1
|
Vande Perre L, Chávez Cerda J, Gochard S, Verstraeten M, Raffoul R, Leonard C, Delbeke J, El Tahry R, Gorza SP, Nonclercq A. Differences in conduction velocities of nerve fibers excited by infrared and electrical stimulation. J Neurosci Methods 2025; 418:110427. [PMID: 40101860 DOI: 10.1016/j.jneumeth.2025.110427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Infrared neural stimulation (INS) uses short optical pulses to activate nerves. While electrical stimulation (ES) activates large-diameter fibers first, light may preferentially activate small-diameter fibers first, which could be valuable for many clinical applications. NEW METHOD This study used a compact diode laser of 1470 nm to perform INS. Conduction velocity (CV) measurements were performed to assess differences in fiber type activation between INS and ES in the rat sciatic nerve and the goat vagus nerve. The rat sciatic nerve was chosen as a standard model because of its well-characterized physiology and extensive use in studies of INS mechanisms. The goat vagus nerve was chosen because of its expected high proportion of small-diameter fibers and its larger size, which allows sufficient separation between recording units to optimize CNAP measurements. RESULTS The results showed that in the rat sciatic nerve, ES-excited fibers had significantly higher CVs (9.81 ± 3.18 m/s) than INS-excited fibers (8.10 ± 2.82 m/s). In the goat vagus nerve, ES produced a mean CV of 6.47 ± 1.25 m/s, but INS did not produce clearly distinguishable compound nerve action potential, highlighting the challenges of applying INS to larger nerves. COMPARISON TO EXISTING METHODS To the best of our knowledge, CV is, for the first time, measured to identify the type of nerve fiber excited by INS. CONCLUSION These results suggest that INS may preferentially activate smaller diameter fibers, providing insight for potential neuromodulation applications.
Collapse
Affiliation(s)
- Louis Vande Perre
- Bio-, Electro, and Mechanical, Systems (BEAMS), Université libre de Bruxelles, Brussels, Belgium; Service Opera-Photonique, Université libre de Bruxelles, Brussels, Belgium.
| | - Javier Chávez Cerda
- Bio-, Electro, and Mechanical, Systems (BEAMS), Université libre de Bruxelles, Brussels, Belgium
| | | | - Maxime Verstraeten
- Bio-, Electro, and Mechanical, Systems (BEAMS), Université libre de Bruxelles, Brussels, Belgium
| | - Romain Raffoul
- Bio-, Electro, and Mechanical, Systems (BEAMS), Université libre de Bruxelles, Brussels, Belgium
| | | | - Jean Delbeke
- Institute of Neurosciences (IoNS), Université Catholique de Louvain, Brussels, Belgium -Department of Neurology, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Riëm El Tahry
- Institute of Neurosciences (IoNS), Université Catholique de Louvain, Brussels, Belgium -Department of Neurology, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Simon-Pierre Gorza
- Service Opera-Photonique, Université libre de Bruxelles, Brussels, Belgium
| | - Antoine Nonclercq
- Bio-, Electro, and Mechanical, Systems (BEAMS), Université libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
2
|
Schuman-Olivier Z, Marin F, Kinder LD, Datko M, Round K, Tohyama S, Garcia RG, Hirschtick RL, Edwards RR, Wells RE, Cheng HT, Barbieri R, Hadjikhani N, Loggia ML, Kaptchuk TJ, Houle TT, Rosen BR, Napadow V. Evaluating brain mechanisms of combined vagus nerve stimulation and mindfulness training for migraine: A randomized 2 × 2 factorial clinical trial protocol. Contemp Clin Trials 2025; 154:107947. [PMID: 40409681 DOI: 10.1016/j.cct.2025.107947] [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: 10/15/2024] [Revised: 04/16/2025] [Accepted: 05/10/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND Migraine is a major cause of disability and efficacious interventions are needed. In this mechanistic study, we investigate the combined, and potentially synergistic, effects of a multimodal intervention combining Mindfulness-Based Stress Reduction (MBSR) and transcutaneous auricular Vagal Nerve Stimulation (taVNS) for migraine. METHODS We utilize a modified double-blinded, placebo-controlled, 2 × 2 factorial randomized longitudinal design to assess the effects of an 8-week MBSR intervention with concomitant Respiratory-gated Auricular Vagal Afferent Nerve Stimulation (RAVANS) taVNS on migraine pathophysiology primary outcomes. We will enroll 150 patients with migraine (4-20 headache days/month). After a run-in month of daily diaries, we expect to randomly assign N = 96 participants to one of four treatment groups: (1) MBSR+RAVANS taVNS, (2) MBSR+Sham taVNS, (3) Nature Education Control (NEC) + RAVANS taVNS, or (4) NEC + Sham taVNS. Before and after intervention, participants have three in-person assessments (a 7 T MRI scan, an autonomic/sensory testing (AST) visit, and a 3 T PET-MRI scan). The primary outcomes for this study assess (1) central sensitization (brainstem/cortical response to trigeminal sensory afference), (2) autonomic dysfunction (High Frequency-Heart Rate Variability (HF-HRV) response to stressors), and (3) neuroinflammation (PET[11C]PBR28 signal). RESULTS Funded by NIH (P01AT009965), registered (NCT03592329). Final longitudinal outcomes will be collected by May 2025. CONCLUSION This mechanistic study is designed to investigate both independent and synergistic neurobiological effects of MBSR and RAVANS taVNS interventions on three distinct pathophysiological mechanisms of migraine. This research will elucidate the mechanistic and potentially synergistic effects of behavioral interventions (e.g., mindfulness) and device-based treatments (e.g., taVNS) for migraine.
Collapse
Affiliation(s)
- Zev Schuman-Olivier
- Center for Mindfulness and Compassion, Department of Psychiatry, Cambridge Health Alliance, Harvard Medical School, Cambridge, MA, USA.
| | - Frances Marin
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA; Center for Mindfulness and Compassion, Department of Psychiatry, Cambridge Health Alliance, Harvard Medical School, Cambridge, MA, USA
| | - Lillian D Kinder
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA; Center for Mindfulness and Compassion, Department of Psychiatry, Cambridge Health Alliance, Harvard Medical School, Cambridge, MA, USA
| | - Michael Datko
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA; Center for Mindfulness and Compassion, Department of Psychiatry, Cambridge Health Alliance, Harvard Medical School, Cambridge, MA, USA
| | - Kassandra Round
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA; Center for Mindfulness and Compassion, Department of Psychiatry, Cambridge Health Alliance, Harvard Medical School, Cambridge, MA, USA
| | - Sarasa Tohyama
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA
| | - Ronald G Garcia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; School of Medicine, Universidad de Santander, Bucaramanga, Colombia
| | - Randy L Hirschtick
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert R Edwards
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rebecca Erwin Wells
- Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Hsinlin T Cheng
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Riccardo Barbieri
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nouchine Hadjikhani
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco L Loggia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Anesthesiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ted J Kaptchuk
- Department of Medicine, Beth Israel Deaconess Hospital, Harvard Medical School, Boston, MA, USA
| | - Timothy T Houle
- Department of Anesthesiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bruce R Rosen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vitaly Napadow
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA; Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
3
|
Jiang X, Yang W, Liu G, Tang H, Zhang R, Zhang L, Li C, Li S. VNS facilitates the neurological function recovery after ischemia/reperfusion injury by regulating the A1/A2 polarization of astrocytes through the NMU-NMUR2 pathway. Neurochem Int 2025; 183:105918. [PMID: 39681141 DOI: 10.1016/j.neuint.2024.105918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/12/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
Stroke is the second leading cause of death worldwide. Although conventional treatments such as thrombolysis and mechanical thrombectomy are effective, their narrow therapeutic window limits long-term neurological recovery. Previous studies have shown that vagus nerve stimulation (VNS) enhances neurological recovery after ischemia/reperfusion (I/R) injury, and neuromedin U (NMU) has neuroprotective effects. This study used a mouse model of cerebral I/R injury to investigate the potential mechanisms of NMU in VNS-mediated neurological improvement. The study consisted of two parts: first, assessing the dynamic expression of NMU and NMUR2, which peaked on day 14 post-I/R. NMUR2 was primarily localized in astrocytes, suggesting that the NMU-NMUR2 signaling pathway plays an important role in astrocyte regulation. Next, interventions with VNS, NMU, and R-PSOP + VNS were conducted to evaluate the role of this pathway in VNS-mediated recovery. The results showed that VNS significantly upregulated NMU and NMUR2 expression, which was blocked by the NMUR2 antagonist R-PSOP. VNS and NMU treatment increased the proportion of A2 astrocytes, reduced A1 astrocytes, and enhanced the expression of VEGF and BDNF, all of which were also blocked by R-PSOP. These findings indicate that the "VNS-NMU-NMUR2-astrocyte A1/A2 polarization-VEGF/BDNF pathway" plays a crucial role in promoting neurovascular remodeling, axonal and dendritic regeneration, and synaptic plasticity, thereby contributing to functional recovery.
Collapse
Affiliation(s)
- Xia Jiang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China.
| | - Wendi Yang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Gang Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Hao Tang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Renzi Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Lina Zhang
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China.
| | - Changqing Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Sheng Li
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
4
|
Tavares-Figueiredo I, Pers YM, Duflos C, Herman F, Sztajnzalc B, Lecoq H, Laffont I, Dupeyron AF, Homs AF. Effect of Transcutaneous Auricular Vagus Nerve Stimulation in Chronic Low Back Pain: A Pilot Study. J Clin Med 2024; 13:7601. [PMID: 39768526 PMCID: PMC11677670 DOI: 10.3390/jcm13247601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 11/28/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Chronic low back pain (CLBP) is a common condition with limited long-term treatment options. Vagus nerve stimulation (VNS) has shown potential for pain improvement, but its use in CLBP remains underexplored. Our aim was to evaluate the efficacy, feasibility and tolerability of transcutaneous auricular vagus nerve stimulation (taVNS) in reducing pain and improving functional outcomes in CLBP patients. Methods: Thirty adults with CLBP (VAS ≥ 40/100) participated in this open-label pilot study (NCT05639270). Patients were treated with a taVNS device on the left ear for 30 min daily over a period of 3 months. The primary outcome was a reduction in pain intensity (VAS) at 1 month. Secondary outcomes included pain intensity at 3 months, disability (Oswestry Disability Index, ODI), quality of life (EQ-5D-5L), catastrophizing and psychological distress. In addition, compliance and adverse events were monitored. Results: After 1 month, 27 patients were evaluated. VAS scores decreased significantly by 16.1 (SD = 17.9) mm (p < 0.001) and by 22.5 (25) mm (p < 0.001) after 3 months (24 patients were analyzed). Functional disability improved with an average reduction in ODI of 11.9 (11.1) points (p < 0.001) after 3 months. Other patient-reported outcomes also improved significantly over the 3-month period. Overall, 51.9% of the patients achieved clinically meaningful pain reduction (≥20 mm), and no serious adverse events were reported. Treatment adherence was good, with half of the patients achieving 80% adherence. Conclusions: This pilot study suggests that taVNS is a feasible, safe and potentially effective treatment for CLBP that warrants further investigation in a randomized controlled trial compared to sham stimulation.
Collapse
Affiliation(s)
- Isabelle Tavares-Figueiredo
- Department of Physical Medicine and Rehabilitation, CHU Montpellier, University of Montpellier, 34295 Montpellier, France; (I.T.-F.); (B.S.); (H.L.); (I.L.)
- Centre d’Investigation Clinique, CHU Montpellier Montpellier, Inserm, CIC 1411, 34295 Montpellier, France
| | - Yves-Marie Pers
- IRMB, University of Montpellier, INSERM, 34295 Montpellier, France;
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Lapeyronie University Hospital, CHU Montpellier, 34295 Montpellier, France
| | - Claire Duflos
- Clinical Research and Epidemiology Unit, CHU Montpellier, University of Montpellier, 34295 Montpellier, France; (C.D.); (F.H.)
| | - Fanchon Herman
- Clinical Research and Epidemiology Unit, CHU Montpellier, University of Montpellier, 34295 Montpellier, France; (C.D.); (F.H.)
| | - Benjamin Sztajnzalc
- Department of Physical Medicine and Rehabilitation, CHU Montpellier, University of Montpellier, 34295 Montpellier, France; (I.T.-F.); (B.S.); (H.L.); (I.L.)
| | - Hugo Lecoq
- Department of Physical Medicine and Rehabilitation, CHU Montpellier, University of Montpellier, 34295 Montpellier, France; (I.T.-F.); (B.S.); (H.L.); (I.L.)
| | - Isabelle Laffont
- Department of Physical Medicine and Rehabilitation, CHU Montpellier, University of Montpellier, 34295 Montpellier, France; (I.T.-F.); (B.S.); (H.L.); (I.L.)
