1
|
Witkin JM, Shafique H, Cerne R, Smith JL, Marini AM, Lipsky RH, Delery E. Mechanistic and therapeutic relationships of traumatic brain injury and γ-amino-butyric acid (GABA). Pharmacol Ther 2024; 256:108609. [PMID: 38369062 DOI: 10.1016/j.pharmthera.2024.108609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/18/2024] [Accepted: 02/01/2024] [Indexed: 02/20/2024]
Abstract
Traumatic brain injury (TBI) is a highly prevalent medical condition for which no medications specific for the prophylaxis or treatment of the condition as a whole exist. The spectrum of symptoms includes coma, headache, seizures, cognitive impairment, depression, and anxiety. Although it has been known for years that the inhibitory neurotransmitter γ-amino-butyric acid (GABA) is involved in TBI, no novel therapeutics based upon this mechanism have been introduced into clinical practice. We review the neuroanatomical, neurophysiological, neurochemical, and neuropharmacological relationships of GABA neurotransmission to TBI with a view toward new potential GABA-based medicines. The long-standing idea that excitatory and inhibitory (GABA and others) balances are disrupted by TBI is supported by the experimental data but has failed to invent novel methods of restoring this balance. The slow progress in advancing new treatments is due to the complexity of the disorder that encompasses multiple dynamically interacting biological processes including hemodynamic and metabolic systems, neurodegeneration and neurogenesis, major disruptions in neural networks and axons, frank brain lesions, and a multitude of symptoms that have differential neuronal and neurohormonal regulatory mechanisms. Although the current and ongoing clinical studies include GABAergic drugs, no novel GABA compounds are being explored. It is suggested that filling the gap in understanding the roles played by specific GABAA receptor configurations within specific neuronal circuits could help define new therapeutic approaches. Further research into the temporal and spatial delivery of GABA modulators should also be useful. Along with GABA modulation, research into the sequencing of GABA and non-GABA treatments will be needed.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA; Departments of Neuroscience and Trauma Research, Ascension St. Vincent Hospital, Indianapolis, IN, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA.
| | | | - Rok Cerne
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA; Department of Anatomy and Cell Biology, Indiana University/Purdue University, Indianapolis, IN, USA
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA
| | - Ann M Marini
- Department of Neurology, Program in Neuroscience, and Molecular and Cellular Biology Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Robert H Lipsky
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Elizabeth Delery
- College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA.
| |
Collapse
|
2
|
Morais A, Chung JY, Wu L, Ayata C, Simon B, Whalen MJ. Non-Invasive Vagal Nerve Stimulation Pre-Treatment Reduces Neurological Dysfunction After Closed Head Injury in Mice. Neurotrauma Rep 2024; 5:150-158. [PMID: 38435077 PMCID: PMC10908330 DOI: 10.1089/neur.2023.0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024] Open
Abstract
Non-invasive vagus nerve stimulation (nVNS) has recently been suggested as a potential therapy for traumatic brain injury (TBI). We previously demonstrated that nVNS inhibits cortical spreading depolarization, the electrophysiological event underlying migraine aura, and is relevant to TBI. Our past work also suggests a role for interleukin-1 beta (IL-1β) in cognitive deficits after closed head injury (CHI) in mice. We show that nVNS pre-treatment suppresses CHI-associated spatial learning and memory impairment and prevents IL-1β activation in injured neurons, but not endothelial cells. In contrast, nVNS administered 10 min after CHI was ineffective. These data suggest that nVNS prophylaxis might ameliorate neuronal dysfunction associated with CHI in populations at high risk for concussive TBI.
Collapse
Affiliation(s)
- Andreia Morais
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Joon Yong Chung
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Limin Wu
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Cenk Ayata
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Bruce Simon
- ElectroCore, Inc., Basking Ridge, New Jersey, USA
| | - Michael J. Whalen
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| |
Collapse
|
3
|
da Silva Fiorin F, do Espírito Santo CC, Da Silva JT, Chung MK. Inflammation, brain connectivity, and neuromodulation in post-traumatic headache. Brain Behav Immun Health 2024; 35:100723. [PMID: 38292321 PMCID: PMC10827408 DOI: 10.1016/j.bbih.2024.100723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 02/01/2024] Open
Abstract
Post-traumatic headache (PTH) is a debilitating condition that affects individuals with different levels of traumatic brain injury (TBI) severity. The difficulties in developing an effective treatment are related to a lack of understanding the complicated mechanisms and neurobiological changes in brain function after a brain injury. Preclinical studies have indicated that peripheral and central sensitization of the trigeminal nociceptive pathways contributes to PTH. While recent brain imaging studies have uncovered widespread changes in brain functional connectivity following trauma, understanding exactly how these networks contribute to PTH after injury remains unknown. Stimulation of peripheral (trigeminal or vagus) nerves show promising efficacies in PTH experimental animals, likely mediated by influencing TBI-induced pathological plasticity by decreasing neuroinflammation and neuronal apoptosis. Non-invasive brain stimulations, such as transcranial magnetic or direct current stimulations, show analgesia for multiple chronic pain conditions, including PTH. Better mechanistic understanding of analgesia achieved by neuromodulations can define peripheral and central mechanisms involved in the development, the resolution, and the management of PTH.
Collapse
Affiliation(s)
- Fernando da Silva Fiorin
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland Baltimore, Program in Neuroscience, Center to Advance Chronic Pain Research, Baltimore, MD, USA
| | - Caroline Cunha do Espírito Santo
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Brazil
| | - Joyce T. Da Silva
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland Baltimore, Program in Neuroscience, Center to Advance Chronic Pain Research, Baltimore, MD, USA
| | - Man-Kyo Chung
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland Baltimore, Program in Neuroscience, Center to Advance Chronic Pain Research, Baltimore, MD, USA
| |
Collapse
|
4
|
Ziesel D, Nowakowska M, Scheruebel S, Kornmueller K, Schäfer U, Schindl R, Baumgartner C, Üçal M, Rienmüller T. Electrical stimulation methods and protocols for the treatment of traumatic brain injury: a critical review of preclinical research. J Neuroeng Rehabil 2023; 20:51. [PMID: 37098582 PMCID: PMC10131365 DOI: 10.1186/s12984-023-01159-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/13/2023] [Indexed: 04/27/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a leading cause of disabilities resulting from cognitive and neurological deficits, as well as psychological disorders. Only recently, preclinical research on electrical stimulation methods as a potential treatment of TBI sequelae has gained more traction. However, the underlying mechanisms of the anticipated improvements induced by these methods are still not fully understood. It remains unclear in which stage after TBI they are best applied to optimize the therapeutic outcome, preferably with persisting effects. Studies with animal models address these questions and investigate beneficial long- and short-term changes mediated by these novel modalities. METHODS In this review, we present the state-of-the-art in preclinical research on electrical stimulation methods used to treat TBI sequelae. We analyze publications on the most commonly used electrical stimulation methods, namely transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), deep brain stimulation (DBS) and vagus nerve stimulation (VNS), that aim to treat disabilities caused by TBI. We discuss applied stimulation parameters, such as the amplitude, frequency, and length of stimulation, as well as stimulation time frames, specifically the onset of stimulation, how often stimulation sessions were repeated and the total length of the treatment. These parameters are then analyzed in the context of injury severity, the disability under investigation and the stimulated location, and the resulting therapeutic effects are compared. We provide a comprehensive and critical review and discuss directions for future research. RESULTS AND CONCLUSION: We find that the parameters used in studies on each of these stimulation methods vary widely, making it difficult to draw direct comparisons between stimulation protocols and therapeutic outcome. Persisting beneficial effects and adverse consequences of electrical simulation are rarely investigated, leaving many questions about their suitability for clinical applications. Nevertheless, we conclude that the stimulation methods discussed here show promising results that could be further supported by additional research in this field.
Collapse
Affiliation(s)
- D Ziesel
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Graz, Austria
| | - M Nowakowska
- Research Unit of Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Graz, Austria
| | - S Scheruebel
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics Division, Medical University of Graz, Graz, Austria
| | - K Kornmueller
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics Division, Medical University of Graz, Graz, Austria
| | - U Schäfer
- Research Unit of Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - R Schindl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics Division, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - C Baumgartner
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - M Üçal
- Research Unit of Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - T Rienmüller
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
5
|
Surendrakumar S, Rabelo TK, Campos ACP, Mollica A, Abrahao A, Lipsman N, Burke MJ, Hamani C. Neuromodulation Therapies in Pre-Clinical Models of Traumatic Brain Injury: Systematic Review and Translational Applications. J Neurotrauma 2023; 40:435-448. [PMID: 35983592 DOI: 10.1089/neu.2022.0286] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) has been associated with several lasting impairments that affect quality of life. Pre-clinical models of TBI have been studied to further our understanding of the underlying short-term and long-term symptomatology. Neuromodulation techniques have become of great interest in recent years as potential rehabilitative therapies after injury because of their capacity to alter neuronal activity and neural circuits in targeted brain regions. This systematic review aims to provide an overlook of the behavioral and neurochemical effects of transcranial direct current stimulation (tDCS), transcranial magnetic stimulation (TMS), deep brain stimulation (DBS), and vagus nerve stimulation (VNS) in pre-clinical TBI models. After screening 629 abstracts, 30 articles were pooled for review. These studies showed that tDCS, TMS, DBS, or VNS delivered to rodents restored TBI-induced deficits in coordination, balance, locomotor activity and improved cognitive impairments in memory, learning, and impulsivity. Potential mechanisms for these effects included neuroprotection, a decrease in apoptosis, neuroplasticity, and the restoration of neural circuit abnormalities. The translational value, potential applicability, and the interpretation of these findings in light of outcome data from clinical trials in patients with TBI are discussed.
Collapse
Affiliation(s)
- Shanan Surendrakumar
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Thallita Kelly Rabelo
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Ana Carolina P Campos
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Adriano Mollica
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Neuropsychiatry Program, Department of Psychiatry, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Agessandro Abrahao
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Hurvitz Brain Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Nir Lipsman
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Hurvitz Brain Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Matthew J Burke
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Hurvitz Brain Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Neuropsychiatry Program, Department of Psychiatry, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Clement Hamani
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Hurvitz Brain Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Arsava EM, Topcuoglu MA, Ay I, Ozdemir AO, Gungor IL, Togay Isikay C, Nazliel B, Kozak HH, Ozturk S, Yilmaz İA, Dora B, Ay H, Ozel T, Sorgun MH, Bahadır EA, Peker E, Aykac O, Mehdiyev Z, Caglayan HZB, Gurses AA, Pektezel MY, Yilmaz E, Oge DD, Parlak S, Bugrul A, Ozguner H, Seker AC, Ozdemir G, Ongun G, Yilmaz SE, Ozguncu C, Turan Isik SM, Ildiz OF, Mammadli A, Yildogan AT, McClure C. Assessment of safety and feasibility of non-invasive vagus nerve stimulation for treatment of acute stroke. Brain Stimul 2022; 15:1467-1474. [PMID: 36356829 DOI: 10.1016/j.brs.2022.10.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 10/16/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Non-invasive vagus nerve stimulation (nVNS) using a hand-held stimulator placed on the neck is an FDA-approved treatment for primary headache disorders. The safety of nVNS is unknown in stroke patients. OBJECTIVE To assess the safety and feasibility of nVNS for the acute treatment of stroke. METHODS TR-VENUS (clinicaltrials.gov identifier NCT03733431) was a randomized, sham-controlled, open-label, multicenter trial conducted in patients with acute ischemic stroke (IS) or intracerebral hemorrhage (ICH). Patients were randomly assigned to standard-dose nVNS, high-dose nVNS, or sham stimulation. The primary endpoint was a composite safety outcome defined as bradycardia or reduction in mean arterial blood pressure during treatment or progression of neurological or death within 24 h of treatment. The feasibility endpoints were the proportion of eligible subjects receiving nVNS within 6 h of symptom onset and the proportion completing all pre-specified treatment doses. Efficacy assessments included infarct growth from baseline to 24 h after treatment. RESULTS Sixty-nine patients (61 IS, 8 ICH) completed the study. The composite safety outcome was achieved in 32.0% in sham and 47.7% in nVNS group (p = 0.203). Treatment was initiated in all but two randomized patients. All dosed subjects received 100% of prespecified stimulations. A non-significant reduction in infarct growth was observed in the high-dose nVNS group (184.2% in sham vs. 63.3% in high-dose nVNS; p = 0.109). CONCLUSIONS The results of this study suggest that nVNS may be safe and feasible in the setting of acute stroke. These findings support further development of nVNS as a potential treatment for acute ischemic stroke.
