural interfaces. Recent advances in active electrode strategies have achieved improved reliability and performance by implementing in-situ control, stimulation, and recording of neural fibers. This paper presents an overview of state-of-the-art neural interface systems that comprise a range of neural electrodes, neurostimulators, and bio-amplifier circuits, with a special focus on interfaces for the peripheral nerves. A discussion on the efficacy of active electrode systems and recommendations for future directions conclude this paper.
Collapse
|
13
|
Ahmed A, Rahman AH, Williams S, Toufiq M, Jamileh E, Chaudhry S, Ahmed K, Ahmed M. Seizures and Epilepsy: An Overview for UK Medical Students. Cureus 2024; 16:e70654. [PMID: 39493075 PMCID: PMC11527550 DOI: 10.7759/cureus.70654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2024] [Indexed: 11/05/2024] Open
Abstract
Epilepsy is a common neurological disorder impacting millions globally, marked by recurrent, unprovoked seizures. This review article, tailored for UK medical students, provides a broad clinical overview of epilepsy, focusing on its pathophysiology, classification, and management strategies. The article clarifies the distinction between epilepsy and seizures and delves into key areas, including risk factors, clinical features, and differential diagnosis. The discussion extends to diagnostic methods, underscoring the importance of conducting a thorough evaluation in diagnosing and managing epilepsy effectively. Data were drawn from the UK National Institute for Health and Care Excellence (NICE) guidelines, World Health Organization (WHO) reports, and key peer-reviewed studies. Particular attention was given to UK-specific data on epilepsy incidence, treatment gaps, and clinical outcomes. A review of relevant literature was conducted, covering epidemiology, pathophysiology, diagnostic protocols, and management strategies based on UK practices. Finally, the article addresses the acute management of seizures and the pharmacological and non-pharmacological management of epilepsy based on the NICE guidelines. The goal is to offer medical students a concise yet comprehensive understanding of epilepsy, preparing them for practical, evidence-based decision-making in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Mubeen Toufiq
- Respiratory Medicine, Princess Alexandra Hospital, London, GBR
| | - Eyad Jamileh
- Gastroenterology, Royal Blackburn Hospital, Blackburn, GBR
| | - Sheema Chaudhry
- Critical Care Medicine, Princess Alexandra Hospital, London, GBR
| | | | - Munir Ahmed
- Physics, Leyton Sixth Form College, London, GBR
| |
Collapse
|
14
|
Kamoga R, Rukundo GZ, Kalungi S, Adriko W, Nakidde G, Obua C, Obongoloch J, Ihunwo AO. Vagus nerve stimulation in dementia: A scoping review of clinical and pre-clinical studies. AIMS Neurosci 2024; 11:398-420. [PMID: 39431268 PMCID: PMC11486617 DOI: 10.3934/neuroscience.2024024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 10/22/2024] Open
Abstract
Background Dementia is a prevalent, progressive, neurodegenerative condition with multifactorial causes. Due to the lack of effective pharmaceutical treatments for dementia, there are growing clinical and research interests in using vagus nerve stimulation (VNS) as a potential non-pharmacological therapy for dementia. However, the extent of the research volume and nature into the effects of VNS on dementia is not well understood. This study aimed to examine the extent and nature of research activities in relation to the use of VNS in dementia and disseminate research findings for the potential utility in dementia care. Methods We performed a scoping review of literature searches in PubMed, HINARI, Google Scholar, and the Cochrane databases from 1980 to November 30th, 2023, including the reference lists of the identified studies. The following search terms were utilized: brain stimulation, dementia, Alzheimer's disease, vagal stimulation, memory loss, Deme*, cognit*, VNS, and Cranial nerve stimulation. The included studies met the following conditions: primary research articles pertaining to both humans and animals for both longitudinal and cross-sectional study designs and published in English from January 1st, 1980, to November 30th, 2023; investigated VNS in either dementia or cognitive impairment; and were not case studies, conference proceedings/abstracts, commentaries, or ordinary review papers. Findings and conclusions We identified 8062 articles, and after screening for eligibility (sequentially by titles, abstracts and full text reading, and duplicate removal), 10 studies were included in the review. All the studies included in this literature review were conducted over the last three decades in high-income geographical regions (i.e., Europe, the United States, the United Kingdom, and China), with the majority of them (7/10) being performed in humans. The main reported outcomes of VNS in the dementia cases were enhanced cognitive functions, an increased functional connectivity of various brain regions involved in learning and memory, microglial structural modifications from neurodestructive to neuroprotective configurations, a reduction of cerebral spinal fluid tau-proteins, and significant evoked brain tissue potentials that could be utilized to diagnose neurodegenerative disorders. The study outcomes highlight the potential for VNS to be used as a non-pharmacological therapy for cognitive impairment in dementia-related diseases such as Alzheimer's disease.
Collapse
Affiliation(s)
- Ronald Kamoga
- Department of Anatomy, Mbarara University of Science and Technology, P.O. Box 1410, Mbarara, Uganda
| | - Godfrey Zari Rukundo
- Department of Psychiatry, Mbarara University of Science and Technology, P.O.Box 1410, Mbarara, Uganda
| | - Samuel Kalungi
- Makerere University, School of health sciences, Department of Pathology. Kampala, Uganda
| | - Wilson Adriko
- Library department, Mbarara University of Science and Technology, P.O. Box 1410, Mbarara Uganda
| | - Gladys Nakidde
- Faculty of Nursing and Health Sciences, Bishop Stuart University, Mbarara, Uganda
| | - Celestino Obua
- Department of Pharmacology, Mbarara University of Science and Technology, P.O.Box 1410, Mbarara, Uganda
| | - Johnes Obongoloch
- Department of Biomedical engineering, Mbarara University of Science and Technology, P.O.Box 1410, Mbarara, Uganda
| | - Amadi Ogonda Ihunwo
- University of the Witwatersrand, School of Anatomical Sciences, Faculty of Health Sciences, Johannesburg, South Africa
| |
Collapse
|
15
|
Chen M, Yang C, Chen Y, Nie K, Wang T, Qu Y. Research hotspots and trends of non-invasive vagus nerve stimulation: a bibliometric analysis from 2004 to 2023. Front Neurol 2024; 15:1429506. [PMID: 39381073 PMCID: PMC11460548 DOI: 10.3389/fneur.2024.1429506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/10/2024] [Indexed: 10/10/2024] Open
Abstract
Objectives Non-invasive vagus nerve stimulation (nVNS) is an emerging neuromodulation technique in recent years, which plays a role in nervous system diseases, psychiatric diseases, and autoimmune diseases. However, there is currently no comprehensive analysis of all the literature published in this field. Therefore, in this article, a bibliometric analysis will be conducted on all the literature published in the field of nVNS in the past 20 years. Methods All articles and reviews published in this field from 2004 to 2023 were extracted from the WOS core database. VOSviewer 1.6.18.0, Scimago Graphica, CiteSpace 6.2.R2, and Excel 2021 were used to analyze the number of publications, participating countries, institutions, authors, references, and research hotspots in this field. Results A total of 843 articles were included in the bibliometric analysis of nVNS. Over the past 20 years, the number of publications in this field has gradually increased, reaching a peak in 2023. The United States and China ranked top two in terms of publication volume, and institutions from these two countries also ranked high in terms of publication volume, citation count, and collaboration intensity. Rong Peijing is the author with the most publications, while Bashar W Badran is the most cited author. Articles in the field of nVNS were most frequently published in Frontiers in Neuroscience, while Brain Stimulation had the most citations. Currently, research hotspots in nVNS mainly focus on its application in diseases and related mechanisms. Conclusion We conducted a comprehensive analysis of the field of nVNS, clarifying the previous research directions, which is helpful to expand its indications and promote clinical application.
Collapse
Affiliation(s)
- Mingyue Chen
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- College of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Sichuan Provincial Key Laboratory of Rehabilitation Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Chunlan Yang
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- College of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Sichuan Provincial Key Laboratory of Rehabilitation Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Yin Chen
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- College of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Sichuan Provincial Key Laboratory of Rehabilitation Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Kailu Nie
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- College of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Sichuan Provincial Key Laboratory of Rehabilitation Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Tingting Wang
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- College of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Sichuan Provincial Key Laboratory of Rehabilitation Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Yun Qu
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- College of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Sichuan Provincial Key Laboratory of Rehabilitation Medicine, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
16
|
Rauhala OJ, Ma L, Wisniewski DJ, Shao S, Schumacher B, Lopez JF, Kaspers M, Zhao Z, Gelinas JN, Khodagholy D. E-Suture: Mixed-Conducting Suture for Medical Devices. Adv Healthc Mater 2024; 13:e2302613. [PMID: 38150402 PMCID: PMC11338356 DOI: 10.1002/adhm.202302613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/17/2023] [Indexed: 12/29/2023]
Abstract
Modern implantable bioelectronics demand soft, biocompatible components that make robust, low-impedance connections with the body and circuit elements. Concurrently, such technologies must demonstrate high efficiency, with the ability to interface between the body's ionic and external electronic charge carriers. Here, a mixed-conducting suture, the e-suture, is presented. Composed of silk, the conducting polymer poly(3,4-ethylenedioxythiophene) polystyrene sulfonate (PEDOT:PSS), and insulating jacketing polymers,the resulting e-suture has mixed-conducting properties at the interface with biological tissue as well as effective insulation along its length. The e-suture can be mechanically integrated into electronics, enabling the acquisition of biopotentials such as electrocardiograms, electromyograms, and local field potentials (LFP). Chronic, in vivo acquisition of LFP with e-sutures remains stable for months with robust brain activity patterns. Furthermore, e-sutures can establish electrophoretic-based local drug delivery, potentially offering enhanced anatomical targeting and decreased side effects associated with systemic administration, while maintaining an electrically conducting interface for biopotential monitoring. E-sutures expand on the conventional role of sutures and wires by providing a soft, biocompatible, and mechanically sound structure that additionally has multifunctional capacity for sensing, stimulation, and drug delivery.
Collapse
Affiliation(s)
- Onni J Rauhala
- Department of Electrical Engineering, Columbia University, New York, 10027, USA
| | - Liang Ma
- Department of Biomedical Engineering, Columbia University, New York, 10027, USA
| | - Duncan J Wisniewski
- Department of Electrical Engineering, Columbia University, New York, 10027, USA
| | - Shan Shao
- Department of Neurology, Columbia University Irving Medical Center, New York, 10032, USA
| | - Brandon Schumacher
- Department of Neurology, Columbia University Irving Medical Center, New York, 10032, USA
| | - Jose Ferrero Lopez
- Department of Neurology, Columbia University Irving Medical Center, New York, 10032, USA
| | - Mara Kaspers
- Department of Biomedical Engineering, Columbia University, New York, 10027, USA
| | - Zifang Zhao
- Department of Electrical Engineering, Columbia University, New York, 10027, USA
| | - Jennifer N Gelinas
- Department of Biomedical Engineering, Columbia University, New York, 10027, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, 10032, USA
| | - Dion Khodagholy
- Department of Electrical Engineering, Columbia University, New York, 10027, USA
| |
Collapse
|
17
|
Burish MJ, Guirguis AB, Schindler EAD. Managing Cluster Headache in Patients with Medical, Psychiatric, and Surgical Comorbidities. Curr Neurol Neurosci Rep 2024; 24:439-452. [PMID: 39017830 DOI: 10.1007/s11910-024-01362-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2024] [Indexed: 07/18/2024]
Abstract
PURPOSE OF REVIEW What should a provider know about medications and other treatments in patients with cluster headache who have medical, psychiatric, and surgical comorbidities? What conversations should providers have with patients about living with and managing cluster headache? RECENT FINDINGS While the number of treatments used in cluster headache is relatively small, numerous considerations were identified related to managing patients with comorbidities. Many of these touch on cardiac, cardiovascular, and cerebrovascular health, but full histories are needed to guide safe and effective treatment. Both older and newer treatments may be contraindicated in certain patients with cluster headache or should be considered carefully. In addition to incorporating medical, psychiatric, and surgical histories in the management plan, collaboration with other providers may be beneficial. Providers should also inquire about patient practices and discuss participation in clinical trials that might be a good fit for the individual.
Collapse
Affiliation(s)
- Mark J Burish
- Department of Neurosurgery, McGovern Medical School at UTHealth Houston, Houston, TX, USA
| | - Alexander B Guirguis
- Veteran Health Administration Headache Center of Excellence, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Emmanuelle A D Schindler
- Veteran Health Administration Headache Center of Excellence, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA.
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.
- VA Connecticut Healthcare System, Neurology Service, MS 127, 950 Campbell Avenue, West Haven, CT, 06516, USA.
| |
Collapse
|
18
|
Huang Y, Yao K, Zhang Q, Huang X, Chen Z, Zhou Y, Yu X. Bioelectronics for electrical stimulation: materials, devices and biomedical applications. Chem Soc Rev 2024; 53:8632-8712. [PMID: 39132912 DOI: 10.1039/d4cs00413b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Bioelectronics is a hot research topic, yet an important tool, as it facilitates the creation of advanced medical devices that interact with biological systems to effectively diagnose, monitor and treat a broad spectrum of health conditions. Electrical stimulation (ES) is a pivotal technique in bioelectronics, offering a precise, non-pharmacological means to modulate and control biological processes across molecular, cellular, tissue, and organ levels. This method holds the potential to restore or enhance physiological functions compromised by diseases or injuries by integrating sophisticated electrical signals, device interfaces, and designs tailored to specific biological mechanisms. This review explains the mechanisms by which ES influences cellular behaviors, introduces the essential stimulation principles, discusses the performance requirements for optimal ES systems, and highlights the representative applications. From this review, we can realize the potential of ES based bioelectronics in therapy, regenerative medicine and rehabilitation engineering technologies, ranging from tissue engineering to neurological technologies, and the modulation of cardiovascular and cognitive functions. This review underscores the versatility of ES in various biomedical contexts and emphasizes the need to adapt to complex biological and clinical landscapes it addresses.
Collapse
Affiliation(s)
- Ya Huang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Kuanming Yao
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Qiang Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Xingcan Huang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Zhenlin Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Yu Zhou
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong, China.
| | - Xinge Yu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
19
|
Wu PY, Caceres AI, Chen J, Sokoloff J, Huang M, Baht GS, Nackley AG, Jordt SE, Terrando N. Vagus nerve stimulation rescues persistent pain following orthopedic surgery in adult mice. Pain 2024; 165:e80-e92. [PMID: 38422485 PMCID: PMC11247455 DOI: 10.1097/j.pain.0000000000003181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/21/2023] [Accepted: 12/13/2023] [Indexed: 03/02/2024]
Abstract
ABSTRACT Postoperative pain is a major clinical problem imposing a significant burden on patients and society. In a survey 2 years after orthopedic surgery, 57% of patients reported persisting postoperative pain. However, only limited progress has been made in the development of safe and effective therapies to prevent the onset and chronification of pain after orthopedic surgery. We established a tibial fracture mouse model that recapitulates clinically relevant orthopedic trauma surgery, which causes changes in neuropeptide levels in dorsal root ganglia and sustained neuroinflammation in the spinal cord. Here, we monitored extended pain behavior in this model, observing chronic bilateral hindpaw mechanical allodynia in both male and female C57BL/6J mice that persisted for >3 months after surgery. We also tested the analgesic effects of a novel, minimally invasive, bioelectronic approach to percutaneously stimulate the vagus nerve (termed percutaneous vagus nerve stimulation [pVNS]). Weekly pVNS treatment for 30 minutes at 10 Hz for 3 weeks after the surgery strongly reduced pain behaviors compared with untreated controls. Percutaneous vagus nerve stimulation also improved locomotor coordination and accelerated bone healing. In the dorsal root ganglia, vagal stimulation inhibited the activation of glial fibrillary acidic protein-positive satellite cells but without affecting microglial activation. Overall, these data provide novel evidence supportive of the use of pVNS to prevent postoperative pain and inform translational studies to test antinociceptive effects of bioelectronic medicine in the clinic.
Collapse
Affiliation(s)
- Pau Yen Wu
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC, United States
| | - Ana Isabel Caceres
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC, United States
| | - Jiegen Chen
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC, United States
| | - Jamie Sokoloff
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC, United States
| | - Mingjian Huang
- Department of Orthopaedic Surgery, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| | - Gurpreet Singh Baht
- Department of Orthopaedic Surgery, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| | - Andrea G. Nackley
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC, United States
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, United States
| | - Sven-Eric Jordt
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC, United States
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, United States
- Integrated Toxicology and Environmental Health Program, Duke University, Durham, United States
| | - Niccolò Terrando
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC, United States
- Department of Cell Biology, Duke University Medical Center, Durham, NC, United States
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
20
|
Wang H, Kwok WH, Yao LQ, Liu XL, Bressington D, Chen ML, Huang HQ, Wang T, Tan JYB. An evidence-based breathing exercise intervention for chronic pain management in breast cancer survivors: A phase II randomized controlled trial. Eur J Oncol Nurs 2024; 71:102625. [PMID: 38897104 DOI: 10.1016/j.ejon.2024.102625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/17/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024]
Abstract
OBJECTIVE Explore the preliminary effects of a breathing exercise (BE) intervention on chronic pain among breast cancer survivors. METHODS This two-parallel-arm, open-label pilot randomized controlled trial recruited 72 breast cancer survivors who were randomly allocated to either the control or intervention group (n = 36 each). Both groups received usual care and a pain information booklet, while the intervention group received 4 weeks of additional BE. The primary clinical outcome was measured using the Brief Pain Inventory (BPI), with secondary clinical outcomes measured by the Hospital Anxiety and Depression Scale (HADS), Quality of Life Patient/Cancer Survivor Version in Chinese (QOLCSV-C), and Functional Assessment of Cancer Therapy- Breast (FACT-B) immediately post-intervention and at 4-week follow-up. Both adjusted and unadjusted Generalized Estimating Equation models were utilized to assess the BE's potential effects, with safety assessed through participant self-report. RESULTS Sixty-eight participants completed the study. Statistical significance was observed in BPI in both adjusted and unadjusted models at post-intervention and follow-up (p < 0.05). BE demonstrated positive effects on anxiety, depression and quality of life improvement across all measures and timepoints in both adjusted and unadjusted models (p < 0.05). The effect sizes were smaller in the adjusted model. Three mild transient discomforts were reported associated with BE practice including dizziness, tiredness and yawning, without requirement of medical treatment. No severe adverse events occurred. CONCLUSION This BE intervention appears effective in alleviating chronic pain, anxiety and depression, and improving quality of life for breast cancer survivors. Fully powered large-scale studies are required to confirm its effects.
Collapse
Affiliation(s)
- Haiying Wang
- School of Nursing (Brisbane Centre), Faculty of Health, Charles Darwin University, Brisbane, QLD, Australia; School of Nursing and Midwifery & Centre for Health Research, University of Southern Queensland, Ipswich, QLD, Australia
| | - Wai Hang Kwok
- School of Nursing and Midwifery, Edith Cowan University, WA, Australia; School of Nursing, Faculty of Health, Charles Darwin University, Ellengowan Drive, Casuarina, NT, Australia
| | - Li-Qun Yao
- College of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou City, Fujian Province, China
| | - Xian-Liang Liu
- School of Nursing and Health Studies, Hong Kong Metropolitan University, Homantin, Kowloon, Hong Kong SAR, China
| | - Daniel Bressington
- School of Nursing, Faculty of Health, Charles Darwin University, Ellengowan Drive, Casuarina, NT, Australia; Faculty of Nursing, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Meng-Ling Chen
- Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Hou-Qiang Huang
- Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Tao Wang
- School of Nursing (Brisbane Centre), Faculty of Health, Charles Darwin University, Brisbane, QLD, Australia.
| | - Jing-Yu Benjamin Tan
- School of Nursing and Midwifery & Centre for Health Research, University of Southern Queensland, Ipswich, QLD, Australia
| |
Collapse
|
21
|
Petersen JCG, Becker R, Petersen LG. Transcutaneous vagal nerve stimulation during lower body negative pressure. Auton Neurosci 2024; 254:103192. [PMID: 38896931 DOI: 10.1016/j.autneu.2024.103192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/04/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Para-sympathetic vagal activation has profound influence on heart rate and other cardiovascular parameters. We tested the hypothesis that transcutaneous Vagal Nerve Stimulation (tVNS) through the auricular branch of the vagus nerve would attenuate the normal sympathetic response to central blood volume reduction by lower body negative pressure (LBNP). METHOD 10 healthy volunteers (6 female; age 21 ± 2 years; weight 62 ± 13 kg; height 167 ± 12 cm) were included in this cross-over design trial. After 15 min rest in supine position, subjects underwent three 15-min periods of 30 mmHg LBNP intervention with and without cyclic tVNS stimulation. Continuous cardiovascular parameters (Nexfin) were recorded. RESULTS Overall tVNS did not convincingly attenuate sympathetic response to central hypovolemia. Deactivation of the tVNS during LBNP resulted in increased MAP at 2.3 ± 0.5 mmHg (P < 0.001). Comparing the cyclic actual active stimulation periods to periods with pause during tVNS intervention showed a decrease in HR by 72.9 ± 11.2 to 70.2 ± 11.6 bpm (mean ± SD; P < 0.05), and concomitant increases in SV (86.0 ± 12.1 to 87.2 ± 12.6 mL; P < 0.05), MAP (82.9 ± 6.3 to 84.0 ± 6.2 mmHg; P < 0.05) and TPR (1116.0 ± 111.1 to 1153 ± 104.8 dyn*s/cm5; P < 0.05). CONCLUSION tVNS in 30 s cycles during LBNP can selectively attenuate HR, prompting a compensatory augmented sympathetic response. It would appear the method used in this study at least, has an isolated cardiac inhibitory effect probably mediated by augmented vagal activity on the sinoatrial or atrio-ventricular node, possibly in combination with reduced activity in the sympathetic cardiac nerve.
