1
|
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
|
2
|
Gentile F, Orlando G, Montuoro S, Ferrari Chen YF, Macefield V, Passino C, Giannoni A, Emdin M. Treating heart failure by targeting the vagus nerve. Heart Fail Rev 2024; 29:1201-1215. [PMID: 39117958 PMCID: PMC11455679 DOI: 10.1007/s10741-024-10430-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/28/2024] [Indexed: 08/10/2024]
Abstract
Increased sympathetic and reduced parasympathetic nerve activity is associated with disease progression and poor outcomes in patients with chronic heart failure. The demonstration that markers of autonomic imbalance and vagal dysfunction, such as reduced heart rate variability and baroreflex sensitivity, hold prognostic value in patients with chronic heart failure despite modern therapies encourages the research for neuromodulation strategies targeting the vagus nerve. However, the approaches tested so far have yielded inconclusive results. This review aims to summarize the current knowledge about the role of the parasympathetic nervous system in chronic heart failure, describing the pathophysiological background, the methods of assessment, and the rationale, limits, and future perspectives of parasympathetic stimulation either by drugs or bioelectronic devices.
Collapse
Affiliation(s)
- Francesco Gentile
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy.
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy.
| | - Giulia Orlando
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
| | - Sabrina Montuoro
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
| | - Yu Fu Ferrari Chen
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy
| | | | - Claudio Passino
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy
| | - Alberto Giannoni
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy
| | - Michele Emdin
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy
| |
Collapse
|
3
|
Evancho A, Do M, Fortenberry D, Billings R, Sartayev A, Tyler WJ. Vagus nerve stimulation in Parkinson's disease: a scoping review of animal studies and human subjects research. NPJ Parkinsons Dis 2024; 10:199. [PMID: 39448636 PMCID: PMC11502766 DOI: 10.1038/s41531-024-00803-1] [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: 03/07/2024] [Accepted: 10/02/2024] [Indexed: 10/26/2024] Open
Abstract
Parkinson's Disease (PD) is a prevalent, progressive neurodegenerative disease with motor and non-motor symptoms. Vagus Nerve Stimulation (VNS) has emerged as a potential therapeutic approach for PD, but published research on this topic varies widely. This scoping review maps existing literature on VNS for PD, highlighting stimulation methods, operational parameters, safety profiles, neurophysiological mechanisms, and clinical outcomes in human and animal models. Online databases were used to identify 788 papers published between 2013 and 2023, from which 17 publications on invasive and non-invasive VNS in PD were selected. Studies showed high variability in VNS parameters and study design. Evidence in animal models and human subjects suggests potential neurophysiological effects on PD-related pathology and motor function improvements. However, significant gaps in the literature remain. Future research should include rigorous reporting of study design, standardization of stimulation parameters, and larger sample sizes to ultimately facilitate translation of VNS into clinical practice.
Collapse
Affiliation(s)
- Alexandra Evancho
- University of Alabama at Birmingham School of Health Professions, Birmingham, AL, USA.
| | - Melissa Do
- University of Alabama at Birmingham School of Engineering, Birmingham, AL, USA
| | | | - Rebecca Billings
- University of Alabama at Birmingham Libraries, Birmingham, AL, USA
| | - Alibek Sartayev
- University of Alabama at Birmingham Graduate Biomedical Sciences, Birmingham, AL, USA
| | - William J Tyler
- University of Alabama at Birmingham School of Health Professions, Birmingham, AL, USA
- University of Alabama at Birmingham School of Engineering, Birmingham, AL, USA
- University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| |
Collapse
|
4
|
Micera S, Menciassi A, Cianferotti L, Gruppioni E, Lionetti V. Organ Neuroprosthetics: Connecting Transplanted and Artificial Organs with the Nervous System. Adv Healthc Mater 2024; 13:e2302896. [PMID: 38656615 DOI: 10.1002/adhm.202302896] [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: 08/30/2023] [Revised: 04/01/2024] [Indexed: 04/26/2024]
Abstract
Implantable neural interfaces with the central and peripheral nervous systems are currently used to restore sensory, motor, and cognitive functions in disabled people with very promising results. They have also been used to modulate autonomic activities to treat diseases such as diabetes or hypertension. Here, this study proposes to extend the use of these technologies to (re-)establish the connection between new (transplanted or artificial) organs and the nervous system in order to increase the long-term efficacy and the effective biointegration of these solutions. In this perspective paper, some clinically relevant applications of this approach are briefly described. Then, the choices that neural engineers must implement about the type, implantation location, and closed-loop control algorithms to successfully realize this approach are highlighted. It is believed that these new "organ neuroprostheses" are going to become more and more valuable and very effective solutions in the years to come.
