1
|
Kang J, Zhou Y, Xiong Q, Dong X. Trigeminal nerve electrical stimulation attenuates early traumatic brain injury through the TLR4/NF-κB/NLRP3 signaling pathway mediated by orexin-A/OX1R system. Aging (Albany NY) 2024; 16:7946-7960. [PMID: 38713160 PMCID: PMC11131994 DOI: 10.18632/aging.205795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/09/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND Traumatic brain injury (TBI) is a significant contributor to global mortality and disability, and emerging evidence indicates that trigeminal nerve electrical stimulation (TNS) is a promising therapeutic intervention for neurological impairment following TBI. However, the precise mechanisms underlying the neuroprotective effects of TNS in TBI are poorly understood. Thus, the objective of this study was to investigate the potential involvement of the orexin-A (OX-A)/orexin receptor 1 (OX1R) mediated TLR4/NF-κB/NLRP3 signaling pathway in the neuroprotective effects of TNS in rats with TBI. METHODS Sprague-Dawley rats were randomly assigned to four groups: sham, TBI, TBI+TNS+SB334867, and TBI+TNS. TBI was induced using a modified Feeney's method, and subsequent behavioral assessments were conducted to evaluate neurological function. The trigeminal nerve trunk was isolated, and TNS was administered following the establishment of the TBI model. The levels of neuroinflammation, brain tissue damage, and proteins associated with the OX1R/TLR4/NF-κB/NLRP3 signaling pathway were assessed using hematoxylin-eosin staining, Nissl staining, western blot analysis, quantitative real-time polymerase chain reaction, and immunofluorescence techniques. RESULTS The findings of our study indicate that TNS effectively mitigated tissue damage, reduced brain edema, and alleviated neurological deficits in rats with TBI. Furthermore, TNS demonstrated the ability to attenuate neuroinflammation levels and inhibit the expression of proteins associated with the TLR4/NF-κB/NLRP3 signaling pathway. However, it is important to note that the aforementioned effects of TNS were reversible upon intracerebroventricular injection of an OX1R antagonist. CONCLUSION TNS may prevent brain damage and relieve neurological deficits after a TBI by inhibiting inflammation, possibly via the TLR4/NF-κB/NLRP3 signaling pathway mediated by OX-A/OX1R.
Collapse
Affiliation(s)
- Junwei Kang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Yifan Zhou
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Qi Xiong
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Xiaoyang Dong
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| |
Collapse
|
2
|
Ren B, Kang J, Wang Y, Meng X, Huang Y, Bai Y, Feng Z. Transcranial direct current stimulation promotes angiogenesis and improves neurological function via the OXA-TF-AKT/ERK signaling pathway in traumatic brain injury. Aging (Albany NY) 2024; 16:6566-6587. [PMID: 38604164 PMCID: PMC11042948 DOI: 10.18632/aging.205724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/19/2024] [Indexed: 04/13/2024]
Abstract
Traumatic brain injury (TBI) and its resulting complications pose a major challenge to global public health, resulting in increased rates of disability and mortality. Cerebrovascular dysfunction is nearly universal in TBI cases and is closely associated with secondary injury after TBI. Transcranial direct current stimulation (tDCS) shows great potential in the treatment of TBI; however, the exact mechanism remains elusive. In this study, we performed in vivo and in vitro experiments to explore the effects and mechanisms of tDCS in a controlled cortical impact (CCI) rat model simulating TBI. In vivo experiments show that tDCS can effectively reduce brain tissue damage, cerebral edema and neurological deficits. The potential mechanism may be that tDCS improves the neurological function of rats by increasing orexin A (OXA) secretion, upregulating the TF-AKT/ERK signaling pathway, and promoting angiogenesis at the injury site. Cellular experiments showed that OXA promoted HUVEC migration and angiogenesis, and these effects were counteracted by the ERK1/2 inhibitor LY3214996. The results of Matrigel experiment in vivo showed that TNF-a significantly reduced the ability of HUVEC to form blood vessels, but OXA could rescue the effect of TNF-a on the ability of HUVEC to form blood vessels. However, LY3214996 could inhibit the therapeutic effect of OXA. In summary, our preliminary study demonstrates that tDCS can induce angiogenesis through the OXA-TF-AKT/ERK signaling pathway, thereby improving neurological function in rats with TBI.
Collapse
Affiliation(s)
- Bingkai Ren
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330003, Jiangxi, China
| | - Junwei Kang
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330003, Jiangxi, China
| | - Yan Wang
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330003, Jiangxi, China
| | - Xiangqiang Meng
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330003, Jiangxi, China
| | - Ying Huang
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330003, Jiangxi, China
| | - Yang Bai
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330003, Jiangxi, China
| | - Zhen Feng
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330003, Jiangxi, China
| |
Collapse
|
3
|
Zhou Y, Yang H, You M, Feng Z, Dong X. Cognition-Enhancement Effect of Median Nerve Electrical Stimulation in Patients with Cognitive Impairment: A Retrospective Cohort Study. World Neurosurg 2024; 184:e537-e545. [PMID: 38320650 DOI: 10.1016/j.wneu.2024.01.165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 02/08/2024]
Abstract
OBJECTIVE People with cognitive impairment often face quality-of-life problems and require ongoing support, which has profound consequences for caregivers and society. Noninvasive brain stimulation techniques, such as median nerve electrical stimulation (MNS), have shown promising potentials in improving cognitive ability in patients with cognitive impairment. Therefore, we aimed to investigate the positive effect and safety of MNS in cognitive impairment. METHODS Patients diagnosed with cognitive impairment from the hospital record management system of the First Affiliated Hospital of Nanchang University from April 1, 2020, to December 31, 2022, were enrolled. Data on patients' basic characteristics, treatment records, and examination results such as the Mini-Mental State Examination (MMSE), activities of daily living (ADL), and P300 event-related potentials before and after treatment were collected. RESULTS Overall, 146 patients with cognitive impairment were enrolled, including 71 patients who underwent conventional therapy (standard treatment group) and 75 patients who underwent conventional therapy and MNS operation (active MNS group). Before treatment, there were no differences between the standard treatment and active MNS groups in terms of age, sex, etiology, duration of symptoms before therapy, hospital stay, whether they had undergone surgery, MMSE score, ADL score, and amplitude and latency of the P300 event-related potentials (P > 0.05). After treatment, we observed significant improvements in the MMSE score, ADL score, amplitude of P300, and decreased latency of P300 event-related potentials in both groups compared with before treatment (P < 0.05). In addition, we observed that the active MNS group showed higher MMSE and ADL scores, higher amplitude of P300 event-related potentials, and lower latency of P300 event-related potentials than the standard treatment group after treatment (P < 0.05). Furthermore, no side effects were associated with MNS operation. CONCLUSIONS These preliminary data provide early evidence that MNS may be a positive effect and safe method for promoting the recovery of cognitive ability in patients with cognitive impairment.
Collapse
Affiliation(s)
- Yifan Zhou
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Haihua Yang
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Mengyu You
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhen Feng
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaoyang Dong
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
4
|
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
|
5
|
Kang J, Ren B, Huang L, Dong X, Xiong Q, Feng Z. Orexin-A alleviates ferroptosis by activating the Nrf2/HO-1 signaling pathway in traumatic brain injury. Aging (Albany NY) 2024; 16:3404-3419. [PMID: 38349868 PMCID: PMC10929813 DOI: 10.18632/aging.205541] [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: 06/15/2023] [Accepted: 11/30/2023] [Indexed: 02/15/2024]
Abstract
BACKGROUND Traumatic Brain Injury (TBI) has high disability and mortality rate. Oxidative stress and ferroptosis are important pathophysiological characteristics after TBI. Orexin-A (OXA) can alleviate neuronal damage in diverse neurological disorders. Nevertheless, the role and mechanism of OXA in TBI stay unknown. OBJECTIVES The research investigated protection influence of OXA on TBI and its potential mechanisms. METHODS Male Sprague-Dawley rats were randomly grouped into: sham, TBI, TBI + normal saline (NS) and TBI+OXA groups. TBI model was constructed in rat via modified Feeney's approach, and OXA treatment was administered following construction of TBI model. RESULTS Relative to TBI+NS group, TBI+OXA group displayed greatly recovered tissue damage and neurological deficits. Additionally, OXA eased oxidative stress as well as ferroptosis in cerebral cortex of rats following TBI. Furthermore, OXA increased Nrf2 expression and regulating factors HO-1 and NQO1 in cerebral cortex of TBI rats. CONCLUSIONS Our research found OXA may restrain ferroptosis via Nrf2/HO-1 signaling pathway activation, thereby reducing brain injury after TBI.
Collapse
Affiliation(s)
- Junwei Kang
- Department of Rehabilitation Medicine, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Bingkai Ren
- Department of Rehabilitation Medicine, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Lianghua Huang
- