1
|
Bremner JD, Russo SJ, Gallagher R, Simon NM. Acute and long-term effects of COVID-19 on brain and mental health: A narrative review. Brain Behav Immun 2025; 123:928-945. [PMID: 39500417 PMCID: PMC11974614 DOI: 10.1016/j.bbi.2024.11.007] [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] [Received: 06/07/2024] [Revised: 09/16/2024] [Accepted: 11/02/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND COVID infection has been associated with long term sequalae (Long COVID) which include neurological and behavioral effects in thousands of patients, but the etiology and scope of symptoms is not well understood. This paper reviews long term sequelae of COVID on brain and mental health in patients with the Long COVID syndrome. METHODS This was a literature review which queried databases for Pubmed, Psychinfo, and Medline for the following topics for January 1, 2020-July 15, 2023: Long COVID, PASC, brain, brain imaging, neurological, neurobiology, mental health, anxiety, depression. RESULTS Tens of thousands of patients have developed Long COVID, with the most common neurobehavioral symptoms anosmia (loss of smell) and fatigue. Anxiety and mood disorders are elevated and seen in about 25% of Long COVID patients. Neuropsychological testing studies show a correlation between symptom severity and cognitive dysfunction, while brain imaging studies show global decreases in gray matter and alterations in olfactory and other brain areas. CONCLUSIONS Studies to date show an increase in neurobehavioral disturbances in patients with Long COVID. Future research is needed to determine mechanisms.
Collapse
Affiliation(s)
- J Douglas Bremner
- Departments of Psychiatry & Behavioral Sciences and Radiology, Emory University School of Medicine, Atlanta Georgia, and the Atlanta VA Medical Center, Decatur, GA, USA; Nash Family Department Neuroscience and Brain-Body Research Center, Icahn School of Medicine at Mt. Sinai, New York, NY, USA; Department of Child and Adolescent Psychiatry, New York University (NYU) Langone Health, New York, NY, USA.
| | - Scott J Russo
- Nash Family Department Neuroscience and Brain-Body Research Center, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Richard Gallagher
- Department of Child and Adolescent Psychiatry, New York University (NYU) Langone Health, New York, NY, USA; Department of Psychiatry, New York University (NYU) Langone Health, New York, NY, USA
| | - Naomi M Simon
- Department of Psychiatry, New York University (NYU) Langone Health, New York, NY, USA
| |
Collapse
|
2
|
Haakana P, Nätkynmäki A, Kirveskari E, Mäkelä JP, Kilgard MP, Tarvainen MP, Shulga A. Effects of auricular vagus nerve stimulation and electrical earlobe stimulation on motor-evoked potential changes induced by paired associative stimulation. Eur J Neurosci 2024; 60:5949-5965. [PMID: 39258329 DOI: 10.1111/ejn.16539] [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: 04/11/2023] [Accepted: 08/29/2024] [Indexed: 09/12/2024]
Abstract
Paired associative stimulation (PAS) is a combination of transcranial magnetic stimulation (TMS) and peripheral nerve stimulation (PNS). PAS can induce long-term potentiation (LTP)-like plasticity in humans, manifested as motor-evoked potential (MEP) enhancement. We have developed a variant of PAS ("high-PAS"), which consists of high-frequency PNS and high-intensity TMS and targets spinal plasticity and promotes rehabilitation after spinal cord injury (SCI). Vagus nerve stimulation (VNS) promotes LTP-like plasticity and enhances recovery in SCI and stroke in humans and animals when combined with repetitive motor training. We combined high-PAS with simultaneous noninvasive transcutaneous auricular VNS (aVNS) to determine if aVNS enhances the extent of PAS-induced MEP amplitude increase. Sixteen healthy participants were stimulated for 20 min in four different sessions (PAS, PAS + aVNS, PAS + shamVNS, and aVNS) in a randomized single-blind setup. MEPs were measured before, immediately after, and at 30, 60, and 90 min post-stimulation. Stimulation protocols with PAS significantly potentiated MEPs (p = 0.005) when compared with aVNS (p = 0.642). Although not significant, MEP enhancement observed after PAS (43.5%) is further increased by aVNS (49.7%) and electrical earlobe stimulation (63.9%). Our aVNS setup failed to significantly enhance the effect of PAS, but sham VNS revealed a trend towards enhanced plasticity. Optimization of auricular VNS stimulation setup is required for possible tests of patients with SCI.
Collapse
Affiliation(s)
- Piia Haakana
- BioMag Laboratory, HUS Diagnostic Center, Helsinki University Hospital, University of Helsinki and Aalto University School of Science, Helsinki, Finland
- Motion Analysis Laboratory, New Children's Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Department of Physiology, University of Helsinki, Helsinki, Finland
| | - Anna Nätkynmäki
- BioMag Laboratory, HUS Diagnostic Center, Helsinki University Hospital, University of Helsinki and Aalto University School of Science, Helsinki, Finland
| | - Erika Kirveskari
- BioMag Laboratory, HUS Diagnostic Center, Helsinki University Hospital, University of Helsinki and Aalto University School of Science, Helsinki, Finland
- HUS Medical Imaging Center, Clinical Neurophysiology; Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Jyrki P Mäkelä
- BioMag Laboratory, HUS Diagnostic Center, Helsinki University Hospital, University of Helsinki and Aalto University School of Science, Helsinki, Finland
| | - Michael P Kilgard
- Texas Biomedical Device Center, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Mika P Tarvainen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Anastasia Shulga
- BioMag Laboratory, HUS Diagnostic Center, Helsinki University Hospital, University of Helsinki and Aalto University School of Science, Helsinki, Finland
- Department of Physical and Rehabilitation Medicine, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
3
|
Du L, He X, Xiong X, Zhang X, Jian Z, Yang Z. Vagus nerve stimulation in cerebral stroke: biological mechanisms, therapeutic modalities, clinical applications, and future directions. Neural Regen Res 2024; 19:1707-1717. [PMID: 38103236 PMCID: PMC10960277 DOI: 10.4103/1673-5374.389365] [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: 06/11/2023] [Revised: 08/31/2023] [Accepted: 09/26/2023] [Indexed: 12/18/2023] Open
Abstract
Stroke is a major disorder of the central nervous system that poses a serious threat to human life and quality of life. Many stroke victims are left with long-term neurological dysfunction, which adversely affects the well-being of the individual and the broader socioeconomic impact. Currently, post-stroke brain dysfunction is a major and difficult area of treatment. Vagus nerve stimulation is a Food and Drug Administration-approved exploratory treatment option for autism, refractory depression, epilepsy, and Alzheimer's disease. It is expected to be a novel therapeutic technique for the treatment of stroke owing to its association with multiple mechanisms such as altering neurotransmitters and the plasticity of central neurons. In animal models of acute ischemic stroke, vagus nerve stimulation has been shown to reduce infarct size, reduce post-stroke neurological damage, and improve learning and memory capacity in rats with stroke by reducing the inflammatory response, regulating blood-brain barrier permeability, and promoting angiogenesis and neurogenesis. At present, vagus nerve stimulation includes both invasive and non-invasive vagus nerve stimulation. Clinical studies have found that invasive vagus nerve stimulation combined with rehabilitation therapy is effective in improving upper limb motor and cognitive abilities in stroke patients. Further clinical studies have shown that non-invasive vagus nerve stimulation, including ear/cervical vagus nerve stimulation, can stimulate vagal projections to the central nervous system similarly to invasive vagus nerve stimulation and can have the same effect. In this paper, we first describe the multiple effects of vagus nerve stimulation in stroke, and then discuss in depth its neuroprotective mechanisms in ischemic stroke. We go on to outline the results of the current major clinical applications of invasive and non-invasive vagus nerve stimulation. Finally, we provide a more comprehensive evaluation of the advantages and disadvantages of different types of vagus nerve stimulation in the treatment of cerebral ischemia and provide an outlook on the developmental trends. We believe that vagus nerve stimulation, as an effective treatment for stroke, will be widely used in clinical practice to promote the recovery of stroke patients and reduce the incidence of disability.
Collapse
Affiliation(s)
- Li Du
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xuan He
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xu Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Zhenxing Yang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
4
|
Berger A, Beckers E, Joris V, Duchêne G, Danthine V, Delinte N, Cakiroglu I, Sherif S, Morrison EIG, Sánchez AT, Macq B, Dricot L, Vandewalle G, El Tahry R. Locus coeruleus features are linked to vagus nerve stimulation response in drug-resistant epilepsy. Front Neurosci 2024; 18:1296161. [PMID: 38469571 PMCID: PMC10926962 DOI: 10.3389/fnins.2024.1296161] [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: 09/18/2023] [Accepted: 01/15/2024] [Indexed: 03/13/2024] Open
Abstract
The locus coeruleus-norepinephrine system is thought to be involved in the clinical effects of vagus nerve stimulation. This system is known to prevent seizure development and induce long-term plastic changes, particularly with the release of norepinephrine in the hippocampus. However, the requisites to become responder to the therapy and the mechanisms of action are still under investigation. Using MRI, we assessed the structural and functional characteristics of the locus coeruleus and microstructural properties of locus coeruleus-hippocampus white matter tracts in patients with drug-resistant epilepsy responding or not to the therapy. Twenty-three drug-resistant epileptic patients with cervical vagus nerve stimulation were recruited for this pilot study, including 13 responders or partial responders and 10 non-responders. A dedicated structural MRI acquisition allowed in vivo localization of the locus coeruleus and computation of its contrast (an accepted marker of LC integrity). Locus coeruleus activity was estimated using functional MRI during an auditory oddball task. Finally, multi-shell diffusion MRI was used to estimate the structural properties of locus coeruleus-hippocampus tracts. These characteristics were compared between responders/partial responders and non-responders and their association with therapy duration was also explored. In patients with a better response to the therapy, trends toward a lower activity and a higher contrast were found in the left medial and right caudal portions of the locus coeruleus, respectively. An increased locus coeruleus contrast, bilaterally over its medial portions, correlated with duration of the treatment. Finally, a higher integrity of locus coeruleus-hippocampus connections was found in patients with a better response to the treatment. These new insights into the neurobiology of vagus nerve stimulation may provide novel markers of the response to the treatment and may reflect neuroplasticity effects occurring in the brain following the implantation.