Collapse
Affiliation(s)
- Silvestro Micera
- The BioRobotics Institute and Department of Excellence in Robotics and AI, Scuola Superiore Sant'Anna, Pisa, 56127, Italy
- Interdisciplinary Research Center Health Science, Scuola Superiore Sant'Anna, Pisa, 56127, Italy
- Bertarelli Foundation Chair in Translational Neuroengineering, Neuro-X Institute, School of Engineering, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, 1015, Switzerland
| | - Arianna Menciassi
- The BioRobotics Institute and Department of Excellence in Robotics and AI, Scuola Superiore Sant'Anna, Pisa, 56127, Italy
- Interdisciplinary Research Center Health Science, Scuola Superiore Sant'Anna, Pisa, 56127, Italy
| | - Luisella Cianferotti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, 50121, Italy
| | | | - Vincenzo Lionetti
- Interdisciplinary Research Center Health Science, Scuola Superiore Sant'Anna, Pisa, 56127, Italy
- UOSVD Anesthesia and Resuscitation, Fondazione Toscana G. Monasterio, Pisa, 56127, Italy
| |
Collapse
|
5
|
Shao M, Yao C, Han Y, Zhou X, Lu Y, Zhang L, Li Y, Tang B. Ablation of myocardial autonomic ganglion plexus in the treatment of bradyarrhythmia A one-arm interventional study. Clinics (Sao Paulo) 2024; 79:100448. [PMID: 39096858 PMCID: PMC11334784 DOI: 10.1016/j.clinsp.2024.100448] [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: 12/05/2023] [Revised: 05/07/2024] [Accepted: 06/30/2024] [Indexed: 08/05/2024] Open
Abstract
OBJECTIVES To study the complications and effectiveness of the treatment of chronic arrhythmias with cardiac Ganglion Plexus (GP) ablation, and to explore the value of the treatment of chronic arrhythmias with GP ablation. METHODS This study was a one-arm interventional study of patients from the first hospital of Xinjiang Medical University and the People's Hospital of Xuancheng City admitted (09/2018-08/2021) because of bradyarrhythmia. The left atrium was modeled using the Carto3 mapping system. The ablation endpoint was the absence of a vagal response under anatomically localized and high-frequency stimulation guidance. Postoperative routine follow-up was conducted. Holter data at 3-, 6-, and 12-months were recorded. RESULTS Fifty patients (25 male, mean age 33.16 ± 7.89 years) were induced vagal response by either LSGP, LIGP, RAGP, or RIGP. The heart rate was stable at 76 bpm, SNRT 1.092s. DC, DR, HR, SDNN, RMSSD values were lower than that before ablation. AC, SSR, TH values were higher than those before ablation, mean heart rate and the slowest heart rate were significantly increased. There were significant differences in follow-up data between the preoperative and postoperative periods (all p < 0.05). All the patients were successfully ablated, and their blood pressure decreased significantly. No complications such as vascular damage, vascular embolism and pericardial effusion occurred. CONCLUSIONS Left Atrial GP ablation has good long-term clinical results and can be used as a treatment option for patients with bradyarrhythmia.
Collapse
Affiliation(s)
- Mingliang Shao
- Department of Cardiovascular, The People's Hospital of Xuancheng City, Xuancheng City, Anhui Province, China; Department of Pacing Electrophysiology, Xinjiang Key Laboratory of Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi City, Xinjiang Uygur Autonomous Region, China
| | - Chenhuan Yao
- Department of Research and Teaching, The People's Hospital of Xuancheng City, Xuancheng City, Anhui Province, China
| | - Yafan Han
- Department of Pacing Electrophysiology, Xinjiang Key Laboratory of Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi City, Xinjiang Uygur Autonomous Region, China
| | - Xianhui Zhou
- Department of Pacing Electrophysiology, Xinjiang Key Laboratory of Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi City, Xinjiang Uygur Autonomous Region, China
| | - Yanmei Lu
- Department of Pacing Electrophysiology, Xinjiang Key Laboratory of Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi City, Xinjiang Uygur Autonomous Region, China
| | - Ling Zhang
- Department of Pacing Electrophysiology, Xinjiang Key Laboratory of Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi City, Xinjiang Uygur Autonomous Region, China
| | - Yaodong Li
- Department of Pacing Electrophysiology, Xinjiang Key Laboratory of Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi City, Xinjiang Uygur Autonomous Region, China
| | - Baopeng Tang
- Department of Pacing Electrophysiology, Xinjiang Key Laboratory of Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi City, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
6
|
Gerhardt T, Huynh P, McAlpine CS. Neuroimmune circuits in the plaque and bone marrow regulate atherosclerosis. Cardiovasc Res 2024:cvae167. [PMID: 39086175 DOI: 10.1093/cvr/cvae167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/02/2024] [Accepted: 06/01/2024] [Indexed: 08/02/2024] Open
Abstract
Atherosclerosis remains the leading cause of death globally. Although its focal pathology is atheroma that develops in arterial walls, atherosclerosis is a systemic disease involving contributions by many organs and tissues. It is now established that the immune system causally contributes to all phases of atherosclerosis. Recent and emerging evidence positions the nervous system as a key modulator of inflammatory processes that underly atherosclerosis. This neuro-immune crosstalk, we are learning, is bidirectional, and immune regulated afferent signaling is becoming increasingly recognized in atherosclerosis. Here, we summarize data and concepts that link the immune and nervous systems in atherosclerosis by focusing on two important sites, the arterial vessel and the bone marrow.