Collapse
Affiliation(s)
| | | | - Ilknur Ay
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Bijen Nazliel
- Department of Neurology, Gazi University, Ankara, Turkey
| | | | | | | | - Babur Dora
- Department of Neurology, Akdeniz University, Antalya, Turkey
| | - Hakan Ay
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Zhang H, Li CL, Qu Y, Yang YX, Du J, Zhao Y. Effects and neuroprotective mechanisms of vagus nerve stimulation on cognitive impairment with traumatic brain injury in animal studies: A systematic review and meta-analysis. Front Neurol 2022; 13:963334. [PMID: 36237612 PMCID: PMC9551312 DOI: 10.3389/fneur.2022.963334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/29/2022] [Indexed: 12/09/2022] Open
Abstract
Introduction Cognitive impairment is the main clinical feature after traumatic brain injury (TBI) and is usually characterized by attention deficits, memory loss, and decreased executive function. Vagus nerve stimulation (VNS) has been reported to show potential improvement in the cognition level after traumatic brain injury in clinical and preclinical studies. However, this topic has not yet been systematically reviewed in published literature. In this study, we present a systematic review and meta-analysis of the effects of VNS on cognitive function in animal models of TBI and their underlying mechanisms. Methods We performed a literature search on PubMed, PsycINFO, Web of Science, Embase, Scopus, and Cochrane Library from inception to December 2021 to identify studies describing the effects of VNS on animal models of TBI. Results Overall, nine studies were identified in animal models (36 mice, 268 rats, and 27 rabbits). An analysis of these studies showed that VNS can improve the performance of TBI animals in behavioral tests (beam walk test: SMD: 4.95; 95% confidence interval [CI]: 3.66, 6.23; p < 0.00001) and locomotor placing tests (SMD: -2.39; 95% CI: -4.07, -0.71; p = 0.005), whereas it reduced brain edema (SMD: -1.58; 95% CI: -2.85, -0.31; p = 0. 01) and decrease TNF-α (SMD: -3.49; 95% CI: -5.78, -1.2; p = 0.003) and IL-1β (SMD: -2.84; 95% CI: -3.96, -1.71; p < 0.00001) expression level in the brain tissue. However, the checklist for SYRCLE showed a moderate risk of bias (quality score between 30% and 60%), mainly because of the lack of sample size calculation, random assignment, and blinded assessment. Conclusion The present review showed that VNS can effectively promote cognitive impairment and neuropathology in animal models of TBI. We hope that the results of this systematic review can be applied to improve the methodological quality of animal experiments on TBI, which will provide more important and conclusive evidence on the clinical value of VNS. To further confirm these results, there is a need for high-quality TBI animal studies with sufficient sample size and a more comprehensive outcome evaluation. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021290797, identifier: CRD42021290797.
Collapse
Affiliation(s)
- Han Zhang
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, China
- Department of Rehabilitation Medicine, Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, China
- College of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, China
- Sichuan Provincial Key Laboratory of Rehabilitation Medicine, Sichuan University, Chengdu, China
| | - Chun-liu Li
- Department of Rehabilitation Medicine, Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, China
| | - Yun Qu
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, China
- College of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, China
- Sichuan Provincial Key Laboratory of Rehabilitation Medicine, Sichuan University, Chengdu, China
| | - Yu-xuan Yang
- Department of Rehabilitation Medicine, Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, China
| | - Juan Du
- Department of Rehabilitation Medicine, Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, China
| | - Yu Zhao
- Department of Rehabilitation Medicine, Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, China
| |
Collapse
|
8
|
Hajtovic S, LoPresti MA, Zhang L, Katlowitz KA, Kizek DJ, Lam S. The role of vagus nerve stimulation in genetic etiologies of drug-resistant epilepsy: a meta-analysis. J Neurosurg Pediatr 2022:1-14. [PMID: 35303699 DOI: 10.3171/2022.1.peds222] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/31/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Drug-resistant epilepsy (DRE) affects many children. Vagus nerve stimulation (VNS) may improve seizure control; however, its role in children with genetic etiologies of epilepsy is not well described. The authors systematically reviewed the literature to examine the effectiveness of VNS in this cohort. METHODS In January 2021, the authors performed a systematic review of the PubMed/MEDLINE, SCOPUS/Embase, Cochrane, and Web of Science databases to investigate the impact of VNS on seizure outcomes in children with genetic etiologies of epilepsy. Primary outcomes included seizure freedom rate, ≥ 90% seizure reduction rate, and ≥ 50% seizure reduction rate. Secondary outcomes were seizure severity and quality of life (QOL), including cognitive, functional, and behavioral outcomes. A random-effects meta-analysis was performed. RESULTS The authors identified 125 articles, of which 47 with 216 nonduplicate patients were analyzed. Common diagnoses were Dravet syndrome (DS) (92/216 patients [42.6%]) and tuberous sclerosis complex (TSC) (63/216 [29.2%]). Seizure freedom was not reported in any patient with DS; the pooled proportion (95% CI) of patients with ≥ 50% seizure reduction was 41% (21%-58%). Secondary cognitive outcomes of VNS were variable in DS patients, but these patients demonstrated benefits in seizure duration and status epilepticus. In TSC patients, the pooled (95% CI) seizure freedom rate was 40% (12%-71%), ≥ 90% seizure reduction rate was 31% (8%-56%), and ≥ 50% reduction rate was 68% (48%-91%). Regarding the secondary outcomes of VNS in TSC patients, several studies reported decreased seizure severity and improved QOL outcomes. There was limited evidence regarding the use of VNS to treat patients with other genetic etiologies of epilepsy, such as mitochondrial disease, Rett syndrome, Doose syndrome, Landau-Kleffner syndrome, Aicardi syndrome, Angelman syndrome, ring chromosome 20 syndrome, and lissencephaly; variable responses were reported in a limited number of cases. CONCLUSIONS The authors conducted a systematic review of VNS outcomes in children with genetic etiologies of DRE. Among the most studied conditions, patients with TSC had substantial seizure reduction and improvements in QOL, whereas those with DS had less robust seizure reduction. Increased testing, diagnosis, and long-term follow-up studies are necessary to better characterize VNS response in these children.
Collapse
Affiliation(s)
- Sabastian Hajtovic
- 1Sophie Davis Biomedical Education Program, City College of New York, City University of New York School of Medicine, New York, New York
| | - Melissa A LoPresti
- 2Department of Neurosurgery, Division of Pediatric Neurosurgery, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas; and
| | - Lu Zhang
- 3Department of Neurosurgery, Division of Pediatric Neurosurgery, Northwestern University, Lurie Children's Hospital, Chicago, Illinois
| | - Kalman A Katlowitz
- 2Department of Neurosurgery, Division of Pediatric Neurosurgery, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas; and
| | - Dominic J Kizek
- 2Department of Neurosurgery, Division of Pediatric Neurosurgery, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas; and
| | - Sandi Lam
- 3Department of Neurosurgery, Division of Pediatric Neurosurgery, Northwestern University, Lurie Children's Hospital, Chicago, Illinois
| |
Collapse
|
9
|
Potential roles of vagus nerve stimulation on traumatic brain injury: Evidence from in vivo and clinical studies. Exp Neurol 2021; 347:113887. [PMID: 34624329 DOI: 10.1016/j.expneurol.2021.113887] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/23/2021] [Accepted: 10/02/2021] [Indexed: 01/08/2023]
Abstract
Traumatic Brain Injury (TBI) is a one of the leading causes of death and disability worldwide. The consequences of TBI can be divided into two stages: 1) the immediate neuronal destruction during the initial trauma, resulting in the primary brain injury and pathophysiologic sequelae, and 2) the secondary brain injury, encompassing mitochondrial dysfunction, inflammation, cellular excitotoxicity, oxidative stress, and cortical edema, resulting in increased intracranial pressure (ICP) with exacerbated brain damage. Although the pathophysiology in TBI has been thoroughly investigated, the effectivity of therapeutic approaches for TBI is still lacking. Vagus nerve stimulation (VNS) has been used for treating medical refractory epilepsy and chronic drug-resistant depression. Several previous studies also demonstrated that VNS has beneficial effects for TBI in animal models and patients. The neuroprotective effects of VNS on TBI are possibly explained through several mechanisms, including a noradrenergic mechanism, anti-inflammatory effects, regulation of neurotransmitters, and attenuation of blood brain barrier breakdown, and brain edema. The aims of this review are to summarize and discuss the current evidence pertinent to the effect of VNS on both primary and secondary brain injury following TBI from both in vivo and clinical studies.
Collapse
|
10
|
Li H, Ni J, Qing H. Gut Microbiota: Critical Controller and Intervention Target in Brain Aging and Cognitive Impairment. Front Aging Neurosci 2021; 13:671142. [PMID: 34248602 PMCID: PMC8267942 DOI: 10.3389/fnagi.2021.671142] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/07/2021] [Indexed: 12/12/2022] Open
Abstract
The current trend for the rapid growth of the global aging population poses substantial challenges for society. The human aging process has been demonstrated to be closely associated with changes in gut microbiota composition, diversity, and functional features. During the first 2 years of life, the gut microbiota undergoes dramatic changes in composition and metabolic functions as it colonizes and develops in the body. Although the gut microbiota is nearly established by the age of three, it continues to mature until adulthood, when it comprises more stable and diverse microbial species. Meanwhile, as the physiological functions of the human body deteriorated with age, which may be a result of immunosenescence and "inflammaging," the guts of elderly people are generally characterized by an enrichment of pro-inflammatory microbes and a reduced abundance of beneficial species. The gut microbiota affects the development of the brain through a bidirectional communication system, called the brain-gut-microbiota (BGM) axis, and dysregulation of this communication is pivotal in aging-related cognitive impairment. Microbiota-targeted dietary interventions and the intake of probiotics/prebiotics can increase the abundance of beneficial species, boost host immunity, and prevent gut-related diseases. This review summarizes the age-related changes in the human gut microbiota based on recent research developments. Understanding these changes will likely facilitate the design of novel therapeutic strategies to achieve healthy aging.