Collapse
Affiliation(s)
- Alexandre Berger
- Department of Clinical Neuroscience, Institute of Neuroscience, Catholic University of Louvain, Brussels, Belgium
- Synergia Medical SA, Mont-Saint-Guibert, Belgium
- Sleep and Chronobiology Laboratory, GIGA-Cyclotron Research Center-in vivo Imaging, University of Liège, Liège, Belgium
| | - Elise Beckers
- Sleep and Chronobiology Laboratory, GIGA-Cyclotron Research Center-in vivo Imaging, University of Liège, Liège, Belgium
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer’s Centre Limburg, Maastricht University, Maastricht, Netherlands
| | - Vincent Joris
- Department of Clinical Neuroscience, Institute of Neuroscience, Catholic University of Louvain, Brussels, Belgium
- Department of Neurosurgery, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Gaëtan Duchêne
- GE Center MR Applications, General Electric Healthcare, Diegem, Belgium
| | - Venethia Danthine
- Department of Clinical Neuroscience, Institute of Neuroscience, Catholic University of Louvain, Brussels, Belgium
| | - Nicolas Delinte
- Department of Clinical Neuroscience, Institute of Neuroscience, Catholic University of Louvain, Brussels, Belgium
- Institute of Information and Communication Technologies, Electronics and Applied Mathematics, Catholic University of Louvain, Louvain-la-Neuve, Belgium
| | - Inci Cakiroglu
- Department of Clinical Neuroscience, Institute of Neuroscience, Catholic University of Louvain, Brussels, Belgium
| | - Siya Sherif
- Sleep and Chronobiology Laboratory, GIGA-Cyclotron Research Center-in vivo Imaging, University of Liège, Liège, Belgium
| | | | - Andres Torres Sánchez
- Department of Clinical Neuroscience, Institute of Neuroscience, Catholic University of Louvain, Brussels, Belgium
- Innoviris, Brussels Institute for Research and Innovation, Brussels, Belgium
| | - Benoit Macq
- Institute of Information and Communication Technologies, Electronics and Applied Mathematics, Catholic University of Louvain, Louvain-la-Neuve, Belgium
| | - Laurence Dricot
- Department of Clinical Neuroscience, Institute of Neuroscience, Catholic University of Louvain, Brussels, Belgium
| | - Gilles Vandewalle
- Sleep and Chronobiology Laboratory, GIGA-Cyclotron Research Center-in vivo Imaging, University of Liège, Liège, Belgium
| | - Riëm El Tahry
- Department of Clinical Neuroscience, Institute of Neuroscience, Catholic University of Louvain, Brussels, Belgium
- Department of Neurology, Center for Refractory Epilepsy, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
5
|
Sperling MR, Wu C, Kang J, Makhalova J, Bartolomei F, Southwell D. The Temporal Lobe Club: Newer Approaches to Treat Temporal Lobe Epilepsy. Epilepsy Curr 2024; 24:10-15. [PMID: 38327532 PMCID: PMC10846515 DOI: 10.1177/15357597231213161] [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] [Indexed: 02/09/2024] Open
Abstract
This brief review summarizes presentations at the Temporal Lobe Club Special Interest Group session held in December 2022 at the American Epilepsy Society meeting. The session addressed newer methods to treat temporal epilepsy, including methods currently in clinical use and techniques under investigation. Brief summaries are provided for each of 4 lectures. Dr Chengyuan Wu discussed ablative techniques such as laser interstitial thermal ablation, radiofrequency ablation, focused ultrasound; Dr Joon Kang reviewed neuromodulation techniques including electrical stimulation and focused ultrasound; Dr Julia Makhalova discussed network effects of the aforementioned techniques; and Dr Derek Southwell reviewed inhibitory interneuron transplantation. These summaries are intended to provide a brief overview and references are provided for the reader to learn more about each topic.
Collapse
Affiliation(s)
| | - Chengyuan Wu
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Joon Kang
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Julia Makhalova
- APHM, Timone Hospital, Epileptology and Cerebral Rhythmology, Marseille, France
- Aix Marseille Univ, INSERM, INS, Inst Neurosci Syst, Marseille, France
- APHM, Timone Hospital, CEMEREM, Marseille, France
| | - Fabrice Bartolomei
- APHM, Timone Hospital, Epileptology and Cerebral Rhythmology, Marseille, France
- Aix Marseille Univ, INSERM, INS, Inst Neurosci Syst, Marseille, France
| | - Derek Southwell
- Department of Neurosurgery, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
| |
Collapse
|
6
|
Yoo HJ, Nashiro K, Dutt S, Min J, Cho C, Thayer JF, Lehrer P, Chang C, Mather M. Daily biofeedback to modulate heart rate oscillations affects structural volume in hippocampal subregions targeted by the locus coeruleus in older adults but not younger adults. Neurobiol Aging 2023; 132:85-99. [PMID: 37769491 PMCID: PMC10840698 DOI: 10.1016/j.neurobiolaging.2023.08.010] [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: 04/26/2022] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 10/03/2023]
Abstract
Using data from a clinical trial, we tested the hypothesis that daily sessions modulating heart rate oscillations affect older adults' volume of a region-of-interest (ROI) comprised of adjacent hippocampal subregions with relatively strong locus coeruleus (LC) noradrenergic input. Younger and older adults were randomly assigned to one of two daily biofeedback practices for 5 weeks: (1) engage in slow-paced breathing to increase the amplitude of oscillations in heart rate at their breathing frequency (Osc+); (2) engage in self-selected strategies to decrease heart rate oscillations (Osc-). The interventions did not significantly affect younger adults' hippocampal volume. Among older adults, the two conditions affected volume in the LC-targeted hippocampal ROI differentially as reflected in a significant condition × time-point interaction on ROI volume. These condition differences were driven by opposing changes in the two conditions (increased volume in Osc+ and decreased volume in Osc-) and were mediated by the degree of heart rate oscillation during training sessions.
Collapse
Affiliation(s)
- Hyun Joo Yoo
- University of Southern California, Los Angeles, CA 90089, USA
| | - Kaoru Nashiro
- University of Southern California, Los Angeles, CA 90089, USA
| | - Shubir Dutt
- University of Southern California, Los Angeles, CA 90089, USA
| | - Jungwon Min
- University of Southern California, Los Angeles, CA 90089, USA
| | - Christine Cho
- University of Southern California, Los Angeles, CA 90089, USA
| | | | - Paul Lehrer
- Rutgers University, New Brunswick, NJ 08852, USA
| | - Catie Chang
- Vanderbilt University, Nashville, TN 37235, USA
| | - Mara Mather
- University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
7
|
Choudhary T, Elliott M, Euliano NR, Gurel NZ, Rivas AG, Wittbrodt MT, Vaccarino V, Shah AJ, Inan OT, Bremner JD. Effect of transcutaneous cervical vagus nerve stimulation on declarative and working memory in patients with Posttraumatic Stress Disorder (PTSD): A pilot study. J Affect Disord 2023; 339:418-425. [PMID: 37442455 PMCID: PMC11940650 DOI: 10.1016/j.jad.2023.07.025] [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] [Received: 02/16/2023] [Revised: 06/10/2023] [Accepted: 07/08/2023] [Indexed: 07/15/2023]
Abstract
BACKGROUND Posttraumatic stress disorder (PTSD) is associated with changes in multiple neurophysiological systems, including verbal declarative memory deficits. Vagus Nerve Stimulation (VNS) has been shown in preliminary studies to enhance function when paired with cognitive and motor tasks. The purpose of this study was to analyze the effect of transcutaneous cervical VNS (tcVNS) on attention, declarative and working memory in PTSD patients. METHODS Fifteen PTSD patients were randomly assigned to active tcVNS (N = 8) or sham (N = 7) stimulation in a double-blinded fashion. Memory assessment tests including paragraph recall and N-back tests were performed to assess declarative and working memory function when paired with active/sham tcVNS once per month in a longitudinal study during which patients self-administered tcVNS/sham twice daily. RESULTS Active tcVNS stimulation resulted in a significant improvement in paragraph recall performance following pairing with paragraph encoding for PTSD patients at two months (p < 0.05). It resulted in a 91 % increase in paragraph recall performance within group (p = 0.03), while sham tcVNS exhibited no such trend in performance improvement. In the N-back study, positive deviations in accuracy, precision and recall measures on different day visits (7,34,64,94) of patients with respect to day 1 revealed a pattern of better performance of the active tcVNS population compared to sham VNS which did not reach statistical significance. LIMITATIONS Our sample size was small. CONCLUSIONS These preliminary results suggest that tcVNS improves attention, declarative and working memory, which may improve quality of life and productivity for patients with PTSD. Future studies are required to confirm these results.
Collapse
Affiliation(s)
- Tilendra Choudhary
- Department of Psychiatry & Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA; Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA; Atlanta VA Medical Center, Decatur, GA, USA.
| | | | | | - Nil Z Gurel
- Reality Labs, Meta Platforms Inc., Menlo Park, CA, USA
| | - Amanda G Rivas
- Department of Psychiatry & Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Matthew T Wittbrodt
- Department of Psychiatry & Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Viola Vaccarino
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA; Department of Medicine, Cardiology Division, Emory University School of Medicine, Atlanta, GA, USA
| | - Amit J Shah
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA; Department of Medicine, Cardiology Division, Emory University School of Medicine, Atlanta, GA, USA; Atlanta VA Medical Center, Decatur, GA, USA
| | - Omer T Inan
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Coulter Department of Bioengineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - J Douglas Bremner
- Department of Psychiatry & Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA; Atlanta VA Medical Center, Decatur, GA, USA; Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
8
|
Yoo HJ, Nashiro K, Dutt S, Min J, Cho C, Thayer JF, Lehrer P, Chang C, Mather M. Daily biofeedback to modulate heart rate oscillations affects structural volume in hippocampal subregions targeted by the locus coeruleus in older adults but not younger adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.02.23286715. [PMID: 37745356 PMCID: PMC10516053 DOI: 10.1101/2023.03.02.23286715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Using data from a clinical trial, we tested the hypothesis that daily sessions modulating heart rate oscillations affect older adults' volume of a region-of-interest (ROI) comprised of adjacent hippocampal subregions with relatively strong locus coeruleus (LC) noradrenergic input. Younger and older adults were randomly assigned to one of two daily biofeedback practices for 5 weeks: 1) engage in slow-paced breathing to increase the amplitude of oscillations in heart rate at their breathing frequency (Osc+); 2) engage in self-selected strategies to decrease heart rate oscillations (Osc-). The interventions did not significantly affect younger adults' hippocampal volume. Among older adults, the two conditions affected volume in the LC-targeted hippocampal ROI differentially as reflected in a significant condition x time-point interaction on ROI volume. These condition differences were driven by opposing changes in the two conditions (increased volume in Osc+ and decreased volume in Osc-) and were mediated by the degree of heart rate oscillation during training sessions.
Collapse
Affiliation(s)
- Hyun Joo Yoo
- University of Southern California, Los Angeles, CA 90089
| | - Kaoru Nashiro
- University of Southern California, Los Angeles, CA 90089
| | - Shubir Dutt
- University of Southern California, Los Angeles, CA 90089
| | - Jungwon Min
- University of Southern California, Los Angeles, CA 90089
| | - Christine Cho
- University of Southern California, Los Angeles, CA 90089
| | | | | | | | - Mara Mather
- University of Southern California, Los Angeles, CA 90089
| |
Collapse
|
9
|
Domenech P. Stimulation du nerf vague pour traiter l’épilepsie et la dépression résistante : vers une physiopathologie commune ? BULLETIN DE L'ACADÉMIE NATIONALE DE MÉDECINE 2023. [DOI: 10.1016/j.banm.2023.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
|
10
|
Kanatome A, Takara T, Umeda S, Ano Y. Effects of matured hop bitter acids on heart rate variability and cognitive performance: A randomized placebo-controlled crossover trial. J Funct Foods 2023. [DOI: 10.1016/j.jff.2022.105383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
11
|
Effect of transcutaneous auricular vagus nerve stimulation on major depressive disorder with peripartum onset: A multicenter, open-label, controlled proof-of-concept clinical trial (DELOS-1). J Affect Disord 2022; 316:34-41. [PMID: 35932937 DOI: 10.1016/j.jad.2022.07.068] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 07/23/2022] [Accepted: 07/30/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Postpartum depression has a high prevalence in the United States (~13 %) and often goes undertreated/untreated. We conducted a multicenter, open-label, proof-of-concept trial to assess the Nēsos wearable, non-invasive, transcutaneous auricular vagus nerve stimulation (taVNS) system for the treatment of major depressive disorder with peripartum onset (PPD). METHODS Women (n = 25), ages 18 to 45, within 9 months postpartum, and diagnosed with PPD were enrolled at 3 sites. The study included 6 weeks open-label therapy and 2 weeks observation. Efficacy outcomes included change from baseline (CFB) in Hamilton Rating Scale for Depression (HAMD17) total scores, HAM-D17 response and remission, and patient and clinician global impression of change (PGIC, CGIC) scores. Analysis included descriptive statistics and mixed-effects models for repeated measures. RESULTS The most common AEs (≥5 %) were discomfort (n = 5), headache (n = 3), and dizziness (n = 2); all resolved without intervention. No serious AEs or deaths occurred. Baseline mean HAM-D17 score was 18.4. Week 6 least squares (LS) mean CFB in HAM-D17 score was -9.7; 74 % achieved response and 61 % achieved remission. At week 6, at least some improvement was reported by 21 of 22 (95 %) clinicians on CGIC and 22 of 23 (96 %) participants on PGIC. LIMITATIONS This was a single-arm, open-label study, and enrollment was limited to participants with mild-to-moderate peripartum depression. CONCLUSION Results from this proof-of-concept study suggest that the Nēsos taVNS system is well tolerated and may be an effective non-invasive, non-pharmacological treatment for major depressive disorder with peripartum onset. Further evaluation in larger sham-controlled studies is needed. CLINICALTRIALS govNCT03972995.