Collapse
Affiliation(s)
- Teresa Gerhardt
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friede Springer Center for Cardiovascular Prevention at Charité, Berlin, Germany
| | - Pacific Huynh
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S McAlpine
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
7
|
Sarikhani P, Hsu HL, Zeydabadinezhad M, Yao Y, Kothare M, Mahmoudi B. Reinforcement learning for closed-loop regulation of cardiovascular system with vagus nerve stimulation: a computational study. J Neural Eng 2024; 21:036027. [PMID: 38718787 PMCID: PMC11145940 DOI: 10.1088/1741-2552/ad48bb] [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: 09/29/2023] [Revised: 04/24/2024] [Accepted: 05/08/2024] [Indexed: 06/04/2024]
Abstract
Objective. Vagus nerve stimulation (VNS) is being investigated as a potential therapy for cardiovascular diseases including heart failure, cardiac arrhythmia, and hypertension. The lack of a systematic approach for controlling and tuning the VNS parameters poses a significant challenge. Closed-loop VNS strategies combined with artificial intelligence (AI) approaches offer a framework for systematically learning and adapting the optimal stimulation parameters. In this study, we presented an interactive AI framework using reinforcement learning (RL) for automated data-driven design of closed-loop VNS control systems in a computational study.Approach.Multiple simulation environments with a standard application programming interface were developed to facilitate the design and evaluation of the automated data-driven closed-loop VNS control systems. These environments simulate the hemodynamic response to multi-location VNS using biophysics-based computational models of healthy and hypertensive rat cardiovascular systems in resting and exercise states. We designed and implemented the RL-based closed-loop VNS control frameworks in the context of controlling the heart rate and the mean arterial pressure for a set point tracking task. Our experimental design included two approaches; a general policy using deep RL algorithms and a sample-efficient adaptive policy using probabilistic inference for learning and control.Main results.Our simulation results demonstrated the capabilities of the closed-loop RL-based approaches to learn optimal VNS control policies and to adapt to variations in the target set points and the underlying dynamics of the cardiovascular system. Our findings highlighted the trade-off between sample-efficiency and generalizability, providing insights for proper algorithm selection. Finally, we demonstrated that transfer learning improves the sample efficiency of deep RL algorithms allowing the development of more efficient and personalized closed-loop VNS systems.Significance.We demonstrated the capability of RL-based closed-loop VNS systems. Our approach provided a systematic adaptable framework for learning control strategies without requiring prior knowledge about the underlying dynamics.
Collapse
Affiliation(s)
- Parisa Sarikhani
- Department of Biomedical Informatics, Emory University, Atlanta, GA, United States of America
| | - Hao-Lun Hsu
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Mahmoud Zeydabadinezhad
- Department of Biomedical Informatics, Emory University, Atlanta, GA, United States of America
| | - Yuyu Yao
- Department of Chemical & Biomolecular Engineering, Lehigh University, Bethlehem, PA, United States of America
| | - Mayuresh Kothare
- Department of Chemical & Biomolecular Engineering, Lehigh University, Bethlehem, PA, United States of America
| | - Babak Mahmoudi
- Department of Biomedical Informatics, Emory University, Atlanta, GA, United States of America
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States of America
| |
Collapse
|
8
|
Haberbusch M, Kronsteiner B, Aigner P, Kiss A, Podesser BK, Moscato F. Importance of cardiac-synchronized vagus nerve stimulation parameters on the provoked chronotropic response for different levels of cardiac innervation. Front Physiol 2024; 15:1379936. [PMID: 38835728 PMCID: PMC11148559 DOI: 10.3389/fphys.2024.1379936] [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: 01/31/2024] [Accepted: 05/02/2024] [Indexed: 06/06/2024] Open
Abstract
Introduction The influence of vagus nerve stimulation (VNS) parameters on provoked cardiac effects in different levels of cardiac innervation is not well understood yet. This study examines the effects of VNS on heart rate (HR) modulation across a spectrum of cardiac innervation states, providing data for the potential optimization of VNS in cardiac therapies. Materials and Methods Utilizing previously published data from VNS experiments on six sheep with intact innervation, and data of additional experiments in five rabbits post bilateral rostral vagotomy, and four isolated rabbit hearts with additionally removed sympathetic influences, the study explored the impact of diverse VNS parameters on HR. Results Significant differences in physiological threshold charges were identified across groups: 0.09 ± 0.06 μC for intact, 0.20 ± 0.04 μC for vagotomized, and 9.00 ± 0.75 μC for isolated hearts. Charge was a key determinant of HR reduction across all innervation states, with diminishing correlations from intact (r = 0.7) to isolated hearts (r = 0.44). An inverse relationship was observed for the number of pulses, with its influence growing in conditions of reduced innervation (intact r = 0.11, isolated r = 0.37). Frequency and stimulation delay showed minimal correlations (r < 0.17) in all conditions. Conclusion Our study highlights for the first time that VNS parameters, including stimulation intensity, pulse width, and pulse number, crucially modulate heart rate across different cardiac innervation states. Intensity and pulse width significantly influence heart rate in innervated states, while pulse number is key in denervated states. Frequency and delay have less impact impact across all innervation states. These findings suggest the importance of customizing VNS therapy based on innervation status, offering insights for optimizing cardiac neuromodulation.
Collapse
Affiliation(s)
- Max Haberbusch
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Bettina Kronsteiner
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Philipp Aigner
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Bruno Karl Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Center for Biomedical Research and Translational Surgery, 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
| |
Collapse
|
9
|
Tamura Y, Nomura A, Kagiyama N, Mizuno A, Node K. Digitalomics, digital intervention, and designing future: The next frontier in cardiology. J Cardiol 2024; 83:318-322. [PMID: 38135148 DOI: 10.1016/j.jjcc.2023.12.002] [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: 09/02/2023] [Revised: 12/10/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
The discipline of cardiology stands at a transformative juncture, primarily influenced by the surge in digital health technologies. These innovations hold the promise to redefine the realms of cardiovascular research and patient care, ushering in an era of individualized and data-driven treatments. This review delves into the heart of this evolution, introducing a comprehensive design for the future of cardiology. Emphasizing the emerging domains of "digitalomics" and "digital intervention", it explores how the integration of patient data, artificial intelligence-enabled diagnostics, and telehealth can lead to more streamlined and personalized cardiovascular health. The "digital-twin" model, a highlight of this approach, encapsulates individual patient characteristics, allowing for targeted treatments. The role of interdisciplinary collaboration in cardiovascular medicine is also underlined, emphasizing the importance of merging traditional cardiology with technological advancements. The convergence of traditional cardiology methods and digital health technologies, facilitated by a transdisciplinary approach, is set to chart a new course in cardiovascular health, emphasizing individualized care and improved clinical outcomes.