Collapse
Affiliation(s)
| | | | - Lonnie G Petersen
- Harvard Medical School, Cambridge, MA, USA; Massachusetts Institute of Technology, Cambridge, MA, USA; University of Copenhagen, Denmark
| |
Collapse
|
22
|
Demircioğlu G, Özden AV, Genç H. Comparison of the efficacy of auricular vagus nerve stimulation and conventional low back rehabilitation in patients with chronic low back pain. Complement Ther Clin Pract 2024; 56:101862. [PMID: 38815433 DOI: 10.1016/j.ctcp.2024.101862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/19/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND In recent years, human and animal studies have provided increasing evidence that vagus nerve stimulation (VNS) can produce analgesic effects as well as alleviating resistant epilepsy and depression. Our study was designed to compare the efficacy of transcutaneous auricular vagus nerve stimulation with conventional low back rehabilitation in patients with chronic low back pain (CLBP). METHODS Sixty patients with LBP were randomly divided into two groups. Group 1 received conventional rehabilitation and home exercise, and Group 2 received transcutaneous auricular VNS and home exercise. Both groups received treatment five days a week for three weeks. Trunk mobility (Modified Schober test, fingertip-to-floor test), muscle strength (CSMI-Cybex Humac-Norm isokinetic dynamometer and Lafayette manual muscle strength measuring device), trunk endurance, balance tests, Visual Analog Scale, Beck Depression Scale, Pittsburgh Sleep Quality Index, Oswestry Disability Index were evaluated. RESULTS At the end of three weeks, within-group assessment results showed positive effects on mobility, functional status, depression and sleep in all groups (p < 0.05). Pain level, endurance time and flexion trunk muscle strength results showed more improvement in Group 2 (p < 0.05). Some parameters of isokinetic lower extremity quadriceps muscle strength and fall risk scores showed a significant improvement in Group 1 (p < 0.05). DISCUSSION VNS has been observed to be more effective on pain, trunk muscle strength and endurance duration and sleep status. Auricular VNS may be included in the treatment of patients with CLBP in whom conventional physical therapy is inadequate or not applicable.
Collapse
Affiliation(s)
- Gamze Demircioğlu
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Atlas University, Turkey; Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Bahçeşehir University, Turkey.
| | - Ali Veysel Özden
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Bahçeşehir University, Turkey.
| | - Hazal Genç
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Bahçeşehir University, Turkey.
| |
Collapse
|
23
|
Likar R, Perruchoud C, Kampusch S, Köstenberger M, Sator S, Stremnitzer C, Wolf A, Neuwersch-Sommeregger S. [Clinical efficacy of auricular vagus nerve stimulation in the treatment of chronic and acute pain : A systematic review]. Schmerz 2024; 38:267-278. [PMID: 36592212 PMCID: PMC11271441 DOI: 10.1007/s00482-022-00686-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2022] [Indexed: 01/03/2023]
Abstract
BACKGROUND Current guidelines recommend a personalized, multimodal, and interdisciplinary approach for the treatment of chronic pain. Already in the acute treatment of postoperative pain, it can be useful to minimize risk factors for chronification. Auricular vagus nerve stimulation (aVNS) could be an effective non-drug therapy for the treatment of chronic and acute pain. AIM OF THE WORK The aim of this systematic review is to evaluate the clinical efficacy of aVNS in chronic and acute pain as well as its effect on medication intake. MATERIALS AND METHODS A systematic literature search was carried out on the application of auricular electrical stimulation in chronic and acute pain. Studies were classified according to their level of evidence and evaluated via the Jadad scale as well as their scientific validity, and then analyzed in terms of indication, method, stimulation parameters, duration of treatment, efficacy, and safety. RESULTS Twenty studies on chronic pain indications, ten studies on acute postoperative pain, as well as seven studies on experimental acute pain were identified and analyzed. The search revealed a total of n = 1105 aVNS-treated patients. The best evidence on the efficacy of aVNS is available for the indications chronic low back pain, chronic cervical syndrome, chronic abdominal pain, and chronic migraine as well as acute postoperative pain in oocyte aspiration, laparoscopic nephrectomy, and open colorectal surgery. Additionally a significant reduction in analgesic or opiate intake was evident in most studies. In three randomized controlled trials in chronic pain patients, a sustainable pain reduction over a period of up to 12 months was shown. Overall, aVNS was very well tolerated. CONCLUSION This review indicates that aVNS can be a complementary and effective non-drug treatment for patients with chronic and acute postoperative pain. Future studies in these indications should focus on standardizing and optimizing treatment parameters, inclusion of quality-of-life outcome parameters, and longer follow-up periods to better understand the sustainable therapeutic effect of aVNS.
Collapse
Affiliation(s)
- Rudolf Likar
- Abteilung für Anästhesiologie und Intensivmedizin, Klinikum Klagenfurt am Wörthersee, Feschnigstraße 11, 9020, Klagenfurt, Österreich.
- Paracelsus Medizinische Privatuniversität, Salzburg, Österreich.
| | | | | | - Markus Köstenberger
- Abteilung für Anästhesiologie und Intensivmedizin, Klinikum Klagenfurt am Wörthersee, Feschnigstraße 11, 9020, Klagenfurt, Österreich
| | - Sabine Sator
- Universitätsklinik für Anästhesie, allgemeine Intensivmedizin und Schmerztherapie, Medizinische Universität Wien, Wien, Österreich
| | | | - Andreas Wolf
- Abteilung für Anästhesie, Krankenhaus St. Vinzenz Zams, Zams, Österreich
| | - Stefan Neuwersch-Sommeregger
- Abteilung für Anästhesiologie und Intensivmedizin, Klinikum Klagenfurt am Wörthersee, Feschnigstraße 11, 9020, Klagenfurt, Österreich
- Medizinische Universität Graz, Graz, Österreich
| |
Collapse
|
24
|
E R, Wang Y. Vagus Nerve Stimulation for Improvement of Vascular Cognitive Impairment. Neuropsychiatr Dis Treat 2024; 20:1445-1451. [PMID: 39072312 PMCID: PMC11283790 DOI: 10.2147/ndt.s465249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/13/2024] [Indexed: 07/30/2024] Open
Abstract
Vagus nerve stimulation (VNS) is acknowledged as a highly effective therapy for various neurological conditions, including refractory epilepsy, depression, Alzheimer's disease (AD), migraine, and stroke. Presently, there is an increasing focus on understanding the impact of VNS on cognitive aspects. Numerous studies suggest that VNS suppresses the body's inflammatory response, leading to enhanced cognitive function in patients. Vascular cognitive impairment (VCI) is a severe cognitive dysfunction syndrome resulting from prolonged chronic cerebral hypoperfusion (CCH), where the primary pathogenesis is CCH-induced neuroinflammation. In this paper, we present a comprehensive overview of the research advancements in using VNS for treating VCI and discuss that VNS improves cognitive function in VCI patients by suppressing neuroinflammation, offering insights into a potential novel approach for addressing this condition.
Collapse
Affiliation(s)
- Ridengnaxi E
- Department of Rehabilitation Medicine, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, People’s Republic of China
| | - Yan Wang
- Department of Rehabilitation Medicine, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, People’s Republic of China
| |
Collapse
|
25
|
Bustamante C, Pinilla Bonilla LB, Restrepo JC. Neurological symphony: post-acute COVID-19 syndrome, an innovative pathophysiological exploration from neuraltherapeutic medicine. Front Integr Neurosci 2024; 18:1417856. [PMID: 39070159 PMCID: PMC11275269 DOI: 10.3389/fnint.2024.1417856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/20/2024] [Indexed: 07/30/2024] Open
Abstract
The SARS-CoV-2 pandemic has affected 771 million people and caused 6.9 million confirmed deaths as of November 2023. Beyond the adversity, a crucial and less-explored chapter unfolds: adaptive sequelae. These have altered social, mental, and emotional conditions, leaving an imprint on biological systems. While some cases fully resolve the pathological process post-acute infection, others persist with symptoms, posing a challenge that underscores the need to comprehend pathophysiology from innovative perspectives. The article delves into “Long COVID” or Post-Acute COVID-19 Syndrome (PACS), where symptoms persist for ≥4 weeks irrespective of initial severity. Risk factors include a history of severe illness, in-hospital management, and intensive care. This article also explores theories, derived from various experimental models, that have demonstrated the involvement of the nervous system in coordination with the psychoneuroimmunoendocrine axes in the expression of inflammation. It is posited that PACS involves processes of peripheral and central sensitization (corticalization), facilitating dishomeostasis and the chronicity of the inflammatory process. In this context, various therapeutic strategies grounded in modulating the inflammatory reflex are reviewed, primarily through the infiltration of local anesthetics via linear and non-linear approaches. Neural therapeutic use is considered to stimulate the regulatory inflammatory circuits coordinated by the neuroimmune-endocrine system.
Collapse
Affiliation(s)
- Carlos Bustamante
- Institute for Advanced Integrative Medicine, Medellin, Colombia
- Faculty of Medicine, National University of Colombia, Bogotá, Colombia
| | - Laura Bibiana Pinilla Bonilla
- Institute for Advanced Integrative Medicine, Medellin, Colombia
- Research and Innovation Center, Integrated Subnetwork of Health Services of South, Bogotá, Colombia
| | | |
Collapse
|
26
|
Natarajan C, Le LHD, Gunasekaran M, Tracey KJ, Chernoff D, Levine YA. Electrical stimulation of the vagus nerve ameliorates inflammation and disease activity in a rat EAE model of multiple sclerosis. Proc Natl Acad Sci U S A 2024; 121:e2322577121. [PMID: 38968104 PMCID: PMC11252997 DOI: 10.1073/pnas.2322577121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/31/2024] [Indexed: 07/07/2024] Open
Abstract
Multiple sclerosis (MS) is a demyelinating central nervous system (CNS) disorder that is associated with functional impairment and accruing disability. There are multiple U.S. Food and Drug Administration (FDA)-approved drugs that effectively dampen inflammation and slow disability progression. However, these agents do not work well for all patients and are associated with side effects that may limit their use. The vagus nerve (VN) provides a direct communication conduit between the CNS and the periphery, and modulation of the inflammatory reflex via electrical stimulation of the VN (VNS) shows efficacy in ameliorating pathology in several CNS and autoimmune disorders. We therefore investigated the impact of VNS in a rat experimental autoimmune encephalomyelitis (EAE) model of MS. In this study, VNS-mediated neuroimmune modulation is demonstrated to effectively decrease EAE disease severity and duration, infiltration of neutrophils and pathogenic lymphocytes, myelin damage, blood-brain barrier disruption, fibrinogen deposition, and proinflammatory microglial activation. VNS modulates expression of genes that are implicated in MS pathogenesis, as well as those encoding myelin proteins and transcription factors regulating new myelin synthesis. Together, these data indicate that neuroimmune modulation via VNS may be a promising approach to treat MS, that not only ameliorates symptoms but potentially also promotes myelin repair (remyelination).