- EuroMov Digital Health in Motion, University of Montpellier, IMT Mines Ales, 34090 Montpellier, France;
| | - Arnaud F. Dupeyron
- EuroMov Digital Health in Motion, University of Montpellier, IMT Mines Ales, 34090 Montpellier, France;
- Department of Physical Medicine and Rehabilitation, CHU Nimes, University of Montpellier, 30900 Nimes, France
| | - Alexis F. Homs
- EuroMov Digital Health in Motion, University of Montpellier, IMT Mines Ales, 34090 Montpellier, France;
- Department of Physical Medicine and Rehabilitation, CHU Nimes, University of Montpellier, 30900 Nimes, France
| |
Collapse
|
5
|
Austelle CW, Cox SS, Wills KE, Badran BW. Vagus nerve stimulation (VNS): recent advances and future directions. Clin Auton Res 2024; 34:529-547. [PMID: 39363044 PMCID: PMC11543756 DOI: 10.1007/s10286-024-01065-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024]
Abstract
PURPOSE Vagus nerve stimulation (VNS) is emerging as a unique and potent intervention, particularly within neurology and psychiatry. The clinical value of VNS continues to grow, while the development of noninvasive options promises to change a landscape that is already quickly evolving. In this review, we highlight recent progress in the field and offer readers a glimpse of the future for this bright and promising modality. METHODS We compiled a narrative review of VNS literature using PubMed and organized the discussion by disease states with approved indications (epilepsy, depression, obesity, post-stroke motor rehabilitation, headache), followed by a section highlighting novel, exploratory areas of VNS research. In each section, we summarized the current role, recent advancements, and future directions of VNS in the treatment of each disease. RESULTS The field continues to gain appreciation for the clinical potential of this modality. VNS was initially developed for treatment-resistant epilepsy, with the first depression studies following shortly thereafter. Overall, VNS has gained approval or clearance in the treatment of medication-refractory epilepsy, treatment-resistant depression, obesity, migraine/cluster headache, and post-stroke motor rehabilitation. CONCLUSION Noninvasive VNS represents an opportunity to bridge the translational gap between preclinical and clinical paradigms and may offer the same therapeutic potential as invasive VNS. Further investigation into how VNS parameters modulate behavior and biology, as well as how to translate noninvasive options into the clinical arena, are crucial next steps for researchers and clinicians studying VNS.
Collapse
Affiliation(s)
- Christopher W Austelle
- Department of Psychiatry and Behavioral Sciences, Stanford University, 401 Quarry Road, Palo Alto, CA, 94305, USA.
- Veterans Affairs Palo Alto Healthcare System, and the Sierra Pacific Mental Illness, Research, Education, and Clinical Center (MIRECC), Palo Alto, CA, USA.
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA.
| | - Stewart S Cox
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Kristin E Wills
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Bashar W Badran
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
6
|
Zhang J, Simoes R, Guo T, Cao YQ. Neuroimmune interactions in the development and chronification of migraine headache. Trends Neurosci 2024; 47:819-833. [PMID: 39271369 DOI: 10.1016/j.tins.2024.08.009] [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: 05/07/2024] [Revised: 07/31/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024]
Abstract
Migraine is highly prevalent and debilitating. The persistent headaches in this condition are thought to arise from the activation and sensitization of the trigeminovascular pathway. Both clinical and animal model studies have suggested that neuroimmune interactions contribute to the pathophysiology of migraine headache. In this review, we first summarize the findings from human studies implicating the dysregulation of the immune system in migraine, including genetic analyses, measurement of circulatory factors, and neuroimaging data. We next discuss recent advances from rodent studies aimed at elucidating the neuroimmune interactions that manifest at various levels of the trigeminovascular pathway and lead to the recruitment of innate and adaptive immune cells as well as immunocompetent glial cells. These cells reciprocally modulate neuronal activity via multiple pro- and anti-inflammatory mediators, thereby regulating peripheral and central sensitization. Throughout the discussions, we highlight the potential clinical and translational implications of the findings.
Collapse
Affiliation(s)
- Jintao Zhang
- Department of Anesthesiology and Pain Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Roli Simoes
- Department of Anesthesiology and Pain Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Tingting Guo
- Department of Anesthesiology and Pain Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Yu-Qing Cao
- Department of Anesthesiology and Pain Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
7
|
Edalati S, Meyer JS, Aravot D, Barac YD. Vagal nerve stimulation potential therapeutic benefits in acute lung rejection and transplantation. Transpl Immunol 2024; 86:102105. [PMID: 39128810 DOI: 10.1016/j.trim.2024.102105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Allograft rejection, accompanied by a rise in proinflammatory cytokines, is a leading cause of morbidity and mortality after lung transplantation. Immunosuppressive treatments are routinely employed as an effective way to prevent rejection, however, there is still an unmet need to develop new strategies to reduce the damage caused to transplanted organs by innate inflammatory responses. Recent research has shown that activating the vagus nerve's efferent arm regulates cytokine production and improves survival in experimental conditions of cytokine excess, such as sepsis, hemorrhagic shock, ischemia-reperfusion injury, among others. The cholinergic anti-inflammatory pathway can provide a localized, fast, and discrete response to inflammation by controlling the neuroimmune response and preventing excessive inflammation. This review intends to assess and discuss, the influence of noninvasive vagal nerve stimulation for prophylactic measures and supporting treatment in patients undergoing organ transplantation rejection with a prominent T-cell mediated immune response as a means of attenuating inflammation and leukocyte infiltration of the graft vessels.
Collapse
Affiliation(s)
- Shaun Edalati
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - J Sam Meyer
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Biomedical Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel; The Division of Cardiovascular and Thoracic Surgery, Rabin Medical Center, Petach-Tikva, Israel
| | - Dan Aravot
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; The Division of Cardiovascular and Thoracic Surgery, Rabin Medical Center, Petach-Tikva, Israel
| | - Yaron D Barac
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; The Division of Cardiovascular and Thoracic Surgery, Rabin Medical Center, Petach-Tikva, Israel
| |
Collapse
|
8
|
Ye D, Vo L, Fairchild TJ, Drummond PD. Temple cooling increases parasympathetic activity and decreases pressure pain on the hand. Eur J Pain 2023; 27:353-365. [PMID: 36451612 DOI: 10.1002/ejp.2061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/26/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND Applying an ice cube to the temple (the conditioning stimulus) inhibits electrically evoked pain in the forearm. The present study aimed to determine whether temple cooling also inhibits pressure- and heat-pain test stimuli in the upper limb and, if so, to investigate the intra-session test-retest reliability of this response. Additional aims were to establish whether pain inhibition evoked by temple cooling was associated with parasympathetic activity; and to explore sex differences in response. METHODS The sample consisted of 40 healthy adults (24 females). Heart rate was recorded continuously throughout the session. An ice cube (3 × 4 cm contact area) was applied for 1 min to the temple on the dominant side. Before and immediately afterwards, the pressure pain threshold was measured from the dorsal hand and sensitivity to heat (individually adjusted at baseline to elicit moderate pain) was measured from the ventral forearm. The procedures were repeated 15 min later. RESULTS Temple cooling inhibited pressure pain on the hand but not heat pain on the forearm. However, test-retest reliability of pressure pain inhibition was poor. Heart rate decreased during temple cooling, consistent with a "diving" reflex. Males had stronger pressure pain inhibition, lower heart rate and higher overall autonomic activity than females. However, cardiac parasympathetic activation during temple cooling was comparable in both sexes and was unrelated to pain inhibition. CONCLUSIONS These findings indicate that temple cooling evokes pain inhibition that is stronger in males than in females. Cardiac parasympathetic activity does not appear to mediate this response. SIGNIFICANCE The conditioning stimulus in the conditioned pain modulation paradigm is often applied to the upper or lower limbs. This may confound pain-inhibitory effects in people with peripheral neuropathy who typically have enhanced or diminished sensation in the extremities. Applying an ice cube at the temple area induces pain-inhibitory effects on the upper limb after the ice is removed. Future research examining pain modulation in people with peripheral neuropathy may consider adopting temple cooling as the conditioning stimulus.
Collapse
Affiliation(s)
- Di Ye
- Discipline of Psychology and Healthy Ageing Research Centre, College of Science, Health, Engineering and Education, Murdoch University, Murdoch, Western Australia, Australia
| | - Lechi Vo
- Discipline of Psychology and Healthy Ageing Research Centre, College of Science, Health, Engineering and Education, Murdoch University, Murdoch, Western Australia, Australia
| | - Timothy J Fairchild
- The Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, Western Australia, Australia
| | - Peter D Drummond
- Discipline of Psychology and Healthy Ageing Research Centre, College of Science, Health, Engineering and Education, Murdoch University, Murdoch, Western Australia, Australia
| |
Collapse
|
9
|
Magara AE, Gallay MN, Moser D, Jeanmonod D. Complete resolution of chronic cluster headache following central lateral thalamotomy using incisionless MRI-guided focused ultrasound with 6 years of follow-up: illustrative case. JOURNAL OF NEUROSURGERY. CASE LESSONS 2022; 4:CASE22259. [PMID: 36443956 PMCID: PMC9705522 DOI: 10.3171/case22259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/29/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND The authors reported the case of a 66-year-old male patient with a 14-year history of right-sided severe episodic and therapy-resistant cluster headache (CH) who underwent bilateral central lateral thalamotomy (CLT) using incisionless transcranial magnetic resonance imaging-guided focused ultrasound (MRgFUS). OBSERVATIONS The patient experienced a single cluster headache attack 5 weeks after the procedure. There were no more pain attacks over the next 6 years of follow-up. LESSONS This treatment success may indicate a common pathophysiology for CH and neurogenic (neuropathic) pain, which has been treated with CLT for more than 30 years. Further experience is needed to assess the reproducibility of this case.
Collapse
|
10
|
Su PYP, Zhang L, He L, Zhao N, Guan Z. The Role of Neuro-Immune Interactions in Chronic Pain: Implications for Clinical Practice. J Pain Res 2022; 15:2223-2248. [PMID: 35957964 PMCID: PMC9359791 DOI: 10.2147/jpr.s246883] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
Chronic pain remains a public health problem and contributes to the ongoing opioid epidemic. Current pain management therapies still leave many patients with poorly controlled pain, thus new or improved treatments are desperately needed. One major challenge in pain research is the translation of preclinical findings into effective clinical practice. The local neuroimmune interface plays an important role in the initiation and maintenance of chronic pain and is therefore a promising target for novel therapeutic development. Neurons interface with immune and immunocompetent cells in many distinct microenvironments along the nociceptive circuitry. The local neuroimmune interface can modulate the activity and property of the neurons to affect peripheral and central sensitization. In this review, we highlight a specific subset of many neuroimmune interfaces. In the central nervous system, we examine the interface between neurons and microglia, astrocytes, and T lymphocytes. In the periphery, we profile the interface between neurons in the dorsal root ganglion with T lymphocytes, satellite glial cells, and macrophages. To bridge the gap between preclinical research and clinical practice, we review the preclinical studies of each neuroimmune interface, discuss current clinical treatments in pain medicine that may exert its action at the neuroimmune interface, and highlight opportunities for future clinical research efforts.
Collapse
Affiliation(s)
- Po-Yi Paul Su
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | - Lingyi Zhang
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
- Department of Anesthesiology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Liangliang He
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
- Department of Pain Management, Xuanwu Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Na Zhao
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
11
|
Epipharyngeal Abrasive Therapy (EAT) Has Potential as a Novel Method for Long COVID Treatment. Viruses 2022; 14:v14050907. [PMID: 35632649 PMCID: PMC9147901 DOI: 10.3390/v14050907] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 01/27/2023] Open
Abstract
COVID-19 often causes sequelae after initial recovery, referred to collectively as long COVID. Long COVID is considered to be caused by the persistence of chronic inflammation after acute COVID-19 infection. We found that all long COVID patients had residual inflammation in the epipharynx, an important site of coronavirus replication, and some long COVID symptoms are similar to those associated with chronic epipharyngitis. Epipharyngeal abrasive therapy (EAT) is a treatment for chronic epipharyngitis in Japan that involves applying zinc chloride as an anti-inflammatory agent to the epipharyngeal mucosa. In this study, we evaluated the efficacy of EAT for the treatment of long COVID. The subjects in this study were 58 patients with long COVID who were treated with EAT in the outpatient department once a week for one month (mean age = 38.4 ± 12.9 years). The intensities of fatigue, headache, and attention disorder, which are reported as frequent symptoms of long COVID, were assessed before and after EAT using the visual analog scale (VAS). EAT reduced inflammation in the epipharynx and significantly improved the intensity of fatigue, headache, and attention disorder, which may be related to myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). These results suggest that EAT has potential as a novel method for long COVID treatment.