Collapse
Affiliation(s)
- Yuichi Tamura
- Pulmonary Hypertension Center, International University of Health and Welfare Mita Hospital, Tokyo, Japan; Department of Cardiology International University of Health and Welfare School of Medicine Narita, Japan; Cardiointelligence Inc., Tokyo, Japan.
| | - Akihiro Nomura
- College of Transdisciplinary Sciences for Innovation, Kanazawa University, Kanazawa, Japan; Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan; Frontier Institute of Tourism Sciences, Kanazawa University, Kanazawa, Japan; Department of Biomedical Informatics, CureApp Institute, Karuizawa, Japan
| | - Nobuyuki Kagiyama
- Department of Digital Health and Telemedicine R&D, Juntendo University, Tokyo, Japan; Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Atsushi Mizuno
- Department of Cardiovascular Medicine, St. Luke's International Hospital, Tokyo, Japan; Leonard Davis Institute for Health Economics, University of Pennsylvania, PA, USA
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University, Saga, Japan
| |
Collapse
|
10
|
Strain MM, Conley NJ, Kauffman LS, Espinoza L, Fedorchak S, Martinez PC, Crook ME, Jalil M, Hodes GE, Abbott SB, Güler AD, Campbell JN, Boychuk CR. Dorsal motor vagal neurons can elicit bradycardia and reduce anxiety-like behavior. iScience 2024; 27:109137. [PMID: 38420585 PMCID: PMC10901094 DOI: 10.1016/j.isci.2024.109137] [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] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/16/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024] Open
Abstract
Cardiovagal neurons (CVNs) innervate cardiac ganglia through the vagus nerve to control cardiac function. Although the cardioinhibitory role of CVNs in nucleus ambiguus (CVNNA) is well established, the nature and functionality of CVNs in dorsal motor nucleus of the vagus (CVNDMV) is less clear. We therefore aimed to characterize CVNDMV anatomically, physiologically, and functionally. Optogenetically activating cholinergic DMV neurons resulted in robust bradycardia through peripheral muscarinic (parasympathetic) and nicotinic (ganglionic) acetylcholine receptors, but not beta-1-adrenergic (sympathetic) receptors. Retrograde tracing from the cardiac fat pad labeled CVNNA and CVNDMV through the vagus nerve. Using whole-cell patch-clamp, CVNDMV demonstrated greater hyperexcitability and spontaneous action potential firing ex vivo despite similar resting membrane potentials, compared to CVNNA. Chemogenetically activating DMV also caused significant bradycardia with a correlated reduction in anxiety-like behavior. Thus, DMV contains uniquely hyperexcitable CVNs and is capable of cardioinhibition and robust anxiolysis.
Collapse
Affiliation(s)
- Misty M. Strain
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | | | - Lily S. Kauffman
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Liliana Espinoza
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Stephanie Fedorchak
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | | | - Maisie E. Crook
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Maira Jalil
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Georgia E. Hodes
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Stephen B.G. Abbott
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Ali D. Güler
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - John N. Campbell
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Carie R. Boychuk
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
11
|
Ajieren H, Fox A, Biggs E, Albors G, Llebaria A, Irazoqui P. Design and validation of a low-cost photomodulator for in vivo photoactivation of a mGluR5 inhibitor. Biomed Eng Lett 2024; 14:245-254. [PMID: 38374907 PMCID: PMC10874351 DOI: 10.1007/s13534-023-00334-3] [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: 06/05/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 02/21/2024] Open
Abstract
Purpose: Severe side effects prevent the utilization of otherwise promising drugs in treatments. These side effects arise when drugs affect untargeted tissues due to poor target specificity. In photopharmacology, light controls the timing and the location of drug delivery, improving treatment specificity and pharmacokinetic control. Photopharmaceuticals have not seen widespread adoption in part because researchers do not always have access to reliable and reproducible light delivery devices at prices which fit within the larger research budget. Method: In this work, we present a customizable photomodulator for use in both wearable and implantable devices. For experimental validation of the photomodulator, we photolyse JF-NP-26 in rats. Results: We successfully drive in vivo photopharmacology with a tethered photomodulator and demonstrate modifications which enable the photomodulator to operate wirelessly. Conclusion: By documenting our photomodulator development, we hope to introduce researchers to a simple solution which significantly lowers the engineering barriers to photopharmacology research. Graphical abstract Researchers present a photomodulator, a device designed to facilitate in vivo photopharmacology. They demonstrate the in vivo capabilities of the photomodulator by photoreleasing raseglurant, an mGluR5 inhibitor, to treat pain in an acute rat model and follow this study by showing how to reconfigure the photomodulator to work wirelessly and interface with other biomedical devices. Supplementary Information The online version contains supplementary material available at 10.1007/s13534-023-00334-3.