Collapse
|
12
|
Guzzetta KE, Cryan JF, O’Leary OF. Microbiota-Gut-Brain Axis Regulation of Adult Hippocampal Neurogenesis. Brain Plast 2022; 8:97-119. [PMID: 36448039 PMCID: PMC9661352 DOI: 10.3233/bpl-220141] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2022] [Indexed: 11/15/2022] Open
Abstract
The birth, maturation, and integration of new neurons in the adult hippocampus regulates specific learning and memory processes, responses to stress, and antidepressant treatment efficacy. This process of adult hippocampal neurogenesis is sensitive to environmental stimuli, including peripheral signals from certain cytokines, hormones, and metabolites, which can promote or hinder the production and survival of new hippocampal neurons. The trillions of microorganisms resident to the gastrointestinal tract, collectively known as the gut microbiota, also demonstrate the ability to modulate adult hippocampal neurogenesis. In doing so, the microbiota-gut-brain axis can influence brain functions regulated by adult hippocampal neurogenesis. Unlike the hippocampus, the gut microbiota is highly accessible to direct interventions, such as prebiotics, probiotics, and antibiotics, and can be manipulated by lifestyle choices including diet. Therefore, understanding the pathways by which the gut microbiota shapes hippocampal neurogenesis may reveal novel targets for non-invasive therapeutics to treat disorders in which alterations in hippocampal neurogenesis have been implicated. This review first outlines the factors which influence both the gut microbiome and adult hippocampal neurogenesis, with cognizance that these effects might happen either independently or due to microbiota-driven mechanisms. We then highlight approaches for investigating the regulation of adult hippocampal neurogenesis by the microbiota-gut-brain axis. Finally, we summarize the current evidence demonstrating the gut microbiota's ability to influence adult hippocampal neurogenesis, including mechanisms driven through immune pathways, microbial metabolites, endocrine signalling, and the nervous system, and postulate implications for these effects in disease onset and treatment.
Collapse
Affiliation(s)
- Katherine E. Guzzetta
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - John F. Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Olivia F. O’Leary
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
13
|
Souza RR, Powers MB, Rennaker RL, McIntyre CK, Hays SA, Kilgard MP. Timing of vagus nerve stimulation during fear extinction determines efficacy in a rat model of PTSD. Sci Rep 2022; 12:16526. [PMID: 36192564 PMCID: PMC9530175 DOI: 10.1038/s41598-022-20301-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/12/2022] [Indexed: 11/09/2022] Open
Abstract
Studies have indicated that vagus nerve stimulation (VNS) enhances extinction learning in rodent models. Here, we investigated if pairing VNS with the conditioned stimulus is required for the enhancing effects of VNS. Adult Sprague-Dawley rats were exposed to intense stress followed by fear conditioning training to produce resistant fear. Rats were then implanted with a cuff electrode around the left vagus. After recovery, rats underwent extinction training paired with VNS (0.5 s, 0.8 mA, 100 µs, and 30 Hz) or with Sham VNS (0 mA). VNS rats were randomized into the following subgroups: During VNS (delivered during presentations of the conditioned stimulus, CS), Between VNS (delivered between CS presentations), Continuous VNS (delivered during the entire extinction session), and Dispersed VNS (delivered at longer inter-stimulation intervals across the extinction session). Sham VNS rats failed to extinguish the conditioned fear response over 5 days of repeated exposure to the CS. Rats that received Between or Dispersed VNS showed modest improvement in conditioned fear at the retention test. During and Continuous VNS groups displayed the greatest reduction in conditioned fear. These findings indicate that delivering VNS paired precisely with CS presentations or continuously throughout extinction promotes the maximum enhancement in extinction learning.
Collapse
Affiliation(s)
- Rimenez R Souza
- Texas Biomedical Device Center, The University of Texas at Dallas, Richardson, TX, 75080, USA.
- School of Behavioral Brain Sciences, The University of Texas at Dallas, Bioengineering and Sciences Building, 14.506, 800 West Campbell Road, Richardson, TX, 75080, USA.
| | - Mark B Powers
- Baylor University Medical Center, Dallas, TX, 75246, USA
| | - Robert L Rennaker
- Texas Biomedical Device Center, The University of Texas at Dallas, Richardson, TX, 75080, USA
- School of Behavioral Brain Sciences, The University of Texas at Dallas, Bioengineering and Sciences Building, 14.506, 800 West Campbell Road, Richardson, TX, 75080, USA
- Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Christa K McIntyre
- Texas Biomedical Device Center, The University of Texas at Dallas, Richardson, TX, 75080, USA
- School of Behavioral Brain Sciences, The University of Texas at Dallas, Bioengineering and Sciences Building, 14.506, 800 West Campbell Road, Richardson, TX, 75080, USA
| | - Seth A Hays
- Texas Biomedical Device Center, The University of Texas at Dallas, Richardson, TX, 75080, USA
- Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Michael P Kilgard
- Texas Biomedical Device Center, The University of Texas at Dallas, Richardson, TX, 75080, USA
- School of Behavioral Brain Sciences, The University of Texas at Dallas, Bioengineering and Sciences Building, 14.506, 800 West Campbell Road, Richardson, TX, 75080, USA
| |
Collapse
|
14
|
Kumaria A, Noah A, Kirkman MA. Does covid-19 impair endogenous neurogenesis? J Clin Neurosci 2022; 105:79-85. [PMID: 36113246 DOI: 10.1016/j.jocn.2022.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/14/2022] [Accepted: 09/03/2022] [Indexed: 10/31/2022]
Abstract
Endogenous neural stem cells are thought to continue to generate new neurons throughout life in the human brain. Endogenous neurogenesis has been proposed to contribute to physiological roles in maintaining and regenerating olfaction, as well as promoting normal cognition, learning and memory. Specific impairments in these processes in COVID-19 - impaired olfaction and cognition - may implicate the SARS-CoV-2 virus in attenuating neurogenesis. Furthermore, neurogenesis has been linked with neuroregeneration; and impaired neuroregeneration has previously been linked with neurodegenerative diseases. Emerging evidence supports an association between COVID-19 infection and accelerated neurodegeneration. Also, structural changes indicating global reduction in brain size and specific reduction in the size of limbic structures - including orbitofrontal cortex, olfactory cortex and parahippocampal gyrus - as a result of SARS-CoV-2 infection have been demonstrated. This paper proposes the hypothesis that SARS-CoV-2 infection may impair endogenous neural stem cell activity. An attenuation of neurogenesis may contribute to reduction in brain size and/or neurodegenerative processes following SARS-CoV-2 infection. Furthermore, as neural stem cells are thought to be the cell of origin in glioma, better understanding of SARS-CoV-2 interaction with tumorigenic stem cells is indicated, with a view to informing therapeutic modulation. The subacute and chronic implications of attenuated endogenous neurogenesis are explored in the context of long COVID. Modulating endogenous neurogenesis may be a novel therapeutic strategy to address specific neurological manifestations of COVID-19 and potential applicability in tumour virotherapy.
Collapse
Affiliation(s)
- Ashwin Kumaria
- Department of Neurosurgery, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Abiodun Noah
- Anaesthesia and Critical Care, Academic Unit of Injury, Inflammation and Recovery Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Matthew A Kirkman
- Department of Neurosurgery, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham, UK
| |
Collapse
|
15
|
Kamel LY, Xiong W, Gott BM, Kumar A, Conway CR. Vagus nerve stimulation: An update on a novel treatment for treatment-resistant depression. J Neurol Sci 2022; 434:120171. [DOI: 10.1016/j.jns.2022.120171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 12/21/2021] [Accepted: 01/21/2022] [Indexed: 12/11/2022]
|
16
|
Berger A, Vespa S, Dricot L, Dumoulin M, Iachim E, Doguet P, Vandewalle G, El Tahry R. How Is the Norepinephrine System Involved in the Antiepileptic Effects of Vagus Nerve Stimulation? Front Neurosci 2021; 15:790943. [PMID: 34924947 PMCID: PMC8675889 DOI: 10.3389/fnins.2021.790943] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/11/2021] [Indexed: 01/09/2023] Open
Abstract
Vagus Nerve Stimulation (VNS) is an adjunctive treatment for patients suffering from inoperable drug-resistant epilepsy. Although a complete understanding of the mediators involved in the antiepileptic effects of VNS and their complex interactions is lacking, VNS is known to trigger the release of neurotransmitters that have seizure-suppressing effects. In particular, norepinephrine (NE) is a neurotransmitter that has been associated with the clinical effects of VNS by preventing seizure development and by inducing long-term plastic changes that could restore a normal function of the brain circuitry. However, the biological requisites to become responder to VNS are still unknown. In this review, we report evidence of the critical involvement of NE in the antiepileptic effects of VNS in rodents and humans. Moreover, we emphasize the hypothesis that the functional integrity of the noradrenergic system could be a determining factor to obtain clinical benefits from the therapy. Finally, encouraging avenues of research involving NE in VNS treatment are discussed. These could lead to the personalization of the stimulation parameters to maximize the antiepileptic effects and potentially improve the response rate to the therapy.
Collapse
Affiliation(s)
- Alexandre Berger
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium.,Synergia Medical SA, Mont-Saint-Guibert, Belgium.,GIGA-Cyclotron Research Center-In Vivo Imaging, University of Liège, Liège, Belgium
| | - Simone Vespa
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Laurence Dricot
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Manon Dumoulin
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Evelina Iachim
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium.,Department of Pediatric Neurology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Gilles Vandewalle
- GIGA-Cyclotron Research Center-In Vivo Imaging, University of Liège, Liège, Belgium
| | - Riëm El Tahry
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium.,Center for Refractory Epilepsy, Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
17
|
Tseng CT, Gaulding SJ, Dancel CLE, Thorn CA. Local activation of α2 adrenergic receptors is required for vagus nerve stimulation induced motor cortical plasticity. Sci Rep 2021; 11:21645. [PMID: 34737352 PMCID: PMC8568982 DOI: 10.1038/s41598-021-00976-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 10/20/2021] [Indexed: 11/09/2022] Open
Abstract
Vagus nerve stimulation (VNS) paired with rehabilitation training is emerging as a potential treatment for improving recovery of motor function following stroke. In rats, VNS paired with skilled forelimb training results in significant reorganization of the somatotopic cortical motor map; however, the mechanisms underlying this form of VNS-dependent plasticity remain unclear. Recent studies have shown that VNS-driven cortical plasticity is dependent on noradrenergic innervation of the neocortex. In the central nervous system, noradrenergic α2 receptors (α2-ARs) are widely expressed in the motor cortex and have been critically implicated in synaptic communication and plasticity. In current study, we examined whether activation of cortical α2-ARs is necessary for VNS-driven motor cortical reorganization to occur. Consistent with previous studies, we found that VNS paired with motor training enlarges the map representation of task-relevant musculature in the motor cortex. Infusion of α2-AR antagonists into M1 blocked VNS-driven motor map reorganization from occurring. Our results suggest that local α2-AR activation is required for VNS-induced cortical reorganization to occur, providing insight into the mechanisms that may underlie the neuroplastic effects of VNS therapy.
Collapse
Affiliation(s)
- Ching-Tzu Tseng
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 W. Campbell Rd, Richardson, TX, 75080, USA
| | - Solomon J Gaulding
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 W. Campbell Rd, Richardson, TX, 75080, USA
| | - Canice Lei E Dancel
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 W. Campbell Rd, Richardson, TX, 75080, USA
| | - Catherine A Thorn
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 W. Campbell Rd, Richardson, TX, 75080, USA.
| |
Collapse
|
18
|
Keute M, Gharabaghi A. Brain plasticity and vagus nerve stimulation. Auton Neurosci 2021; 236:102876. [PMID: 34537681 DOI: 10.1016/j.autneu.2021.102876] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/01/2021] [Accepted: 08/29/2021] [Indexed: 01/01/2023]
Abstract
After damage to the central nervous system, caused by traumatic injury or ischemia, plasticity becomes critically important for functional recovery. When this inherent capacity to adapt is limited despite training, external stimulation may support this process. Vagus nerve stimulation (VNS) is an effective method to enhance the effect of motor rehabilitation training on functional recovery. However, the mechanisms by which VNS exerts beneficial effects on cortical plasticity are not completely understood. Experimental work suggests that VNS fosters a neurochemical milieu that facilitates synaptic plasticity and supports reinforcement mechanisms. Animal studies, furthermore, suggest that VNS delivery is time-critical and that optima in the parameter space need to be titrated for effect maximization. Human studies suggest that VNS modifies corticospinal excitability. First studies in stroke patients show positive results for invasive, and also promising findings for non-invasive VNS.
Collapse
Affiliation(s)
- Marius Keute
- Institute for Neuromodulation and Neurotechnology, Department of Neurosurgery and Neurotechnology, University of Tuebingen, Tuebingen, Germany.
| | - Alireza Gharabaghi
- Institute for Neuromodulation and Neurotechnology, Department of Neurosurgery and Neurotechnology, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
19
|
Kumaria A, Basu S, Tolias CM. Vagus nerve stimulation in ischaemic stroke: further possibilities. Br J Neurosurg 2021; 36:425-426. [PMID: 34281451 DOI: 10.1080/02688697.2021.1950633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Ashwin Kumaria
- Department of Neurosurgery, Queen's Medical Centre, Nottingham, UK
| | - Surajit Basu
- Department of Neurosurgery, Queen's Medical Centre, Nottingham, UK.,School of Medicine, University of Nottingham, Nottingham, UK
| | | |
Collapse
|
20
|
Kucuker MU, Almorsy AG, Sonmez AI, Ligezka AN, Doruk Camsari D, Lewis CP, Croarkin PE. A Systematic Review of Neuromodulation Treatment Effects on Suicidality. Front Hum Neurosci 2021; 15:660926. [PMID: 34248523 PMCID: PMC8267816 DOI: 10.3389/fnhum.2021.660926] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/17/2021] [Indexed: 12/15/2022] Open
Abstract
Introduction: Neuromodulation is an important group of therapeutic modalities for neuropsychiatric disorders. Prior studies have focused on efficacy and adverse events associated with neuromodulation. Less is known regarding the influence of neuromodulation treatments on suicidality. This systematic review sought to examine the effects of various neuromodulation techniques on suicidality. Methods: A systematic review of the literature from 1940 to 2020 following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guideline was conducted. Any reported suicide-related outcome, including suicidal ideation, suicide intent, suicide attempt, completed suicide in reports were considered as a putative measure of treatment effect on suicidality. Results: The review identified 129 relevant studies. An exploratory analysis of a randomized controlled trial comparing the effects of sertraline and transcranial direct-current stimulation (tDCS) for treating depression reported a decrease in suicidal ideation favoring tDCS vs. placebo and tDCS combined with sertraline vs. placebo. Several studies reported an association between repetitive transcranial magnetic stimulation and improvements in suicidal ideation. In 12 of the studies, suicidality was the primary outcome, ten of which showed a significant improvement in suicidal ideation. Electroconvulsive therapy (ECT) and magnetic seizure therapy was also shown to be associated with lower suicidal ideation and completed suicide rates. There were 11 studies which suicidality was the primary outcome and seven of these showed an improvement in suicidal ideation or suicide intent and fewer suicide attempts or completed suicides in patients treated with ECT. There was limited literature focused on the potential protective effect of vagal nerve stimulation with respect to suicidal ideation. Data were mixed regarding the potential effects of deep brain stimulation on suicidality. Conclusions: Future prospective studies of neuromodulation that focus on the primary outcome of suicidality are urgently needed. Systematic Review Registration: https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=125599, identifier: CRD42019125599.
Collapse
Affiliation(s)
- Mehmet Utku Kucuker
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
| | - Ammar G. Almorsy
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
| | - Ayse Irem Sonmez
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, United States
| | - Anna N. Ligezka
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, United States
| | - Deniz Doruk Camsari
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
| | - Charles P. Lewis
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, United States
| | - Paul E. Croarkin
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
21
|
Venkatasamy L, Nizamutdinov D, Jenkins J, Shapiro LA. Vagus Nerve Stimulation Ameliorates Cognitive Impairment and Increased Hippocampal Astrocytes in a Mouse Model of Gulf War Illness. Neurosci Insights 2021; 16:26331055211018456. [PMID: 34104886 PMCID: PMC8165814 DOI: 10.1177/26331055211018456] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/29/2021] [Indexed: 01/17/2023] Open
Abstract
Gulf war illness (GWI), is a chronic multi-symptom illness that has impacted approximately one-third of the veterans who served in the 1990 to 1991 Gulf War. GWI symptoms include cognitive impairments (eg, memory and concentration problems), headaches, migraines, fatigue, gastrointestinal and respiratory issues, as well as emotional deficits. The exposure to neurological chemicals such as the anti-nerve gas drug, pyridostigmine bromide (PB), and the insecticide permethrin (PER), may contribute to the etiologically related factors of GWI. Various studies utilizing mouse models of GWI have reported the interplay of these chemical agents in increasing neuroinflammation and cognitive dysfunction. Astrocytes are involved in the secretion of neuroinflammatory cytokines and chemokines in pathological conditions and have been implicated in GWI symptomology. We hypothesized that exposure to PB and PER causes lasting changes to hippocampal astrocytes, concurrent with chronic cognitive deficits that can be reversed by cervical vagus nerve stimulation (VNS). GWI was induced in CD1 mice by injecting the mixture of PER (200 mg/kg) and PB (2 mg/kg), i.p. for 10 consecutive days. VNS stimulators were implanted at 33 weeks after GWI induction. The results show age-related cognitive alterations at approximately 9 months after exposure to PB and PER. The results also showed an increased number of GFAP-labeled astrocytes in the hippocampus and dentate gyrus that was ameliorated by VNS.
Collapse
Affiliation(s)
- Lavanya Venkatasamy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University, Bryan, TX, USA
| | - Damir Nizamutdinov
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University, Bryan, TX, USA
| | - Jaclyn Jenkins
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University, Bryan, TX, USA
| | - Lee A Shapiro
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University, Bryan, TX, USA
| |
Collapse
|
22
|
Vlaicu A, Bustuchina Vlaicu M. Vagus nerve stimulation for treatment-resistant depression: is this therapy distinct from other antidepressant treatments? Int J Psychiatry Clin Pract 2020; 24:349-356. [PMID: 32677482 DOI: 10.1080/13651501.2020.1779751] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND The treatment-resistant depression (TRD) is a very disabling disease. OBJECTIVE The aim of this article is to provide an overview of the therapeutic activity of vagus nerve stimulation (VNS) therapy system in TRD. We summarised the progress made during the last decade in this area. METHODS We conducted a non-systematic review on the efficacy and safety of the VNS therapy for this disease. We analysed the results from acute and long-term studies that utilised this technique. Major electronic databases were searched. RESULTS The patients with TRD may show acute and long-term benefit when treated with this technique. There are promising results for VNS therapy for these patients. The level of evidence as an acute treatment option is only 3, but as chronic treatment is 2. This therapy should be offered as an added long-term treatment option for patients with chronic and recurrent difficult to treat depression. CONCLUSIONS The antidepressant effects of this procedure remain controversial. The clinical trials have produced mixed results, but VNS therapy for TRD has two distinct features that differentiate it from other antidepressant treatments: a sustained therapeutic response obtained in highly resistant depressive disorders, a favourable safety profile and guaranteed compliance.
Collapse
Affiliation(s)
- Andrei Vlaicu
- Service of Psychiatry, Hospital Andre Breton, Saint-Dizier, France
| | - Mihaela Bustuchina Vlaicu
- Neurosurgery Department, Hospital Pitié Salpêtrière, Paris, France.,INSERM, U955, The Translational Psychiatry Laboratory, Créteil, France
| |
Collapse
|
23
|
Cambiaghi M, Crupi R, Bautista EL, Elsamadisi A, Malik W, Pozdniakova H, Han Z, Buffelli M, Battaglia F. The Effects of 1-Hz rTMS on Emotional Behavior and Dendritic Complexity of Mature and Newly Generated Dentate Gyrus Neurons in Male Mice. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17114074. [PMID: 32521613 PMCID: PMC7312937 DOI: 10.3390/ijerph17114074] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/14/2022]
Abstract
Low-frequency repetitive transcranial magnetic stimulation (1-Hz rTMS) is a promising noninvasive tool for the treatment of depression. Hippocampal neuronal plasticity is thought to play a pivotal role in the pathophysiology of depressive disorders and the mechanism of action of antidepressant treatments. We investigated the effect of 1-Hz rTMS treatment on hippocampal dentate gyrus structural plasticity and related emotional behaviors modifications. Experimentally, adult male mice received either five days of 1-Hz rTMS or Sham stimulation. After stimulation, the mice underwent a battery of tests for anxiety-like and depression-like behaviors. We also tested the effect of treatment on mature and newly generated granule cell dendritic complexity. Our data showed that 1-Hz rTMS induced structural plasticity in mature granule cells, as evidenced by increased dendritic length and number of intersections. However, the stimulation did not increase the proliferation of the dentate gyrus progenitor cells. On the contrary, the stimulated mice showed increased dendritic complexity of newly generated neurons. Moreover, 1-Hz rTMS resulted in antidepressant-like effects in the tail suspension test, but it did not affect anxiety-like behaviors. Therefore, our results indicate that 1-Hz rTMS modulates dentate gyrus morphological plasticity in mature and newly generated neurons. Furthermore, our data provide some evidence of an association between the antidepressant-like activity of 1-Hz rTMS and structural plasticity in the hippocampus.
Collapse
Affiliation(s)
- Marco Cambiaghi
- Department of Neurosciences, Biomedicine and Movement Sciences-University of Verona, 37134 Verona, Italy; (M.C.); (M.B.)
| | - Rosalia Crupi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98125 Messina, Italy;
| | - Erick Larios Bautista
- Department of Medical Sciences, Neurology and Psychiatry, Hackensack Meridian School of Medicine, Seton Hall University, Nutley, NJ 07110, USA; (E.L.B.); (A.E.); (W.M.); (H.P.); (Z.H.)
| | - Amir Elsamadisi
- Department of Medical Sciences, Neurology and Psychiatry, Hackensack Meridian School of Medicine, Seton Hall University, Nutley, NJ 07110, USA; (E.L.B.); (A.E.); (W.M.); (H.P.); (Z.H.)
| | - Wasib Malik
- Department of Medical Sciences, Neurology and Psychiatry, Hackensack Meridian School of Medicine, Seton Hall University, Nutley, NJ 07110, USA; (E.L.B.); (A.E.); (W.M.); (H.P.); (Z.H.)
| | - Helen Pozdniakova
- Department of Medical Sciences, Neurology and Psychiatry, Hackensack Meridian School of Medicine, Seton Hall University, Nutley, NJ 07110, USA; (E.L.B.); (A.E.); (W.M.); (H.P.); (Z.H.)
| | - Zhiyong Han
- Department of Medical Sciences, Neurology and Psychiatry, Hackensack Meridian School of Medicine, Seton Hall University, Nutley, NJ 07110, USA; (E.L.B.); (A.E.); (W.M.); (H.P.); (Z.H.)
| | - Mario Buffelli
- Department of Neurosciences, Biomedicine and Movement Sciences-University of Verona, 37134 Verona, Italy; (M.C.); (M.B.)
| | - Fortunato Battaglia
- Department of Medical Sciences, Neurology and Psychiatry, Hackensack Meridian School of Medicine, Seton Hall University, Nutley, NJ 07110, USA; (E.L.B.); (A.E.); (W.M.); (H.P.); (Z.H.)
- Correspondence: ; Tel.: +97-3761-9605
| |
Collapse
|
24
|
Adair D, Truong D, Esmaeilpour Z, Gebodh N, Borges H, Ho L, Bremner JD, Badran BW, Napadow V, Clark VP, Bikson M. Electrical stimulation of cranial nerves in cognition and disease. Brain Stimul 2020; 13:717-750. [PMID: 32289703 PMCID: PMC7196013 DOI: 10.1016/j.brs.2020.02.019] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 02/13/2020] [Accepted: 02/17/2020] [Indexed: 02/06/2023] Open
Abstract
The cranial nerves are the pathways through which environmental information (sensation) is directly communicated to the brain, leading to perception, and giving rise to higher cognition. Because cranial nerves determine and modulate brain function, invasive and non-invasive cranial nerve electrical stimulation methods have applications in the clinical, behavioral, and cognitive domains. Among other neuromodulation approaches such as peripheral, transcranial and deep brain stimulation, cranial nerve stimulation is unique in allowing axon pathway-specific engagement of brain circuits, including thalamo-cortical networks. In this review we amalgamate relevant knowledge of 1) cranial nerve anatomy and biophysics; 2) evidence of the modulatory effects of cranial nerves on cognition; 3) clinical and behavioral outcomes of cranial nerve stimulation; and 4) biomarkers of nerve target engagement including physiology, electroencephalography, neuroimaging, and behavioral metrics. Existing non-invasive stimulation methods cannot feasibly activate the axons of only individual cranial nerves. Even with invasive stimulation methods, selective targeting of one nerve fiber type requires nuance since each nerve is composed of functionally distinct axon-types that differentially branch and can anastomose onto other nerves. None-the-less, precisely controlling stimulation parameters can aid in affecting distinct sets of axons, thus supporting specific actions on cognition and behavior. To this end, a rubric for reproducible dose-response stimulation parameters is defined here. Given that afferent cranial nerve axons project directly to the brain, targeting structures (e.g. thalamus, cortex) that are critical nodes in higher order brain networks, potent effects on cognition are plausible. We propose an intervention design framework based on driving cranial nerve pathways in targeted brain circuits, which are in turn linked to specific higher cognitive processes. State-of-the-art current flow models that are used to explain and design cranial-nerve-activating stimulation technology require multi-scale detail that includes: gross anatomy; skull foramina and superficial tissue layers; and precise nerve morphology. Detailed simulations also predict that some non-invasive electrical or magnetic stimulation approaches that do not intend to modulate cranial nerves per se, such as transcranial direct current stimulation (tDCS) and transcranial magnetic stimulation (TMS), may also modulate activity of specific cranial nerves. Much prior cranial nerve stimulation work was conceptually limited to the production of sensory perception, with individual titration of intensity based on the level of perception and tolerability. However, disregarding sensory emulation allows consideration of temporal stimulation patterns (axon recruitment) that modulate the tone of cortical networks independent of sensory cortices, without necessarily titrating perception. For example, leveraging the role of the thalamus as a gatekeeper for information to the cerebral cortex, preventing or enhancing the passage of specific information depending on the behavioral state. We show that properly parameterized computational models at multiple scales are needed to rationally optimize neuromodulation that target sets of cranial nerves, determining which and how specific brain circuitries are modulated, which can in turn influence cognition in a designed manner.
Collapse
Affiliation(s)
- Devin Adair
- Department of Biomedical Engineering, City College of New York, New York, NY, USA
| | - Dennis Truong
- Department of Biomedical Engineering, City College of New York, New York, NY, USA
| | - Zeinab Esmaeilpour
- Department of Biomedical Engineering, City College of New York, New York, NY, USA.
| | - Nigel Gebodh
- Department of Biomedical Engineering, City College of New York, New York, NY, USA
| | - Helen Borges
- Department of Biomedical Engineering, City College of New York, New York, NY, USA
| | - Libby Ho
- Department of Biomedical Engineering, City College of New York, New York, NY, USA
| | - J Douglas Bremner
- Department of Psychiatry & Behavioral Sciences and Radiology, Emory University School of Medicine, Atlanta, GA, USA; Atlanta VA Medical Center, Decatur, GA, USA
| | - Bashar W Badran
- Department of Psychiatry & Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Vitaly Napadow
- Martinos Center for Biomedical Imaging, Department of Radiology, MGH, Harvard medical school, Boston, MA, USA
| | - Vincent P Clark
- Psychology Clinical Neuroscience Center, Dept. Psychology, MSC03-2220, University of New Mexico, Albuquerque, NM, 87131, USA; Department of Psychology, University of New Mexico, Albuquerque, NM, 87131, USA; The Mind Research Network of the Lovelace Biomedical Research Institute, 1101 Yale Blvd. NE, Albuquerque, NM, 87106, USA
| | - Marom Bikson
- Department of Biomedical Engineering, City College of New York, New York, NY, USA.
| |
Collapse
|
25
|
Souza RR, Robertson NM, Mathew E, Tabet MN, Bucksot JE, Pruitt DT, Rennaker RL, Hays SA, McIntyre CK, Kilgard MP. Efficient parameters of vagus nerve stimulation to enhance extinction learning in an extinction-resistant rat model of PTSD. Prog Neuropsychopharmacol Biol Psychiatry 2020; 99:109848. [PMID: 31863872 DOI: 10.1016/j.pnpbp.2019.109848] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 12/11/2019] [Accepted: 12/16/2019] [Indexed: 12/20/2022]
Abstract
Vagus nerve stimulation (VNS) has shown promise as an adjuvant treatment for posttraumatic stress disorder (PTSD), as it enhances fear extinction and reduces anxiety symptoms in multiple rat models of this condition. Yet, identification of the optimal stimulation paradigm is needed to facilitate clinical translation of this potential therapy. Using an extinction-resistant rat model of PTSD, we tested whether varying VNS intensity and duration could maximize extinction learning while minimizing the total amount of stimulation. We confirmed that sham rats failed to extinguish after a week of extinction training. Delivery of the standard LONG VNS trains (30 s) at 0.4 mA enhanced extinction and reduced anxiety but did not prevent fear return. Increasing the intensity of LONG VNS trains to 0.8 mA prevented fear return and attenuated anxiety symptoms. Interestingly, delivering 1, 4 or 16 SHORT VNS bursts (0.5 s) at 0.8 mA during each cue presentation in extinction training also enhanced extinction. LONG VNS trains or multiple SHORT VNS bursts at 0.8 mA attenuated fear renewal and reinstatement, promoted extinction generalization and reduced generalized anxiety. Delivering 16 SHORT VNS bursts also facilitated extinction in fewer trials. This study provides the first evidence that brief bursts of VNS can enhance extinction training, reduce relapse and support symptom remission using much less VNS than previous protocols. These findings suggest that VNS parameters can be adjusted in order to minimize total charge delivery and maximize therapeutic effectiveness.
Collapse
Affiliation(s)
- Rimenez R Souza
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America; School of Behavioral Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America.
| | - Nicole M Robertson
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America
| | - Ezek Mathew
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America; School of Behavioral Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America
| | - Michel N Tabet
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America; School of Behavioral Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America
| | - Jesse E Bucksot
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America; Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America
| | - David T Pruitt
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America; School of Behavioral Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America; Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America
| | - Robert L Rennaker
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America; School of Behavioral Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America; Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America
| | - Seth A Hays
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America; Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America
| | - Christa K McIntyre
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America; School of Behavioral Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America
| | - Michael P Kilgard
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America; School of Behavioral Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, United States of America
| |
Collapse
|
26
|
Rosso P, Iannitelli A, Pacitti F, Quartini A, Fico E, Fiore M, Greco A, Ralli M, Tirassa P. Vagus nerve stimulation and Neurotrophins: a biological psychiatric perspective. Neurosci Biobehav Rev 2020; 113:338-353. [PMID: 32278791 DOI: 10.1016/j.neubiorev.2020.03.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 02/06/2023]
Abstract
Since 2004, vagus nerve stimulation (VNS) has been used in treatment-resistant or treatment-intolerant depressive episodes. Today, VNS is suggested as possible therapy for a larger spectrum of psychiatric disorders, including schizophrenia, obsessive compulsive disorders, and panic disorders. Despite a large body of literature supports the application of VNS in patients' treatment, the exact mechanism of action of VNS remains not fully understood. In the present study, the major knowledges on the brain areas and neuronal pathways regulating neuroimmune and autonomic response subserving VNS effects are reviewed. Furthermore, the involvement of the neurotrophins (NTs) Nerve Growth Factor (NGF) and Brain Derived Neurotrophic Factor (BDNF) in vagus nerve (VN) physiology and stimulation is revised. The data on brain NGF/BDNF synthesis and in turn on the activity-dependent plasticity, connectivity rearrangement and neurogenesis, are presented and discussed as potential biomarkers for optimizing stimulatory parameters for VNS. A vagus nerve-neurotrophin interaction model in the brain is finally proposed as a working hypothesis for future studies addressed to understand pathophysiology of psychiatric disturbance.