Collapse
|
12
|
The trigeminal pathways. J Neurol 2022; 269:3443-3460. [DOI: 10.1007/s00415-022-11002-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/29/2022] [Accepted: 01/29/2022] [Indexed: 12/14/2022]
|
13
|
Hamdi H, Spatola G, Lagarde S, McGonigal A, Paz-Paredes A, Bizeau A, Bartolomei F, Carron R. Use of Polyvinyl Alcohol Sponge Cubes for Vagal Nerve Stimulation: A Suggestion for the Wrapping Step. Technical Note and Step-by-Step Operative Technique. Oper Neurosurg (Hagerstown) 2021; 18:487-495. [PMID: 31386756 PMCID: PMC7594176 DOI: 10.1093/ons/opz227] [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: 07/26/2018] [Accepted: 05/29/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Vagal nerve stimulation (VNS) is an approved treatment for epilepsy and depression. Wrapping the helical electrodes around the nerve can prove technically challenging. However, a quick and efficient method to slightly elevate the nerve can highly facilitate this part of the procedure. OBJECTIVE To provide useful surgical tips to facilitate the procedure. METHODS Based on experience of more than 150 adult cases for mainly epilepsy (primary lead implant), the authors share their surgical technique to provide the experienced surgeons or newcomers to the field of VNS with some useful tips. All patients signed informed consent according to the local ethics committee guidelines. RESULTS The article consists of a detailed step-by-step description of the whole procedure illustrated through high-resolution colored photographs of the surgical field. Special reference is made to the usefulness of polyvinyl alcohol (PVA) sponge cubes to elevate the nerve instead of the commonly used silicon vessel loops. CONCLUSION The use of surgical microscope and PVA sponge cubes to elevate the nerve constitute key points to make VNS an easy surgery.
Collapse
Affiliation(s)
- Hussein Hamdi
- Department of Functional and Stereotactic Neurosurgery, Timone University Hospital, Marseille, France.,Aix Marseille Univ, APHM, INSERM, INS, Inst Neurosci Syst, Timone Hospital, Marseille, France.,Functional Neurosurgery and Stereotaxy Unit, Neurological Surgery Department, Tanta University, Egypt
| | - Giorgio Spatola
- Department of Functional and Stereotactic Neurosurgery, Timone University Hospital, Marseille, France.,Aix Marseille Univ, APHM, INSERM, INS, Inst Neurosci Syst, Timone Hospital, Marseille, France
| | - Stanislas Lagarde
- Epileptology Department, Aix Marseille Univ, APHM, INSERM, INS, Inst Neurosci Syst, Timone Hospital, Marseille, France
| | - Aileen McGonigal
- Epileptology Department, Aix Marseille Univ, APHM, INSERM, INS, Inst Neurosci Syst, Timone Hospital, Marseille, France
| | - Armando Paz-Paredes
- Department of Functional and Stereotactic Neurosurgery, Timone University Hospital, Marseille, France
| | - Alain Bizeau
- Department of Cervico-Facial Surgery, Sainte Musse Hospital, Toulon, France
| | - Fabrice Bartolomei
- Epileptology Department, Aix Marseille Univ, APHM, INSERM, INS, Inst Neurosci Syst, Timone Hospital, Marseille, France
| | - Romain Carron
- Department of Functional and Stereotactic Neurosurgery, Timone University Hospital, Marseille, France.,Aix Marseille Univ, APHM, INSERM, INS, Inst Neurosci Syst, Timone Hospital, Marseille, France
| |
Collapse
|
14
|
Sharif ZI, Galand V, Hucker WJ, Singh JP. Evolving Cardiac Electrical Therapies for Advanced Heart Failure Patients. Circ Arrhythm Electrophysiol 2021; 14:e009668. [PMID: 33858178 DOI: 10.1161/circep.120.009668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Symptomatic heart failure (HF) patients despite optimal medical therapy and advances such as invasive hemodynamic monitoring remain challenging to manage. While cardiac resynchronization therapy remains a highly effective therapy for a subset of HF patients with wide QRS, a majority of symptomatic HF patients are poor candidates for such. Recently, cardiac contractility modulation, neuromodulation based on carotid baroreceptor stimulation, and phrenic nerve stimulation have been approved by the US Food and Drug Administration and are emerging as therapeutic options for symptomatic HF patients. This state-of-the-art review examines the role of these evolving electrical therapies in advanced HF.
Collapse
Affiliation(s)
- Zain I Sharif
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (Z.I.S., V.G., W.J.H., J.P.S.)
| | - Vincent Galand
- Division of Cardiology, Université de Rennes, CHU Rennes, INSERM, LTSI-UMR 1099, France (V.G.).,Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (Z.I.S., V.G., W.J.H., J.P.S.)
| | - William J Hucker
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (Z.I.S., V.G., W.J.H., J.P.S.)
| | - Jagmeet P Singh
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (Z.I.S., V.G., W.J.H., J.P.S.)
| |
Collapse
|
15
|
Grazzi L, Toppo C, D’Amico D, Leonardi M, Martelletti P, Raggi A, Guastafierro E. Non-Pharmacological Approaches to Headaches: Non-Invasive Neuromodulation, Nutraceuticals, and Behavioral Approaches. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:1503. [PMID: 33562487 PMCID: PMC7914516 DOI: 10.3390/ijerph18041503] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/27/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023]
Abstract
Significant side effects or drug interactions can make pharmacological management of headache disorders very difficult. Non-conventional and non-pharmacological treatments are becoming increasingly used to overcome these issues. In particular, non-invasive neuromodulation, nutraceuticals, and behavioral approaches are well tolerated and indicated for specific patient categories such as adolescents and pregnant women. This paper aims to present the main approaches reported in the literature in the management of headache disorders. We therefore reviewed the available literature published between 2010 and 2020 and performed a narrative presentation for each of the three categories (non-invasive neuromodulation, nutraceuticals, and behavioral therapies). Regarding non-invasive neuromodulation, we selected transcranial magnetic stimulation, supraorbital nerve stimulation, transcranial direct current stimulation, non-invasive vagal nerve stimulation, and caloric vestibular stimulation. For nutraceuticals, we selected Feverfew, Butterbur, Riboflavin, Magnesium, and Coenzyme Q10. Finally, for behavioral approaches, we selected biofeedback, cognitive behavioral therapy, relaxation techniques, mindfulness-based therapy, and acceptance and commitment therapy. These approaches are increasingly seen as a valid treatment option in headache management, especially for patients with medication overuse or contraindications to drug treatment. However, further investigations are needed to consider the effectiveness of these approaches also with respect to the long-term effects.
Collapse
Affiliation(s)
- Licia Grazzi
- UOC Neuroalgologia, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy;
| | - Claudia Toppo
- UOC Neurologia, Salute Pubblica e Disabilità, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (C.T.); (M.L.); (A.R.); (E.G.)
| | - Domenico D’Amico
- UOC Neuroalgologia, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy;
| | - Matilde Leonardi
- UOC Neurologia, Salute Pubblica e Disabilità, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (C.T.); (M.L.); (A.R.); (E.G.)
| | - Paolo Martelletti
- Department of Clinical and Molecular Medicine, Sapienza University, 00185 Rome, Italy;
| | - Alberto Raggi
- UOC Neurologia, Salute Pubblica e Disabilità, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (C.T.); (M.L.); (A.R.); (E.G.)
| | - Erika Guastafierro
- UOC Neurologia, Salute Pubblica e Disabilità, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (C.T.); (M.L.); (A.R.); (E.G.)
| |
Collapse
|
16
|
Courties A, Berenbaum F, Sellam J. Vagus nerve stimulation in musculoskeletal diseases. Joint Bone Spine 2021; 88:105149. [PMID: 33548494 DOI: 10.1016/j.jbspin.2021.105149] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/21/2021] [Indexed: 01/21/2023]
Abstract
The vagus nerve is the main nerve of the parasympathetic autonomic nervous system. Beyond its vegetative functions, the vagus nerve possesses anti-inflammatory and analgesic properties. Initially developed in the treatment of refractory epilepsy, vagus nerve stimulation (VNS) is currently being evaluated in several musculoskeletal diseases. VNS can be invasive by placing an electrode around the cervical vagus nerve and connected to a generator implanted subcutaneously or non-invasive stimulating the cervical vagus nerve branch percutaneously (auricular or cervical). In rheumatoid arthritis (RA) patients, VNS has been shown to dampen the inflammatory response of circulatory peripheral cells. Several open-labeled small pilot studies have demonstrated that VNS, either invasive or transcutaneous, is associated with a significant decrease of RA disease activity. As well, other studies have shown that VNS could limit fatigue in Sjogren's syndrome and systemic lupus, or decrease pain in fibromyalgia as well as in erosive hand osteoarthritis. However, some questions remain, such as the settings of stimulation, the duration of treatment, or the optimal stimulation route. Finally, randomized controlled trials versus sham stimulation with large samples of patients are mandatory to definitively conclude about the efficacy of VNS.
Collapse
Affiliation(s)
- Alice Courties
- INSERM UMRS 938, service de rhumatologie, Sorbonne Université, hôpital Saint-Antoine, AP-HP, Paris, France
| | - Francis Berenbaum
- INSERM UMRS 938, service de rhumatologie, Sorbonne Université, hôpital Saint-Antoine, AP-HP, Paris, France
| | - Jérémie Sellam
- INSERM UMRS 938, service de rhumatologie, Sorbonne Université, hôpital Saint-Antoine, AP-HP, Paris, France.
| |
Collapse
|
17
|
Noninvasive vagus nerve stimulation and morphine transiently inhibit trigeminal pain signaling in a chronic headache model. Pain Rep 2020; 5:e881. [PMID: 33364541 PMCID: PMC7752694 DOI: 10.1097/pr9.0000000000000881] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 12/02/2022] Open
Abstract
Noninvasive vagus nerve stimulation suppressed persistent trigeminal nociception in a chronic headache model similarly to morphine and may provide a safe, nonaddictive abortive therapy for chronic headache. Introduction: Chronic headache conditions are characterized by persistent sensitization of the trigeminal system, which involves dysfunction of descending pain modulation. We previously reported that noninvasive vagus nerve stimulation (nVNS) inhibits trigeminal nociception in models of episodic migraine through a mechanism involving enhanced serotonergic and GABAergic descending pain signaling. Objectives: The analgesic effectiveness of nVNS and morphine were investigated in an animal model of chronic headache mediated by the combination of the 3 migraine risk factors of neck muscle tension, paradoxical sleep deprivation, and pungent odors. Methods: Sprague–Dawley rats were injected with complete Freund's adjuvant in the trapezius and sleep deprived for 1 night to promote trigeminal sensitization. After 7 days, animals were exposed to a pungent odor, and mechanical nocifensive head withdrawal responses were determined using von Frey filaments. Beginning on day 3 after odor exposure, animals were treated daily with either nVNS or morphine for 7 days. Results: Exposure of animals sensitized by neck inflammation and sleep deprivation to a pungent odor resulted in a prolonged state of trigeminal nociception. Daily administration of nVNS or morphine significantly repressed the nocifensive response; however, cessation resulted in a return to heightened pretreatment nocifensive levels. Conclusions: The combination of reported migraine risk factors promotes a state of sustained trigeminal hypersensitivity characteristic of chronic headache. Daily nVNS was similarly effective as morphine in inhibiting nociception and may represent a safer, opioid-sparing therapeutic option for other chronic pain disorders involving sensitization of the trigeminal system by promoting descending pain modulation.