Collapse
Affiliation(s)
- Hans Ajieren
- Department of Electrical Engineering, Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218 USA
| | - Andrew Fox
- Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Ethan Biggs
- Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Gabriel Albors
- The Margolis Center for Health Policy, Duke University, Durham, NC 27708 USA
| | - Amadeu Llebaria
- Institute for Advanced Chemistry of Catalonia, Spanish National Research Council, Jordi Girona, 18-26, 08034 Barcelona, Spain
| | - Pedro Irazoqui
- Department of Electrical Engineering, Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218 USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, 733 N. Broadway, Baltimore, MD 21205 USA
| |
Collapse
|
12
|
Chen L, Tang C, Wang Z, Zhang L, Gu B, Liu X, Ming D. Enhancing Motor Sequence Learning via Transcutaneous Auricular Vagus Nerve Stimulation (taVNS): An EEG Study. IEEE J Biomed Health Inform 2024; 28:1285-1296. [PMID: 38109248 DOI: 10.1109/jbhi.2023.3344176] [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: 12/20/2023]
Abstract
Motor learning plays a crucial role in human life, and various neuromodulation methods have been utilized to strengthen or improve it. Transcutaneous auricular vagus nerve stimulation (taVNS) has gained increasing attention due to its non-invasive nature, affordability and ease of implementation. Although the potential of taVNS on regulating motor learning has been suggested, its actual regulatory effect has yet been fully explored. Electroencephalogram (EEG) analysis provides an in-depth understanding of cognitive processes involved in motor learning so as to offer methodological support for regulation of motor learning. To investigate the effect of taVNS on motor learning, this study recruited 22 healthy subjects to participate a single-blind, sham-controlled, and within-subject serial reaction time task (SRTT) experiment. Every subject involved in two sessions at least one week apart and received a 20-minute active/sham taVNS in each session. Behavioral indicators as well as EEG characteristics during the task state, were extracted and analyzed. The results revealed that compared to the sham group, the active group showed higher learning performance. Additionally, the EEG results indicated that after taVNS, the motor-related cortical potential amplitudes and alpha-gamma modulation index decreased significantly and functional connectivity based on partial directed coherence towards frontal lobe was enhanced. These findings suggest that taVNS can improve motor learning, mainly through enhancing cognitive and memory functions rather than simple movement learning. This study confirms the positive regulatory effect of taVNS on motor learning, which is particularly promising as it offers a potential avenue for enhancing motor skills and facilitating rehabilitation.
Collapse
|
13
|
de Faria GM, Lopes EG, Tobaldini E, Montano N, Cunha TS, Casali KR, de Amorim HA. Advances in Non-Invasive Neuromodulation: Designing Closed-Loop Devices for Respiratory-Controlled Transcutaneous Vagus Nerve Stimulation. Healthcare (Basel) 2023; 12:31. [PMID: 38200937 PMCID: PMC10778699 DOI: 10.3390/healthcare12010031] [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: 11/01/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024] Open
Abstract
Studies suggest non-invasive transcutaneous auricular vagus nerve stimulation (taVNS) as a potential therapeutic option for various pathological conditions, such as epilepsy and depression. Exhalation-controlled taVNS, which synchronizes stimulation with internal body rhythms, holds promise for enhanced neuromodulation, but there is no closed-loop system in the literature capable of performing such integration in real time. In this context, the objective was to develop real-time signal processing techniques and an integrated closed-loop device with sensors to acquire physiological data. After a conditioning stage, the signal is processed and delivers synchronized electrical stimulation during the patient's expiratory phase. Additional modules were designed for processing, software-controlled selectors, remote and autonomous operation, improved analysis, and graphical visualization. The signal processing method effectively extracted respiratory cycles and successfully attenuated signal noise. Heart rate variability was assessed in real time, using linear statistical evaluation. The prototype feedback stimulator device was physically constructed. Respiratory peak detection achieved an accuracy of 90%, and the real-time processing resulted in a small delay of up to 150 ms in the detection of the expiratory phase. Thus, preliminary results show promising accuracy, indicating the need for additional tests to optimize real-time processing and the application of the prototype in clinical studies.
Collapse
Affiliation(s)
- Gabriella Maria de Faria
- Institute of Science and Technology, Universidade Federal de São Paulo, São José dos Campos 12231-280, Brazil; (G.M.d.F.); (E.G.L.); (T.S.C.); (H.A.d.A.)
| | - Eugênia Gonzales Lopes
- Institute of Science and Technology, Universidade Federal de São Paulo, São José dos Campos 12231-280, Brazil; (G.M.d.F.); (E.G.L.); (T.S.C.); (H.A.d.A.)
| | - Eleonora Tobaldini
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (E.T.); (N.M.)
| | - Nicola Montano
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (E.T.); (N.M.)
| | - Tatiana Sousa Cunha
- Institute of Science and Technology, Universidade Federal de São Paulo, São José dos Campos 12231-280, Brazil; (G.M.d.F.); (E.G.L.); (T.S.C.); (H.A.d.A.)
| | - Karina Rabello Casali
- Institute of Science and Technology, Universidade Federal de São Paulo, São José dos Campos 12231-280, Brazil; (G.M.d.F.); (E.G.L.); (T.S.C.); (H.A.d.A.)
| | - Henrique Alves de Amorim
- Institute of Science and Technology, Universidade Federal de São Paulo, São José dos Campos 12231-280, Brazil; (G.M.d.F.); (E.G.L.); (T.S.C.); (H.A.d.A.)
| |
Collapse
|
14
|
Strain MM, Conley NJ, Kauffman LS, Espinoza L, Fedorchak S, Martinez PC, Crook ME, Jalil M, Hodes GE, Abbott SBG, Güler AD, Campbell JN, Boychuk CR. Dorsal Motor Vagal Neurons Can Elicit Bradycardia and Reduce Anxiety-Like Behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.566855. [PMID: 38014247 PMCID: PMC10680764 DOI: 10.1101/2023.11.14.566855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Cardiovagal neurons (CVNs) innervate cardiac ganglia through the vagus nerve to control cardiac function. Although the cardioinhibitory role of CVNs in nucleus ambiguus (CVNNA) is well established, the nature and functionality of CVNs in dorsal motor nucleus of the vagus (CVNDMV) is less clear. We therefore aimed to characterize CVNDMV anatomically, physiologically, and functionally. Optogenetically activating cholinergic DMV neurons resulted in robust bradycardia through peripheral muscarinic (parasympathetic) and nicotinic (ganglionic) acetylcholine receptors, but not beta-1-adrenergic (sympathetic) receptors. Retrograde tracing from the cardiac fat pad labeled CVNNA and CVNDMV through the vagus nerve. Using whole cell patch clamp, CVNDMV demonstrated greater hyperexcitability and spontaneous action potential firing ex vivo despite similar resting membrane potentials, compared to CVNNA. Chemogenetically activating DMV also caused significant bradycardia with a correlated reduction in anxiety-like behavior. Thus, DMV contains uniquely hyperexcitable CVNs capable of cardioinhibition and robust anxiolysis.
Collapse
Affiliation(s)
- Misty M. Strain
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX
| | | | - Lily S. Kauffman
- Department of Biology, University of Virginia, Charlottesville, VA
| | - Liliana Espinoza
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX
| | - Stephanie Fedorchak
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX
| | | | - Maisie E. Crook
- Department of Biology, University of Virginia, Charlottesville, VA
| | - Maira Jalil
- Department of Biology, University of Virginia, Charlottesville, VA
| | - Georgia E. Hodes
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | | | - Ali D. Güler
- Department of Biology, University of Virginia, Charlottesville, VA
| | - John N. Campbell
- Department of Biology, University of Virginia, Charlottesville, VA
| | - Carie R. Boychuk
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO
| |
Collapse
|
15
|
Mylavarapu RV, Kanumuri VV, de Rivero Vaccari JP, Misra A, McMillan DW, Ganzer PD. Importance of timing optimization for closed-loop applications of vagus nerve stimulation. Bioelectron Med 2023; 9:8. [PMID: 37101239 PMCID: PMC10134677 DOI: 10.1186/s42234-023-00110-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 04/06/2023] [Indexed: 04/28/2023] Open
Abstract
In recent decades, vagus nerve stimulation (VNS) therapy has become widely used for clinical applications including epilepsy, depression, and enhancing the effects of rehabilitation. However, several questions remain regarding optimization of this therapy to maximize clinical outcomes. Although stimulation parameters such as pulse width, amplitude, and frequency are well studied, the timing of stimulation delivery both acutely (with respect to disease events) and chronically (over the timeline of a disease's progression) has generally received less attention. Leveraging such information would provide a framework for the implementation of next generation closed-loop VNS therapies. In this mini-review, we summarize a number of VNS therapies and discuss (1) general timing considerations for these applications and (2) open questions that could lead to further therapy optimization.
Collapse
Affiliation(s)
| | - Vivek V Kanumuri
- Department of Otolaryngology, University of Miami, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA
- Department of Neurological Surgery, University of Miami, Miami, FL, USA
| | - Amrit Misra
- Newton Wellesley Neurology Associates, Newton, MA, USA
| | - David W McMillan
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA
- Department of Neurological Surgery, University of Miami, Miami, FL, USA
| | - Patrick D Ganzer
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA.
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA.
| |
Collapse
|
16
|
Elamin ABA, Forsat K, Senok SS, Goswami N. Vagus Nerve Stimulation and Its Cardioprotective Abilities: A Systematic Review. J Clin Med 2023; 12:jcm12051717. [PMID: 36902505 PMCID: PMC10003006 DOI: 10.3390/jcm12051717] [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: 12/29/2022] [Revised: 02/10/2023] [Accepted: 02/18/2023] [Indexed: 02/24/2023] Open
Abstract
Despite the vagus nerve stimulator (VNS) being used in neuroscience, it has recently been highlighted that it has cardioprotective functions. However, many studies related to VNS are not mechanistic in nature. This systematic review aims to focus on the role of VNS in cardioprotective therapy, selective vagus nerve stimulators (sVNS), and their functional capabilities. A systemic review of the current literature was conducted on VNS, sVNS, and their ability to induce positive effects on arrhythmias, cardiac arrest, myocardial ischemia/reperfusion injury, and heart failure. Both experimental and clinical studies were reviewed and assessed separately. Of 522 research articles retrieved from literature archives, 35 met the inclusion criteria and were included in the review. Literature analysis proves that combining fiber-type selectivity with spatially-targeted vagus nerve stimulation is feasible. The role of VNS as a tool for modulating heart dynamics, inflammatory response, and structural cellular components was prominently seen across the literature. The application of transcutaneous VNS, as opposed to implanted electrodes, provides the best clinical outcome with minimal side effects. VNS presents a method for future cardiovascular treatment that can modulate human cardiac physiology. However, continued research is needed for further insight.