Collapse
Affiliation(s)
| | | | | | - Kevin J. Tracey
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY11030
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY11549
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY11549
| | | | - Yaakov A. Levine
- SetPoint Medical, Valencia, CA91355
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY11549
- Division of Cardiovascular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm171 76, Sweden
| |
Collapse
|
27
|
Liu F, Habibollahi M, Wu Y, Neshatvar N, Zhang J, Zinno C, Akouissi O, Bernini F, Alibrandi L, Gabisonia K, Lionetti V, Carpaneto J, Lancashire H, Jiang D, Micera S, Demosthenous A. A multi-channel stimulator with an active electrode array implant for vagal-cardiac neuromodulation studies. Bioelectron Med 2024; 10:16. [PMID: 38970083 PMCID: PMC11227238 DOI: 10.1186/s42234-024-00148-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/21/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Implantable vagus nerve stimulation is a promising approach for restoring autonomic cardiovascular functions after heart transplantation. For successful treatment a system should have multiple electrodes to deliver precise stimulation and complex neuromodulation patterns. METHODS This paper presents an implantable multi-channel stimulation system for vagal-cardiac neuromodulation studies in swine species. The system comprises an active electrode array implant percutaneously connected to an external wearable controller. The active electrode array implant has an integrated stimulator ASIC mounted on a ceramic substrate connected to an intraneural electrode array via micro-rivet bonding. The implant is silicone encapsulated for biocompatibility and implanted lifetime. The stimulation parameters are remotely transmitted via a Bluetooth telemetry link. RESULTS The size of the encapsulated active electrode array implant is 8 mm × 10 mm × 3 mm. The stimulator ASIC has 10-bit current amplitude resolution and 16 independent output channels, each capable of delivering up to 550 µA stimulus current and a maximum voltage of 20 V. The active electrode array implant was subjected to in vitro accelerated lifetime testing at 70 °C for 7 days with no degradation in performance. After over 2 h continuous stimulation, the surface temperature change of the implant was less than 0.5 °C. In addition, in vivo testing on the sciatic nerve of a male Göttingen minipig demonstrated that the implant could effectively elicit an EMG response that grew progressively stronger on increasing the amplitude of the stimulation. CONCLUSIONS The multi-channel stimulator is suitable for long term implantation. It shows potential as a useful tool in vagal-cardiac neuromodulation studies in animal models for restoring autonomic cardiovascular functions after heart transplantation.
Collapse
Affiliation(s)
- Fangqi Liu
- Department of Electronic and Electrical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
| | - Maryam Habibollahi
- Department of Electronic and Electrical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
| | - Yu Wu
- Department of Electronic and Electrical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
| | - Nazanin Neshatvar
- Department of Electronic and Electrical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
| | - Jiaxing Zhang
- Department of Electronic and Electrical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
| | - Ciro Zinno
- BioRobotics Institute, Scuola Superiore Sant'Anna (SSSA), 56025, Pisa, Italy
| | | | - Fabio Bernini
- BioMedLab, Scuola Superiore Sant'Anna (SSSA), Pisa, Italy
| | - Lisa Alibrandi
- BioMedLab, Scuola Superiore Sant'Anna (SSSA), Pisa, Italy
| | | | | | - Jacopo Carpaneto
- BioRobotics Institute, Scuola Superiore Sant'Anna (SSSA), 56025, Pisa, Italy
| | - Henry Lancashire
- Department of Medical Physics and Bioengineering, University College London, Gower Street, London, WC1E 6BT, UK
| | - Dai Jiang
- Department of Electronic and Electrical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
| | - Silvestro Micera
- BioRobotics Institute, Scuola Superiore Sant'Anna (SSSA), 56025, Pisa, Italy
| | - Andreas Demosthenous
- Department of Electronic and Electrical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK.
| |
Collapse
|
28
|
Zafeiropoulos S, Ahmed U, Bikou A, Mughrabi IT, Stavrakis S, Zanos S. Vagus nerve stimulation for cardiovascular diseases: Is there light at the end of the tunnel? Trends Cardiovasc Med 2024; 34:327-337. [PMID: 37506989 DOI: 10.1016/j.tcm.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/12/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023]
Abstract
Autonomic dysfunction and chronic inflammation contribute to the pathogenesis and progression of several cardiovascular diseases (CVD), such as heart failure with preserved ejection fraction, atherosclerotic CVD, pulmonary arterial hypertension, and atrial fibrillation. The vagus nerve provides parasympathetic innervation to the heart, vessels, and lungs, and is also implicated in the neural control of inflammation through a neuroimmune pathway involving the spleen. Stimulation of the vagus nerve (VNS) can in principle restore autonomic balance and suppress inflammation, with potential therapeutic benefits in these diseases. Although VNS ameliorated CVD in several animal models, early human studies have demonstrated variable efficacy. The purpose of this review is to discuss the rationale behind the use of VNS in the treatment of CVD, to critically review animal and human studies of VNS in CVD, and to propose possible means to overcome the challenges in the clinical translation of VNS in CVD.
Collapse
Affiliation(s)
- Stefanos Zafeiropoulos
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, NY, USA; Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Umair Ahmed
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Alexia Bikou
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Ibrahim T Mughrabi
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Stavros Stavrakis
- Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stavros Zanos
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, NY, USA; Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA.
| |
Collapse
|
29
|
Trevizan-Baú P, McAllen RM. What is the Vagal-Adrenal Axis? J Comp Neurol 2024; 532:e25656. [PMID: 38980012 DOI: 10.1002/cne.25656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024]
Abstract
Some recent publications have used the term "vagal-adrenal axis" to account for mechanisms involved in the regulation of inflammation by electroacupuncture. This concept proposes that efferent parasympathetic nerve fibers in the vagus directly innervate the adrenal glands to influence catecholamine secretion. Here, we discuss evidence for anatomical and functional links between the vagi and adrenal glands that may be relevant in the context of inflammation and its neural control by factors, including acupuncture. First, we find that evidence for any direct vagal parasympathetic efferent innervation of the adrenal glands is weak and likely artifactual. Second, we find good evidence that vagal afferent fibers directly innervate the adrenal gland, although their function is uncertain. Third, we highlight a wealth of evidence for indirect pathways, whereby vagal afferent signals act via the central nervous system to modify adrenal-dependent anti-inflammatory responses. Vagal afferents, not efferents, are thus the likely key to these phenomena.
Collapse
Affiliation(s)
- Pedro Trevizan-Baú
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA
| | - Robin M McAllen
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
30
|
Sévoz-Couche C, Liao W, Foo HYC, Bonne I, Lu TB, Tan Qi Hui C, Azhar SH, Peh WYX, Yen SC, Wong WSF. Direct vagus nerve stimulation: A new tool to control allergic airway inflammation through α7 nicotinic acetylcholine receptor. Br J Pharmacol 2024; 181:1916-1934. [PMID: 38430056 DOI: 10.1111/bph.16334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 11/24/2023] [Accepted: 01/08/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND AND PURPOSE Asthma is characterized by airway inflammation, mucus hypersecretion, and airway hyperresponsiveness. The use of nicotinic agents to mimic the cholinergic anti-inflammatory pathway (CAP) controls experimental asthma. Yet, the effects of vagus nerve stimulation (VNS)-induced CAP on allergic inflammation remain unknown. EXPERIMENTAL APPROACH BALB/c mice were sensitized and challenged with house dust mite (HDM) extract and treated with active VNS (5 Hz, 0.5 ms, 0.05-1 mA). Bronchoalveolar lavage (BAL) fluid was assessed for total and differential cell counts and cytokine levels. Lungs were examined by histopathology and electron microscopy. KEY RESULTS In the HDM mouse asthma model, VNS at intensities equal to or above 0.1 mA (VNS 0.1) but not sham VNS reduced BAL fluid differential cell counts and alveolar macrophages expressing α7 nicotinic receptors (α7nAChR), goblet cell hyperplasia, and collagen deposition. Besides, VNS 0.1 also abated HDM-induced elevation of type 2 cytokines IL-4 and IL-5 and was found to block the phosphorylation of transcription factor STAT6 and expression level of IRF4 in total lung lysates. Finally, VNS 0.1 abrogated methacholine-induced hyperresponsiveness in asthma mice. Prior administration of α-bungarotoxin, a specific inhibitor of α7nAChR, but not propranolol, a specific inhibitor of β2-adrenoceptors, abolished the therapeutic effects of VNS 0.1. CONCLUSION AND IMPLICATIONS Our data revealed the protective effects of VNS on various clinical features in allergic airway inflammation model. VNS, a clinically approved therapy for depression and epilepsy, appears to be a promising new strategy for controlling allergic asthma.
Collapse
Affiliation(s)
- Caroline Sévoz-Couche
- INSERM, UMRS1158 Neurophysiologie Respiratoire et Clinique, Sorbonne Université, Paris, France
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wupeng Liao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Singapore-HUJ Alliance for Research and Enterprise (SHARE), National University of Singapore, Singapore
| | - Hazel Y C Foo
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Singapore-HUJ Alliance for Research and Enterprise (SHARE), National University of Singapore, Singapore
| | - Isabelle Bonne
- Electron Microscopy Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Thong Beng Lu
- Electron Microscopy Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Caris Tan Qi Hui
- Advanced imaging and Histology Core, Immunology Program, Life Science Institute, National University of Singapore, Singapore
| | - Syaza Hazwany Azhar
- Advanced imaging and Histology Core, Immunology Program, Life Science Institute, National University of Singapore, Singapore
| | - Wendy Yen Xian Peh
- The N.1 Institute for Health, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
| | - Shih-Cheng Yen
- The N.1 Institute for Health, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Singapore-HUJ Alliance for Research and Enterprise (SHARE), National University of Singapore, Singapore
- Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
31
|
Santi S, Fuggetta F, Colicchio G, D’Ercole M, Izzo A, D’Alessandris QG, Burattini B, Martinelli R, Montano N. Long-Term Outcome in Adult Patients with Drug-Resistant Epilepsy Submitted to Vagus Nerve Stimulation. Brain Sci 2024; 14:639. [PMID: 39061382 PMCID: PMC11274406 DOI: 10.3390/brainsci14070639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Epilepsy treatment primarily involves antiseizure medications (ASMs) to eliminate seizures and improve the quality of life, but many patients develop drug-resistant epilepsy (DRE), necessitating alternative interventions. This study aimed to evaluate the long-term efficacy and safety of vagus nerve stimulation (VNS) in managing DRE. We retrospectively analyzed data from 105 adult patients treated at Agostino Gemelli Hospital from 1994 to 2022. Among the 73 patients with follow-up data, 80.8% were responders, experiencing significant reductions in seizure frequency over an average follow-up period of 9.4 years. Although 19.2% were non-responders, many of these patients still opted for generator replacements due to improvements in quality of life, such as fewer falls and shorter post-ictal periods. The overall complication rate was 12.3%, with most complications being mild and manageable. These findings suggest that VNS offers substantial long-term benefits for patients with DRE, improving seizure control and quality of life. This study underscores the importance of VNS as a viable long-term treatment option for DRE, highlighting its potential to significantly enhance patient outcomes and quality of life.