Collapse
Affiliation(s)
- Pamela Rosso
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy
| | - Angela Iannitelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Francesca Pacitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy; Psychiatry Unit San Salvatore Hospital, L'Aquila, Italy
| | - Adele Quartini
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Elena Fico
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy
| | - Marco Fiore
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy
| | - Antonio Greco
- Department of Sense Organs, Sapienza University of Rome, Italy
| | - Massimo Ralli
- Department of Sense Organs, Sapienza University of Rome, Italy
| | - Paola Tirassa
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy.
| |
Collapse
|
27
|
Rodenkirch C, Wang Q. Rapid and transient enhancement of thalamic information transmission induced by vagus nerve stimulation. J Neural Eng 2020; 17:026027. [PMID: 31935689 DOI: 10.1088/1741-2552/ab6b84] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Vagus nerve stimulation (VNS) has been FDA-approved as a long-term, therapeutic treatment for multiple disorders, including pharmacoresistant epilepsy and depression. Here we elucidate the short-term effects of VNS on sensory processing. APPROACH We employed an information theoretic approach to examine the effects of VNS on thalamocortical transmission of sensory-related information along the somatosensory pathway. MAIN RESULTS We found that VNS enhanced the selectivity of the response of thalamic neurons to specific kinetic features in the stimuli, resulting in a significant increase in the efficiency and rate of stimulus-related information conveyed by thalamic spikes. VNS-induced improvements in thalamic sensory processing coincided with a decrease in thalamic burst firing. Importantly, we found VNS-induced enhancement of sensory processing had a rapid onset and offset, completely disappearing one minute after cessation of VNS. The timescales of these effects indicate against an underlying mechanism involving long-term neuroplasticity. We found several patterns of VNS (tonic, standard duty-cycle, and fast duty-cycle) all induced similar improvements in sensory processing. Under closer inspection we noticed that due to the fast timescale of VNS effects on sensory processing, standard duty-cycle VNS induced a fluctuating sensory processing state which may be sub-optimal for perceptual behavior. Fast duty-cycle VNS and continuous, tonic VNS induced quantitatively similar improvements in thalamic information transmission as standard duty-cycle VNS without inducing a fluctuating thalamic state. Further, we found the strength of VNS-induced improvements in sensory processing increased monotonically with amplitude and frequency of VNS. SIGNIFICANCE These results demonstrate, for the first time, the feasibility of utilizing specific patterns of VNS to rapidly improve sensory processing and confirm fast duty-cycle and tonic patterns as optimal for this purpose, while showing standard duty-cycle VNS causes non-optimal fluctuations in thalamic state.
Collapse
Affiliation(s)
- Charles Rodenkirch
- Department of Biomedical Engineering, Columbia University, ET351, 500 W. 120th Street, New York, NY 10027, United States of America
| | | |
Collapse
|
28
|
Vagus nerve stimulation as a promising adjunctive treatment for ischemic stroke. Neurochem Int 2019; 131:104539. [DOI: 10.1016/j.neuint.2019.104539] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/03/2019] [Accepted: 08/21/2019] [Indexed: 12/26/2022]
|
29
|
Cruz-Pereira JS, Rea K, Nolan YM, O'Leary OF, Dinan TG, Cryan JF. Depression's Unholy Trinity: Dysregulated Stress, Immunity, and the Microbiome. Annu Rev Psychol 2019; 71:49-78. [PMID: 31567042 DOI: 10.1146/annurev-psych-122216-011613] [Citation(s) in RCA: 201] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Depression remains one of the most prevalent psychiatric disorders, with many patients not responding adequately to available treatments. Chronic or early-life stress is one of the key risk factors for depression. In addition, a growing body of data implicates chronic inflammation as a major player in depression pathogenesis. More recently, the gut microbiota has emerged as an important regulator of brain and behavior and also has been linked to depression. However, how this holy trinity of risk factors interact to maintain physiological homeostasis in the brain and body is not fully understood. In this review, we integrate the available data from animal and human studies on these three factors in the etiology and progression of depression. We also focus on the processes by which this microbiota-immune-stress matrix may influence centrally mediated events and on possible therapeutic interventions to correct imbalances in this triune.
Collapse
Affiliation(s)
- Joana S Cruz-Pereira
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland; , , , , , .,Department of Anatomy and Neuroscience, University College Cork, Cork T12 K8AF, Ireland
| | - Kieran Rea
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland; , , , , ,
| | - Yvonne M Nolan
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland; , , , , , .,Department of Anatomy and Neuroscience, University College Cork, Cork T12 K8AF, Ireland
| | - Olivia F O'Leary
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland; , , , , , .,Department of Anatomy and Neuroscience, University College Cork, Cork T12 K8AF, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland; , , , , , .,Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork T12 K8AF, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland; , , , , , .,Department of Anatomy and Neuroscience, University College Cork, Cork T12 K8AF, Ireland
| |
Collapse
|
30
|
The effect of transcutaneous vagus nerve stimulation on fear generalization and subsequent fear extinction. Neurobiol Learn Mem 2019; 161:192-201. [DOI: 10.1016/j.nlm.2019.04.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/06/2019] [Accepted: 04/11/2019] [Indexed: 12/27/2022]
|
31
|
Markert MS, Fisher RS. Neuromodulation - Science and Practice in Epilepsy: Vagus Nerve Stimulation, Thalamic Deep Brain Stimulation, and Responsive NeuroStimulation. Expert Rev Neurother 2018; 19:17-29. [DOI: 10.1080/14737175.2019.1554433] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Matthew S. Markert
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert S. Fisher
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
32
|
Cavarsan CF, Malheiros J, Hamani C, Najm I, Covolan L. Is Mossy Fiber Sprouting a Potential Therapeutic Target for Epilepsy? Front Neurol 2018; 9:1023. [PMID: 30555406 PMCID: PMC6284045 DOI: 10.3389/fneur.2018.01023] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 11/13/2018] [Indexed: 11/13/2022] Open
Abstract
Mesial temporal lobe epilepsy (MTLE) caused by hippocampal sclerosis is one of the most frequent focal epilepsies in adults. It is characterized by focal seizures that begin in the hippocampus, sometimes spread to the insulo-perisylvian regions and may progress to secondary generalized seizures. Morphological alterations in hippocampal sclerosis are well defined. Among them, hippocampal sclerosis is characterized by prominent cell loss in the hilus and CA1, and abnormal mossy fiber sprouting (granular cell axons) into the dentate gyrus inner molecular layer. In this review, we highlight the role of mossy fiber sprouting in seizure generation and hippocampal excitability and discuss the response of alternative treatment strategies in terms of MFS and spontaneous recurrent seizures in models of TLE (temporal lobe epilepsy).
Collapse
Affiliation(s)
- Clarissa F Cavarsan
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Jackeline Malheiros
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Clement Hamani
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil.,Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Imad Najm
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Luciene Covolan
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil.,Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
33
|
Huffman WJ, Subramaniyan S, Rodriguiz RM, Wetsel WC, Grill WM, Terrando N. Modulation of neuroinflammation and memory dysfunction using percutaneous vagus nerve stimulation in mice. Brain Stimul 2018; 12:19-29. [PMID: 30337243 DOI: 10.1016/j.brs.2018.10.005] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The vagus nerve is involved in regulating immunity and resolving inflammation. Current strategies aimed at modulating neuroinflammation and cognitive decline, in many cases, are limited and ineffective. OBJECTIVE We sought to develop a minimally invasive, targeted, vagus nerve stimulation approach (pVNS), and we tested its efficacy with respect to microglial activation and amelioration of cognitive dysfunction following lipopolysaccharide (LPS) endotoxemia in mice. METHODS We stimulated the cervical vagus nerve in mice using an ultrasound-guided needle electrode under sevoflurane anesthesia. The concentric bipolar needle electrode was percutaneously placed adjacent to the carotid sheath and stimulation was verified in real-time using bradycardia as a biomarker. Activation of vagal fibers was confirmed with immunostaining in relevant brainstem structures, including the dorsal motor nucleus and nucleus tractus solitarius. Efficacy of pVNS was evaluated following administration of LPS and analyses of changes in inflammation and behavior. RESULTS pVNS enabled stimulation of the vagus nerve as demonstrated by changes in bradycardia and histological evaluation of c-Fos and choline acetyltransferase expression in brainstem nuclei. Following LPS administration, pVNS significantly reduced plasma levels of tumor necrosis factor-α at 3 h post-injection. pVNS prevented LPS-induced hippocampal microglial activation as analyzed by changes in Iba-1 immunoreactivity, including cell body enlargement and shortened ramifications. Cognitive dysfunction following endotoxemia was also restored by pVNS. CONCLUSION Targeted cervical VNS using this novel percutaneous approach reduced LPS-induced systemic and brain inflammation and significantly improved cognitive responses. These results provide a novel therapeutic approach using bioelectronic medicine to modulate neuro-immune interactions that affect cognition.
Collapse
Affiliation(s)
- William J Huffman
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA; Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Saraswathi Subramaniyan
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Ramona M Rodriguiz
- Department of Psychiatry and Behavioral Sciences, Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical Center, Durham, NC, 27710, USA
| | - William C Wetsel
- Department of Psychiatry and Behavioral Sciences, Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical Center, Durham, NC, 27710, USA; Department of Neurobiology and Cell Biology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Warren M Grill
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA; Department of Electrical and Computer Engineering, Neurobiology, and Neurosurgery, Duke University, Durham, NC, 27708, USA
| | - Niccolò Terrando
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|
34
|
The Mechanism of Action of Vagus Nerve Stimulation in Treatment-Resistant Depression: Current Conceptualizations. Psychiatr Clin North Am 2018; 41:395-407. [PMID: 30098653 DOI: 10.1016/j.psc.2018.04.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Stimulation of the left cervical vagus nerve, or vagus nerve stimulation (VNS), brings about an antidepressant response in a subset of treatment-resistant depression (TRD) patients. How this occurs is poorly understood; however, knowledge of the neuroanatomic vagal pathways, in conjunction with functional brain imaging studies, suggests several brain regions associated with mood regulation are critical: brainstem nuclei (locus coeruleus, dorsal raphe, and ventral tegmental area), thalamus, and insular and prefrontal cortex. Furthermore, animal studies suggest that VNS enhances neuroplasticity and changes in neuronal firing patterns. Continued study to better understand the mechanism of action of VNS in TRD is warranted.
Collapse
|
35
|
Hachem LD, Wong SM, Ibrahim GM. The vagus afferent network: emerging role in translational connectomics. Neurosurg Focus 2018; 45:E2. [DOI: 10.3171/2018.6.focus18216] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Vagus nerve stimulation (VNS) is increasingly considered for the treatment of intractable epilepsy and holds potential for the management of a variety of neuropsychiatric conditions. The emergence of the field of connectomics and the introduction of large-scale modeling of neural networks has helped elucidate the underlying neurobiology of VNS, which may be variably expressed in patient populations and related to responsiveness to stimulation. In this report, the authors outline current data on the underlying neural circuitry believed to be implicated in VNS responsiveness in what the authors term the “vagus afferent network.” The emerging role of biomarkers to predict treatment effect is further discussed and important avenues for future work are highlighted.