Collapse
Affiliation(s)
| | - Kowthar Forsat
- College of Medicine, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Solomon Silas Senok
- College of Medicine, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Nandu Goswami
- Institute of Physiology (Gravitational Physiology and Medicine), Medical University of Graz, 8036 Graz, Austria
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates
- Correspondence:
| |
Collapse
|
17
|
Donahue MJ, Ejneby MS, Jakešová M, Caravaca AS, Andersson G, Sahalianov I, Đerek V, Hult H, Olofsson PS, Głowacki ED. Wireless optoelectronic devices for vagus nerve stimulation in mice. J Neural Eng 2022; 19. [PMID: 36356313 DOI: 10.1088/1741-2552/aca1e3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 11/10/2022] [Indexed: 11/12/2022]
Abstract
Objective.Vagus nerve stimulation (VNS) is a promising approach for the treatment of a wide variety of debilitating conditions, including autoimmune diseases and intractable epilepsy. Much remains to be learned about the molecular mechanisms involved in vagus nerve regulation of organ function. Despite an abundance of well-characterized rodent models of common chronic diseases, currently available technologies are rarely suitable for the required long-term experiments in freely moving animals, particularly experimental mice. Due to challenging anatomical limitations, many relevant experiments require miniaturized, less invasive, and wireless devices for precise stimulation of the vagus nerve and other peripheral nerves of interest. Our objective is to outline possible solutions to this problem by using nongenetic light-based stimulation.Approach.We describe how to design and benchmark new microstimulation devices that are based on transcutaneous photovoltaic stimulation. The approach is to use wired multielectrode cuffs to test different stimulation patterns, and then build photovoltaic stimulators to generate the most optimal patterns. We validate stimulation through heart rate analysis.Main results.A range of different stimulation geometries are explored with large differences in performance. Two types of photovoltaic devices are fabricated to deliver stimulation: photocapacitors and photovoltaic flags. The former is simple and more compact, but has limited efficiency. The photovoltaic flag approach is more elaborate, but highly efficient. Both can be used for wireless actuation of the vagus nerve using light impulses.Significance.These approaches can enable studies in small animals that were previously challenging, such as long-termin vivostudies for mapping functional vagus nerve innervation. This new knowledge may have potential to support clinical translation of VNS for treatment of select inflammatory and neurologic diseases.
Collapse
Affiliation(s)
- Mary J Donahue
- Laboratory of Organic Electronics, Campus Norrköping, Linköping University, SE-60174 Norrköping, Sweden
| | - Malin Silverå Ejneby
- Laboratory of Organic Electronics, Campus Norrköping, Linköping University, SE-60174 Norrköping, Sweden.,Wallenberg Centre for Molecular Medicine, Linköping University, SE-58185 Linköping, Sweden
| | - Marie Jakešová
- Bioelectronics Materials and Devices Laboratory, Central European Institute of Technology, Brno University of Technology, Purkyňova 123, 61200 Brno, Czech Republic
| | - April S Caravaca
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Stockholm Center for Bioelectronic Medicine, MedTechLabs, Karolinska University Hospital, Solna, Sweden
| | | | - Ihor Sahalianov
- Bioelectronics Materials and Devices Laboratory, Central European Institute of Technology, Brno University of Technology, Purkyňova 123, 61200 Brno, Czech Republic
| | - Vedran Đerek
- Department of Physics, Faculty of Science, University of Zagreb, Bijenička c. 32, 10000 Zagreb, Croatia
| | - Henrik Hult
- Stockholm Center for Bioelectronic Medicine, MedTechLabs, Karolinska University Hospital, Solna, Sweden.,Department of Mathematics, KTH, 11428 Stockholm, Sweden
| | - Peder S Olofsson
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Stockholm Center for Bioelectronic Medicine, MedTechLabs, Karolinska University Hospital, Solna, Sweden.,Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, United States of America
| | - Eric Daniel Głowacki
- Laboratory of Organic Electronics, Campus Norrköping, Linköping University, SE-60174 Norrköping, Sweden.,Bioelectronics Materials and Devices Laboratory, Central European Institute of Technology, Brno University of Technology, Purkyňova 123, 61200 Brno, Czech Republic
| |
Collapse
|
18
|
Wang C, Su T, Xiao L, Wang Y, Huo X, Li W, Ding J, Sun T. Right vagus nerve stimulation improves motor behavior by exerting neuroprotective effects in Parkinson's disease rats. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1314. [PMID: 36660708 PMCID: PMC9843310 DOI: 10.21037/atm-22-5366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Background Parkinson's disease (PD) is a common movement disorder disease. Left vagus nerve stimulation (LVNS) is a potential treatment option for PD. Compared with the left vagus nerve, the right vagus nerve is more closely connected with the midbrain dopaminergic neurons, which are the lesion locations of PD. However, whether right vagus nerve stimulation (RVNS) has a therapeutic effect on PD has not yet been studied. Therefore, in this study, we studied the therapeutic effect and underlying mechanism of RVNS using a PD rat model. Methods To establish the PD rat model, 8-week-old male Sprague-Dawley rats were intraperitoneally injected with rotenone for 21 days. The cuff electrodes were implanted into the right cervical vagal carotid sheaths of the rats. The right vagus nerve was continuously stimulated for 14 days using a radio stimulation system. Behavioral tests were performed before and after stimulation. Finally, tyrosine hydroxylase (TH), vesicular monoamine transporter 2 (VMAT2), and α-synuclein in the midbrain, including the substantia nigra (SN) and ventral tegmental area (VTA), were detected by immunofluorescence. Results A markedly lower distance traveled and rearing number was observed in the rotenone, rotenone + sham, and rotenone + RVNS groups compared to the vehicle group. After the stimulation days, the distance traveled and rearing number were both higher in the rotenone + RVNS group compared to the rotenone and rotenone + sham groups (P<0.01, P<0.0001). A remarkable increase in distance traveled and rearing number was observed in the rotenone + RVNS group after stimulation. TH expression in the vehicle group was significantly up-regulated than the other groups. RVNS markedly up-regulated TH expression level. A significantly higher expression of α-synuclein was observed in the rotenone, rotenone + sham, and rotenone + RVNS groups compared to the vehicle group. The expression of α-synuclein was lower in the rotenone + RVNS group compared to the rotenone and rotenone + sham groups. A markedly higher VMAT2 expression was observed in the vehicle group compared to other groups. RVNS significantly up-regulated VMAT2 expression. Conclusions The improved motor behavior and neuroprotective effects on the midbrain dopaminergic neurons in the PD rat model suggest that RVNS could be used as a potential treatment for PD.