Collapse
Affiliation(s)
- Samuele Santi
- Department of Neuroscience, Neurosurgery Section, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.S.); (G.C.); (Q.G.D.); (B.B.); (R.M.)
| | - Filomena Fuggetta
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (F.F.); (M.D.); (A.I.)
| | - Gabriella Colicchio
- Department of Neuroscience, Neurosurgery Section, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.S.); (G.C.); (Q.G.D.); (B.B.); (R.M.)
| | - Manuela D’Ercole
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (F.F.); (M.D.); (A.I.)
| | - Alessandro Izzo
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (F.F.); (M.D.); (A.I.)
| | - Quintino Giorgio D’Alessandris
- Department of Neuroscience, Neurosurgery Section, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.S.); (G.C.); (Q.G.D.); (B.B.); (R.M.)
| | - Benedetta Burattini
- Department of Neuroscience, Neurosurgery Section, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.S.); (G.C.); (Q.G.D.); (B.B.); (R.M.)
| | - Renata Martinelli
- Department of Neuroscience, Neurosurgery Section, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.S.); (G.C.); (Q.G.D.); (B.B.); (R.M.)
| | - Nicola Montano
- Department of Neuroscience, Neurosurgery Section, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.S.); (G.C.); (Q.G.D.); (B.B.); (R.M.)
| |
Collapse
|
32
|
Berthon A, Wernisch L, Stoukidi M, Thornton M, Tessier-Lariviere O, Fortier-Poisson P, Mamen J, Pinkney M, Lee S, Sarkans E, Annecchino L, Appleton B, Garsed P, Patterson B, Gonshaw S, Jakopec M, Shunmugam S, Edwards T, Tukiainen A, Jennings J, Lajoie G, Hewage E, Armitage O. Using neural biomarkers to personalize dosing of vagus nerve stimulation. Bioelectron Med 2024; 10:15. [PMID: 38880906 PMCID: PMC11181600 DOI: 10.1186/s42234-024-00147-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Vagus nerve stimulation (VNS) is an established therapy for treating a variety of chronic diseases, such as epilepsy, depression, obesity, and for stroke rehabilitation. However, lack of precision and side-effects have hindered its efficacy and extension to new conditions. Achieving a better understanding of the relationship between VNS parameters and neural and physiological responses is therefore necessary to enable the design of personalized dosing procedures and improve precision and efficacy of VNS therapies. METHODS We used biomarkers from recorded evoked fiber activity and short-term physiological responses (throat muscle, cardiac and respiratory activity) to understand the response to a wide range of VNS parameters in anaesthetised pigs. Using signal processing, Gaussian processes (GP) and parametric regression models we analyse the relationship between VNS parameters and neural and physiological responses. RESULTS Firstly, we illustrate how considering multiple stimulation parameters in VNS dosing can improve the efficacy and precision of VNS therapies. Secondly, we describe the relationship between different VNS parameters and the evoked fiber activity and show how spatially selective electrodes can be used to improve fiber recruitment. Thirdly, we provide a detailed exploration of the relationship between the activations of neural fiber types and different physiological effects. Finally, based on these results, we discuss how recordings of evoked fiber activity can help design VNS dosing procedures that optimize short-term physiological effects safely and efficiently. CONCLUSION Understanding of evoked fiber activity during VNS provide powerful biomarkers that could improve the precision, safety and efficacy of VNS therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Guillaume Lajoie
- Université de Montréal and Mila-Quebec AI Institute, Montréal, Canada
| | | | | |
Collapse
|
33
|
Lei J, Tang LL, You HJ. Pathological pain: Non-motor manifestations in Parkinson disease and its treatment. Neurosci Biobehav Rev 2024; 161:105646. [PMID: 38569983 DOI: 10.1016/j.neubiorev.2024.105646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/13/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024]
Abstract
In addition to motor symptoms, non-motor manifestations of Parkinson's disease (PD), i.e. pain, depression, sleep disturbance, and autonomic disorders, have received increasing attention. As one of the non-motor symptoms, pain has a high prevalence and is considered an early pre-motor symptom in the development of PD. In relation to pathological pain and its management in PD, particularly in the early stages, it is hypothesized that the loss of dopaminergic neurons causes a functional deficit in supraspinal structures, leading to an imbalance in endogenous descending modulation. Deficits in dopaminergic-dependent pathways also affect non-dopaminergic neurotransmitter systems that contribute to the pathological processing of nociceptive input, the integration, and modulation of pain in PD. This review examines the onset and progression of pain in PD, with a particular focus on alterations in the central modulation of nociception. The discussion highlights the importance of abnormal endogenous descending facilitation and inhibition in PD pain, which may provide potential clues to a better understanding of the nature of pathological pain and its effective clinical management.
Collapse
Affiliation(s)
- Jing Lei
- Center for Translational Medicine Research on Sensory-Motor Diseases, Yan'an University, Yan'an 716000, China; Key Laboratory of Yan'an Sports Rehabilitation Medicine, Yan'an 716000, China
| | - Lin-Lin Tang
- Center for Translational Medicine Research on Sensory-Motor Diseases, Yan'an University, Yan'an 716000, China
| | - Hao-Jun You
- Center for Translational Medicine Research on Sensory-Motor Diseases, Yan'an University, Yan'an 716000, China; Key Laboratory of Yan'an Sports Rehabilitation Medicine, Yan'an 716000, China.
| |
Collapse
|
34
|
Kang C, Zhang J, Li X, Liu T. Optimal stimulating site and the minimum current intensity of subdiaphragmatic vagal stimulation to promote gastric peristalsis in swine. Asian J Surg 2024; 47:2219-2221. [PMID: 38331615 DOI: 10.1016/j.asjsur.2024.01.120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/19/2024] [Indexed: 02/10/2024] Open
Affiliation(s)
- Chunbo Kang
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital of Capital Medical University, Beijing, 100144, China
| | - Jie Zhang
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital of Capital Medical University, Beijing, 100144, China
| | - Xiaowei Li
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital of Capital Medical University, Beijing, 100144, China
| | - Tiejun Liu
- Department of General Surgery, Beijing Rehabilitation Hospital of Capital Medical University, Beijing, 100144, China.
| |
Collapse
|
35
|
Kronsteiner B, Carrero-Rojas G, Reissig LF, Moghaddam AS, Schwendt KM, Gerges S, Maierhofer U, Aszmann OC, Pastor AM, Kiss A, Podesser BK, Birkfellner W, Moscato F, Blumer R, Weninger WJ. Characterization, number, and spatial organization of nerve fibers in the human cervical vagus nerve and its superior cardiac branch. Brain Stimul 2024; 17:510-524. [PMID: 38677543 DOI: 10.1016/j.brs.2024.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/11/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Electrical stimulation of the vagus nerve (VN) is a therapy for epilepsy, obesity, depression, and heart diseases. However, whole nerve stimulation leads to side effects. We examined the neuroanatomy of the mid-cervical segment of the human VN and its superior cardiac branch to gain insight into the side effects of VN stimulation and aid in developing targeted stimulation strategies. METHODS Nerve specimens were harvested from eight human body donors, then subjected to immunofluorescence and semiautomated quantification to determine the signature, quantity, and spatial distribution of different axonal categories. RESULTS The right and left cervical VN (cVN) contained a total of 25,489 ± 2781 and 23,286 ± 3164 fibers, respectively. Two-thirds of the fibers were unmyelinated and one-third were myelinated. About three-quarters of the fibers in the right and left cVN were sensory (73.9 ± 7.5 % versus 72.4 ± 5.6 %), while 13.2 ± 1.8 % versus 13.3 ± 3.0 % were special visceromotor and parasympathetic, and 13 ± 5.9 % versus 14.3 ± 4.0 % were sympathetic. Special visceromotor and parasympathetic fibers formed clusters. The superior cardiac branches comprised parasympathetic, vagal sensory, and sympathetic fibers with the left cardiac branch containing more sympathetic fibers than the right (62.7 ± 5.4 % versus 19.8 ± 13.3 %), and 50 % of the left branch contained sensory and sympathetic fibers only. CONCLUSION The study indicates that selective stimulation of vagal sensory and motor fibers is possible. However, it also highlights the potential risk of activating sympathetic fibers in the superior cardiac branch, especially on the left side.
Collapse
Affiliation(s)
- Bettina Kronsteiner
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria; Division of Anatomy, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Genova Carrero-Rojas
- Division of Anatomy, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Lukas F Reissig
- Division of Anatomy, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Atieh Seyedian Moghaddam
- Division of Anatomy, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Karoline M Schwendt
- Division of Anatomy, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Sylvia Gerges
- Division of Anatomy, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Udo Maierhofer
- Clinical Laboratory for Bionic Extremity Reconstruction, Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Oskar C Aszmann
- Clinical Laboratory for Bionic Extremity Reconstruction, Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria; Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Angel M Pastor
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012, Sevilla, Spain
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria; Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Austria
| | - Bruno K Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria; Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Austria
| | - Wolfgang Birkfellner
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Francesco Moscato
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Roland Blumer
- Division of Anatomy, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria.
| | - Wolfgang J Weninger
- Division of Anatomy, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
36
|
Sprunks TE, McLeod KJ, Staelin R. Pulsed shortwave electromagnetic field therapy increases quality of life in canines with symptoms of osteoarthritics. Vet Med Sci 2024; 10:e1408. [PMID: 38516818 PMCID: PMC10958403 DOI: 10.1002/vms3.1408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Joint stiffness, lameness and reduced activity levels are common inflammatory responses observed in canines and have significant impact on quality of life (QOL). The symptoms are often ascribed to osteoarthritis (OA), for which the standard treatment is systemic anti-inflammatories, but pharmacologic intervention can have significant short-term and long-term side effects. OBJECTIVES Test the efficacy of a Food and Drug Administration (FDA)-cleared pulsed shortwave therapy (PSWT) device as a means to modulate vagus nerve activity and initiate a systemic anti-inflammatory response to determine its ability to improve functionality and the QOL of canines with inflammatory symptoms commonly associated with OA. METHODS A randomized, double-blinded, placebo-controlled 14-day study of 60 dogs with a presumptive prior diagnosis of OA in at least one limb joint. Two outcomes assessing changes in the dog's QOL and functionality were measured: subjectively determined changes in eight behaviours associated with discomfort and objectively determined changes in passive range of motion (PROM). The device was secured near the cervico-thoracic region of the dog's spine. PROM measures were taken at baseline and at the end of study. Behavioural measures were taken daily. RESULTS Forty-nine animals completed the study. No negative side effects were reported. Average subjective discomfort scores for the treatment group (N = 26) were reduced from 3.74 to 2.10 (44%), compared to no improvement in the placebo group (N = 23) over the study period (p = 0.0001). Average PROM scores increased by 5.51 (4.59-6.23) degrees relative to the placebo group (p < 0.01). Ninety-six per cent of the treatment group showed either increased PROM or improved behavioural changes or both, compared to 4% for the placebo group (p < 0.01). Most changes occurred within the first 8 days of treatment. CONCLUSIONS PSWT applied at the level of the cervico-thoracic spine to target the vagus nerve may have the potential to improve QOL in dogs manifesting behaviours commonly associated with OA.
Collapse
Affiliation(s)
| | - Kenneth J. McLeod
- Department of Systems Science and Industrial EngineeringBinghamton UniversityBinghamtonNew YorkUSA
| | - Richard Staelin
- Fuqua School of BusinessDuke UniversityDurhamNorth CarolinaUSA
| |
Collapse
|
37
|
Davidson B, Bhattacharya A, Sarica C, Darmani G, Raies N, Chen R, Lozano AM. Neuromodulation techniques - From non-invasive brain stimulation to deep brain stimulation. Neurotherapeutics 2024; 21:e00330. [PMID: 38340524 PMCID: PMC11103220 DOI: 10.1016/j.neurot.2024.e00330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/14/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
Over the past 30 years, the field of neuromodulation has witnessed remarkable advancements. These developments encompass a spectrum of techniques, both non-invasive and invasive, that possess the ability to both probe and influence the central nervous system. In many cases neuromodulation therapies have been adopted into standard care treatments. Transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), and transcranial ultrasound stimulation (TUS) are the most common non-invasive methods in use today. Deep brain stimulation (DBS), spinal cord stimulation (SCS), and vagus nerve stimulation (VNS), are leading surgical methods for neuromodulation. Ongoing active clinical trials using are uncovering novel applications and paradigms for these interventions.
Collapse
Affiliation(s)
- Benjamin Davidson
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada
| | | | - Can Sarica
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada; Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Ghazaleh Darmani
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Nasem Raies
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Robert Chen
- Krembil Research Institute, University Health Network, Toronto, ON, Canada; Edmond J. Safra Program in Parkinson's Disease Morton and Gloria Shulman Movement Disorders Clinic, Division of Neurology, University of Toronto, Toronto, ON, Canada
| | - Andres M Lozano
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada; Krembil Research Institute, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
38
|
Mensah-Brown KG, Naylor RM, Graepel S, Brinjikji W. Neuromodulation: What the neurointerventionalist needs to know. Interv Neuroradiol 2024:15910199231224554. [PMID: 38454831 PMCID: PMC11569746 DOI: 10.1177/15910199231224554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 12/02/2023] [Indexed: 03/09/2024] Open
Abstract
Neuromodulation is the alteration of neural activity in the central, peripheral, or autonomic nervous systems. Consequently, this term lends itself to a variety of organ systems including but not limited to the cardiac, nervous, and even gastrointestinal systems. In this review, we provide a primer on neuromodulation, examining the various technological systems employed and neurological disorders targeted with this technology. Ultimately, we undergo a historical analysis of the field's development, pivotal discoveries and inventions gearing this review to neuro-adjacent subspecialties with a specific focus on neurointerventionalists.
Collapse
Affiliation(s)
| | - Ryan M. Naylor
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
39
|
Hofmann GC, Gama de Barcellos Filho P, Khodadadi F, Ostrowski D, Kline DD, Hasser EM. Vagotomy blunts cardiorespiratory responses to vagal afferent stimulation via pre- and postsynaptic effects in the nucleus tractus solitarii. J Physiol 2024; 602:1147-1174. [PMID: 38377124 DOI: 10.1113/jp285854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/29/2024] [Indexed: 02/22/2024] Open
Abstract
Viscerosensory information travels to the brain via vagal afferents, where it is first integrated within the brainstem nucleus tractus solitarii (nTS), a critical contributor to cardiorespiratory function and site of neuroplasticity. We have shown that decreasing input to the nTS via unilateral vagus nerve transection (vagotomy) induces morphological changes in nTS glia and reduces sighs during hypoxia. The mechanisms behind post-vagotomy changes are not well understood. We hypothesized that chronic vagotomy alters cardiorespiratory responses to vagal afferent stimulation via blunted nTS neuronal activity. Male Sprague-Dawley rats (6 weeks old) underwent right cervical vagotomy caudal to the nodose ganglion, or sham surgery. After 1 week, rats were anaesthetized, ventilated and instrumented to measure mean arterial pressure (MAP), heart rate (HR), and splanchnic sympathetic and phrenic nerve activity (SSNA and PhrNA, respectively). Vagal afferent stimulation (2-50 Hz) decreased cardiorespiratory parameters and increased neuronal Ca2+ measured by in vivo photometry and in vitro slice imaging of nTS GCaMP8m. Vagotomy attenuated both these reflex and neuronal Ca2+ responses compared to shams. Vagotomy also reduced presynaptic Ca2+ responses to stimulation (Cal-520 imaging) in the nTS slice. The decrease in HR, SSNA and PhrNA due to nTS nanoinjection of exogenous glutamate also was tempered following vagotomy. This effect was not restored by blocking excitatory amino acid transporters. However, the blunted responses were mimicked by NMDA, not AMPA, nanoinjection and were associated with reduced NR1 subunits in the nTS. Altogether, these results demonstrate that vagotomy induces multiple changes within the nTS tripartite synapse that influence cardiorespiratory reflex responses to afferent stimulation. KEY POINTS: Multiple mechanisms within the nucleus tractus solitarii (nTS) contribute to functional changes following vagal nerve transection. Vagotomy results in reduced cardiorespiratory reflex responses to vagal afferent stimulation and nTS glutamate nanoinjection. Blunted responses occur via reduced presynaptic Ca2+ activation and attenuated NMDA receptor expression and function, leading to a reduction in nTS neuronal activation. These results provide insight into the control of autonomic and respiratory function, as well as the plasticity that can occur in response to nerve damage and cardiorespiratory disease.
Collapse
Affiliation(s)
- Gabrielle C Hofmann
- Comparative Medicine, University of Missouri, Columbia, Missouri, USA
- Area Pathobiology, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Procopio Gama de Barcellos Filho
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Fateme Khodadadi
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Daniela Ostrowski
- Department of Pharmacology, A.T. Still University, Kirksville, Missouri, USA
| | - David D Kline
- Area Pathobiology, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
- Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - Eileen M Hasser
- Area Pathobiology, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
- Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
40
|
Long M, Li Y, He H, Gu N. The Story of Ferumoxytol: Synthesis Production, Current Clinical Applications, and Therapeutic Potential. Adv Healthc Mater 2024; 13:e2302773. [PMID: 37931150 DOI: 10.1002/adhm.202302773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/01/2023] [Indexed: 11/08/2023]
Abstract
Ferumoxytol, approved by the U.S. Food and Drug Administration in 2009, is one of the intravenous iron oxide nanoparticles authorized for the treatment of iron deficiency in chronic kidney disease and end-stage renal disease. With its exceptional magnetic properties, catalytic activity, and immune activity, as well as good biocompatibility and safety, ferumoxytol has gained significant recognition in various biomedical diagnoses and treatments. Unlike most existing reviews on this topic, this review primarily focuses on the recent clinical and preclinical advances of ferumoxytol in disease treatment, spanning anemia, cancer, infectious inflammatory diseases, regenerative medicine application, magnetic stimulation for neural modulation, etc. Additionally, the newly discovered mechanisms associated with the biological effects of ferumoxytol are discussed, including its magnetic, catalytic, and immunomodulatory properties. Finally, the summary and future prospects concerning the treatment and application of ferumoxytol-based nanotherapeutics are presented.
Collapse
Affiliation(s)
- Mengmeng Long
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Yan Li
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Hongliang He
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Ning Gu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
- Medical School, Nanjing University, Nanjing, 210008, P. R. China
| |
Collapse
|
41
|
Cai Y, Zhang Y, Fang Y, Hu H, Li X, Fang L. Evaluating the efficacy and acceptability of vagus nerve stimulation for fibromyalgia: a PRISMA-compliant protocol for a systematic review and meta-analysis. Front Neurol 2024; 15:1367295. [PMID: 38450074 PMCID: PMC10914987 DOI: 10.3389/fneur.2024.1367295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/12/2024] [Indexed: 03/08/2024] Open
Abstract
Background Fibromyalgia has imposed substantial burdens on patients' health and well-being, yet effective therapeutic options for this condition remain limited. Recently, vagus nerve stimulation (VNS) has emerged as a promising therapy for fibromyalgia. Nonetheless, despite the increasing number of randomized clinical trials (RCTs), current evidence remains inconclusive. Therefore, this protocol of a systematic review and meta-analysis aims to synthesize the existing evidence to clarify the efficacy and acceptability of VNS for treating fibromyalgia. Methods A comprehensive search for eligible RCTs will be conducted across nine bibliographic databases, namely PubMed, Cochrane Library, Embase, AMED, PsycINFO, PEDro, Chinese BioMedical Literature Database, Chinese National Knowledge Infrastructure, and Wangfang database. Data obtained from the included studies will be synthesized quantitively using RevMan 5.4.1 for meta-analyses. The methodological soundness of included RCTs will be assessed via the Cochrane's updated risk of bias tool (version 2.0). Additionally, sensitivity analyses, publication bias assessment, and subgroup analyses will be conducted as appropriate. Finally, we will utilize the Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) system to evaluate the certainty for the body of evidence. Conclusion The findings of our study are anticipated to ascertain the efficacy and acceptability of VNS as a promising treatment option for fibromyalgia. This will not only fill current research gap but also identify potential areas for future research. The findings will provide essential guidance for evidence-based treatment decisions for fibromyalgia, benefiting both patients and clinicians.
Collapse
Affiliation(s)
- Yunhuo Cai
- Department of Rehabilitation, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yajun Zhang
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiyan Fang
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hantong Hu
- Department of Acupuncture and Moxibustion, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xingling Li
- Department of Acupuncture and Moxibustion, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Lianqiang Fang
- Department of Acupuncture and Moxibustion, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
42
|
González-González MA, Conde SV, Latorre R, Thébault SC, Pratelli M, Spitzer NC, Verkhratsky A, Tremblay MÈ, Akcora CG, Hernández-Reynoso AG, Ecker M, Coates J, Vincent KL, Ma B. Bioelectronic Medicine: a multidisciplinary roadmap from biophysics to precision therapies. Front Integr Neurosci 2024; 18:1321872. [PMID: 38440417 PMCID: PMC10911101 DOI: 10.3389/fnint.2024.1321872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/10/2024] [Indexed: 03/06/2024] Open
Abstract
Bioelectronic Medicine stands as an emerging field that rapidly evolves and offers distinctive clinical benefits, alongside unique challenges. It consists of the modulation of the nervous system by precise delivery of electrical current for the treatment of clinical conditions, such as post-stroke movement recovery or drug-resistant disorders. The unquestionable clinical impact of Bioelectronic Medicine is underscored by the successful translation to humans in the last decades, and the long list of preclinical studies. Given the emergency of accelerating the progress in new neuromodulation treatments (i.e., drug-resistant hypertension, autoimmune and degenerative diseases), collaboration between multiple fields is imperative. This work intends to foster multidisciplinary work and bring together different fields to provide the fundamental basis underlying Bioelectronic Medicine. In this review we will go from the biophysics of the cell membrane, which we consider the inner core of neuromodulation, to patient care. We will discuss the recently discovered mechanism of neurotransmission switching and how it will impact neuromodulation design, and we will provide an update on neuronal and glial basis in health and disease. The advances in biomedical technology have facilitated the collection of large amounts of data, thereby introducing new challenges in data analysis. We will discuss the current approaches and challenges in high throughput data analysis, encompassing big data, networks, artificial intelligence, and internet of things. Emphasis will be placed on understanding the electrochemical properties of neural interfaces, along with the integration of biocompatible and reliable materials and compliance with biomedical regulations for translational applications. Preclinical validation is foundational to the translational process, and we will discuss the critical aspects of such animal studies. Finally, we will focus on the patient point-of-care and challenges in neuromodulation as the ultimate goal of bioelectronic medicine. This review is a call to scientists from different fields to work together with a common endeavor: accelerate the decoding and modulation of the nervous system in a new era of therapeutic possibilities.
Collapse
Affiliation(s)
- María Alejandra González-González
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatric Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Silvia V. Conde
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NOVA University, Lisbon, Portugal
| | - Ramon Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Stéphanie C. Thébault
- Laboratorio de Investigación Traslacional en salud visual (D-13), Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| | - Marta Pratelli
- Neurobiology Department, Kavli Institute for Brain and Mind, UC San Diego, La Jolla, CA, United States
| | - Nicholas C. Spitzer
- Neurobiology Department, Kavli Institute for Brain and Mind, UC San Diego, La Jolla, CA, United States
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- International Collaborative Center on Big Science Plan for Purinergic Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Cuneyt G. Akcora
- Department of Computer Science, University of Central Florida, Orlando, FL, United States
| | | | - Melanie Ecker
- Department of Biomedical Engineering, University of North Texas, Denton, TX, United States
| | | | - Kathleen L. Vincent
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, United States
| | - Brandy Ma
- Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
43
|
Mondal B, Choudhury S, Banerjee R, Roy A, Chatterjee K, Basu P, Singh R, Halder S, Shubham S, Baker SN, Baker MR, Kumar H. Effects of non-invasive vagus nerve stimulation on clinical symptoms and molecular biomarkers in Parkinson's disease. Front Aging Neurosci 2024; 15:1331575. [PMID: 38384731 PMCID: PMC10879328 DOI: 10.3389/fnagi.2023.1331575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/20/2023] [Indexed: 02/23/2024] Open
Abstract
Non-invasive vagus nerve stimulation (nVNS) is an established neurostimulation therapy used in the treatment of epilepsy, migraine and cluster headache. In this randomized, double-blind, sham-controlled trial we explored the role of nVNS in the treatment of gait and other motor symptoms in Parkinson's disease (PD) patients. In a subgroup of patients, we measured selected neurotrophins, inflammatory markers and markers of oxidative stress in serum. Thirty-three PD patients with freezing of gait (FOG) were randomized to either active nVNS or sham nVNS. After baseline assessments, patients were instructed to deliver six 2 min stimulations (12 min/day) of the active nVNS/sham nVNS device for 1 month at home. Patients were then re-assessed. After a one-month washout period, they were allocated to the alternate treatment arm and the same process was followed. Significant improvements in key gait parameters (speed, stance time and step length) were observed with active nVNS. While serum tumor necrosis factor- α decreased, glutathione and brain-derived neurotrophic factor levels increased significantly (p < 0.05) after active nVNS treatment. Here we present the first evidence of the efficacy and safety of nVNS in the treatment of gait in PD patients, and propose that nVNS can be used as an adjunctive therapy in the management of PD patients, especially those suffering from FOG. Clinical trial registration: identifier ISRCTN14797144.
Collapse
Affiliation(s)
| | | | | | - Akash Roy
- Institute of Neurosciences Kolkata, Kolkata, India
| | | | - Purba Basu
- Institute of Neurosciences Kolkata, Kolkata, India
| | - Ravi Singh
- Institute of Neurosciences Kolkata, Kolkata, India
| | | | | | - Stuart N. Baker
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Mark R. Baker
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- Department of Neurology, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
- Department of Clinical Neurophysiology, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | | |
Collapse
|
44
|
Sun K, Li Y, Zhai Z, Yin H, Liang S, Zhai F, Cui Y, Zhang G. Effects of transcutaneous auricular vagus nerve stimulation and exploration of brain network mechanisms in children with high-functioning autism spectrum disorder: study protocol for a randomized controlled trial. Front Psychiatry 2024; 15:1337101. [PMID: 38374975 PMCID: PMC10875019 DOI: 10.3389/fpsyt.2024.1337101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 01/19/2024] [Indexed: 02/21/2024] Open
Abstract
Background Autism Spectrum Disorders (ASD) are a collection of neurodevelopmental diseases characterized by poor social interaction and communication, a limited range of interests, and stereotyped behavior. High-functioning autism (HFA) indicates a subgroup of individuals with autism who possess cognitive and/or language skills that are within the average to above-normal range for their age. Transcutaneous auricular vagus nerve stimulation (taVNS) holds promise in children with HFA. However, few studies have used randomized controlled trials to validate the effectiveness of taVNS. Therefore, in this study, we intend to provide a study protocol to examine the therapeutic effects of taVNS in individuals diagnosed with HFA and to investigate the process of brain network remodeling in individuals with ASD using functional imaging techniques to observe alterations in large-scale neural networks. Methods and design We planned to employ a randomized, double-blind experimental design, including 40 children receiving sham stimulation and 40 children receiving real stimulation. We will assess clinical scales and perform functional imaging examinations before and after the stimulation. Additionally, we will include age- and gender-matched healthy children as controls and conduct functional imaging examinations. We plan first to observe the therapeutic effects of taVNS. Furthermore, we will observe the impact of taVNS stimulation on the brain network. Discussion taVNS was a low-risk, easy-to-administer, low-cost, and portable option to modulate the vagus system. taVNS may improve the social performance of HFA. Changes in the network properties of the large-scale brain network may be related to the efficacy of taVNS. Clinical trial registration http://www.chictr.org.cn, identifier ChiCTR2300074035.
Collapse
Affiliation(s)
- Ke Sun
- Functional Neurosurgery Department, National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Ying Li
- Department of Psychiatry, Beijing Children’s Hospital, Beijing, China
| | - Zhenhang Zhai
- Functional Neurosurgery Department, National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Heqing Yin
- Functional Neurosurgery Department, National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Shuli Liang
- Functional Neurosurgery Department, National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Feng Zhai
- Functional Neurosurgery Department, National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Yonghua Cui
- Department of Psychiatry, Beijing Children’s Hospital, Beijing, China
| | - Guojun Zhang
- Functional Neurosurgery Department, National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
45
|
Morales AW, Du J, Warren DJ, Fernández-Jover E, Martinez-Navarrete G, Bouteiller JMC, McCreery DC, Lazzi G. Machine learning enables non-Gaussian investigation of changes to peripheral nerves related to electrical stimulation. Sci Rep 2024; 14:2795. [PMID: 38307915 PMCID: PMC10837107 DOI: 10.1038/s41598-024-53284-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 01/30/2024] [Indexed: 02/04/2024] Open
Abstract
Electrical stimulation of the peripheral nervous system (PNS) is becoming increasingly important for the therapeutic treatment of numerous disorders. Thus, as peripheral nerves are increasingly the target of electrical stimulation, it is critical to determine how, and when, electrical stimulation results in anatomical changes in neural tissue. We introduce here a convolutional neural network and support vector machines for cell segmentation and analysis of histological samples of the sciatic nerve of rats stimulated with varying current intensities. We describe the methodologies and present results that highlight the validity of the approach: machine learning enabled highly efficient nerve measurement collection, while multivariate analysis revealed notable changes to nerves' anatomy, even when subjected to levels of stimulation thought to be safe according to the Shannon current limits.
Collapse
Affiliation(s)
- Andres W Morales
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA.
| | - Jinze Du
- Department of Electrical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - David J Warren
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| | | | | | - Jean-Marie C Bouteiller
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
- Institute for Technology and Medical Systems (ITEMS), Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
| | | | - Gianluca Lazzi
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Electrical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Ophthalmology, University of Southern California, Los Angeles, CA, 90089, USA
- Institute for Technology and Medical Systems (ITEMS), Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
| |
Collapse
|
46
|
Jelinek M, Lipkova J, Duris K. Vagus nerve stimulation as immunomodulatory therapy for stroke: A comprehensive review. Exp Neurol 2024; 372:114628. [PMID: 38042360 DOI: 10.1016/j.expneurol.2023.114628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/20/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
Stroke is a devastating cerebrovascular pathology with high morbidity and mortality. Inflammation plays a central role in the pathophysiology of stroke. Vagus nerve stimulation (VNS) is a promising immunomodulatory method that has shown positive effects in stroke treatment, including neuroprotection, anti-apoptosis, anti-inflammation, antioxidation, reduced infarct volume, improved neurological scores, and promotion of M2 microglial polarization. In this review, we summarize the current knowledge about the vagus nerve's immunomodulatory effects through the cholinergic anti-inflammatory pathway (CAP) and provide a comprehensive assessment of the available experimental literature focusing on the use of VNS in stroke treatment.