Collapse
Affiliation(s)
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
11
|
Transcutaneous vagus nerve stimulation in patients with attention-deficit/hyperactivity disorder: A viable option? PROGRESS IN BRAIN RESEARCH 2021; 264:171-190. [PMID: 34167655 DOI: 10.1016/bs.pbr.2021.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Individuals with attention-deficit/hyperactivity disorder (ADHD) suffer from a range of cognitive and behavioral problems that severely impair their educational and occupational attainment. ADHD symptoms have been linked to structural and functional changes within and between different brain regions, particularly the prefrontal cortex. At the system level, reduced availability of the neurotransmitters dopamine (DA) and norepinephrine (NE) but also γ-aminobutyric acid (GABA) have been repeatedly demonstrated. Recently, non-invasive brain stimulation (NIBS) techniques have been explored as treatment alternatives to alter dysfunctional activation patterns in specified brain areas or networks. In the current paper, we introduce transcutaneous vagus nerve stimulation (tVNS) as a systemic approach to directly affect NE and GABA neurotransmission. TVNS is a non-drug intervention with low risk and proven efficacy in improving cognitive particularly executive functions. It is easy to apply and therefore well-suited to provide home-based or mobile treatment options allowing a significant increase in treatment intensity and providing easier access to medical care for individuals who are unable to regularly visit a clinician. We describe in detail the underlying mechanisms of tVNS and current fields of application and discuss its potential as an adjuvant treatment for ADHD.
Collapse
|
12
|
Sackeim HA. Staging and Combining Brain Stimulation Interventions: Vagus Nerve Stimulation and Electroconvulsive Therapy. J ECT 2021; 37:80-83. [PMID: 34029304 DOI: 10.1097/yct.0000000000000745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Harold A Sackeim
- From the Departments of Psychiatry and Radiology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| |
Collapse
|
13
|
Wang Y, Zhan G, Cai Z, Jiao B, Zhao Y, Li S, Luo A. Vagus nerve stimulation in brain diseases: Therapeutic applications and biological mechanisms. Neurosci Biobehav Rev 2021; 127:37-53. [PMID: 33894241 DOI: 10.1016/j.neubiorev.2021.04.018] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 04/12/2021] [Accepted: 04/18/2021] [Indexed: 12/21/2022]
Abstract
Brain diseases, including neurodegenerative, cerebrovascular and neuropsychiatric diseases, have posed a deleterious threat to human health and brought a great burden to society and the healthcare system. With the development of medical technology, vagus nerve stimulation (VNS) has been approved by the Food and Drug Administration (FDA) as an alternative treatment for refractory epilepsy, refractory depression, cluster headaches, and migraines. Furthermore, current evidence showed promising results towards the treatment of more brain diseases, such as Parkinson's disease (PD), autistic spectrum disorder (ASD), traumatic brain injury (TBI), and stroke. Nonetheless, the biological mechanisms underlying the beneficial effects of VNS in brain diseases remain only partially elucidated. This review aims to delve into the relevant preclinical and clinical studies and update the progress of VNS applications and its potential mechanisms underlying the biological effects in brain diseases.
Collapse
Affiliation(s)
- Yue Wang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Gaofeng Zhan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ziwen Cai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Bo Jiao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yilin Zhao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Shiyong Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ailin Luo
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
14
|
Tang Y, Dong X, Chen G, Ye W, Kang J, Tang Y, Feng Z. Vagus Nerve Stimulation Attenuates Early Traumatic Brain Injury by Regulating the NF-κB/NLRP3 Signaling Pathway. Neurorehabil Neural Repair 2020; 34:831-843. [PMID: 32772884 DOI: 10.1177/1545968320948065] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Traumatic brain injury (TBI) is a major cause of death and disability worldwide. Oxidative stress, inflammation, and apoptosis are vital pathophysiological features post-TBI. OBJECTIVES Research has shown that vagus nerve stimulation (VNS) can attenuate oxidative stress in various diseases. However, the critical role of VNS in TBI is still not completely understood. This study investigated the protective effects and potential mechanism of VNS on TBI. METHODS Male Sprague-Dawley rats were randomized into 3 groups: sham, TBI, and TBI + VNS. The TBI model was induced in rats by the free-fall drop method. The vagal nerve trunk was separated, and VNS was performed after establishing the TBI model. RESULTS The results showed that VNS significantly ameliorated tissue damage, neurological deficits, and cerebral edema, compared with the sham VNS group. Additionally, VNS alleviated oxidative stress, inflammation, and apoptosis in the pericontusive cortex of rats after TBI. VNS also significantly suppressed expression of the nuclear factor-κB (NF-κB) protein in the nucleus and activation of the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome. CONCLUSIONS Taken together, the present study indicates that VNS may attenuate brain damage after TBI by inhibiting oxidative stress, inflammation, and apoptosis, possibly through the NF-κB/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Yunliang Tang
- Department of Rehabilitation Medicine, First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Xiaoyang Dong
- Department of Rehabilitation Medicine, First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Gengfa Chen
- Department of Rehabilitation Medicine, First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Wen Ye
- Department of Rehabilitation Medicine, First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Junwei Kang
- Department of Rehabilitation Medicine, First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Yang Tang
- Department of Rehabilitation Medicine, First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Zhen Feng
- Department of Rehabilitation Medicine, First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
15
|
Rosso P, Iannitelli A, Pacitti F, Quartini A, Fico E, Fiore M, Greco A, Ralli M, Tirassa P. Vagus nerve stimulation and Neurotrophins: a biological psychiatric perspective. Neurosci Biobehav Rev 2020; 113:338-353. [PMID: 32278791 DOI: 10.1016/j.neubiorev.2020.03.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 02/06/2023]
Abstract
Since 2004, vagus nerve stimulation (VNS) has been used in treatment-resistant or treatment-intolerant depressive episodes. Today, VNS is suggested as possible therapy for a larger spectrum of psychiatric disorders, including schizophrenia, obsessive compulsive disorders, and panic disorders. Despite a large body of literature supports the application of VNS in patients' treatment, the exact mechanism of action of VNS remains not fully understood. In the present study, the major knowledges on the brain areas and neuronal pathways regulating neuroimmune and autonomic response subserving VNS effects are reviewed. Furthermore, the involvement of the neurotrophins (NTs) Nerve Growth Factor (NGF) and Brain Derived Neurotrophic Factor (BDNF) in vagus nerve (VN) physiology and stimulation is revised. The data on brain NGF/BDNF synthesis and in turn on the activity-dependent plasticity, connectivity rearrangement and neurogenesis, are presented and discussed as potential biomarkers for optimizing stimulatory parameters for VNS. A vagus nerve-neurotrophin interaction model in the brain is finally proposed as a working hypothesis for future studies addressed to understand pathophysiology of psychiatric disturbance.
Collapse
Affiliation(s)
- Pamela Rosso
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy
| | - Angela Iannitelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Francesca Pacitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy; Psychiatry Unit San Salvatore Hospital, L'Aquila, Italy
| | - Adele Quartini
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Elena Fico
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy
| | - Marco Fiore
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy
| | - Antonio Greco
- Department of Sense Organs, Sapienza University of Rome, Italy
| | - Massimo Ralli
- Department of Sense Organs, Sapienza University of Rome, Italy
| | - Paola Tirassa
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy.
| |
Collapse
|
16
|
Colzato L, Beste C. A literature review on the neurophysiological underpinnings and cognitive effects of transcutaneous vagus nerve stimulation: challenges and future directions. J Neurophysiol 2020; 123:1739-1755. [PMID: 32208895 DOI: 10.1152/jn.00057.2020] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Brain stimulation approaches are important to gain causal mechanistic insights into the relevance of functional brain regions and/or neurophysiological systems for human cognitive functions. In recent years, transcutaneous vagus nerve stimulation (tVNS) has attracted considerable popularity. It is a noninvasive brain stimulation technique based on the stimulation of the vagus nerve. The stimulation of this nerve activates subcortical nuclei, such as the locus coeruleus and the nucleus of the solitary tract, and from there, the activation propagates to the cortex. Since tVNS is a novel stimulation technique, this literature review outlines a brief historical background of tVNS, before detailing underlying neurophysiological mechanisms of action, stimulation parameters, cognitive effects of tVNS on healthy humans, and, lastly, current challenges and future directions of tVNS research in cognitive functions. Although more research is needed, we conclude that tVNS, by increasing norepineprine (NE) and gamma-aminobutyric acid (GABA) levels, affects NE- and GABA-related cognitive performance. The review provides detailed background information how to use tVNS as a neuromodulatory tool in cognitive neuroscience and outlines important future leads of research on tVNS.
Collapse
Affiliation(s)
- Lorenza Colzato
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany.,Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany.,Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan, China
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany.,Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan, China
| |
Collapse
|
17
|
Sun J, Yang X, Zhang Y, Zhang W, Lu J, Hu Q, Liu R, Zhou C, Chen C. Salvinorin A attenuates early brain injury through PI3K/Akt pathway after subarachnoid hemorrhage in rat. Brain Res 2019; 1719:64-70. [PMID: 31125530 DOI: 10.1016/j.brainres.2019.05.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 02/01/2023]
Abstract
Early brain injury (EBI) refers to the direct injury to the brain during the first 72 h after subarachnoid hemorrhage (SAH), which is one of the major causes for the poor clinical outcome after SAH. In this study, we investigated the effect and the related mechanism of Salvinorin A (SA), a selective kappa opioid receptor agonist, on EBI after SAH. SA was administered by intraperitoneal injection at 24 h, 48 h and 72 h after SAH. The volume of lateral ventricle was measured by magnetic resonance imaging (MRI). The neuronal morphological changes and the apoptotic level in CA1 area of hippocampus were observed by Nissl and TUNEL staining respectively. Protein expression of p-PI3K, p-Akt, p-IKKα/β, p-NF-κB, FoxO1, Bim, Bax and Cleaved-caspase-3 was measured to explore the potential mechanism. We found that SA alleviated the neuronal morphological changes and apoptosis in CA1 area of hippocampus. The mechanism might be related to the increased protein expression of p-PI3K/p-Akt, which accompanied by decreased expression of p-IKKα/β, p-NF-κB, FoxO1, Bim, Bax and Cleaved-caspase-3 in the hippocampus. Thus, therapeutic interventions of SA targeting the PI3K/Akt pathway might be a novel approach to ameliorate EBI via reducing the apoptosis and inflammation after SAH.