Collapse
Affiliation(s)
- Chaofan Wang
- Ningxia Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yinchuan, China
| | - Tong Su
- Department of Radiology, Ningxia Hui Autonomous Region Hospital and Research Institute of traditional Chinese Medicine, Ningxia Medical University, Yinchuan, China
| | - Lifei Xiao
- Ningxia Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yinchuan, China
| | - Yangyang Wang
- Ningxia Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yinchuan, China
| | - Xianhao Huo
- Ningxia Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yinchuan, China
| | - Wenchao Li
- Ningxia Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yinchuan, China
| | - Jiangwei Ding
- Ningxia Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yinchuan, China
| | - Tao Sun
- Ningxia Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
19
|
Ottaviani MM, Macefield VG. Structure and Functions of the Vagus Nerve in Mammals. Compr Physiol 2022; 12:3989-4037. [PMID: 35950655 DOI: 10.1002/cphy.c210042] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
We review the structure and function of the vagus nerve, drawing on information obtained in humans and experimental animals. The vagus nerve is the largest and longest cranial nerve, supplying structures in the neck, thorax, and abdomen. It is also the only cranial nerve in which the vast majority of its innervation territory resides outside the head. While belonging to the parasympathetic division of the autonomic nervous system, the nerve is primarily sensory-it is dominated by sensory axons. We discuss the macroscopic and microscopic features of the nerve, including a detailed description of its extensive territory. Histochemical and genetic profiles of afferent and efferent axons are also detailed, as are the central nuclei involved in the processing of sensory information conveyed by the vagus nerve and the generation of motor (including parasympathetic) outflow via the vagus nerve. We provide a comprehensive review of the physiological roles of vagal sensory and motor neurons in control of the cardiovascular, respiratory, and gastrointestinal systems, and finish with a discussion on the interactions between the vagus nerve and the immune system. © 2022 American Physiological Society. Compr Physiol 12: 1-49, 2022.
Collapse
Affiliation(s)
- Matteo M Ottaviani
- Department of Neurosurgery, Università Politecnica delle Marche, Ancona, Italy
| | - Vaughan G Macefield
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia.,Department of Anatomy & Physiology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
20
|
Pollina L, Vallone F, Ottaviani MM, Strauss I, Carlucci L, Recchia FA, Micera S, Moccia S. A lightweight learning-based decoding algorithm for intraneural vagus nerve activity classification in pigs. J Neural Eng 2022; 19. [PMID: 35896098 DOI: 10.1088/1741-2552/ac84ab] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/27/2022] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Bioelectronic medicine is an emerging field that aims at developing closed-loop neuromodulation protocols for the autonomic nervous system (ANS) to treat a wide range of disorders. When designing a closed-loop protocol for real time modulation of the ANS, the computational execution time and the memory and power demands of the decoding step are important factors to consider. In the context of cardiovascular and respiratory diseases, these requirements may partially explain why closed-loop clinical neuromodulation protocols that adapt stimulation parameters on patient's clinical characteristics are currently missing. APPROACH Here, we developed a lightweight learning-based decoder for the classification of cardiovascular and respiratory functional challenges from neural signals acquired through intraneural electrodes implanted in the cervical vagus nerve (VN) of 5 anaesthetized pigs. Our algorithm is based on signal temporal windowing, 9 handcrafted features, and Random Forest (RF) model for classification. Temporal windowing ranging from 50 ms to 1 sec, compatible in duration with cardio-respiratory dynamics, was applied to the data in order to mimic a pseudo real-time scenario. MAIN RESULTS We were able to achieve high balanced accuracy (BA) values over the whole range of temporal windowing duration. We identified 500 ms as the optimal temporal windowing duration for both BA values and computational execution time processing, achieving more than 86% for BA and a computational execution time of only ∼6.8 ms. Our algorithm outperformed in terms of balanced accuracy and computational execution time a state of the art decoding algorithm tested on the same dataset [1]. We found that RF outperformed other machine learning models such as Support Vector Machines, K-Nearest Neighbors, and Multi-Layer Perceptrons. SIGNIFICANCE Our approach could represent an important step towards the implementation of a closed-loop neuromodulation protocol relying on a single intraneural interface able to perform real-time decoding tasks and selective modulation of the VN.
Collapse
Affiliation(s)
- Leonardo Pollina
- Sant'Anna School of Advanced Studies, P.za Martiri della Liberta', 33, Pisa, 56127, ITALY
| | - Fabio Vallone
- Sant'Anna School of Advanced Studies, P.za Martiri della Liberta', 33, Pisa, 56127, ITALY
| | - Matteo M Ottaviani
- Scuola Superiore Sant'Anna, Istituto di Scienze Della Vita (ISV), P.za Martiri della Liberta', 33, Pisa, 56127, ITALY
| | - Ivo Strauss
- Scuola Superiore Sant'Anna, P.za Martiri della Libertà 33, Pisa, 56127, ITALY
| | - Lucia Carlucci
- Scuola Superiore Sant'Anna, Istituto di Scienze Della Vita (ISV), P.zza Martiri della Libertà 33, Pisa, 56127, ITALY
| | - Fabio A Recchia
- Scuola Superiore Sant'Anna, Istituto di Scienze Della Vita (ISV), P.za Martiri della Libertà 33, Pisa, 56127, ITALY
| | - Silvestro Micera
- Scuola Superiore Sant'Anna, P.za Martiri della Liberta', 33, Pisa, Toscana, 56127, ITALY
| | - Sara Moccia
- Scuola Superiore Sant'Anna, P.za Martiri della Liberta', 33, Pisa, 56127, ITALY
| |
Collapse
|