Collapse
Affiliation(s)
- Matyas Jelinek
- Department of Pathophysiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Jolana Lipkova
- Department of Pathophysiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Kamil Duris
- Department of Pathophysiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Department of Neurosurgery, The University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
47
|
Kellett DO, Aziz Q, Humphries JD, Korsak A, Braga A, Gutierrez Del Arroyo A, Crescente M, Tinker A, Ackland GL, Gourine AV. Transcriptional response of the heart to vagus nerve stimulation. Physiol Genomics 2024; 56:167-178. [PMID: 38047311 PMCID: PMC11281814 DOI: 10.1152/physiolgenomics.00095.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/30/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023] Open
Abstract
Heart failure is a major clinical problem, with treatments involving medication, devices, and emerging neuromodulation therapies such as vagus nerve stimulation (VNS). Considering the ongoing interest in using VNS to treat cardiovascular disease, it is important to understand the genetic and molecular changes developing in the heart in response to this form of autonomic neuromodulation. This experimental animal (rat) study investigated the immediate transcriptional response of the ventricular myocardium to selective stimulation of vagal efferent activity using an optogenetic approach. Vagal preganglionic neurons in the dorsal motor nucleus of the vagus nerve were genetically targeted to express light-sensitive chimeric channelrhodopsin variant ChIEF and stimulated using light. RNA sequencing of the left ventricular myocardium identified 294 differentially expressed genes (false discovery rate < 0.05). Qiagen Ingenuity Pathway Analysis (IPA) highlighted 118 canonical pathways that were significantly modulated by vagal activity, of which 14 had a z score of ≥2/≤-2, including EIF-2, IL-2, integrin, and NFAT-regulated cardiac hypertrophy. IPA revealed the effect of efferent vagus stimulation on protein synthesis, autophagy, fibrosis, autonomic signaling, inflammation, and hypertrophy. IPA further predicted that the identified differentially expressed genes were the targets of 50 upstream regulators, including transcription factors (e.g., MYC and NRF1) and microRNAs (e.g., miR-335-3p and miR-338-3p). These data demonstrate that the vagus nerve has a major impact on the myocardial expression of genes involved in the regulation of key biological pathways. The transcriptional response of the ventricular myocardium induced by stimulation of vagal efferents is consistent with the beneficial effect of maintained/increased vagal activity on the heart.NEW & NOTEWORTHY This experimental animal study investigated the immediate transcriptional response of the ventricular myocardium to selective stimulation of vagal efferent activity. Vagal stimulation induced significant transcriptional changes in the heart involving the pathways controlling autonomic signaling, inflammation, fibrosis, and hypertrophy. This study provides the first direct evidence that myocardial gene expression is modulated by the activity of the autonomic nervous system.
Collapse
Affiliation(s)
- Daniel O Kellett
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Qadeer Aziz
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Jonathan D Humphries
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Alla Korsak
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Alice Braga
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Ana Gutierrez Del Arroyo
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Marilena Crescente
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Andrew Tinker
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Gareth L Ackland
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Alexander V Gourine
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
48
|
Kellett DO, Aziz Q, Humphries JD, Korsak A, Braga A, Del Arroyo AG, Crescente M, Tinker A, Ackland GL, Gourine AV. Transcriptional response of the heart to vagus nerve stimulation. Physiol Genomics 2024; 56:167-178. [PMID: 39071113 PMCID: PMC7616044 DOI: 10.6084/m9.figshare.24449590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024] Open
Abstract
Heart failure is a major clinical problem, with treatments involving medication, devices, and emerging neuromodulation therapies such as vagus nerve stimulation (VNS). Considering the ongoing interest in using VNS to treat cardiovascular disease it is important to understand the genetic and molecular changes developing in the heart in response to this form of autonomic neuromodulation. This experimental animal (rat) study investigated the immediate transcriptional response of the ventricular myocardium to selective stimulation of vagal efferent activity using an optogenetic approach. Vagal preganglionic neurons in the dorsal motor nucleus of the vagus nerve were genetically targeted to express light-sensitive chimeric channelrhodopsin variant ChIEF, and stimulated using light. RNA sequencing of left ventricular myocardium identified 294 differentially expressed genes (DEGs, false discovery rate <0.05). Qiagen Ingenuity Pathway Analysis (IPA) highlighted 118 canonical pathways that were significantly modulated by vagal activity, of which 14 had a z-score of ≥2/≤-2, including EIF-2, IL-2, Integrin, and NFAT-regulated cardiac hypertrophy. IPA revealed the effect of efferent vagus stimulation on protein synthesis, autophagy, fibrosis, autonomic signalling, inflammation, and hypertrophy. IPA further predicted that the identified DEGs were the targets of 50 upstream regulators, including transcription factors (e.g., MYC, NRF1) and microRNAs (e.g., miR-335-3p, miR-338-3p). These data demonstrate that the vagus nerve has a major impact on myocardial expression of genes involved in regulation of key biological pathways. The transcriptional response of the ventricular myocardium induced by stimulation of vagal efferents is consistent with the beneficial effect of maintained/increased vagal activity on the heart.
Collapse
Affiliation(s)
- Daniel O Kellett
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Qadeer Aziz
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | - Alla Korsak
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Alice Braga
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Ana Gutierrez Del Arroyo
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Marilena Crescente
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Andrew Tinker
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Gareth L Ackland
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Alexander V Gourine
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| |
Collapse
|
49
|
Li XY, Liu JQ, Wang Y, Chen Y, Hu WH, Lv YX, Wu Y, Lv J, Tang JM, Kong D. VNS improves VSMC metabolism and arteriogenesis in infarcted hearts through m/n-AChR-Akt-SDF-1α in adult male rats. J Mol Histol 2024; 55:51-67. [PMID: 38165566 PMCID: PMC10830782 DOI: 10.1007/s10735-023-10171-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 10/21/2023] [Indexed: 01/04/2024]
Abstract
Vagal nerve stimulation (VNS) provides a novel therapeutic strategy for injured hearts by activating cholinergic anti-inflammatory pathways. However, little information is available on the metabolic pattern and arteriogenesis of VSMCs after MI. VNS has been shown to stimulate the expression of CPT1α, CPT1β, Glut1, Glut4 and SDF-1α in coronary VSMCs, decreasing the number of CD68-positive macrophages while increasing CD206-positive macrophages in the infarcted hearts, leading to a decrease in TNF-α and IL-1β accompanied by a reduced ratio of CD68- and CD206-positive cells, which were dramatically abolished by atropine and mecamylamine in vivo. Knockdown of SDF-1α substantially abrogated the effect of VNS on macrophagecell alteration and inflammatory factors in infarcted hearts. Mechanistically, ACh induced SDF-1α expression in VSMCs in a dose-dependent manner. Conversely, atropine, mecamylamine, and a PI3K/Akt inhibitor completely eliminated the effect of ACh on SDF-1α expression. Functionally, VNS promoted arteriogenesis and improved left ventricular performance, which could be abolished by Ad-shSDF-1α. Thus, VNS altered the VSMC metabolism pattern and arteriogenesis to repair the infarcted heart by inducing SDF-1α expression, which was associated with the m/nAChR-Akt signaling pathway.
Collapse
Affiliation(s)
- Xing-Yuan Li
- Department of Physiology, Faculty of Basic Medical Sciences, Zunyi Medicical University, Zunyi, 563006, Guizhou, People's Republic of China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Jia-Qi Liu
- Nursing College, Hubei Province Chinese Medicine Hospital, Hubei University of Traditional Chinese Medicine, Wuhan, 430065, Hubei, People's Republic of China
| | - Yan Wang
- Department of Physiology, Faculty of Basic Medical Sciences, Zunyi Medicical University, Zunyi, 563006, Guizhou, People's Republic of China
| | - Yan Chen
- Department of Physiology, Faculty of Basic Medical Sciences, Zunyi Medicical University, Zunyi, 563006, Guizhou, People's Republic of China
| | - Wen-Hui Hu
- Department of Physiology, Faculty of Basic Medical Sciences, Zunyi Medicical University, Zunyi, 563006, Guizhou, People's Republic of China
| | - Yan-Xia Lv
- Hubei Key Laboratory of Embryonic Stem Cell Research, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Yan Wu
- Hubei Key Laboratory of Embryonic Stem Cell Research, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Jing Lv
- Institute of Basic Medical Sciences, Institute of Biomedicine, Hubei University of Medicine, Hubei, 442000, People's Republic of China.
| | - Jun-Ming Tang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.
- Institute of Basic Medical Sciences, Institute of Biomedicine, Hubei University of Medicine, Hubei, 442000, People's Republic of China.
| | - Deying Kong
- Department of Physiology, Faculty of Basic Medical Sciences, Zunyi Medicical University, Zunyi, 563006, Guizhou, People's Republic of China.
| |
Collapse
|
50
|
Guo Y, Gharibani P. Analgesic Effects of Vagus Nerve Stimulation on Visceral Hypersensitivity: A Direct Comparison Between Invasive and Noninvasive Methods in Rats. Neuromodulation 2024; 27:284-294. [PMID: 37191611 DOI: 10.1016/j.neurom.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/20/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023]
Abstract
OBJECTIVES The aims of this study were to investigate analgesic effects of vagus nerve stimulation (VNS) on visceral hypersensitivity (VH) in a rodent model of functional dyspepsia (FD) and to compare invasive VNS with noninvasive auricular VNS (aVNS). MATERIALS AND METHODS Eighteen ten-day-old male rats were gavaged with 0.1% iodoacetamide (IA) or 2% sucrose solution for six days. After eight weeks, IA-treated rats were implanted with electrodes for VNS or aVNS (n = 6 per group). Different parameters, varying in frequency and stimulation duty cycle, were tested to find the best parameter based on the improvement of VH assessed by electromyogram (EMG) during gastric distension. RESULTS Compared with sucrose-treated rats, visceral sensitivity was increased significantly in IA-treated "FD" rats and ameliorated remarkably by VNS (at 40, 60, and 80 mm Hg; p ≤ 0.02, respectively) and aVNS (at 60 and 80 mm Hg; p ≤ 0.05, respectively) with the parameter of 100 Hz and 20% duty cycle. There was no significant difference in area under the curve of EMG responses between VNS and aVNS (at 60 and 80 mm Hg, both p > 0.05). Spectral analysis of heart rate variability revealed a significant enhancement in vagal efferent activity while applying VNS/aVNS compared with sham stimulation (p < 0.01). In the presence of atropine, no significant differences were noted in EMG after VNS/aVNS. Naloxone blocked the analgesic effects of VNS/aVNS. CONCLUSIONS VNS/aVNS with optimized parameter elicits ameliorative effects on VH, mediated by autonomic and opioid mechanisms. aVNS is as effective as direct VNS and has great potential for treating visceral pain in patients with FD.
Collapse
Affiliation(s)
- Yu Guo
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Payam Gharibani
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Division of Neuroimmunology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|