Collapse
Affiliation(s)
- Juan Sun
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Department of Neurology, Affiliated Hospital of Qinghai University, China
| | - Xiaomei Yang
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yan Zhang
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Weiguang Zhang
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jianfei Lu
- Discipline of Neuroscience, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, China
| | - Qin Hu
- Discipline of Neuroscience, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, China
| | - Renyu Liu
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Changman Zhou
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Chunhua Chen
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
18
|
Yakunina N, Kim SS, Nam EC. BOLD fMRI effects of transcutaneous vagus nerve stimulation in patients with chronic tinnitus. PLoS One 2018; 13:e0207281. [PMID: 30485375 PMCID: PMC6261575 DOI: 10.1371/journal.pone.0207281] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 10/29/2018] [Indexed: 12/25/2022] Open
Abstract
Objective Vagus nerve stimulation (VNS) is a neuromodulation method used for treatment of epilepsy and depression. Transcutaneous VNS (tVNS) has been gaining popularity as a noninvasive alternative to VNS. Previous tVNS neuroimaging studies revealed brain (de)activation patterns that involved multiple areas implicated in tinnitus generation and perception. In this study, functional magnetic resonance imaging (fMRI) was used to explore the effects of tVNS on brain activity in patients with tinnitus. Methods Thirty-six patients with chronic tinnitus received tVNS to the inner tragus, cymba conchae, and earlobe (sham stimulation). Results The locus coeruleus and nucleus of the solitary tract in the brainstem were activated in response to stimulation of both locations compared with the sham stimulation. The cochlear nuclei were also activated, which was not observed in healthy subjects with normal hearing. Multiple auditory and limbic structures, as well as other brain areas associated with generation and perception of tinnitus, were deactivated by tVNS, particularly the parahippocampal gyrus, which was recently speculated to cause tinnitus in hearing-impaired patients. Conclusions tVNS via the inner tragus or cymba conchae suppressed neural activity in the auditory, limbic, and other tinnitus-related non-auditory areas through auditory and vagal ascending pathways in tinnitus patients. The results from this study are discussed in the context of several existing models of tinnitus. They indicate that the mechanism of action of tVNS might be involved in multiple brain areas responsible for the generation of tinnitus, tinnitus-related emotional annoyance, and their mutual reinforcement.
Collapse
Affiliation(s)
- Natalia Yakunina
- Institute of Medical Science, Kangwon National University, School of Medicine, Chuncheon, Republic of Korea
- Neuroscience Research Institute, Kangwon National University Hospital, Chuncheon, Republic of Korea
| | - Sam Soo Kim
- Neuroscience Research Institute, Kangwon National University Hospital, Chuncheon, Republic of Korea
- Department of Radiology, Kangwon National University, School of Medicine, Chuncheon, Republic of Korea
| | - Eui-Cheol Nam
- Neuroscience Research Institute, Kangwon National University Hospital, Chuncheon, Republic of Korea
- Department of Otolaryngology, Kangwon National University, School of Medicine, Chuncheon, Republic of Korea
- * E-mail:
| |
Collapse
|
19
|
Yang Y, Yang LY, Orban L, Cuylear D, Thompson J, Simon B, Yang Y. Non-invasive vagus nerve stimulation reduces blood-brain barrier disruption in a rat model of ischemic stroke. Brain Stimul 2018; 11:689-698. [PMID: 29496430 PMCID: PMC6019567 DOI: 10.1016/j.brs.2018.01.034] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 12/19/2017] [Accepted: 01/31/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Vagus nerve stimulation (VNS) significantly reduces infarct volume in rat models of cerebral ischemia, but the mechanism of this protective effect remains open. HYPOTHESIS This study tested the hypothesis that non-invasive VNS (nVNS), during transient middle cerebral artery occlusion (MCAO), protects the blood-brain barrier (BBB), leading to reduced infarct size in ischemic brain. METHODS Spontaneous hypertensive rats (SHRs) were subjected to a 90 min MCAO. nVNS treated rats received 5 stimulations (duration: 2 min; every 10 min) on the skin overlying the cervical vagus nerve in the neck beginning 30 min after MCAO onset. Control rats received the same stimulations on the quadriceps femoris muscle. Twenty-four hours after MCAO onset, MRI and immunohistochemistry (IHC) were performed for analyses of infarct size and BBB leakage. RESULTS Compared with the control group, anatomic MRI T2-weighted images showed significantly smaller infarct sizes in the nVNS group. Dynamic contrast-enhanced (DCE)-MRI showed a significantly decreased BBB transfer rate (Ki map) in the lesion area in the nVNS group, which was spatially correlated with the attenuation of the infarct size. Furthermore, significantly lower serum IgG leakage, visualized by IHC, was seen in the ischemic hemisphere in nVNS treated rats. nVNS also protected vascular tight junction proteins from disruption in microvessels, and reduced expression of matrix metalloproteinases-2/9 in reactive astrocytes surrounding the compromised vessels in the ischemic hemispheres. CONCLUSION Our data suggest that the neuroprotective role of a series of nVNS administrations during MCA occlusion, spatially correlates with protection of BBB integrity from damage and reduction of infarct extent induced by ischemic stroke.
Collapse
Affiliation(s)
- Yirong Yang
- College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Lisa Y Yang
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Lilla Orban
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Darnell Cuylear
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Jeffrey Thompson
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Bruce Simon
- ElectroCore LLC, Basking Ridge, NJ 07920, USA
| | - Yi Yang
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
20
|
Vagus Nerve Stimulation and Other Neuromodulation Methods for Treatment of Traumatic Brain Injury. Neurocrit Care 2017; 24:308-19. [PMID: 26399249 DOI: 10.1007/s12028-015-0203-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The objective of this paper is to review the current literature regarding the use of vagus nerve stimulation (VNS) in preclinical models of traumatic brain injury (TBI) as well as discuss the potential role of VNS along with alternative neuromodulation approaches in the treatment of human TBI. Data from previous studies have demonstrated VNS-mediated improvement following TBI in animal models. In these cases, VNS was observed to enhance motor and cognitive recovery, attenuate cerebral edema and inflammation, reduce blood brain barrier breakdown, and confer neuroprotective effects. Yet, the underlying mechanisms by which VNS enhances recovery following TBI remain to be fully elucidated. Several hypotheses have been offered including: a noradrenergic mechanism, reduction in post-TBI seizures and hyper-excitability, anti-inflammatory effects, attenuation of blood-brain barrier breakdown, and cerebral edema. We present other potential mechanisms by which VNS acts including enhancement of synaptic plasticity and recruitment of endogenous neural stem cells, stabilization of intracranial pressure, and interaction with the ghrelin system. In addition, alternative methods for the treatment of TBI including deep brain stimulation, transcranial magnetic stimulation, transcranial direct current stimulation, and focused ultrasound stimulation are discussed. Although the primary source data show that VNS improves TBI outcomes, it remains to be determined if these findings can be translated to clinical settings.
Collapse
|
21
|
Chen W, Meng FG. Ictal heart rate changes and the effects of vagus nerve stimulation for patients with refractory epilepsy. Neuropsychiatr Dis Treat 2017; 13:2351-2356. [PMID: 28919768 PMCID: PMC5592906 DOI: 10.2147/ndt.s142714] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Vagus nerve stimulation (VNS) shows long-term efficiency worldwide in most pharmacoresistant patients with epilepsy; however, there are still a small number of patients who are non-responders to VNS therapy. It has been shown that VNS treatment outcomes for drug-resistant epilepsy may be predicted by preoperative heart-rate variability measurements and that patients with epilepsy with ictal tachycardia (IT) during seizures have good responses to VNS. However, few studies have reported the efficacy of VNS in patients with epilepsy with ictal bradycardia (IB) or normal heart rate (HR), and none have explored the possible mechanisms of VNS efficacy based on different HR types. HR during seizures varies, and we presume that different HRs during seizures may impact the effects of VNS. It has been shown that blood pressure in the human body needs to be maintained through the arterial baroreflex (ABR). VNS efficacy in patients with epilepsy with IT, IB, and normal HR during seizures may be related to ABR. Mechanical signals generated by VNS are similar to the autonomic nerve pathways and, thus, we propose the hypothesis that different HRs during seizures can predict VNS efficacy in patients. If VNS is highly efficient in patients with IT during seizures, VNS in patients with a normal HR during seizures may be less efficient, and may even be inefficient in patients with IB during seizures.
Collapse
Affiliation(s)
- Wei Chen
- Department of Neurology, Liaocheng People's Hospital, Liaocheng
| | - Fan-Gang Meng
- Beijing Neurosurgical Institute, Capital Medical University.,Beijing Key Laboratory of Neuromodulation, Beijing Municipal Science and Technology Commission, Beijing, People's Republic of China
| |
Collapse
|
22
|
Activation of Alpha 7 Cholinergic Nicotinic Receptors Reduce Blood-Brain Barrier Permeability following Experimental Traumatic Brain Injury. J Neurosci 2016; 36:2809-18. [PMID: 26937017 DOI: 10.1523/jneurosci.3197-15.2016] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
UNLABELLED Traumatic brain injury (TBI) is a major human health concern that has the greatest impact on young men and women. The breakdown of the blood-brain barrier (BBB) is an important pathological consequence of TBI that initiates secondary processes, including infiltration of inflammatory cells, which can exacerbate brain inflammation and contribute to poor outcome. While the role of inflammation within the injured brain has been examined in some detail, the contribution of peripheral/systemic inflammation to TBI pathophysiology is largely unknown. Recent studies have implicated vagus nerve regulation of splenic cholinergic nicotinic acetylcholine receptor α7 (nAChRa7) signaling in the regulation of systemic inflammation. However, it is not known whether this mechanism plays a role in TBI-triggered inflammation and BBB breakdown. Following TBI, we observed that plasma TNF-α and IL-1β levels, as well as BBB permeability, were significantly increased in nAChRa7 null mice (Chrna7(-/-)) relative to wild-type mice. The administration of exogenous IL-1β and TNF-α to brain-injured animals worsened Evans Blue dye extravasation, suggesting that systemic inflammation contributes to TBI-triggered BBB permeability. Systemic administration of the nAChRa7 agonist PNU-282987 or the positive allosteric modulator PNU-120596 significantly attenuated TBI-triggered BBB compromise. Supporting a role for splenic nAChRa7 receptors, we demonstrate that splenic injection of the nicotinic receptor blocker α-bungarotoxin increased BBB permeability in brain-injured rats, while PNU-282987 injection decreased such permeability. These effects were not seen when α-bungarotoxin or PNU-282987 were administered to splenectomized, brain-injured rats. Together, these findings support the short-term use of nAChRa7-activating agents as a strategy to reduce TBI-triggered BBB permeability. SIGNIFICANCE STATEMENT Breakdown of the blood-brain barrier (BBB) in response to traumatic brain injury (TBI) allows for the accumulation of circulating fluids and proinflammatory cells in the injured brain. These processes can exacerbate TBI pathology and outcome. While the role of inflammation in the injured tissue has been examined in some detail, the contribution of peripheral inflammation in BBB breakdown and ensuing pathology has not been well defined. We present experimental evidence to indicate that the stimulation of nicotinic acetylcholine α7 receptors (nAChRa7s) can reduce peripheral inflammation and BBB breakdown after TBI. These results suggest that activators of nAChRa7 may have therapeutic utility for the treatment of TBI.
Collapse
|
23
|
Xiang YX, Wang WX, Xue Z, Zhu L, Wang SB, Sun ZH. Electrical stimulation of the vagus nerve protects against cerebral ischemic injury through an anti-infammatory mechanism. Neural Regen Res 2015; 10:576-82. [PMID: 26170817 PMCID: PMC4424749 DOI: 10.4103/1673-5374.155430] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2015] [Indexed: 11/17/2022] Open
Abstract
Vagus nerve stimulation exerts protective effects against ischemic brain injury; however, the underlying mechanisms remain unclear. In this study, a rat model of focal cerebral ischemia was established using the occlusion method, and the right vagus nerve was given electrical stimulation (constant current of 0.5 mA; pulse width, 0.5 ms; frequency, 20 Hz; duration, 30 seconds; every 5 minutes for a total of 60 minutes) 30 minutes, 12 hours, and 1, 2, 3, 7 and 14 days after surgery. Electrical stimulation of the vagus nerve substantially reduced infarct volume, improved neurological function, and decreased the expression levels of tumor necrosis factor-α and interleukin-6 in rats with focal cerebral ischemia. The experimental findings indicate that the neuroprotective effect of vagus nerve stimulation following cerebral ischemia may be associated with the inhibition of tumor necrosis factor-α and interleukin-6 expression.
Collapse
Affiliation(s)
- Yao-Xian Xiang
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Wen-Xin Wang
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Zhe Xue
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Lei Zhu
- Department of Radiology, Beijing Electric Power Hospit, Beijng, China
| | - Sheng-Bao Wang
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Zheng-Hui Sun
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
24
|
Zheng X, Wang W, Liu R, Huang H, Zhang R, Sun L. Effect of p62 on tau hyperphosphorylation in a rat model of Alzheimer's disease. Neural Regen Res 2015; 7:1304-11. [PMID: 25657660 PMCID: PMC4308800 DOI: 10.3969/j.issn.1673-5374.2012.17.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 05/07/2012] [Indexed: 12/31/2022] Open
Abstract
Tau hyperphosphorylation is a main cause of neuronal loss in Alzheimer's disease, which can be caused by many factors, including oxidative stress. The multifunctional protein p62, which exists in neurofibrillary tangles and causes aggregation of hyperphosphorylated tau, not only serves as a receptor in selective autophagy, but also regulates oxidative stress. However, whether p62 participates in oxidative stress-induced tau hyperphosphorylation remains unclear. In this study, we produced an Alzheimer's disease rat model by injecting β-amyloid protein into the hippocampus and β-galactose intraperitoneally. Hematoxylin-eosin staining was used for morphological analysis of brain tissue, and western blotting, immunohistochemistry and reverse transcription-PCR were employed to study p62 and autophagy related proteins, antioxidant defense system kelch-like ECH-associated protein 1-NF-E2-related factor 2 related proteins and hyperphosphorylated tau, respectively. The number of neurons in the brain decreased in Alzheimer's disease rats, and the autophagy related proteins Atg12-Atg5, microtubule-associated protein 1 light chain 3-phosphatidylethanolamine and Beclin1 increased significantly, while p62 expression reduced. Expression of kelch-like ECH-associated protein 1 increased, NF-E2-related factor 2 protein and the downstream gene products of glutamate cysteine ligase catalytic subunit and glutamate cysteine ligase modulatory subunit decreased, and hyperphosphorylated tau increased. These findings demonstrate that autophagy levels increased and p62 levels decreased in the brains of Alzheimer's disease rats. Moreover, the anti-oxidative capability of the NF-E2-related factor 2-antioxidant response element pathway was decreased, which may be the cause of tau hyperphosphorylation in Alzheimer's disease brain tissue and the subsequent structural and functional damage to neurons.
Collapse
Affiliation(s)
- Xianhong Zheng
- Department of Cytobiology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, Jilin Province, China
| | - Weiwei Wang
- Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, Jilin Province, China
| | - Ruizhi Liu
- Department of Cytobiology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, Jilin Province, China
| | - Honglan Huang
- Department of Pathogen Biology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, Jilin Province, China
| | - Rihui Zhang
- Department of Physiology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, Jilin Province, China
| | - Liankun Sun
- Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
25
|
Li Y, Korgaonkar AA, Swietek B, Wang J, Elgammal FS, Elkabes S, Santhakumar V. Toll-like receptor 4 enhancement of non-NMDA synaptic currents increases dentate excitability after brain injury. Neurobiol Dis 2014; 74:240-53. [PMID: 25497689 DOI: 10.1016/j.nbd.2014.11.021] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 11/21/2014] [Accepted: 11/26/2014] [Indexed: 11/18/2022] Open
Abstract
Concussive brain injury results in neuronal degeneration, microglial activation and enhanced excitability in the hippocampal dentate gyrus, increasing the risk for epilepsy and memory dysfunction. Endogenous molecules released during injury can activate innate immune responses including toll-like receptor 4 (TLR4). Recent studies indicate that immune mediators can modulate neuronal excitability. Since non-specific agents that reduce TLR4 signaling can limit post-traumatic neuropathology, we examined whether TLR4 signaling contributes to early changes in dentate excitability after brain injury. Concussive brain injury caused a transient increase in hippocampal TLR4 expression within 4h, which peaked at 24h. Post-injury increase in TLR4 expression in the dentate gyrus was primarily neuronal and persisted for one week. Acute, in vitro treatment with TLR4 ligands caused bidirectional modulation of dentate excitability in control and brain-injured rats, with a reversal in the direction of modulation after brain injury. TLR4 antagonists decreased, and agonist increased, afferent-evoked dentate excitability one week after brain injury. NMDA receptor antagonist did not occlude the ability of LPS-RS, a TLR4 antagonist, to decrease post-traumatic dentate excitability. LPS-RS failed to modulate granule cell NMDA EPSCs but decreased perforant path-evoked non-NMDA EPSC peak amplitude and charge transfer in both granule cells and mossy cells. Our findings indicate an active role for TLR4 signaling in early post-traumatic dentate hyperexcitability. The novel TLR4 modulation of non-NMDA glutamatergic currents, identified herein, could represent a general mechanism by which immune activation influences neuronal excitability in neurological disorders that recruit sterile inflammatory responses.
Collapse
Affiliation(s)
- Ying Li
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Akshata A Korgaonkar
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; Graduate School of Biomedical Sciences, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Bogumila Swietek
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Jianfeng Wang
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Fatima S Elgammal
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Stella Elkabes
- Graduate School of Biomedical Sciences, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Vijayalakshmi Santhakumar
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; Graduate School of Biomedical Sciences, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; Department of Pharmacology and Physiology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA.
| |
Collapse
|
26
|
Zhou L, Lin J, Lin J, Kui G, Zhang J, Yu Y. Neuroprotective effects of vagus nerve stimulation on traumatic brain injury. Neural Regen Res 2014; 9:1585-91. [PMID: 25368644 PMCID: PMC4211199 DOI: 10.4103/1673-5374.141783] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2014] [Indexed: 11/15/2022] Open
Abstract
Previous studies have shown that vagus nerve stimulation can improve the prognosis of traumatic brain injury. The aim of this study was to elucidate the mechanism of the neuroprotective effects of vagus nerve stimulation in rabbits with brain explosive injury. Rabbits with brain explosive injury received continuous stimulation (10 V, 5 Hz, 5 ms, 20 minutes) of the right cervical vagus nerve. Tumor necrosis factor-α, interleukin-1β and interleukin-10 concentrations were detected in serum and brain tissues, and water content in brain tissues was measured. Results showed that vagus nerve stimulation could reduce the degree of brain edema, decrease tumor necrosis factor-α and interleukin-1β concentrations, and increase interleukin-10 concentration after brain explosive injury in rabbits. These data suggest that vagus nerve stimulation may exert neuroprotective effects against explosive injury via regulating the expression of tumor necrosis factor-α, interleukin-1β and interleukin-10 in the serum and brain tissue.
Collapse
Affiliation(s)
- Long Zhou
- Affiliated Dongnan Hospital of Xiamen University, the 175 Hospital of Chinese PLA, Trauma Neurosurgery Center of Nanjing Military Region, Xiamen, Fujian Province, China
| | - Jinhuang Lin
- Affiliated Dongnan Hospital of Xiamen University, the 175 Hospital of Chinese PLA, Trauma Neurosurgery Center of Nanjing Military Region, Xiamen, Fujian Province, China
| | - Junming Lin
- Affiliated Dongnan Hospital of Xiamen University, the 175 Hospital of Chinese PLA, Trauma Neurosurgery Center of Nanjing Military Region, Xiamen, Fujian Province, China
| | - Guoju Kui
- Affiliated Dongnan Hospital of Xiamen University, the 175 Hospital of Chinese PLA, Trauma Neurosurgery Center of Nanjing Military Region, Xiamen, Fujian Province, China
| | - Jianhua Zhang
- The 73131 Corps of Chinese PLA, Xiamen, Fujian Province, China
| | - Yigang Yu
- Affiliated Dongnan Hospital of Xiamen University, the 175 Hospital of Chinese PLA, Trauma Neurosurgery Center of Nanjing Military Region, Xiamen, Fujian Province, China
| |
Collapse
|
27
|
Chen C, Cui H, Li Z, Wang R, Zhou C. Normobaric oxygen for cerebral ischemic injury. Neural Regen Res 2014; 8:2885-94. [PMID: 25206609 PMCID: PMC4146175 DOI: 10.3969/j.issn.1673-5374.2013.31.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 09/16/2013] [Indexed: 12/12/2022] Open
Abstract
Oxygen inhalation has been shown to increase oxygen supply to tissues after cerebral ischemia/ reperfusion injury, protecting injured neural cells. However, hyperbaric oxygen may aggravate oxidative stress. By contrast, normobaric oxygen has the rapid and non-invasive characteristics and may have therapeutic effects on ischemic/hypoxic disease. Rats inhaled normobaric oxygen (95% O2) for 6 consecutive days, and then a rat model of focal cerebral ischemia was established. Nissl and 2,3,5-triphenyltetrazolium chloride (TTC) staining revealed that normobaric oxygen pretreatment improved neurological deficits and reduced infarct volume. Immunohistochemical staining and western blot assay revealed that the expression of hypoxia-inducible factor-1α, Notch-1, vascular endothelial growth factor and erythropoietin were increased. Behavioral studies also verified that neurological deficit scores increased. The hypoxia-inducible factor inhibitor 2-methoxyestradiol treatment at 1 hour before administration of normobaric oxygen could suppress the protective effect of normobaric oxygen. Given these observations, normobaric oxygen pretreatment may alleviate cerebral ischemic injury via the hypoxia-inducible factor signal pathway.
Collapse
Affiliation(s)
- Chunhua Chen
- Department of Anatomy and Histoembryology, Peking University Health Science Center, Beijing 100191, China
| | - Haimeng Cui
- Department of Anatomy and Histoembryology, Peking University Health Science Center, Beijing 100191, China
| | - Zihe Li
- Department of Anatomy and Histoembryology, Peking University Health Science Center, Beijing 100191, China
| | - Ruifeng Wang
- Department of Anatomy and Histoembryology, Peking University Health Science Center, Beijing 100191, China
| | - Changman Zhou
- Department of Anatomy and Histoembryology, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
28
|
Abstract
Over the past 35 years or so, PET brain imaging has allowed powerful and unique insights into brain function under normal conditions and in disease states. Initially, as PET instrumentation continued to develop, studies were focused on brain perfusion and glucose metabolism. This permitted refinement of brain imaging for important, non-oncologic clinical indications. The ability of PET to not only provide spatial localization of metabolic changes but also to accurately and consistently quantify their distribution proved valuable for applications in the clinical setting. Specifically, glucose metabolism brain imaging using (F-18) fluorodeoxyglucose continues to be invaluable for evaluating patients with intractable seizures for identifying seizure foci and operative planning. Cerebral glucose metabolism also contributes to diagnosis of neurodegenerative diseases that cause dementia. Alzheimer disease, dementia with Lewy bodies, and the several variants of frontotemporal lobar degeneration have differing typical patterns of hypometabolism. In Alzheimer disease, hypometabolism has furthermore been associated with poorer cognitive performance and ensuing cognitive and functional decline. As the field of radiochemistry evolved, novel radioligands including radiolabeled flumazenil, dopamine transporter ligands, nicotine receptor ligands, and others have allowed for further understanding of molecular changes in the brain associated with various diseases. Recently, PET brain imaging reached another milestone with the approval of (F-18) florbetapir imaging by the United States Federal Drug Administration for detection of amyloid plaque accumulation in brain, the major histopathologic hallmark of Alzheimer disease, and efforts have been made to define the clinical role of this imaging agent in the setting of the currently limited treatment options. Hopefully, this represents the first of many new radiopharmaceuticals that would allow improved diagnostic and prognostic information in these and other clinical applications, including Parkinson disease and traumatic brain injury.
Collapse
Affiliation(s)
- Ilya Nasrallah
- Division of Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
29
|
Wang W, Kang J, Li H, Su J, Wu J, Xu Y, Yu H, Xiang X, Yi H, Lu Y, Sun L. Regulation of endoplasmic reticulum stress in rat cortex by p62/ZIP through the Keap1-Nrf2-ARE signalling pathway after transient focal cerebral ischaemia. Brain Inj 2013; 27:924-33. [PMID: 23782269 DOI: 10.3109/02699052.2013.793397] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
PRIMARY OBJECTIVE p62/ZIP as the autophagy receptor can transport the misfolded proteins to a macroautophagy-lysosome system for degradation and also create a positive feedback loop between p62/ZIP and Nrf2. However, the role of p62/ZIP on cerebral ischaemia is unclear. The aim of this study was to evaluate the role of p62/ZIP in the regulation of endoplasmic reticulum(ER) stress induced by cerebral ischaemia/reperfusion. RESEARCH DESIGN Different ischemic periods were designed by transient middle cerebral artery occlusion (tMCAO) using the suture method. METHODS AND PROCEDURES At 24 hours after reperfusion, the ischaemic brain tissue was studied histologically and biochemically for autophagic, ER stress and Keap1-Nrf2-ARE signalling pathway markers. MAIN OUTCOMES AND RESULTS Prolongation of ischaemia significantly increased the cortical injury observed in rats and was associated with a gradual increase in the protein expression of ubiquitin-aggregates, Grp78, GADD153/CHOP and p62/ZIP. Autophagy marker Atg12-Atg5 and LC3-PE increased and then decreased. Moreover, p62/ZIP mRNA expression increased and then decreased and was consistent with Nrf2 activation. CONCLUSIONS p62/ZIP not only plays a key role in scavenging protein aggregates during autophagy, but it may also be involved in preventing oxidative injury and alleviating ER stress through the Keap1-Nrf2-ARE signalling pathway during cerebral ischaemia/reperfusion injury.
Collapse
Affiliation(s)
- Weiwei Wang
- Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, 126 Xinmin Street, Changchun, Jilin, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Rocha L. Interaction between electrical modulation of the brain and pharmacotherapy to control pharmacoresistant epilepsy. Pharmacol Ther 2013; 138:211-28. [DOI: 10.1016/j.pharmthera.2013.01.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 01/07/2013] [Indexed: 12/15/2022]
|
31
|
Shi C, Flanagan SR, Samadani U. Vagus nerve stimulation to augment recovery from severe traumatic brain injury impeding consciousness: a prospective pilot clinical trial. Neurol Res 2013; 35:263-76. [PMID: 23485054 PMCID: PMC4568744 DOI: 10.1179/1743132813y.0000000167] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Traumatic brain injury (TBI) has high morbidity and mortality in both civilian and military populations. Blast and other mechanisms of TBI damage the brain by causing neurons to disconnect and atrophy. Such traumatic axonal injury can lead to persistent vegetative and minimally conscious states (VS and MCS), for which limited treatment options exist, including physical, occupational, speech, and cognitive therapies. More than 60 000 patients have received vagus nerve stimulation (VNS) for epilepsy and depression. In addition to decreased seizure frequency and severity, patients report enhanced mood, reduced daytime sleepiness independent of seizure control, increased slow wave sleep, and improved cognition, memory, and quality of life. Early stimulation of the vagus nerve accelerates the rate and extent of behavioral and cognitive recovery after fluid percussion brain injury in rats. METHODS We recently obtained Food and Drug Administration (FDA) approval for a pilot prospective randomized crossover trial to demonstrate objective improvement in clinical outcome by placement of a vagus nerve stimulator in patients who are recovering from severe TBI. Our hypothesis is that stimulation of the vagus nerve results in increased cerebral blood flow and metabolism in the forebrain, thalamus, and reticular formation, which promotes arousal and improved consciousness, thereby improving outcome after TBI resulting in MCS or VS. DISCUSSION If this study demonstrates that VNS can safely and positively impact outcome, then a larger randomized prospective crossover trial will be proposed.
Collapse
Affiliation(s)
- Chen Shi
- Department of Neurosurgery, New York University School of Medicine and NYU Langone Medical Center, 550 First Ave. New York, NY 10016
| | - Steven R. Flanagan
- Department of Rehabilitation Medicine, New York University School of Medicine and NYU Langone Medical Center, 240 E. 38 St. New York, NY 10016
| | - Uzma Samadani
- Department of Neurosurgery, New York University School of Medicine and NYU Langone Medical Center, 550 First Ave. New York, NY 10016
- Division of Neurosurgery, New York Harbor Healthcare System Manhattan Veterans Hospital, 423 E. 23 St. New York, NY 10010
| |
Collapse
|
32
|
Kraus T, Kiess O, Hösl K, Terekhin P, Kornhuber J, Forster C. CNS BOLD fMRI effects of sham-controlled transcutaneous electrical nerve stimulation in the left outer auditory canal - a pilot study. Brain Stimul 2013; 6:798-804. [PMID: 23453934 DOI: 10.1016/j.brs.2013.01.011] [Citation(s) in RCA: 205] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2012] [Revised: 01/03/2013] [Accepted: 01/15/2013] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND It has recently been shown that electrical stimulation of sensory afferents within the outer auditory canal may facilitate a transcutaneous form of central nervous system stimulation. Functional magnetic resonance imaging (fMRI) blood oxygenation level dependent (BOLD) effects in limbic and temporal structures have been detected in two independent studies. In the present study, we investigated BOLD fMRI effects in response to transcutaneous electrical stimulation of two different zones in the left outer auditory canal. It is hypothesized that different central nervous system (CNS) activation patterns might help to localize and specifically stimulate auricular cutaneous vagal afferents. METHODOLOGY 16 healthy subjects aged between 20 and 37 years were divided into two groups. 8 subjects were stimulated in the anterior wall, the other 8 persons received transcutaneous vagus nervous stimulation (tVNS) at the posterior side of their left outer auditory canal. For sham control, both groups were also stimulated in an alternating manner on their corresponding ear lobe, which is generally known to be free of cutaneous vagal innervation. Functional MR data from the cortex and brain stem level were collected and a group analysis was performed. RESULTS In most cortical areas, BOLD changes were in the opposite direction when comparing anterior vs. posterior stimulation of the left auditory canal. The only exception was in the insular cortex, where both stimulation types evoked positive BOLD changes. Prominent decreases of the BOLD signals were detected in the parahippocampal gyrus, posterior cingulate cortex and right thalamus (pulvinar) following anterior stimulation. In subcortical areas at brain stem level, a stronger BOLD decrease as compared with sham stimulation was found in the locus coeruleus and the solitary tract only during stimulation of the anterior part of the auditory canal. CONCLUSIONS The results of the study are in line with previous fMRI studies showing robust BOLD signal decreases in limbic structures and the brain stem during electrical stimulation of the left anterior auditory canal. BOLD signal decreases in the area of the nuclei of the vagus nerve may indicate an effective stimulation of vagal afferences. In contrast, stimulation at the posterior wall seems to lead to unspecific changes of the BOLD signal within the solitary tract, which is a key relay station of vagal neurotransmission. The results of the study show promise for a specific novel method of cranial nerve stimulation and provide a basis for further developments and applications of non-invasive transcutaneous vagus stimulation in psychiatric patients.
Collapse
Affiliation(s)
- Thomas Kraus
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University, Erlangen-Nuremberg, Germany; Frankenalb-Klinik Engelthal, Clinic for Psychiatry, Psychotherapy, Psychosomatic Medicine, and Addiction Rehabilitation, Germany.
| | | | | | | | | | | |
Collapse
|
33
|
Englot DJ, Rolston JD, Wang DD, Hassnain KH, Gordon CM, Chang EF. Efficacy of vagus nerve stimulation in posttraumatic versus nontraumatic epilepsy. J Neurosurg 2012; 117:970-7. [PMID: 22978542 DOI: 10.3171/2012.8.jns122] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT In the US, approximately 500,000 individuals are hospitalized yearly for traumatic brain injury (TBI), and posttraumatic epilepsy (PTE) is a common sequela of TBI. Improved treatment strategies for PTE are critically needed, as patients with the disorder are often resistant to antiepileptic medications and are poor candidates for definitive resection. Vagus nerve stimulation (VNS) is an adjunctive treatment for medically refractory epilepsy that results in a ≥ 50% reduction in seizure frequency in approximately 50% of patients after 1 year of therapy. The role of VNS in PTE has been poorly studied. The aim of this study was to determine whether patients with PTE attain more favorable seizure outcomes than individuals with nontraumatic epilepsy etiologies. METHODS Using a case-control study design, the authors retrospectively compared seizure outcomes after VNS therapy in patients with PTE versus those with nontraumatic epilepsy (non-PTE) who were part of a large prospectively collected patient registry. RESULTS After VNS therapy, patients with PTE demonstrated a greater reduction in seizure frequency (50% fewer seizures at the 3-month follow-up; 73% fewer seizures at 24 months) than patients with non-PTE (46% fewer seizures at 3 months; 57% fewer seizures at 24 months). Overall, patients with PTE had a 78% rate of clinical response to VNS therapy at 24 months (that is, ≥ 50% reduction in seizure frequency) as compared with a 61% response rate among patients with non-PTE (OR 1.32, 95% CI 1.07-1.61), leading to improved outcomes according to the Engel classification (p < 0.0001, Cochran-Mantel-Haenszel statistic). CONCLUSIONS Vagus nerve stimulation should be considered in patients with medically refractory PTE who are not good candidates for resection. A controlled prospective trial is necessary to further examine seizure outcomes as well as neuropsychological outcomes after VNS therapy in patients with intractable PTE.
Collapse
Affiliation(s)
- Dario J Englot
- Comprehensive Epilepsy Center, University of California, San Francisco, California 94143-0112, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Darrah SD, Miller MA, Ren D, Hoh NZ, Scanlon JM, Conley YP, Wagner AK. Genetic variability in glutamic acid decarboxylase genes: associations with post-traumatic seizures after severe TBI. Epilepsy Res 2012; 103:180-94. [PMID: 22840783 DOI: 10.1016/j.eplepsyres.2012.07.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 07/02/2012] [Accepted: 07/03/2012] [Indexed: 11/18/2022]
Abstract
Post traumatic seizures (PTS) occur frequently after traumatic brain injury (TBI). Since gamma-amino butyric acid (GABA) neurotransmission is central to excitotoxicity and seizure development across multiple models, we investigated how genetic variability for glutamic acid decarboxylase (GAD) influences risk for PTS. Using both a tagging and functional single nucleotide polymorphism (SNP) approach, we genotyped the GAD1 and GAD2 genes and linked them with PTS data, regarding time to first seizure, obtained for 257 adult subjects with severe TBI. No significant associations were found for GAD2. In the GAD1 gene, the tagging SNP (tSNP) rs3828275 was associated with an increased risk for PTS occurring <1 wk. The tSNP rs769391 and the functional SNP rs3791878 in the GAD1 gene were associated with increased PTS risk occurring 1 wk-6 mo post-injury. Both risk variants conferred an increased susceptibility to PTS compared to subjects with 0-1 risk variant. Also, those with haplotypes having both risk variants had a higher PTS risk 1 wk-6 mo post-injury than those without these haplotypes. Similarly, diplotype analysis showed those with 2 copies of the haplotype containing both risk alleles were at the highest PTS risk. These results implicate genetic variability within the GABA system in modulating the development of PTS.
Collapse
Affiliation(s)
- Shaun D Darrah
- University of Pittsburgh, Department of Physical Medicine & Rehabilitation, 3471 Fifth Avenue, Suite 202, Pittsburgh, PA 15213, United States.
| | | | | | | | | | | | | |
Collapse
|
35
|
Kumaria A, Tolias CM. Is there a role for vagus nerve stimulation therapy as a treatment of traumatic brain injury? Br J Neurosurg 2012; 26:316-20. [PMID: 22404761 DOI: 10.3109/02688697.2012.663517] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
This paper aims to review the current literature on vagus nerve stimulation (VNS) use in animal models of traumatic brain injury (TBI) and explore its potential role in treatment of human TBI. A MEDLINE search yielded four primary papers from the same group that demonstrated VNS mediated improvement following fluid percussion models of TBI in rats, seen as motor and cognitive improvements, reduction of cortical oedema and neuroprotective effects. The underlying mechanisms are elusive and authors attribute these to attenuation of post traumatic seizures, a noradrenergic mechanism and as yet undetermined mechanisms. Reviewing and elaborating on these ideas, we speculate other potential mechanisms including attenuation of peri-infarct depolarisations, attenuation of glutamate mediated excitotoxicity, stabilisation of intracranial pressure, enhancement of synaptic plasticity, upregulation of endogenous neurogenesis and anti-inflammatory effects may have a role. Although this data unequivocally shows that VNS improves outcome from TBI in animal models, it remains to be determined if these findings translate clinically. Further studies are warranted.
Collapse
Affiliation(s)
- Ashwin Kumaria
- Department of Neurosurgery, Wessex Neurological Centre, Southampton, UK.
| | | |
Collapse
|
36
|
Vonck K, de Herdt V, Sprengers M, Ben-Menachem E. Neurostimulation for epilepsy. HANDBOOK OF CLINICAL NEUROLOGY 2012; 108:955-970. [PMID: 22939078 DOI: 10.1016/b978-0-444-52899-5.00040-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Affiliation(s)
- Kristl Vonck
- Department of Neurology, Ghent University Hospital, Ghent, Belgium.
| | | | | | | |
Collapse
|
37
|
Liu H, Liu Y, Yu J, Lai M, Zhu H, Sun A, Chen W, Zhou W. Vagus nerve stimulation inhibits heroin-seeking behavior induced by heroin priming or heroin-associated cues in rats. Neurosci Lett 2011; 494:70-4. [DOI: 10.1016/j.neulet.2011.02.059] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 02/18/2011] [Accepted: 02/18/2011] [Indexed: 10/18/2022]
|
38
|
Sun Z, Baker W, Hiraki T, Greenberg JH. The effect of right vagus nerve stimulation on focal cerebral ischemia: an experimental study in the rat. Brain Stimul 2011; 5:1-10. [PMID: 22037134 DOI: 10.1016/j.brs.2011.01.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 12/21/2010] [Accepted: 01/24/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The aim of this study was to determine the effect of vagus nerve stimulation (VNS) on infarct size after transient and after permanent focal cerebral ischemia in rats and to test the hypothesis that VNS-induced neuroprotection is due to changes in cerebral blood flow. METHODS Ischemia was produced by either temporary proximal middle cerebral artery occlusion (TMCAO) or permanent distal middle cerebral artery occlusion (PMCAO). Stimulating electrodes were implanted on the cervical part of the right vagus nerve, and electrical stimulation was initiated 30 minutes after the induction of ischemia and delivered for 30 seconds every 5 minutes for 1 hour. All the procedures were duplicated but no stimulus was delivered in control groups. Cerebral blood flow in the MCA territory was continuously monitored with laser speckle contrast imaging. A neurologic evaluation was undertaken after 24 hours of ischemia, and animals were euthanized and neuronal damage evaluated. RESULTS Ischemic lesion volume was smaller in VNS-treated animals in both the temporary and permanent ischemic groups (P<.01). VNS-treated animals in TMCAO had better functional scores at 24 hours as compared with control animals (P<.01), but there were no statistically significant differences in the neurobehavioral scores in PMCAO (P=.089). Cerebral blood flow changes in the MCA territory during ischemia did not differ between the VNS-treated animals and control animals in either group. CONCLUSIONS VNS offers neuroprotection against stroke in both temporary and permanent ischemia. Although the precise mechanism of this effect remains to be determined, alterations in cerebral blood flow do not appear to play a role. VNS could readily be translated to clinical practice.
Collapse
Affiliation(s)
- Zhenghui Sun
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6063, USA
| | | | | | | |
Collapse
|
39
|
Wager-Smith K, Markou A. Depression: a repair response to stress-induced neuronal microdamage that can grade into a chronic neuroinflammatory condition? Neurosci Biobehav Rev 2011; 35:742-64. [PMID: 20883718 PMCID: PMC3777427 DOI: 10.1016/j.neubiorev.2010.09.010] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 09/17/2010] [Accepted: 09/21/2010] [Indexed: 12/19/2022]
Abstract
Depression is a major contributor to the global burden of disease and disability, yet it is poorly understood. Here we review data supporting a novel theoretical model for the biology of depression. In this model, a stressful life event leads to microdamage in the brain. This damage triggers an injury repair response consisting of a neuroinflammatory phase to clear cellular debris and a spontaneous tissue regeneration phase involving neurotrophins and neurogenesis. During healing, released inflammatory mediators trigger sickness behavior and psychological pain via mechanisms similar to those that produce physical pain during wound healing. The depression remits if the neuronal injury repair process resolves successfully. Importantly, however, the acute psychological pain and neuroinflammation often transition to chronicity and develop into pathological depressive states. This hypothesis for depression explains substantially more data than alternative models, including why emerging data show that analgesic, anti-inflammatory, pro-neurogenic and pro-neurotrophic treatments have antidepressant effects. Thus, an acute depressive episode can be conceptualized as a normally self-limiting but highly error-prone process of recuperation from stress-triggered neuronal microdamage.
Collapse
Affiliation(s)
- Karen Wager-Smith
- Department of Psychiatry, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0603, USA.
| | | |
Collapse
|
40
|
Zakharova EI, Dudchenko AM, Svinov MM, Fedorova MM, Germanova EL. Cholinergic systems of the rat brain and neuronal reorganization under conditions of acute hypoxia. NEUROCHEM J+ 2010. [DOI: 10.1134/s1819712410040082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
41
|
De Herdt V, De Waele J, Raedt R, Wyckhuys T, El Tahry R, Vonck K, Wadman W, Boon P. Modulation of seizure threshold by vagus nerve stimulation in an animal model for motor seizures. Acta Neurol Scand 2010; 121:271-6. [PMID: 20003088 DOI: 10.1111/j.1600-0404.2009.01223.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The precise mechanism of action of vagus nerve stimulation (VNS) in suppressing epileptic seizures remains to be elucidated. This study investigates whether VNS modulates cortical excitability by determining the threshold for provoking focal motor seizures by cortical electrical stimulation before and after VNS. MATERIAL AND METHODS Male Wistar rats (n = 8) were implanted with a cuff-electrode around the left vagus nerve and with stimulation electrodes placed bilaterally on the rat motor cortex. Motor seizure threshold (MST) was assessed for each rat before and immediately after 1 h of VNS with standard stimulation parameters, during two to three sessions on different days. RESULTS An overall significant increase of the MST was observed following 1 h of VNS compared to the baseline value (1420 microA and 1072 microA, respectively; P < 0.01). The effect was reproducible over time with an increase in MST in each experimental session. CONCLUSIONS VNS significantly increases the MST in a cortical stimulation model for motor seizures. These data indicate that VNS is capable of modulating cortical excitability.
Collapse
|
42
|
George MS, Aston-Jones G. Noninvasive techniques for probing neurocircuitry and treating illness: vagus nerve stimulation (VNS), transcranial magnetic stimulation (TMS) and transcranial direct current stimulation (tDCS). Neuropsychopharmacology 2010; 35:301-16. [PMID: 19693003 PMCID: PMC3055429 DOI: 10.1038/npp.2009.87] [Citation(s) in RCA: 236] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2009] [Revised: 06/12/2009] [Accepted: 06/17/2009] [Indexed: 01/19/2023]
Abstract
Although the preceding chapters discuss much of the new knowledge of neurocircuitry of neuropsychiatric diseases, and an invasive approach to treatment, this chapter describes and reviews the noninvasive methods of testing circuit-based theories and treating neuropsychiatric diseases that do not involve implanting electrodes into the brain or on its surface. These techniques are transcranial magnetic stimulation, vagus nerve stimulation, and transcranial direct current stimulation. Two of these approaches have FDA approval as therapies.
Collapse
Affiliation(s)
- Mark S George
- Departments of Psychiatry, Radiology and Neuroscience, Institute of Psychiatry, MUSC Center for Advanced Imaging Research, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | |
Collapse
|
43
|
Aalbers M, Rijkers K, van Winden L, Hoogland G, Vles J, Majoie H. Horner's syndrome: A complication of experimental carotid artery surgery in rats. Auton Neurosci 2009; 147:64-9. [DOI: 10.1016/j.autneu.2009.01.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Revised: 01/08/2009] [Accepted: 01/14/2009] [Indexed: 11/30/2022]
|
44
|
Boon P, Raedt R, de Herdt V, Wyckhuys T, Vonck K. Electrical stimulation for the treatment of epilepsy. Neurotherapeutics 2009; 6:218-27. [PMID: 19332313 PMCID: PMC5084197 DOI: 10.1016/j.nurt.2008.12.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2008] [Accepted: 12/31/2008] [Indexed: 01/24/2023] Open
Abstract
Despite the advent of new pharmacological treatments and the high success rate of many surgical treatments for epilepsy, a substantial number of patients either do not become seizure-free or they experience major adverse events (or both). Neurostimulation-based treatments have gained considerable interest in the last decade. Vagus nerve stimulation (VNS) is an alternative treatment for patients with medically refractory epilepsy, who are unsuitable candidates for conventional epilepsy surgery, or who have had such surgery without optimal outcome. Although responder identification studies are lacking, long-term VNS studies show response rates between 40% and 50% and long-term seizure freedom in 5% to 10% of patients. Surgical complications and perioperative morbidity are low. Research into the mechanism of action of VNS has revealed a crucial role for the thalamus and cortical areas that are important in the epileptogenic process. Acute deep brain stimulation (DBS) in various thalamic nuclei and medial temporal lobe structures has recently been shown to be efficacious in small pilot studies. There is little evidence-based information on rational targets and stimulation parameters. Amygdalohippocampal DBS has yielded a significant decrease of seizure counts and interictal EEG abnormalities during long-term follow-up. Data from pilot studies suggest that chronic DBS for epilepsy may be a feasible, effective, and safe procedure. Further trials with larger patient populations and with controlled, randomized, and closed-loop designs should now be initiated. Further progress in understanding the mechanism of action of DBS for epilepsy is a necessary step to making this therapy more efficacious and established.
Collapse
Affiliation(s)
- Paul Boon
- Reference Center for Refractory Epilepsy and Laboratory for Clinical and Experimental Neurophysiology (LCEN), Department of Neurology, Ghent University Hospital, Ghent, Belgium.
| | | | | | | | | |
Collapse
|
45
|
Vonck K, De Herdt V, Boon P. Vagal nerve stimulation--a 15-year survey of an established treatment modality in epilepsy surgery. Adv Tech Stand Neurosurg 2009; 34:111-46. [PMID: 19368083 DOI: 10.1007/978-3-211-78741-0_5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Neurostimulation is an emerging treatment for neurological diseases. Electrical stimulation of the tenth cranial nerve or vagus nerve stimulation (VNS) has become a valuable option in the therapeutic armamentarium for patients with refractory epilepsy. It is indicated in patients with refractory epilepsy who are unsuitable candidates for epilepsy surgery or who have had insufficient benefit from such a treatment. Vagus nerve stimulation reduces seizure frequency with > 50% in 1/3 of patients and has a mild side effects profile. Research to elucidate the mechanism of action of vagus nerve stimulation has shown that effective stimulation in humans is primarily mediated by afferent vagal A- and B-fibers. Crucial brainstem and intracranial structures include the locus coeruleus, the nucleus of the solitary tract, the thalamus and limbic structures. Neurotransmitters playing a role may involve the major inhibitory neurotransmitter GABA but also serotoninergic and adrenergic systems. This manuscript reviews the clinical studies investigating efficacy and side effects in patients and the experimental studies aiming to elucidate the mechanims of action.
Collapse
Affiliation(s)
- K Vonck
- Department of Neurology, Ghent University Hospital, Gent, Belgium
| | | | | |
Collapse
|
46
|
Chen C, Hu Q, Yan J, Yang X, Shi X, Lei J, Chen L, Huang H, Han J, Zhang JH, Zhou C. Early inhibition of HIF-1alpha with small interfering RNA reduces ischemic-reperfused brain injury in rats. Neurobiol Dis 2008; 33:509-17. [PMID: 19166937 DOI: 10.1016/j.nbd.2008.12.010] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2008] [Revised: 11/14/2008] [Accepted: 12/18/2008] [Indexed: 12/23/2022] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) plays an essential role in cerebral ischemia as a proapoptotic factor. We hypothesized that HIF-1alpha siRNA can protect the brain from ischemic damage by inhibiting HIF-1alpha induced apoptotic pathway at the RNA level in a rat focal ischemic model. Results showed that treatment with HIF-1alpha siRNA reduced the infarct volume, decreased mortality, improved neurological deficits and reduced Evans blue extravasation. The expression of HIF-1alpha mRNA (Real-Time PCR) and protein were significantly silenced and the immunohistochemistry and Western blot revealed the suppression of HIF-1alpha, VEGF, p53 and Caspase-3. Double fluorescence labeling showed HIF-1alpha positive immunoreactive materials were partly colocalized with NeuN, p53 and Caspase-3 in the injured cerebral cortex. This study showed that HIF-1alpha siRNA may protect the ischemic-reperfused neurons in vivo via inhibition of HIF-1alpha, its downstream VEGF and other apoptotic-related proteins such as p53 and Caspase-3 and may have potentials for the early treatment of ischemic cerebral stroke.
Collapse
Affiliation(s)
- Chunhua Chen
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing 100083, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Follesa P, Biggio F, Gorini G, Caria S, Talani G, Dazzi L, Puligheddu M, Marrosu F, Biggio G. Vagus nerve stimulation increases norepinephrine concentration and the gene expression of BDNF and bFGF in the rat brain. Brain Res 2007; 1179:28-34. [PMID: 17920573 DOI: 10.1016/j.brainres.2007.08.045] [Citation(s) in RCA: 246] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2007] [Revised: 08/10/2007] [Accepted: 08/17/2007] [Indexed: 11/28/2022]
Abstract
Vagus nerve stimulation therapy, effective for treatment-resistant epilepsy, has recently been approved also for treatment-resistant depression; nevertheless, the molecular mechanism(s) underlying its therapeutic action remains unclear. Given that neurotrophic factors and monoamines could play a crucial role in the pathophysiology of depression, we tested whether vagus nerve stimulation increases the expression of brain-derived neurotrophic factor, fibroblast growth factor, and nerve growth factor as well as the concentration of norepinephrine in the rat brain. Rats were implanted with a vagus nerve stimulator device and the effects of acute stimulation were evaluated on the growth factors mRNA levels and norepinephrine concentration by ribonuclease protection assay and microdialysis, respectively. We found that acute vagus nerve stimulation increased the expression of brain-derived neurotrophic factor and fibroblast growth factor in the hippocampus and cerebral cortex, decreased the abundance of nerve growth factor mRNA in the hippocampus, and, similar to the antidepressant drug venlafaxine, increased the norepinephrine concentration in the prefrontal cortex. This study demonstrates that acute vagus nerve stimulation triggers neurochemical and molecular changes in the rat brain involving neurotransmitters and growth factors known to play a crucial role in neuronal trophism. These new findings contribute to the elucidation of the molecular mechanisms underlying the therapeutic actions of vagus nerve stimulation in both treatment-resistant depression and epilepsy.
Collapse
Affiliation(s)
- Paolo Follesa
- Department of Experimental Biology, Section of Neuroscience, University of Cagliari, Cagliari, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Clough RW, Neese SL, Sherill LK, Tan AA, Duke A, Roosevelt RW, Browning RA, Smith DC. Cortical edema in moderate fluid percussion brain injury is attenuated by vagus nerve stimulation. Neuroscience 2007; 147:286-93. [PMID: 17543463 DOI: 10.1016/j.neuroscience.2007.04.043] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Revised: 04/23/2007] [Accepted: 04/27/2007] [Indexed: 11/16/2022]
Abstract
Development of cerebral edema (intracellular and/or extracellular water accumulation) following traumatic brain injury contributes to mortality and morbidity that accompanies brain injury. Chronic intermittent vagus nerve stimulation (VNS) initiated at either 2 h or 24 h (VNS: 30 s train of 0.5 mA, 20 Hz, biphasic pulses every 30 min) following traumatic brain injury enhances recovery of motor and cognitive function in rats in the weeks following brain injury; however, the mechanisms of facilitated recovery are unknown. The present study examines the effects of VNS on development of acute cerebral edema following unilateral fluid percussion brain injury (FPI) in rats, concomitant with assessment of their behavioral recovery. Two hours following FPI, VNS was initiated. Behavioral testing, using both beam walk and locomotor placing tasks, was conducted at 1 and 2 days following FPI. Edema was measured 48 h post-FPI by the customary method of region-specific brain weights before and after complete dehydration. Results of this study replicated that VNS initiated at 2 h after FPI: 1) effectively facilitated the recovery of vestibulomotor function at 2 days after FPI assessed by beam walk performance (P<0.01); and 2) tended to improve locomotor placing performance at the same time point (P=0.18). Most interestingly, results of this study showed that development of edema within the cerebral cortex ipsilateral to FPI was significantly attenuated at 48 h in FPI rats receiving VNS compared with non-VNS FPI rats (P<0.04). Finally, a correlation analysis between beam walk performance and cerebral edema following FPI revealed a significant inverse correlation between behavior performance and cerebral edema. Together, these results suggest that VNS facilitation of motor recovery following experimental brain injury in rats is associated with VNS-mediated attenuation of cerebral edema.
Collapse
Affiliation(s)
- R W Clough
- Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL 62901-6503, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Chen C, Hu Q, Yan J, Lei J, Qin L, Shi X, Luan L, Yang L, Wang K, Han J, Nanda A, Zhou C. Multiple effects of 2ME2 and D609 on the cortical expression of HIF-1alpha and apoptotic genes in a middle cerebral artery occlusion-induced focal ischemia rat model. J Neurochem 2007; 102:1831-1841. [PMID: 17532791 DOI: 10.1111/j.1471-4159.2007.04652.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Despite 2-methoxyestradiol (2ME2) and tricyclodecan-9-yl-xanthogenate (D609) having multiple effects on cancer cells, mechanistically, both of them down-regulate hypoxia-inducible factor-1alpha (HIF-1alpha) and vascular endothelial growth factor (VEGF). We hypothesize HIF-1alpha plays an essential role in cerebral ischemia as a pro-apoptosis regulator; 2ME2 and D609 decrease the levels of HIF-1alpha and VEGF, that might contribute to protecting brain from ischemia injury. A total of 102 male Sprague-Dawley rats were split into five groups: sham, middle cerebral artery occlusion (MCAO), MCAO + dimethyl sulfoxide, MCAO + 2ME2, and MCAO + D609. 2ME2 and D609 were injected intraperitoneally 1 h after reperfusion. Rats were killed at 24 h and 7 days. At 24 h, 2ME2 and D609 reduce the levels of HIF-1alpha and VEGF (enzyme-linked immunosorbent assay), depress the expression of HIF-1alpha, VEGF, BCL2/adenovirus E1B 19 kDa interacting protein 3 (BNIP3) and cleaved caspase 3 (western blot and immunohistochemistry) in the brain infarct area. Double fluorescence labeling shows HIF-1alpha positive immunoreactive materials are co-localized with BNIP3 and terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling inside the nuclei of neurons. At 7 days, 2ME2 and D609 reduce the infarct volume (2,3,7-triphenyltetrazolium chloride) and blood-brain barrier extravasation, decrease the mortality and improve the neurological deficits. In conclusion, 2ME2 and D609 are powerful agents to protect brain from cerebral ischemic injury by inhibiting HIF-1alpha expression, attenuating the superfluous expression of VEGF to avoid blood-brain barrier disruption and suppressing neuronal apoptosis via BNIP3 pathway.
Collapse
Affiliation(s)
- Chunhua Chen
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Qin Hu
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Junhao Yan
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Jiliang Lei
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Lihua Qin
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Xianzhong Shi
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Liju Luan
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Lei Yang
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Ke Wang
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Jingyan Han
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Anil Nanda
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Changman Zhou
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| |
Collapse
|