Collapse
Affiliation(s)
- Laureen D. Hachem
- 1Division of Neurosurgery, Department of Surgery, University of Toronto
| | - Simeon M. Wong
- 2Department of Diagnostic Imaging, Hospital for Sick Children, Toronto; and
| | - George M. Ibrahim
- 1Division of Neurosurgery, Department of Surgery, University of Toronto
- 3Division of Neurosurgery, Hospital for Sick Children, Program in Neuroscience and Mental Health, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
36
|
Mercante B, Ginatempo F, Manca A, Melis F, Enrico P, Deriu F. Anatomo-Physiologic Basis for Auricular Stimulation. Med Acupunct 2018; 30:141-150. [PMID: 29937968 DOI: 10.1089/acu.2017.1254] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Introduction: Stimulation of cranial nerves modulates central nervous system (CNS) activity via the extensive connections of their brainstem nuclei to higher-order structures. Clinical experience with vagus-nerve stimulation (VNS) demonstrates that it produces robust therapeutic effects, however, posing concerns related to its invasiveness and side-effects. Discussion: Trigeminal nerve stimulation (TNS) has been recently proposed as a valid alternative to VNS. The ear presents afferent vagus and trigeminal-nerve distribution; its innervation is the theoretical basis of different reflex therapies, including auriculotherapy. An increasing number of studies have shown that several therapeutic effects induced by invasive VNS and TNS, can be reproduced by noninvasive auricular-nerve stimulation. However, the sites and neurobiologic mechanisms by which VNS and TNS produce their therapeutic effects are not clear yet. Conclusions: Accumulating evidence suggests that VNS and TNS share multiple levels and mechanisms of action in the CNS.
Collapse
Affiliation(s)
- Beniamina Mercante
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari Italy
| | - Francesca Ginatempo
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari Italy
| | - Andrea Manca
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari Italy
| | - Francesco Melis
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari Italy
| | - Paolo Enrico
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari Italy
| | - Franca Deriu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari Italy
| |
Collapse
|
37
|
Liu CM, Kanoski SE. Homeostatic and non-homeostatic controls of feeding behavior: Distinct vs. common neural systems. Physiol Behav 2018; 193:223-231. [PMID: 29421588 DOI: 10.1016/j.physbeh.2018.02.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/02/2018] [Accepted: 02/03/2018] [Indexed: 02/06/2023]
Abstract
Understanding the neurobiological controls of feeding behavior is critical in light of the growing obesity pandemic, a phenomenon largely based on excessive caloric consumption. Feeding behavior and its underlying biological substrates are frequently divided in the literature into two separate categories: [1] homeostatic processes involving energy intake based on caloric and other metabolic deficits, and [2] non-homeostatic processes that involve feeding driven by environmental and cognitive factors. The present review summarizes both historic and recent research examining the homeostatic regulation of feeding with specific emphasis on hypothalamic and hindbrain circuitry that monitor and regulate various metabolic signals. Regarding non-homeostatic controls, we highlight higher-order brain structures that integrate feeding-relevant external, interoceptive, and cognitive factors, including sensory cortical processing, learned associations in the hippocampus, and reward-based processing in the nucleus accumbens and interconnected mesolimbic circuitry. Finally, the current review focuses on recent evidence that challenges the traditional view that distinct neural systems regulate homeostatic vs. non-homeostatic controls of feeding behavior. Specifically, we highlight several feeding-related endocrine systems that act on both lower- and higher-order substrates, present evidence for the modulation of learned and cognitive feeding-relevant behaviors by lower-order brain regions, and highlight data showing that apparent homeostatic-based feeding behavior is modulated by higher-order brain regions. Our concluding perspective is that the classic dissociation between homeostatic and non-homeostatic constructs in relation to feeding behavior is limited with regards to understanding the complex integrated neurobiological systems that control energy balance.
Collapse
Affiliation(s)
- Clarissa M Liu
- University of Southern California, Neuroscience Graduate Program, Los Angeles, CA, United States; University of Southern California, Department of Biological Sciences, Human and Evolutionary Biology Section, Los Angeles, CA, United States
| | - Scott E Kanoski
- University of Southern California, Neuroscience Graduate Program, Los Angeles, CA, United States; University of Southern California, Department of Biological Sciences, Human and Evolutionary Biology Section, Los Angeles, CA, United States.
| |
Collapse
|
38
|
Mercante B, Deriu F, Rangon CM. Auricular Neuromodulation: The Emerging Concept beyond the Stimulation of Vagus and Trigeminal Nerves. MEDICINES (BASEL, SWITZERLAND) 2018; 5:10. [PMID: 29361732 PMCID: PMC5874575 DOI: 10.3390/medicines5010010] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/12/2018] [Accepted: 01/19/2018] [Indexed: 12/16/2022]
Abstract
Neuromodulation, thanks to intrinsic and extrinsic brain feedback loops, seems to be the best way to exploit brain plasticity for therapeutic purposes. In the past years, there has been tremendous advances in the field of non-pharmacological modulation of brain activity. This review of different neurostimulation techniques will focus on sites and mechanisms of both transcutaneous vagus and trigeminal nerve stimulation. These methods are scientifically validated non-invasive bottom-up brain modulation techniques, easily implemented from the outer ear. In the light of this, auricles could transpire to be the most affordable target for non-invasive manipulation of central nervous system functions.
Collapse
Affiliation(s)
- Beniamina Mercante
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy.
| | - Franca Deriu
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy.
| | - Claire-Marie Rangon
- Head of Scientific Auriculotherapy Diploma, Faculty of Medicine, University of Paris-Saclay, Saclay 94270, France.
| |
Collapse
|
39
|
De Cicco V, Tramonti Fantozzi MP, Cataldo E, Barresi M, Bruschini L, Faraguna U, Manzoni D. Trigeminal, Visceral and Vestibular Inputs May Improve Cognitive Functions by Acting through the Locus Coeruleus and the Ascending Reticular Activating System: A New Hypothesis. Front Neuroanat 2018; 11:130. [PMID: 29358907 PMCID: PMC5766640 DOI: 10.3389/fnana.2017.00130] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 12/15/2017] [Indexed: 12/25/2022] Open
Abstract
It is known that sensory signals sustain the background discharge of the ascending reticular activating system (ARAS) which includes the noradrenergic locus coeruleus (LC) neurons and controls the level of attention and alertness. Moreover, LC neurons influence brain metabolic activity, gene expression and brain inflammatory processes. As a consequence of the sensory control of ARAS/LC, stimulation of a sensory channel may potential influence neuronal activity and trophic state all over the brain, supporting cognitive functions and exerting a neuroprotective action. On the other hand, an imbalance of the same input on the two sides may lead to an asymmetric hemispheric excitability, leading to an impairment in cognitive functions. Among the inputs that may drive LC neurons and ARAS, those arising from the trigeminal region, from visceral organs and, possibly, from the vestibular system seem to be particularly relevant in regulating their activity. The trigeminal, visceral and vestibular control of ARAS/LC activity may explain why these input signals: (1) affect sensorimotor and cognitive functions which are not directly related to their specific informational content; and (2) are effective in relieving the symptoms of some brain pathologies, thus prompting peripheral activation of these input systems as a complementary approach for the treatment of cognitive impairments and neurodegenerative disorders.
Collapse
Affiliation(s)
- Vincenzo De Cicco
- Laboratory of Sensorimotor Integration, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Maria P Tramonti Fantozzi
- Laboratory of Sensorimotor Integration, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | | | - Massimo Barresi
- Institut des Maladie Neurodégénératives, University of Bordeaux, Bordeaux, France
| | - Luca Bruschini
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Ugo Faraguna
- Laboratory of Sensorimotor Integration, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy.,Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Pisa, Italy
| | - Diego Manzoni
- Laboratory of Sensorimotor Integration, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| |
Collapse
|
40
|
Breit S, Kupferberg A, Rogler G, Hasler G. Vagus Nerve as Modulator of the Brain-Gut Axis in Psychiatric and Inflammatory Disorders. Front Psychiatry 2018; 9:44. [PMID: 29593576 PMCID: PMC5859128 DOI: 10.3389/fpsyt.2018.00044] [Citation(s) in RCA: 585] [Impact Index Per Article: 83.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 02/01/2018] [Indexed: 12/13/2022] Open
Abstract
The vagus nerve represents the main component of the parasympathetic nervous system, which oversees a vast array of crucial bodily functions, including control of mood, immune response, digestion, and heart rate. It establishes one of the connections between the brain and the gastrointestinal tract and sends information about the state of the inner organs to the brain via afferent fibers. In this review article, we discuss various functions of the vagus nerve which make it an attractive target in treating psychiatric and gastrointestinal disorders. There is preliminary evidence that vagus nerve stimulation is a promising add-on treatment for treatment-refractory depression, posttraumatic stress disorder, and inflammatory bowel disease. Treatments that target the vagus nerve increase the vagal tone and inhibit cytokine production. Both are important mechanism of resiliency. The stimulation of vagal afferent fibers in the gut influences monoaminergic brain systems in the brain stem that play crucial roles in major psychiatric conditions, such as mood and anxiety disorders. In line, there is preliminary evidence for gut bacteria to have beneficial effect on mood and anxiety, partly by affecting the activity of the vagus nerve. Since, the vagal tone is correlated with capacity to regulate stress responses and can be influenced by breathing, its increase through meditation and yoga likely contribute to resilience and the mitigation of mood and anxiety symptoms.
Collapse
Affiliation(s)
- Sigrid Breit
- Division of Molecular Psychiatry, Translational Research Center, University Hospital of Psychiatry, University of Bern, Bern, Switzerland
| | - Aleksandra Kupferberg
- Division of Molecular Psychiatry, Translational Research Center, University Hospital of Psychiatry, University of Bern, Bern, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Gregor Hasler
- Division of Molecular Psychiatry, Translational Research Center, University Hospital of Psychiatry, University of Bern, Bern, Switzerland
| |
Collapse
|
41
|
|
42
|
Boku S, Nakagawa S, Toda H, Hishimoto A. Neural basis of major depressive disorder: Beyond monoamine hypothesis. Psychiatry Clin Neurosci 2018; 72:3-12. [PMID: 28926161 DOI: 10.1111/pcn.12604] [Citation(s) in RCA: 239] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2017] [Indexed: 12/14/2022]
Abstract
The monoamine hypothesis has been accepted as the most common hypothesis of major depressive disorder (MDD) for a long period because of its simplicity and understandability. Actually, most currently used antidepressants have been considered to act based on the monoamine hypothesis. However, an important problem of the monoamine hypothesis has been pointed out as follows: it fails to explain the latency of response to antidepressants. In addition, many patients with MDD have remained refractory to currently used antidepressants. Therefore, monoamine-alternate hypotheses are required to explain the latency of response to antidepressants. Such hypotheses have been expected to contribute to identifying hopeful new therapeutic targets for MDD. Past studies have revealed that the volume of the hippocampus is decreased in patients with MDD, which is likely caused by the failure of the hypothalamic-pituitary-adrenal axis and following elevation of glucocorticoids. Two hypotheses have been proposed to explain the volume of the hippocampus: (i) the neuroplasticity hypothesis; and (ii) the neurogenesis hypothesis. The neuroplasticity hypothesis explains how the hippocampal volume is decreased by the morphological changes of hippocampal neurons, such as the shortening length of dendrites and the decreased number and density of spines. The neurogenesis hypothesis explains how the hippocampal volume is decreased by the decrease of neurogenesis in the hippocampal dentate gyrus. These hypotheses are able to explain the latency of response to antidepressants. In this review, we first overview how the neuroplasticity and neurogenesis hypotheses have been developed. We then describe the details of these hypotheses.
Collapse
Affiliation(s)
- Shuken Boku
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shin Nakagawa
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hiroyuki Toda
- Department of Psychiatry, National Defense Medical College, Tokorozawa, Japan
| | - Akitoyo Hishimoto
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
43
|
Loerwald KW, Borland MS, Rennaker RL, Hays SA, Kilgard MP. The interaction of pulse width and current intensity on the extent of cortical plasticity evoked by vagus nerve stimulation. Brain Stimul 2017; 11:271-277. [PMID: 29174302 DOI: 10.1016/j.brs.2017.11.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 11/10/2017] [Accepted: 11/11/2017] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Repeatedly pairing a tone with a brief burst of vagus nerve stimulation (VNS) results in a reorganization of primary auditory cortex (A1). The plasticity-enhancing and memory-enhancing effects of VNS follow an inverted-U response to stimulation intensity, in which moderate intensity currents yield greater effects than low or high intensity currents. It is not known how other stimulation parameters effect the plasticity-enhancing effects of VNS. OBJECTIVE We sought to investigate the effect of pulse-width and intensity on VNS efficacy. Here, we used the extent of plasticity induced by VNS-tone pairing to assess VNS efficacy. METHODS Rats were exposed to a 9 kHz tone paired to VNS with varying current intensities and pulse widths. Cortical plasticity was measured as changes in the percent of area of primary auditory cortex responding to a range of sounds in VNS-treated rats relative to naïve rats. RESULTS We find that a combination of low current intensity (200 μA) and short pulse duration (100 μs) is insufficient to drive cortical plasticity. Increasing the pulse duration to 500 μs results in a reorganization of receptive fields in A1 auditory cortex. The extent of plasticity engaged under these conditions is less than that driven by conditions previously reported to drive robust plasticity (800 μA with 100 μs wide pulses). CONCLUSION These results suggest that the plasticity-enhancing and memory-enhancing effects of VNS follow an inverted-U response of stimulation current that is influenced by pulse width. Furthermore, shorter pulse widths may offer a clinical advantage when determining optimal stimulation current. These findings may facilitate determination of optimal VNS parameters for clinical application.
Collapse
Affiliation(s)
| | - Michael S Borland
- Texas Biomedical Device Center, Richardson, TX 75080, United States; The University of Texas at Dallas, School of Behavioral Brain Sciences, 800 West Campbell Road, GR 41, Richardson, TX 75080-3021, United States
| | - Robert L Rennaker
- Texas Biomedical Device Center, Richardson, TX 75080, United States; The University of Texas at Dallas, School of Behavioral Brain Sciences, 800 West Campbell Road, GR 41, Richardson, TX 75080-3021, United States; The University of Texas at Dallas, Erik Jonsson School of Engineering and Computer Science, BSB 11, Richardson, TX 75080, United States
| | - Seth A Hays
- Texas Biomedical Device Center, Richardson, TX 75080, United States; The University of Texas at Dallas, Erik Jonsson School of Engineering and Computer Science, BSB 11, Richardson, TX 75080, United States
| | - Michael P Kilgard
- Texas Biomedical Device Center, Richardson, TX 75080, United States; The University of Texas at Dallas, School of Behavioral Brain Sciences, 800 West Campbell Road, GR 41, Richardson, TX 75080-3021, United States
| |
Collapse
|
44
|
Abstract
BACKGROUND Recently, the US FDA has approved "vagal blocking therapy or vBLoc® therapy" as a new treatment for obesity. The aim of the present study was to study the mechanism-of-action of "VBLOC" in rat models. METHODS Rats were implanted with VBLOC, an intra-abdominal electrical device with leads placed around gastric vagal trunks through an abdominal incision and controlled by wireless device. Body weight, food intake, hunger/satiety, and metabolic parameters were monitored by a comprehensive laboratory animal monitoring system. Brain-gut responses were analyzed physiologically. RESULTS VBLOC reduced body weight and food intake, which was associated with increased satiety but not with decreased hunger. Brain activities in response to VBLOC included increased gene expression of leptin and CCKb receptors, interleukin-1β, tumor necrosis factor, and transforming growth factor β1 in the brainstem; increased CCK, somatostatin, and tyrosine hydroxylase in the hippocampus; increased NPY, AgRP, and Foxa2 in the hypothalamus; and reduced CCKb receptor, melanocortin 4 receptor, and insulin receptor in the hypothalamus. Plasma concentrations of CCK, gastrin, glucagon, GLP-1, and PYY and gastric acid secretion were unchanged in response to VBLOC. CONCLUSIONS Based on the present study, we may suggest that VBLOC induces satiety through vagal signaling, leading to reduced food intake and loss of body weight.
Collapse
|
45
|
Yasuhara T, Date I, Liska MG, Kaneko Y, Vale FL. Translating regenerative medicine techniques for the treatment of epilepsy. Brain Circ 2017; 3:156-162. [PMID: 30276318 PMCID: PMC6057691 DOI: 10.4103/bc.bc_21_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 09/05/2017] [Accepted: 09/11/2017] [Indexed: 01/03/2023] Open
Abstract
Epilepsy is considered a chronic neurological disorder and is accompanied by persistent and diverse disturbances in electrical brain activity. While antiepileptic pharmaceuticals are still the predominant treatment for epilepsy, the advent of numerous surgical interventions has further improved outcomes for patients. Despite these advancements, a subpopulation continues to experience intractable seizures which are resistant to current conventional and nonconventional therapeutic options. In this review, we begin with an introduction to the clinical presentation of epilepsy before discussing the clinically relevant laboratory models of epilepsy. Finally, we explore the implications of regenerative medicine – including cell therapy, neuroprotective agents, and electrical stimulation – for epilepsy, supplemented with our laboratory's data. This paper is a review article. Referred literature in this paper has been listed in the references section. The datasets supporting the conclusions of this article are available online by searching various databases, including PubMed. Some original points in this article come from the laboratory practice in our research center and the authors’ experiences.
Collapse
Affiliation(s)
- Takao Yasuhara
- Department of Neurological Surgery, Okayama University, Graduate School of Medicine, Okayama, Japan
| | - Isao Date
- Department of Neurological Surgery, Okayama University, Graduate School of Medicine, Okayama, Japan
| | - M Grant Liska
- Department of Neurosurgery and Brain Repair, USF Morsani College of Medicine, Tampa, FL 33612, USA
| | - Yuji Kaneko
- Department of Neurosurgery and Brain Repair, USF Morsani College of Medicine, Tampa, FL 33612, USA
| | - Fernando L Vale
- Department of Neurosurgery and Brain Repair, USF Morsani College of Medicine, Tampa, FL 33612, USA
| |
Collapse
|
46
|
Mercante B, Enrico P, Floris G, Quartu M, Boi M, Serra MP, Follesa P, Deriu F. Trigeminal nerve stimulation induces Fos immunoreactivity in selected brain regions, increases hippocampal cell proliferation and reduces seizure severity in rats. Neuroscience 2017; 361:69-80. [DOI: 10.1016/j.neuroscience.2017.08.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/06/2017] [Accepted: 08/03/2017] [Indexed: 12/11/2022]
|
47
|
Burger A, Verkuil B, Fenlon H, Thijs L, Cools L, Miller H, Vervliet B, Van Diest I. Mixed evidence for the potential of non-invasive transcutaneous vagal nerve stimulation to improve the extinction and retention of fear. Behav Res Ther 2017; 97:64-74. [DOI: 10.1016/j.brat.2017.07.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 07/07/2017] [Accepted: 07/07/2017] [Indexed: 12/22/2022]
|
48
|
Abstract
BACKGROUND Changes in hippocampal gray matter volumes are proposed to be involved in pathogenesis, course, and treatment response of major depressive disorder. Converging evidence suggests that reduced neurogenesis may occur in treatment-resistant depression (TRD). Vagus nerve stimulation (VNS) is a well-defined, long-term brain stimulation treatment for TRD. However, its in vivo positive effect on hippocampal modulation as mechanism of action has never been investigated before in clinical studies. In this study, we intended to explore hippocampal volumetric changes and clinical antidepressant responses in patients with TRD after 6 and 12 months of treatment with VNS. METHODS The TRD outpatients were evaluated for VNS implantation. Right and left hippocampal volumes in 6 TRD patients, who met the criteria for VNS treatment, were measured at baseline before the implantation and after 6 and 12 months. The patients were assessed using Beck Depression Inventory and Hamilton Depression Rating Scale at baseline and at follow-up visits. RESULTS There was a statistically significant and progressive increase in right and left hippocampal volumes during the follow up (P < 0.05). Furthermore, patients showed a significant improvement on Hamilton Depression Rating Scale and Beck Depression Inventory scores (P < 0.05). CONCLUSIONS Our data suggest a VNS modulatory effect on hippocampal plasticity as measured by hippocampal gray volume increase in TRD patients. These preliminary findings indicate the fundamental role of hippocampal remodeling as a marker of response to VNS in TRD.
Collapse
|
49
|
Abstract
Major depressive disorder (MDD) is prevalent. Although standards antidepressants are more effective than placebo, up to 35% of patients do not respond to 4 or more conventional treatments and are considered to have treatment-resistant depression (TRD). Considerable effort has been devoted to trying to find effective treatments for TRD. This review focuses on vagus nerve stimulation (VNS), approved for TRD in 2005 by the Food and Drugs Administration. Stimulation is carried by bipolar electrodes on the left cervical vagus nerve, which are attached to an implanted stimulator generator. The vagus bundle contains about 80% of afferent fibers terminating in the medulla, from which there are projections to many areas of brain, including the limbic forebrain. Various types of brain imaging studies reveal widespread functional effects in brain after either acute or chronic VNS. Although more randomized control trials of VNS need to be carried out before a definitive conclusion can be reached about its efficacy, the results of open studies, carried out over period of 1 to 2 years, show much more efficacy when compared with results from treatment as usual studies. There is an increase in clinical response to VNS between 3 and 12 months, which is quite different from that seen with standard antidepressant treatment of MDD. Preclinically, VNS affects many of the same brain areas, neurotransmitters (serotonin, norepinephrine) and signal transduction mechanisms (brain-derived neurotrophic factor-tropomyosin receptor kinase B) as those found with traditional antidepressants. Nevertheless, the mechanisms by which VNS benefits patients nonresponsive to conventional antidepressants is unclear, with further research needed to clarify this.
Collapse
Affiliation(s)
- Flavia R Carreno
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| | - Alan Frazer
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- South Texas Veterans Health Care System, San Antonio, TX, USA
| |
Collapse
|
50
|
Vagus Nerve Stimulation and Other Neuromodulation Methods for Treatment of Traumatic Brain Injury. Neurocrit Care 2017; 24:308-19. [PMID: 26399249 DOI: 10.1007/s12028-015-0203-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The objective of this paper is to review the current literature regarding the use of vagus nerve stimulation (VNS) in preclinical models of traumatic brain injury (TBI) as well as discuss the potential role of VNS along with alternative neuromodulation approaches in the treatment of human TBI. Data from previous studies have demonstrated VNS-mediated improvement following TBI in animal models. In these cases, VNS was observed to enhance motor and cognitive recovery, attenuate cerebral edema and inflammation, reduce blood brain barrier breakdown, and confer neuroprotective effects. Yet, the underlying mechanisms by which VNS enhances recovery following TBI remain to be fully elucidated. Several hypotheses have been offered including: a noradrenergic mechanism, reduction in post-TBI seizures and hyper-excitability, anti-inflammatory effects, attenuation of blood-brain barrier breakdown, and cerebral edema. We present other potential mechanisms by which VNS acts including enhancement of synaptic plasticity and recruitment of endogenous neural stem cells, stabilization of intracranial pressure, and interaction with the ghrelin system. In addition, alternative methods for the treatment of TBI including deep brain stimulation, transcranial magnetic stimulation, transcranial direct current stimulation, and focused ultrasound stimulation are discussed. Although the primary source data show that VNS improves TBI outcomes, it remains to be determined if these findings can be translated to clinical settings.
Collapse
|