Collapse
|
18
|
Drewes AM, Brock C, Rasmussen SE, Møller HJ, Brock B, Deleuran BW, Farmer AD, Pfeiffer-Jensen M. Short-term transcutaneous non-invasive vagus nerve stimulation may reduce disease activity and pro-inflammatory cytokines in rheumatoid arthritis: results of a pilot study. Scand J Rheumatol 2020; 50:20-27. [PMID: 33047630 DOI: 10.1080/03009742.2020.1764617] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Objective: Rheumatoid arthritis (RA) is a chronic, autoimmune, inflammatory disease. Studies suggest that pro-inflammatory cytokines may be attenuated by the vagus nerve through the cholinergic anti-inflammatory pathway. We aimed to evaluate the anti-inflammatory effects of short-term transcutaneous non-invasive vagus nerve stimulation (n-VNS) applied to the cervical vagus nerve in patients with RA. Method: We conducted a single-centre, open-label, preliminary proof-of-concept study of n-VNS in two cohorts of participants with RA: one with high disease activity (n = 16) and one with low disease activity (n = 20). Disease Activity Score based on 28-joint count-C-reactive protein (DAS28-CRP), cardiac vagal tone, and pro-inflammatory cytokines were measured at baseline and after 1 and 4 days of n-VNS. Results: In the high disease activity group, n-VNS resulted in reductions in DAS28-CRP (4.1 to 3.8, p = 0.02), CRP (8.2 to 6 mg/mL, p = 0.01), and interferon-γ (29.8 to 22.5 pg/mL, p = 0.02). In the low disease activity group, there was no effect on DAS28-CRP, and n-VNS was associated with a decrease in cardiac vagal tone (p = 0.03) and a reduction in interleukin-10 (0.8 to 0.6 pg/mL, p = 0.02). Participants with high disease activity had lower baseline cardiac vagal tone than those with low disease activity (3.6 ± 2 vs 4.9 ± 3 linear vagal scale, p = 0.03). Cardiac vagal tone was negatively associated with DAS28-CRP (r = -0.37, p = 0.03). Overall, n-VNS was well tolerated. Conclusion: This study provides preliminary support for an anti-inflammatory effect of n-VNS in patients with RA. These findings warrant further investigation in larger placebo-controlled trials.
Collapse
Affiliation(s)
- A M Drewes
- Department of Rheumatology, Aarhus University Hospital , Aarhus, Denmark
| | - C Brock
- Mech-Sense, Department of Gastroenterology and Hepatology, Clinical Institute, Aalborg University Hospital , Aalborg, Denmark
| | - S E Rasmussen
- Department of Rheumatology, Aarhus University Hospital , Aarhus, Denmark
| | - H J Møller
- Department of Clinical Biochemistry, Aarhus University Hospital , Aarhus, Denmark
| | - B Brock
- Steno Diabetes Center Copenhagen , Gentofte, Denmark
| | - B W Deleuran
- Department of Rheumatology, Aarhus University Hospital , Aarhus, Denmark
| | - A D Farmer
- Mech-Sense, Department of Gastroenterology and Hepatology, Clinical Institute, Aalborg University Hospital , Aalborg, Denmark.,Centre for Trauma and Neuroscience, Blizard Institute, Wingate Institute of Neurogastroenterology, Barts and the London School of Medicine and Dentistry, Queen Mary University of London , London, UK.,Institute of Applied Clinical Sciences, University of Keele , Stoke on Trent, UK
| | - M Pfeiffer-Jensen
- Department of Rheumatology, Aarhus University Hospital , Aarhus, Denmark
| |
Collapse
|
19
|
Silberstein SD, Yuan H, Najib U, Ailani J, Morais ALD, Mathew PG, Liebler E, Tassorelli C, Diener HC. Non-invasive vagus nerve stimulation for primary headache: A clinical update. Cephalalgia 2020; 40:1370-1384. [PMID: 32718243 DOI: 10.1177/0333102420941864] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background Non-invasive vagus nerve stimulation (nVNS) is a proven treatment for cluster headache and migraine. Several possible mechanisms of action by which nVNS mitigates headache have been identified. Methods We conducted a narrative review of recent scientific and clinical research into nVNS for headache, including findings from mechanistic studies and their possible relationships to the clinical effects of nVNS. Results Findings from animal and human studies have provided possible mechanistic explanations for nVNS efficacy in headache involving four core areas: Autonomic nervous system functions; cortical spreading depression inhibition; neurotransmitter regulation; and nociceptive modulation. We discuss how overlap and interplay among these areas may underlie the utility of nVNS in the context of clinical evidence supporting its safety and efficacy as acute and preventive therapy for both cluster headache and migraine. Possible future nVNS applications are also discussed. Conclusion Significant progress over the past several years has yielded valuable mechanistic and clinical evidence that, combined with the excellent safety and tolerability profile of nVNS, suggests that it should be considered a first-line treatment for both acute and preventive treatment of cluster headache, an effective option for acute treatment of migraine, and a highly relevant, practical option for migraine prevention.
Collapse
Affiliation(s)
- Stephen D Silberstein
- Jefferson Headache Center, Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Hsiangkuo Yuan
- Jefferson Headache Center, Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Umer Najib
- Department of Neurology, West Virginia University, Morgantown, West Virginia, USA
| | - Jessica Ailani
- Medstar Georgetown University Hospital, Washington, DC, USA
| | - Andreia Lopes de Morais
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Paul G Mathew
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Brigham & Women's Hospital, Boston, Massachusetts, USA
- Department of Neurology, Harvard Vanguard Medical Associates, Braintree, Massachusetts, USA
| | - Eric Liebler
- electroCore, Inc., Basking Ridge, New Jersey, USA
| | - Cristina Tassorelli
- Headache Science Center, IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Hans-Christoph Diener
- Institute for Medical Informatics, Biometry and Epidemiology, Medical Faculty of the University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
20
|
Fishman MA, Antony A, Esposito M, Deer T, Levy R. The Evolution of Neuromodulation in the Treatment of Chronic Pain: Forward-Looking Perspectives. PAIN MEDICINE 2020; 20:S58-S68. [PMID: 31152176 PMCID: PMC6600066 DOI: 10.1093/pm/pnz074] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background The field of neuromodulation is continually evolving, with the past decade showing significant advancement in the therapeutic efficacy of neuromodulation procedures. The continued evolution of neuromodulation technology brings with it the promise of addressing the needs of both patients and physicians, as current technology improves and clinical applications expand. Design This review highlights the current state of the art of neuromodulation for treating chronic pain, describes key areas of development including stimulation patterns and neural targets, expanding indications and applications, feedback-controlled systems, noninvasive approaches, and biomarkers for neuromodulation and technology miniaturization. Results and Conclusions The field of neuromodulation is undergoing a renaissance of technology development with potential for profoundly improving the care of chronic pain patients. New and emerging targets like the dorsal root ganglion, as well as high-frequency and patterned stimulation methodologies such as burst stimulation, are paving the way for better clinical outcomes. As we look forward to the future, neural sensing, novel target-specific stimulation patterns, and approaches combining neuromodulation therapies are likely to significantly impact how neuromodulation is used. Moreover, select biomarkers may influence and guide the use of neuromodulation and help objectively demonstrate efficacy and outcomes.
Collapse
Affiliation(s)
| | | | | | - Timothy Deer
- The Spine and Nerve Center of the Virginias, Charleston, West Virginia
| | - Robert Levy
- Institute for Neuromodulation, Boca Raton, Florida, USA
| |
Collapse
|
21
|
Myers DE. The receptive field for visceral pain referred orofacially by the vagus nerves. Clin Anat 2020; 34:24-29. [PMID: 32279338 DOI: 10.1002/ca.23604] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND The nociceptive receptive field of the vagus nerves in animals includes virtually the entire thoracic, abdominal and laryngopharyngeal regions. However, the role of the vagus nerves in the transmission of visceral pain in humans, with the exception of pain from coronary artery diseases, is believed to be insignificant. AIM The purpose of this report is to map out the clinical visceral pain receptive field of the vagus nerves relative to its nociceptive counterpart in animals. MATERIALS AND METHODS The PubMed database and PMC were searched for case reports of patients with orofacial pain believed by the author(s) of the article to be referred from underlying non-cardiac thoracic, laryngopharyngeal or abdominal diseases. Reports of diseases for which non-neural explanations for the orofacial spread of pain were suggested were excluded. RESULTS A total of 52 case reports of jaw pain and/or otalgia referred from laryngopharyngeal and noncardiac thoracic sources were discovered. In addition, a multicenter prospective study found that 25.8% of more than 3,000 patients with thoracic aortic dissection experienced pain in the head and neck region. In stark contrast, no case reports of orofacially referred pain from abdominal diseases were found. DISCUSSION The results indicate that the laryngopharyngeal and thoracic portions of the vagal receptive field are capable of referring pain orofacially while the abdominal portion is not. The roles of the somatotopic organization of the trigeminal sub nucleus caudalis and neuromodulation in this referral of pain were discussed. CONCLUSION Referred orofacial pain can lead to delayed diagnosis and poorer outcome in visceral diseases.
Collapse
|
22
|
Mogilevski T, Burgell R, Aziz Q, Gibson PR. Review article: the role of the autonomic nervous system in the pathogenesis and therapy of IBD. Aliment Pharmacol Ther 2019; 50:720-737. [PMID: 31418887 DOI: 10.1111/apt.15433] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/25/2019] [Accepted: 07/01/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND There is a growing body of evidence implicating a role for the brain-gut axis in the pathogenesis of inflammation in patients with IBD. AIMS To perform a narrative review of published literature regarding the association of the autonomic nervous system and intestinal inflammation and to describe the rationale for and emerging use of autonomic manipulation as a therapeutic agent METHODS: Current relevant literature was summarised and critically examined. RESULTS There is substantial pre-clinical and clinical evidence for a multifaceted anti-inflammatory effect of the vagus at both systemic and local intestinal levels. It acts via acetylcholine-mediated activation of α-7-acetylcholine receptors involving multiple cell types in innate and adaptive immunity and the enteric nervous system with subsequent protective influences on the intestinal barrier, inflammatory mechanisms and the microbiome. In patients with IBD, there is evidence for a sympatho-vagal imbalance, functional enteric neuronal depletion and hyporeactivity of the hypothalamic-pituitary-adrenal axis. Direct or transcutaneous vagal neuromodulation up-regulates the cholinergic anti-inflammatory pathway in pre-clinical and clinical models with down-regulation of systemic and local intestinal inflammation. This is supported by two small studies in Crohn's disease although remains to be investigated in ulcerative colitis. CONCLUSIONS Modulating the cholinergic anti-inflammatory pathway influences inflammation both systemically and at a local intestinal level. It represents a potentially underutilised anti-inflammatory therapeutic strategy. Given the likely pathogenic role of the autonomic nervous system in patients with IBD, vagal neuromodulation, an apparently safe and successful means of increasing vagal tone, warrants further clinical exploration.
Collapse
Affiliation(s)
- Tamara Mogilevski
- Centre for Neuroscience, Surgery and Trauma, Barts and the London School of Medicine and Dentistry, Blizard Institute, Wingate Institute of Neurogastroenterology, London, UK.,Barts Health NHS Trust, London, UK.,Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Australia
| | - Rebecca Burgell
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Australia
| | - Qasim Aziz
- Centre for Neuroscience, Surgery and Trauma, Barts and the London School of Medicine and Dentistry, Blizard Institute, Wingate Institute of Neurogastroenterology, London, UK.,Barts Health NHS Trust, London, UK
| | - Peter R Gibson
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Australia
| |
Collapse
|
23
|
Ginn C, Patel B, Walker R. Existing and emerging applications for the neuromodulation of nerve activity through targeted delivery of electric stimuli. Int J Neurosci 2019; 129:1013-1023. [PMID: 31092102 DOI: 10.1080/00207454.2019.1609473] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The effective treatment of many diseases requires the use of multiple treatment strategies among which neuromodulation is playing an increasingly important role. Neuromodulation devices that act to normalize or modulate nerve activity through the targeted delivery of electrical stimuli will be the focus of this review. These devices encompass deep brain stimulators, vagus nerve stimulators, spinal cord simulators and sacral nerve stimulators. Already neuromodulation has proven successful in the treatment of a broad range of conditions from Parkinson's disease to chronic pain and urinary incontinence. Many of these approaches seek to exploit the activities of the autonomic nervous system, which influences organ function through the release of neurotransmitters and associated signalling cascades. This review will outline existing and emerging applications for each of these neuromodulation devices, proposed mechanisms of action and clinical studies evaluating both their safety and therapeutic efficacy.
Collapse
Affiliation(s)
- Claire Ginn
- ElectronRx Ltd., Eagle Labs , Cambridge , UK
| | - Bipin Patel
- ElectronRx Ltd., Eagle Labs , Cambridge , UK
| | - Robert Walker
- School of Biological Sciences, University of Southampton , Southampton , UK
| |
Collapse
|
24
|
Brainstem neuroimaging of nociception and pain circuitries. Pain Rep 2019; 4:e745. [PMID: 31579846 PMCID: PMC6727990 DOI: 10.1097/pr9.0000000000000745] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/22/2019] [Accepted: 03/24/2019] [Indexed: 01/09/2023] Open
Abstract
The brainstem is known to be an important brain area for nociception and pain processing, and both relaying and coordinating signaling between the cerebrum, cerebellum, and spinal cord. Although preclinical models of pain have characterized the many roles that brainstem nuclei play in nociceptive processing, the degree to which these circuitries extend to humans is not as well known. Unfortunately, the brainstem is also a very challenging region to evaluate in humans with neuroimaging. The challenges for human brainstem imaging arise from the location of this elongated brain structure, proximity to cardiorespiratory noise sources, and the size of its constituent nuclei. These challenges can require dedicated approaches to brainstem imaging, which should be adopted when study hypotheses are focused on brainstem processing of nociception or modulation of pain perception. In fact, our review will highlight many pain neuroimaging studies that have reported some brainstem involvement in nociceptive processing and chronic pain pathology. However, we note that with recent advances in neuroimaging leading to improved spatial and temporal resolution, more studies are needed that take advantage of data collection and analysis methods focused on the challenges of brainstem neuroimaging.
Collapse
|
25
|
Halker Singh RB, Ailani J, Robbins MS. Neuromodulation for the Acute and Preventive Therapy of Migraine and Cluster Headache. Headache 2019; 59 Suppl 2:33-49. [DOI: 10.1111/head.13586] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2019] [Indexed: 12/15/2022]
Affiliation(s)
| | - Jessica Ailani
- Department of Neurology Georgetown University Washington DC USA
| | | |
Collapse
|
26
|
Lerman I, Davis B, Huang M, Huang C, Sorkin L, Proudfoot J, Zhong E, Kimball D, Rao R, Simon B, Spadoni A, Strigo I, Baker DG, Simmons AN. Noninvasive vagus nerve stimulation alters neural response and physiological autonomic tone to noxious thermal challenge. PLoS One 2019; 14:e0201212. [PMID: 30759089 PMCID: PMC6373934 DOI: 10.1371/journal.pone.0201212] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/12/2018] [Indexed: 11/23/2022] Open
Abstract
The mechanisms by which noninvasive vagal nerve stimulation (nVNS) affect central and peripheral neural circuits that subserve pain and autonomic physiology are not clear, and thus remain an area of intense investigation. Effects of nVNS vs sham stimulation on subject responses to five noxious thermal stimuli (applied to left lower extremity), were measured in 30 healthy subjects (n = 15 sham and n = 15 nVNS), with fMRI and physiological galvanic skin response (GSR). With repeated noxious thermal stimuli a group × time analysis showed a significantly (p < .001) decreased response with nVNS in bilateral primary and secondary somatosensory cortices (SI and SII), left dorsoposterior insular cortex, bilateral paracentral lobule, bilateral medial dorsal thalamus, right anterior cingulate cortex, and right orbitofrontal cortex. A group × time × GSR analysis showed a significantly decreased response in the nVNS group (p < .0005) bilaterally in SI, lower and mid medullary brainstem, and inferior occipital cortex. Finally, nVNS treatment showed decreased activity in pronociceptive brainstem nuclei (e.g. the reticular nucleus and rostral ventromedial medulla) and key autonomic integration nuclei (e.g. the rostroventrolateral medulla, nucleus ambiguous, and dorsal motor nucleus of the vagus nerve). In aggregate, noninvasive vagal nerve stimulation reduced the physiological response to noxious thermal stimuli and impacted neural circuits important for pain processing and autonomic output.
Collapse
Affiliation(s)
- Imanuel Lerman
- VA Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, La Jolla, CA, United States of America
- Department of Anesthesiology, Center for Pain Medicine, University of California San Diego School of Medicine, La Jolla, CA, United States of America
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, United States of America
- * E-mail:
| | - Bryan Davis
- Department of Anesthesiology, Center for Pain Medicine, University of California San Diego School of Medicine, La Jolla, CA, United States of America
| | - Mingxiong Huang
- Department of Radiology, University of California San Diego School of Medicine, La Jolla, CA, United States of America
- Department of Radiology, VA San Diego Healthcare System, La Jolla, CA, United States of America
| | - Charles Huang
- Department of Radiology, University of California San Diego School of Medicine, La Jolla, CA, United States of America
- Department of Radiology, VA San Diego Healthcare System, La Jolla, CA, United States of America
| | - Linda Sorkin
- Department of Anesthesiology, Center for Pain Medicine, University of California San Diego School of Medicine, La Jolla, CA, United States of America
| | - James Proudfoot
- Department of Anesthesiology, Center for Pain Medicine, University of California San Diego School of Medicine, La Jolla, CA, United States of America
| | - Edward Zhong
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, United States of America
| | - Donald Kimball
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, United States of America
| | - Ramesh Rao
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, United States of America
| | - Bruce Simon
- electroCore LLC, Basking Ridge NJ, United States of America
| | - Andrea Spadoni
- VA Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, La Jolla, CA, United States of America
- Department of Psychiatry University of California San Diego School of Medicine, La Jolla, CA, United States of America
| | - Irina Strigo
- Department of Psychiatry, VA San Francisco Healthcare System, San Francisco, CA, United States of America
| | - Dewleen G. Baker
- VA Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, La Jolla, CA, United States of America
- Department of Psychiatry University of California San Diego School of Medicine, La Jolla, CA, United States of America
| | - Alan N. Simmons
- VA Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, La Jolla, CA, United States of America
- Department of Psychiatry University of California San Diego School of Medicine, La Jolla, CA, United States of America
| |
Collapse
|
27
|
Jungmann M, Vencatachellum S, Van Ryckeghem D, Vögele C. Effects of Cold Stimulation on Cardiac-Vagal Activation in Healthy Participants: Randomized Controlled Trial. JMIR Form Res 2018; 2:e10257. [PMID: 30684416 PMCID: PMC6334714 DOI: 10.2196/10257] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/14/2018] [Accepted: 07/28/2018] [Indexed: 11/20/2022] Open
Abstract
Background The experience of psychological stress has not yet been adequately tackled with digital technology by catering to healthy individuals who wish to reduce their acute stress levels. For the design of digitally mediated solutions, physiological mechanisms need to be investigated that have the potential to induce relaxation with the help of technology. Research has shown that physiological mechanisms embodied in the face and neck regions are effective for diminishing stress-related symptoms. Our study expands on these areas with the design for a wearable in mind. As this study charts new territory in research, it also is a first evaluation of the viability for a wearables concept to reduce stress. Objective The objectives of this study were to assess whether (1) heart rate variability would increase and (2) heart rate would decrease during cold stimulation using a thermode device compared with a (nonstimulated) control condition. We expected effects in particular in the neck and cheek regions and less in the forearm area. Methods The study was a fully randomized, within-participant design. Volunteer participants were seated in a laboratory chair and tested with cold stimulation on the right side of the body. A thermode was placed on the neck, cheek, and forearm. We recorded and subsequently analyzed participants’ electrocardiogram. The cold stimulation was applied in 16-second intervals over 4 trials per testing location. The control condition proceeded exactly like the cold condition, except we manipulated the temperature variable to remain at the baseline temperature. We measured heart rate as interbeat intervals in milliseconds and analyzed root mean square of successive differences to index heart rate variability. We analyzed data using a repeated-measures ANOVA (analysis of variance) approach with 2 repeated-measures factors: body location (neck, cheek, forearm) and condition (cold, control). Results Data analysis of 61 participants (after exclusion of outliers) showed a main effect and an interaction effect for body location and for condition, for both heart rate and heart rate variability. The results demonstrate a pattern of cardiovascular reactivity to cold stimulation, suggesting an increase in cardiac-vagal activation. The effect was significant for cold stimulation in the lateral neck area. Conclusions The results confirmed our main hypothesis that cold stimulation at the lateral neck region would result in higher heart rate variability and lower heart rate than in the control condition. This sets the stage for further investigations of stress reduction potential in the neck region by developing a wearable prototype that can be used for cold application. Future studies should include a stress condition, test for a range of temperatures and durations, and collect self-report data on perceived stress levels to advance findings.
Collapse
Affiliation(s)
- Manuela Jungmann
- Institute for Health and Behaviour, Faculty of Language and Literature, Humanities, Arts and Education, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Shervin Vencatachellum
- Institute for Health and Behaviour, Faculty of Language and Literature, Humanities, Arts and Education, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Dimitri Van Ryckeghem
- Institute for Health and Behaviour, Faculty of Language and Literature, Humanities, Arts and Education, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Claus Vögele
- Institute for Health and Behaviour, Faculty of Language and Literature, Humanities, Arts and Education, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
28
|
Tassorelli C, Grazzi L, de Tommaso M, Pierangeli G, Martelletti P, Rainero I, Dorlas S, Geppetti P, Ambrosini A, Sarchielli P, Liebler E, Barbanti P. Noninvasive vagus nerve stimulation as acute therapy for migraine: The randomized PRESTO study. Neurology 2018; 91:e364-e373. [PMID: 29907608 PMCID: PMC6070381 DOI: 10.1212/wnl.0000000000005857] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/11/2018] [Indexed: 01/03/2023] Open
Abstract
Objective To evaluate the efficacy, safety, and tolerability of noninvasive vagus nerve stimulation (nVNS; gammaCore; electroCore, LLC, Basking Ridge, NJ) for the acute treatment of migraine in a multicenter, double-blind, randomized, sham-controlled trial. Methods A total of 248 participants with episodic migraine with/without aura were randomized to receive nVNS or sham within 20 minutes from pain onset. Participants were to repeat treatment if pain had not improved in 15 minutes. Results nVNS (n = 120) was superior to sham (n = 123) for pain freedom at 30 minutes (12.7% vs 4.2%; p = 0.012) and 60 minutes (21.0% vs 10.0%; p = 0.023) but not at 120 minutes (30.4% vs 19.7%; p = 0.067; primary endpoint; logistic regression) after the first treated attack. A post hoc repeated-measures test provided further insight into the therapeutic benefit of nVNS through 30, 60, and 120 minutes (odds ratio 2.3; 95% confidence interval 1.2, 4.4; p = 0.012). nVNS demonstrated benefits across other endpoints including pain relief at 120 minutes and was safe and well-tolerated. Conclusion This randomized sham-controlled trial supports the abortive efficacy of nVNS as early as 30 minutes and up to 60 minutes after an attack. Findings also suggest effective pain relief, tolerability, and practicality of nVNS for the acute treatment of episodic migraine. ClinicalTrials.gov identifier NCT02686034. Classification of evidence This study provides Class I evidence that for patients with an episodic migraine, nVNS significantly increases the probability of having mild pain or being pain-free 2 hours poststimulation (absolute difference 13.2%).
Collapse
Affiliation(s)
- Cristina Tassorelli
- From the Headache Science Centre (C.T.), IRCCS C. Mondino Foundation, Pavia; University of Pavia (C.T.); Headache Center (L.G.), Carlo Besta Neurological Institute and Foundation, Milan; Neurophysiology and Pain Unit (M.d.T.), University of Bari Aldo Moro; Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) (G.P.), Istituto delle Scienze Neurologiche di Bologna; Department of Clinical and Molecular Medicine (P.M.), Sapienza University, Rome; Department of Neuroscience (I.R.), University of Turin, Italy; MedLogix Communications, LLC (S.D.), Itasca IL; Headache Centre (P.G.), University Hospital of Careggi, Florence; IRCCS Neuromed (A.A.), Pozzilli (IS); Neurologic Clinic (P.S.), Santa Maria della Misericordia Hospital, Perugia, Italy; electroCore, LLC (E.L.), Basking Ridge, NJ; and Headache and Pain Unit (P.B.), IRCCS San Raffaele Pisana, Rome, Italy.
| | - Licia Grazzi
- From the Headache Science Centre (C.T.), IRCCS C. Mondino Foundation, Pavia; University of Pavia (C.T.); Headache Center (L.G.), Carlo Besta Neurological Institute and Foundation, Milan; Neurophysiology and Pain Unit (M.d.T.), University of Bari Aldo Moro; Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) (G.P.), Istituto delle Scienze Neurologiche di Bologna; Department of Clinical and Molecular Medicine (P.M.), Sapienza University, Rome; Department of Neuroscience (I.R.), University of Turin, Italy; MedLogix Communications, LLC (S.D.), Itasca IL; Headache Centre (P.G.), University Hospital of Careggi, Florence; IRCCS Neuromed (A.A.), Pozzilli (IS); Neurologic Clinic (P.S.), Santa Maria della Misericordia Hospital, Perugia, Italy; electroCore, LLC (E.L.), Basking Ridge, NJ; and Headache and Pain Unit (P.B.), IRCCS San Raffaele Pisana, Rome, Italy
| | - Marina de Tommaso
- From the Headache Science Centre (C.T.), IRCCS C. Mondino Foundation, Pavia; University of Pavia (C.T.); Headache Center (L.G.), Carlo Besta Neurological Institute and Foundation, Milan; Neurophysiology and Pain Unit (M.d.T.), University of Bari Aldo Moro; Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) (G.P.), Istituto delle Scienze Neurologiche di Bologna; Department of Clinical and Molecular Medicine (P.M.), Sapienza University, Rome; Department of Neuroscience (I.R.), University of Turin, Italy; MedLogix Communications, LLC (S.D.), Itasca IL; Headache Centre (P.G.), University Hospital of Careggi, Florence; IRCCS Neuromed (A.A.), Pozzilli (IS); Neurologic Clinic (P.S.), Santa Maria della Misericordia Hospital, Perugia, Italy; electroCore, LLC (E.L.), Basking Ridge, NJ; and Headache and Pain Unit (P.B.), IRCCS San Raffaele Pisana, Rome, Italy
| | - Giulia Pierangeli
- From the Headache Science Centre (C.T.), IRCCS C. Mondino Foundation, Pavia; University of Pavia (C.T.); Headache Center (L.G.), Carlo Besta Neurological Institute and Foundation, Milan; Neurophysiology and Pain Unit (M.d.T.), University of Bari Aldo Moro; Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) (G.P.), Istituto delle Scienze Neurologiche di Bologna; Department of Clinical and Molecular Medicine (P.M.), Sapienza University, Rome; Department of Neuroscience (I.R.), University of Turin, Italy; MedLogix Communications, LLC (S.D.), Itasca IL; Headache Centre (P.G.), University Hospital of Careggi, Florence; IRCCS Neuromed (A.A.), Pozzilli (IS); Neurologic Clinic (P.S.), Santa Maria della Misericordia Hospital, Perugia, Italy; electroCore, LLC (E.L.), Basking Ridge, NJ; and Headache and Pain Unit (P.B.), IRCCS San Raffaele Pisana, Rome, Italy
| | - Paolo Martelletti
- From the Headache Science Centre (C.T.), IRCCS C. Mondino Foundation, Pavia; University of Pavia (C.T.); Headache Center (L.G.), Carlo Besta Neurological Institute and Foundation, Milan; Neurophysiology and Pain Unit (M.d.T.), University of Bari Aldo Moro; Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) (G.P.), Istituto delle Scienze Neurologiche di Bologna; Department of Clinical and Molecular Medicine (P.M.), Sapienza University, Rome; Department of Neuroscience (I.R.), University of Turin, Italy; MedLogix Communications, LLC (S.D.), Itasca IL; Headache Centre (P.G.), University Hospital of Careggi, Florence; IRCCS Neuromed (A.A.), Pozzilli (IS); Neurologic Clinic (P.S.), Santa Maria della Misericordia Hospital, Perugia, Italy; electroCore, LLC (E.L.), Basking Ridge, NJ; and Headache and Pain Unit (P.B.), IRCCS San Raffaele Pisana, Rome, Italy
| | - Innocenzo Rainero
- From the Headache Science Centre (C.T.), IRCCS C. Mondino Foundation, Pavia; University of Pavia (C.T.); Headache Center (L.G.), Carlo Besta Neurological Institute and Foundation, Milan; Neurophysiology and Pain Unit (M.d.T.), University of Bari Aldo Moro; Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) (G.P.), Istituto delle Scienze Neurologiche di Bologna; Department of Clinical and Molecular Medicine (P.M.), Sapienza University, Rome; Department of Neuroscience (I.R.), University of Turin, Italy; MedLogix Communications, LLC (S.D.), Itasca IL; Headache Centre (P.G.), University Hospital of Careggi, Florence; IRCCS Neuromed (A.A.), Pozzilli (IS); Neurologic Clinic (P.S.), Santa Maria della Misericordia Hospital, Perugia, Italy; electroCore, LLC (E.L.), Basking Ridge, NJ; and Headache and Pain Unit (P.B.), IRCCS San Raffaele Pisana, Rome, Italy
| | - Stefanie Dorlas
- From the Headache Science Centre (C.T.), IRCCS C. Mondino Foundation, Pavia; University of Pavia (C.T.); Headache Center (L.G.), Carlo Besta Neurological Institute and Foundation, Milan; Neurophysiology and Pain Unit (M.d.T.), University of Bari Aldo Moro; Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) (G.P.), Istituto delle Scienze Neurologiche di Bologna; Department of Clinical and Molecular Medicine (P.M.), Sapienza University, Rome; Department of Neuroscience (I.R.), University of Turin, Italy; MedLogix Communications, LLC (S.D.), Itasca IL; Headache Centre (P.G.), University Hospital of Careggi, Florence; IRCCS Neuromed (A.A.), Pozzilli (IS); Neurologic Clinic (P.S.), Santa Maria della Misericordia Hospital, Perugia, Italy; electroCore, LLC (E.L.), Basking Ridge, NJ; and Headache and Pain Unit (P.B.), IRCCS San Raffaele Pisana, Rome, Italy
| | - Pierangelo Geppetti
- From the Headache Science Centre (C.T.), IRCCS C. Mondino Foundation, Pavia; University of Pavia (C.T.); Headache Center (L.G.), Carlo Besta Neurological Institute and Foundation, Milan; Neurophysiology and Pain Unit (M.d.T.), University of Bari Aldo Moro; Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) (G.P.), Istituto delle Scienze Neurologiche di Bologna; Department of Clinical and Molecular Medicine (P.M.), Sapienza University, Rome; Department of Neuroscience (I.R.), University of Turin, Italy; MedLogix Communications, LLC (S.D.), Itasca IL; Headache Centre (P.G.), University Hospital of Careggi, Florence; IRCCS Neuromed (A.A.), Pozzilli (IS); Neurologic Clinic (P.S.), Santa Maria della Misericordia Hospital, Perugia, Italy; electroCore, LLC (E.L.), Basking Ridge, NJ; and Headache and Pain Unit (P.B.), IRCCS San Raffaele Pisana, Rome, Italy
| | - Anna Ambrosini
- From the Headache Science Centre (C.T.), IRCCS C. Mondino Foundation, Pavia; University of Pavia (C.T.); Headache Center (L.G.), Carlo Besta Neurological Institute and Foundation, Milan; Neurophysiology and Pain Unit (M.d.T.), University of Bari Aldo Moro; Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) (G.P.), Istituto delle Scienze Neurologiche di Bologna; Department of Clinical and Molecular Medicine (P.M.), Sapienza University, Rome; Department of Neuroscience (I.R.), University of Turin, Italy; MedLogix Communications, LLC (S.D.), Itasca IL; Headache Centre (P.G.), University Hospital of Careggi, Florence; IRCCS Neuromed (A.A.), Pozzilli (IS); Neurologic Clinic (P.S.), Santa Maria della Misericordia Hospital, Perugia, Italy; electroCore, LLC (E.L.), Basking Ridge, NJ; and Headache and Pain Unit (P.B.), IRCCS San Raffaele Pisana, Rome, Italy
| | - Paola Sarchielli
- From the Headache Science Centre (C.T.), IRCCS C. Mondino Foundation, Pavia; University of Pavia (C.T.); Headache Center (L.G.), Carlo Besta Neurological Institute and Foundation, Milan; Neurophysiology and Pain Unit (M.d.T.), University of Bari Aldo Moro; Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) (G.P.), Istituto delle Scienze Neurologiche di Bologna; Department of Clinical and Molecular Medicine (P.M.), Sapienza University, Rome; Department of Neuroscience (I.R.), University of Turin, Italy; MedLogix Communications, LLC (S.D.), Itasca IL; Headache Centre (P.G.), University Hospital of Careggi, Florence; IRCCS Neuromed (A.A.), Pozzilli (IS); Neurologic Clinic (P.S.), Santa Maria della Misericordia Hospital, Perugia, Italy; electroCore, LLC (E.L.), Basking Ridge, NJ; and Headache and Pain Unit (P.B.), IRCCS San Raffaele Pisana, Rome, Italy
| | - Eric Liebler
- From the Headache Science Centre (C.T.), IRCCS C. Mondino Foundation, Pavia; University of Pavia (C.T.); Headache Center (L.G.), Carlo Besta Neurological Institute and Foundation, Milan; Neurophysiology and Pain Unit (M.d.T.), University of Bari Aldo Moro; Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) (G.P.), Istituto delle Scienze Neurologiche di Bologna; Department of Clinical and Molecular Medicine (P.M.), Sapienza University, Rome; Department of Neuroscience (I.R.), University of Turin, Italy; MedLogix Communications, LLC (S.D.), Itasca IL; Headache Centre (P.G.), University Hospital of Careggi, Florence; IRCCS Neuromed (A.A.), Pozzilli (IS); Neurologic Clinic (P.S.), Santa Maria della Misericordia Hospital, Perugia, Italy; electroCore, LLC (E.L.), Basking Ridge, NJ; and Headache and Pain Unit (P.B.), IRCCS San Raffaele Pisana, Rome, Italy
| | - Piero Barbanti
- From the Headache Science Centre (C.T.), IRCCS C. Mondino Foundation, Pavia; University of Pavia (C.T.); Headache Center (L.G.), Carlo Besta Neurological Institute and Foundation, Milan; Neurophysiology and Pain Unit (M.d.T.), University of Bari Aldo Moro; Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) (G.P.), Istituto delle Scienze Neurologiche di Bologna; Department of Clinical and Molecular Medicine (P.M.), Sapienza University, Rome; Department of Neuroscience (I.R.), University of Turin, Italy; MedLogix Communications, LLC (S.D.), Itasca IL; Headache Centre (P.G.), University Hospital of Careggi, Florence; IRCCS Neuromed (A.A.), Pozzilli (IS); Neurologic Clinic (P.S.), Santa Maria della Misericordia Hospital, Perugia, Italy; electroCore, LLC (E.L.), Basking Ridge, NJ; and Headache and Pain Unit (P.B.), IRCCS San Raffaele Pisana, Rome, Italy
| | | |
Collapse
|
29
|
Modulation of brainstem activity and connectivity by respiratory-gated auricular vagal afferent nerve stimulation in migraine patients. Pain 2018; 158:1461-1472. [PMID: 28541256 DOI: 10.1097/j.pain.0000000000000930] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Migraine pathophysiology includes altered brainstem excitability, and recent neuromodulatory approaches aimed at controlling migraine episodes have targeted key brainstem relay and modulatory nuclei. In this study, we evaluated the impact of respiratory-gated auricular vagal afferent nerve stimulation (RAVANS), a novel neuromodulatory intervention based on an existing transcutaneous vagus nerve stimulation approach, in the modulation of brainstem activity and connectivity in migraine patients. We applied 3T-functional magnetic resonance imaging with improved in-plane spatial resolution (2.62 × 2.62 mm) in episodic migraine (interictal) and age- and sex-matched healthy controls to evaluate brain response to RAVANS (gated to either inhalation or exhalation) and sham stimulation. We further investigated RAVANS modulation of tactile trigeminal sensory afference response in the brainstem using air-puff stimulation directed to the forehead during functional magnetic resonance imaging. Compared with sham and inhalatory-gated RAVANS (iRAVANS), exhalatory-gated RAVANS (eRAVANS) activated an ipsilateral pontomedullary region consistent with nucleus tractus solitarii (NTS). During eRAVANS, NTS connectivity was increased to anterior insula and anterior midcingulate cortex, compared with both sham and iRAVANS, in migraine patients. Increased connectivity was inversely correlated with relative time to the next migraine attack, suggesting clinical relevance to this change in connectivity. Poststimulation effects were also noted immediately after eRAVANS, as we found increased activation in putative pontine serotonergic (ie, nucleus raphe centralis) and noradrenergic (ie, locus coeruleus) nuclei in response to trigeminal sensory afference. Regulation of activity and connectivity of brainstem and cortical regions involved in serotonergic and noradrenergic regulation and pain modulation may constitute an underlying mechanism supporting beneficial clinical outcomes for eRAVANS applied for episodic migraine.
Collapse
|
30
|
Mourdoukoutas AP, Truong DQ, Adair DK, Simon BJ, Bikson M. High-Resolution Multi-Scale Computational Model for Non-Invasive Cervical Vagus Nerve Stimulation. Neuromodulation 2018; 21:261-268. [PMID: 29076212 PMCID: PMC5895480 DOI: 10.1111/ner.12706] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/26/2017] [Accepted: 08/25/2017] [Indexed: 12/28/2022]
Abstract
OBJECTIVES To develop the first high-resolution, multi-scale model of cervical non-invasive vagus nerve stimulation (nVNS) and to predict vagus fiber type activation, given clinically relevant rheobase thresholds. METHODS An MRI-derived Finite Element Method (FEM) model was developed to accurately simulate key macroscopic (e.g., skin, soft tissue, muscle) and mesoscopic (cervical enlargement, vertebral arch and foramen, cerebral spinal fluid [CSF], nerve sheath) tissue components to predict extracellular potential, electric field (E-Field), and activating function along the vagus nerve. Microscopic scale biophysical models of axons were developed to compare axons of varying size (Aα-, Aβ- and Aδ-, B-, and C-fibers). Rheobase threshold estimates were based on a step function waveform. RESULTS Macro-scale accuracy was found to determine E-Field magnitudes around the vagus nerve, while meso-scale precision determined E-field changes (activating function). Mesoscopic anatomical details that capture vagus nerve passage through a changing tissue environment (e.g., bone to soft tissue) profoundly enhanced predicted axon sensitivity while encapsulation in homogenous tissue (e.g., nerve sheath) dulled axon sensitivity to nVNS. CONCLUSIONS These findings indicate that realistic and precise modeling at both macroscopic and mesoscopic scales are needed for quantitative predictions of vagus nerve activation. Based on this approach, we predict conventional cervical nVNS protocols can activate A- and B- but not C-fibers. Our state-of-the-art implementation across scales is equally valuable for models of spinal cord stimulation, cortex/deep brain stimulation, and other peripheral/cranial nerve models.
Collapse
Affiliation(s)
- Antonios P. Mourdoukoutas
- Department of Biomedical Engineering, The City College of New York, City University of New York, New York, NY
| | - Dennis Q. Truong
- Department of Biomedical Engineering, The City College of New York, City University of New York, New York, NY
| | - Devin K. Adair
- Department of Psychology, The Graduate Center, City University of New York, New York, New York
| | | | - Marom Bikson
- Department of Biomedical Engineering, The City College of New York, City University of New York, New York, NY
| |
Collapse
|
31
|
Grazzi L, Sansone E, Rizzoli P. A Short Review of the Non-invasive Transcutaneous Pericranial Electrical Stimulation Techniques and their Application in Headache. Curr Pain Headache Rep 2018; 22:4. [PMID: 29350303 DOI: 10.1007/s11916-018-0654-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW In this short review, the most common non-invasive neuromodulatory techniques will be described, along with their advantages and disadvantages and their application in headache. Available preventive treatments can be unhelpful or may have unpleasant side effects; moreover, the rate of response to preventive drugs does not exceed 50%, lower in chronic migraine; alternative options would be welcome. Though the concept of neuromodulation was originally developed with invasive methods, newer non-invasive techniques are appearing. RECENT FINDINGS The novel neuromodulatory techniques have been developed with encouraging results: compared with traditional pharmacotherapy, advantages of non-invasive neuromodulation include reduced incidence of adverse effects, improved adherence, and safety and ease of use. The results are encouraging for acute or preventive treatment of different kinds of headache. A variety of neuromodulatory approaches is expanding fastly and has opened new possibilities for treatment of patients suffering from many forms of headache, especially those who have failed traditional pharmacotherapy. The non-invasive treatments can be seen as supplementing traditional management in refractory patients. Current study results are encouraging but preliminary and larger and more rigorous trials are needed to clarify benefit and mode of action.
Collapse
Affiliation(s)
- Licia Grazzi
- 3rd Neurology Unit, Neuroalgology, Fondazione I.R.C.C.S. Istituto Neurologico "Carlo Besta", Via Celoria 11, 20133, Milan, Italy.
| | - Emanuela Sansone
- 3rd Neurology Unit, Neuroalgology, Fondazione I.R.C.C.S. Istituto Neurologico "Carlo Besta", Via Celoria 11, 20133, Milan, Italy
| | - Paul Rizzoli
- John Graham Headache Centre/Faulkner Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
32
|
Dong XY, Feng Z. Wake-promoting effects of vagus nerve stimulation after traumatic brain injury: upregulation of orexin-A and orexin receptor type 1 expression in the prefrontal cortex. Neural Regen Res 2018; 13:244-251. [PMID: 29557373 PMCID: PMC5879895 DOI: 10.4103/1673-5374.226395] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Orexins, produced in the lateral hypothalamus, are important neuropeptides that participate in the sleep/wake cycle, and their expression coincides with the projection area of the vagus nerve in the brain. Vagus nerve stimulation has been shown to decrease the amounts of daytime sleep and rapid eye movement in epilepsy patients with traumatic brain injury. In the present study, we investigated whether vagus nerve stimulation promotes wakefulness and affects orexin expression. A rat model of traumatic brain injury was established using the free fall drop method. In the stimulated group, rats with traumatic brain injury received vagus nerve stimulation (frequency, 30 Hz; current, 1.0 mA; pulse width, 0.5 ms; total stimulation time, 15 minutes). In the antagonist group, rats with traumatic brain injury were intracerebroventricularly injected with the orexin receptor type 1 (OX1R) antagonist SB334867 and received vagus nerve stimulation. Changes in consciousness were observed after stimulation in each group. Enzyme-linked immunosorbent assay, western blot assay and immunohistochemistry were used to assess the levels of orexin-A and OX1R expression in the prefrontal cortex. In the stimulated group, consciousness was substantially improved, orexin-A protein expression gradually increased within 24 hours after injury and OX1R expression reached a peak at 12 hours, compared with rats subjected to traumatic brain injury only. In the antagonist group, the wake-promoting effect of vagus nerve stimulation was diminished, and orexin-A and OX1R expression were decreased, compared with that of the stimulated group. Taken together, our findings suggest that vagus nerve stimulation promotes the recovery of consciousness in comatose rats after traumatic brain injury. The upregulation of orexin-A and OX1R expression in the prefrontal cortex might be involved in the wake-promoting effects of vagus nerve stimulation.
Collapse
Affiliation(s)
- Xiao-Yang Dong
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhen Feng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
33
|
Silberstein SD, Mechtler LL, Kudrow DB, Calhoun AH, McClure C, Saper JR, Liebler EJ, Rubenstein Engel E, Tepper SJ. Non-Invasive Vagus Nerve Stimulation for the ACute Treatment of Cluster Headache: Findings From the Randomized, Double-Blind, Sham-Controlled ACT1 Study. Headache 2017; 56:1317-32. [PMID: 27593728 PMCID: PMC5113831 DOI: 10.1111/head.12896] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 05/11/2016] [Accepted: 06/10/2016] [Indexed: 01/03/2023]
Abstract
Objective To evaluate non‐invasive vagus nerve stimulation (nVNS) as an acute cluster headache (CH) treatment. Background Many patients with CH experience excruciating attacks at a frequency that is not sufficiently addressed by current symptomatic treatments. Methods One hundred fifty subjects were enrolled and randomized (1:1) to receive nVNS or sham treatment for ≤1 month during a double‐blind phase; completers could enter a 3‐month nVNS open‐label phase. The primary end point was response rate, defined as the proportion of subjects who achieved pain relief (pain intensity of 0 or 1) at 15 minutes after treatment initiation for the first CH attack without rescue medication use through 60 minutes. Secondary end points included the sustained response rate (15‐60 minutes). Subanalyses of episodic cluster headache (eCH) and chronic cluster headache (cCH) cohorts were prespecified. Results The intent‐to‐treat population comprised 133 subjects: 60 nVNS‐treated (eCH, n = 38; cCH, n = 22) and 73 sham‐treated (eCH, n = 47; cCH, n = 26). A response was achieved in 26.7% of nVNS‐treated subjects and 15.1% of sham‐treated subjects (P = .1). Response rates were significantly higher with nVNS than with sham for the eCH cohort (nVNS, 34.2%; sham, 10.6%; P = .008) but not the cCH cohort (nVNS, 13.6%; sham, 23.1%; P = .48). Sustained response rates were significantly higher with nVNS for the eCH cohort (P = .008) and total population (P = .04). Adverse device effects (ADEs) were reported by 35/150 (nVNS, 11; sham, 24) subjects in the double‐blind phase and 18/128 subjects in the open‐label phase. No serious ADEs occurred. Conclusions In one of the largest randomized sham‐controlled studies for acute CH treatment, the response rate was not significantly different (vs sham) for the total population; nVNS provided significant, clinically meaningful, rapid, and sustained benefits for eCH but not for cCH, which affected results in the total population. This safe and well‐tolerated treatment represents a novel and promising option for eCH. ClinicalTrials.gov identifier: NCT01792817.
Collapse
Affiliation(s)
| | - Laszlo L Mechtler
- Department of Neurology and Neuro-Oncology, Dent Neurologic Headache Center, Amherst, NY, USA
| | - David B Kudrow
- California Medical Clinic for Headache, Santa Monica, CA, USA
| | | | | | - Joel R Saper
- Michigan Head Pain and Neurological Institute, Ann Arbor, MI, USA
| | - Eric J Liebler
- Department of Scientific, Medical and Governmental Affairs, electroCore, LLC, Basking Ridge, NJ, USA
| | | | - Stewart J Tepper
- Department of Neurology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Dr. Tepper was at Cleveland Clinic Headache Center, Cleveland, OH, at the time of study completion
| | | |
Collapse
|
34
|
Sclocco R, Beissner F, Bianciardi M, Polimeni JR, Napadow V. Challenges and opportunities for brainstem neuroimaging with ultrahigh field MRI. Neuroimage 2017; 168:412-426. [PMID: 28232189 DOI: 10.1016/j.neuroimage.2017.02.052] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/30/2017] [Accepted: 02/19/2017] [Indexed: 12/19/2022] Open
Abstract
The human brainstem plays a central role in connecting the cerebrum, the cerebellum and the spinal cord to one another, hosting relay nuclei for afferent and efferent signaling, and providing source nuclei for several neuromodulatory systems that impact central nervous system function. While the investigation of the brainstem with functional or structural magnetic resonance imaging has been hampered for years due to this brain structure's physiological and anatomical characteristics, the field has seen significant advances in recent years thanks to the broader adoption of ultrahigh-field (UHF) MRI scanning. In the present review, we focus on the advantages offered by UHF in the context of brainstem imaging, as well as the challenges posed by the investigation of this complex brain structure in terms of data acquisition and analysis. We also illustrate how UHF MRI can shed new light on the neuroanatomy and neurophysiology underlying different brainstem-based circuitries, such as the central autonomic network and neurotransmitter/neuromodulator systems, discuss existing and foreseeable clinical applications to better understand diseases such as chronic pain and Parkinson's disease, and explore promising future directions for further improvements in brainstem imaging using UHF MRI techniques.
Collapse
Affiliation(s)
- Roberta Sclocco
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, CNY 149-2301, 13th St. Charlestown, Boston, MA 02129, USA; Department of Radiology, Logan University, Chesterfield, MO, USA.
| | - Florian Beissner
- Somatosensory and Autonomic Therapy Research, Institute for Neuroradiology, Hannover Medical School, Hannover, Germany
| | - Marta Bianciardi
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, CNY 149-2301, 13th St. Charlestown, Boston, MA 02129, USA
| | - Jonathan R Polimeni
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, CNY 149-2301, 13th St. Charlestown, Boston, MA 02129, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Vitaly Napadow
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, CNY 149-2301, 13th St. Charlestown, Boston, MA 02129, USA; Department of Radiology, Logan University, Chesterfield, MO, USA
| |
Collapse
|
35
|
Abstract
Chronic pain is an issue encountered by many health care providers in their routine clinical practice. In addition to generalized patient suffering, this condition has significant clinical, psychological, and socioeconomic impact due to its widespread occurrence. The landscape of chronic pain management has been changing rapidly with an array of treatment innovations, better understanding of established therapies, and care coordination across specialties. In this article, we have reviewed emerging new modalities as well as transformation of established therapies by interventional, pharmacologic, rehabilitative, psychological, complimentary, and interdisciplinary approaches.
Collapse
|
36
|
Pintea B, Hampel K, Boström J, Surges R, Vatter H, Lendvai IS, Kinfe TM. Extended Long-Term Effects of Cervical Vagal Nerve Stimulation on Headache Intensity/Frequency and Affective/Cognitive Headache Perception in Drug Resistant Complex-Partial Seizure Patients. Neuromodulation 2016; 20:375-382. [PMID: 27873376 DOI: 10.1111/ner.12540] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/28/2016] [Accepted: 09/28/2016] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Invasive vagal nerve stimulation (iVNS) is an established treatment option for drug-resistant focal seizures and has been assumed to diminish frequent co-incidental daily headache/migraine. However, long-term effects on cognitive/affective head pain perception, headache intensity/frequency are lacking. We therefore investigated potential iVNS-induced effects in patients with drug-resistant focal seizure and daily headache/migraine. MATERIALS AND METHODS A clinical database was used to select 325 patients with drug-resistant epilepsy treated by either iVNS plus best medical treatment (BMT) or BMT alone, compared to a healthy control group (HC). We assessed headache intensity (VAS), headache frequency, affective/cognitive pain perception (PASS; FSVA), migraine disability scores (MIDAS), sleep architecture (PSQI), depressive symptoms (BDI), and body weight (BMI). RESULTS Nineteen patients with daily headache/migraine composed the clinical groups (10 iVNS and 9 BMT; iVNS mean age 49 years, range 36-61 years; BMT mean age 45 years, range 23-63 years; equally distributed gender). Cervical iVNS was applied from 5-13 years (mean 8 years) with following stimulation patterns: 1.3 mA (0.5-2 mA), 20 Hz, 250 μsec, 30 sec on/1.9 min off (0.5-5 min). The iVNS group had significantly lower VAS scores (iVNS 5.4; BMT 7.8; p = 0.03) and PASS cognitive/anxiety subscores (iVNS 21; BMT 16; p = 0.02) compared to BMT and HC. Global PASS (p = 0.07), FSVA, PSQI, BDI, and BMI scores did not differ significantly between groups. CONCLUSIONS iVNS appears to have positive modulatory long-term effects on headache and affective/cognitive head pain perception in patients with drug-resistant focal epilepsy, thus deserving further attention.
Collapse
Affiliation(s)
- Bogdan Pintea
- Department of Neurosurgery, Rheinische Friedrich Wilhelms University Hospital, Bonn, Germany
| | - Kevin Hampel
- Department of Epileptology, Rheinische Friedrich Wilhelms University Hospital, Bonn, Germany
| | - Jan Boström
- Department of Neurosurgery, Rheinische Friedrich Wilhelms University Hospital, Bonn, Germany
| | - Rainer Surges
- Department of Epileptology, Rheinische Friedrich Wilhelms University Hospital, Bonn, Germany
| | - Hartmut Vatter
- Department of Neurosurgery, Rheinische Friedrich Wilhelms University Hospital, Bonn, Germany
| | - Ilana S Lendvai
- Department of Neurosurgery, Rheinische Friedrich Wilhelms University Hospital, Bonn, Germany
- Division of Functional Neurosurgery, Stereotaxy and Neuromodulation, Rheinische Friedrich Wilhelms University Hospital, Bonn, Germany
| | - Thomas M Kinfe
- Department of Neurosurgery, Rheinische Friedrich Wilhelms University Hospital, Bonn, Germany
- Division of Functional Neurosurgery, Stereotaxy and Neuromodulation, Rheinische Friedrich Wilhelms University Hospital, Bonn, Germany
| |
Collapse
|
37
|
Lee S, Abd-Elsayed A. Some Non-FDA Approved Uses for Neuromodulation in Treating Autonomic Nervous System Disorders: A Discussion of the Preliminary Support. Neuromodulation 2016; 19:791-803. [PMID: 27339825 DOI: 10.1111/ner.12456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/25/2016] [Accepted: 04/22/2016] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Neuromodulation, including cavernous nerve stimulation, gastric electrical stimulation, deep brain stimulation, and vagus nerve stimulation, has been used with success in treating several functional disease conditions. The FDA has approved the use of neuromodulation for a few indications. We discuss in our review article the evidence of using neuromodulation for treating some important disorders involving the autonomic nervous system that are not currently FDA approved. METHODS This was a review article that included a systematic online web search for human clinical studies testing the efficacy of neuromodulation in treating erectile dysfunction, gastroparesis, gastroesophageal reflux disease, obesity, asthma, and heart failure. Our review includes all feasibility studies, nonrandomized clinical trials, and randomized controlled trials. RESULTS Our systematic literature search found 3, 4, 5, 4, 1, and 4 clinical studies relating to erectile dysfunction, gastroparesis, gastroesophageal reflux disease, obesity, asthma, and heart failure, respectively. CONCLUSION This review article shows preliminary support based on clinical studies that neuromodulation can be of benefit for patients with important autonomic nervous system disease conditions that are not currently approved by the FDA. All of these investigational uses are encouraging; further studies are necessary and warranted for all indications discussed in this review before achieving FDA approval.
Collapse
Affiliation(s)
- Samuel Lee
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Alaa Abd-Elsayed
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
38
|
Morris J, Straube A, Diener HC, Ahmed F, Silver N, Walker S, Liebler E, Gaul C. Cost-effectiveness analysis of non-invasive vagus nerve stimulation for the treatment of chronic cluster headache. J Headache Pain 2016; 17:43. [PMID: 27102120 PMCID: PMC4840129 DOI: 10.1186/s10194-016-0633-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 04/07/2016] [Indexed: 01/03/2023] Open
Abstract
Background Cluster headache (CH) is a debilitating condition that is generally associated with substantial health care costs. Few therapies are approved for abortive or prophylactic treatment. Results from the prospective, randomised, open-label PREVA study suggested that adjunctive treatment with a novel non-invasive vagus nerve stimulation (nVNS) device led to decreased attack frequency and abortive medication use in patients with chronic CH (cCH). Herein, we evaluate whether nVNS is cost-effective compared with the current standard of care (SoC) for cCH. Methods A pharmacoeconomic model from the German statutory health insurance perspective was developed to estimate the 1-year cost-effectiveness of nVNS + SoC (versus SoC alone) using data from PREVA. Short-term treatment response data were taken from the clinical trial; longer-term response was modelled under scenarios of response maintenance, constant rate of response loss, and diminishing rate of response loss. Health-related quality of life was estimated by modelling EQ-5D™ data from PREVA; benefits were defined as quality-adjusted life-years (QALY). Abortive medication use data from PREVA, along with costs for the nVNS device and abortive therapies (i.e. intranasal zolmitriptan, subcutaneous sumatriptan, and inhaled oxygen), were used to assess health care costs in the German setting. Results The analysis resulted in mean expected yearly costs of €7096.69 for nVNS + SoC and €7511.35 for SoC alone and mean QALY of 0.607 for nVNS + SoC and 0.522 for SoC alone, suggesting that nVNS generates greater health benefits for lower overall cost. Abortive medication costs were 23 % lower with nVNS + SoC than with SoC alone. In the alternative scenarios (i.e. constant rate of response loss and diminishing rate of response loss), nVNS + SoC was more effective and cost saving than SoC alone. Conclusions In all scenarios modelled from a German perspective, nVNS was cost-effective compared with current SoC, which suggests that adjunctive nVNS therapy provides economic benefits in the treatment of cCH. Notably, the current analysis included only costs associated with abortive treatments. Treatment with nVNS will likely promote further economic benefit when other potential sources of cost savings (e.g. reduced frequency of clinic visits) are considered. Trial registration Clinicaltrials.gov identifier NCT01701245, 03OCT2012.
Collapse
Affiliation(s)
- James Morris
- Cogentia Healthcare Consulting Ltd., Richmond House, 16-20 Regent Street, Cambridge, CB2 1DB, UK.
| | - Andreas Straube
- Ludwig Maximilian University of Munich, Marchioninistr 15, Munich, D81377, Germany
| | - Hans-Christoph Diener
- Department of Neurology and Headache Center, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Fayyaz Ahmed
- Hull and Yorkshire Hospitals, Hull Royal Infirmary, Anlaby Road, Hull, HU3 2JZ, UK
| | - Nicholas Silver
- The Walton Centre for Neurology and Neurosurgery, Lower Lane, Liverpool, L9 7LJ, UK
| | - Simon Walker
- Cogentia Healthcare Consulting Ltd., Richmond House, 16-20 Regent Street, Cambridge, CB2 1DB, UK
| | - Eric Liebler
- electroCore, LLC, 150 Allen Road, Suite 201, Basking Ridge, NJ, 07920, USA
| | - Charly Gaul
- Department of Neurology and Headache Center, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany.,Migraine and Headache Clinic Königstein, Ölmühlweg 31, 61462, Königstein im Taunus, Germany
| |
Collapse
|
39
|
Holle-Lee D, Gaul C. Noninvasive vagus nerve stimulation in the management of cluster headache: clinical evidence and practical experience. Ther Adv Neurol Disord 2016; 9:230-4. [PMID: 27134678 DOI: 10.1177/1756285616636024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The efficacy of invasive vagal nerve stimulation as well as other invasive neuromodulatory approaches such as deep brain stimulation, occipital nerve stimulation, and ganglion sphenopalatine stimulation has been shown in the treatment of headache disorders in several studies in the past. However, these invasive treatment options were quite costly and often associated with perioperative and postoperative side effects, some severe. As such, they were predominantly restricted to chronic and therapy refractory patients. Transcutaneous vagal nerve stimulation now offers a new, noninvasive neuromodulatory treatment approach. Recently published studies showed encouraging results of noninvasive vagus nerve stimulation (nVNS), especially with respect to cluster headache, with high tolerability and a low rate of side effects; however, randomized controlled trials are needed to prove its efficacy. Further data also indicate therapeutic benefits regarding treatment of migraine and medication overuse headache. This review summarizes current knowledge and personal experiences of nVNS in the treatment of cluster headache.
Collapse
Affiliation(s)
- Dagny Holle-Lee
- Department of Neurology and Westgerman Headache Center Essen, University Hospital Essen, Hufelandstr. 55, 45127 Essen, Germany
| | - Charly Gaul
- Migräne- und Kopfschmerzklinik Königstein, Ölmühlweg 31, 61462 Königstein im Taunus, Germany
| |
Collapse
|
40
|
Cook IA, Abrams M, Leuchter AF. Trigeminal Nerve Stimulation for Comorbid Posttraumatic Stress Disorder and Major Depressive Disorder. Neuromodulation 2016; 19:299-305. [PMID: 26818103 DOI: 10.1111/ner.12399] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/27/2015] [Accepted: 12/28/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVES External stimulation of the trigeminal nerve (eTNS) is an emerging neuromodulation therapy for epilepsy and depression. Preliminary studies suggest it has an excellent safety profile and is associated with significant improvements in seizures and mood. Neuroanatomical projections of the trigeminal system suggest eTNS may alter activity in structures regulating mood, anxiety, and sleep. In this proof-of-concept trial, the effects of eTNS were evaluated in adults with posttraumatic stress disorder (PTSD) and comorbid unipolar major depressive disorder (MDD) as an adjunct to pharmacotherapy for these commonly co-occurring conditions. MATERIALS AND METHODS Twelve adults with PTSD and MDD were studied in an eight-week open outpatient trial (age 52.8 [13.7 sd], 8F:4M). Stimulation was applied to the supraorbital and supratrochlear nerves for eight hours each night as an adjunct to pharmacotherapy. Changes in symptoms were monitored using the PTSD Patient Checklist (PCL), Hamilton Depression Rating Scale (HDRS-17), Quick Inventory of Depressive Symptomatology (QIDS-C), and the Quality of Life Enjoyment and Satisfaction Questionnaire (Q-LES-Q). RESULTS Over the eight weeks, eTNS treatment was associated with significant decreases in PCL (p = 0.003; median decrease of 15 points; effect size d 1.5), HDRS-17 (p < 0.001; 42% response rate, 25% remission; d 2.1), and QIDS-C scores (p < 0.001; d 1.8), as well as an improvement in quality of life (Q-LES-Q, p < 0.01). eTNS was well tolerated with few treatment emergent adverse events. CONCLUSIONS Significant improvements in PTSD and depression severity were achieved in the eight weeks of acute eTNS treatment. This novel approach to wearable brain stimulation may have use as an adjunct to pharmacotherapy in these disorders if efficacy and tolerability are confirmed with additional studies.
Collapse
Affiliation(s)
- Ian A Cook
- Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA.,Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.,Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences at UCLA, Los Angeles, CA, USA.,NeuroSigma, Inc, Los Angeles, CA, USA
| | - Michelle Abrams
- Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA.,Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Andrew F Leuchter
- Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA.,Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|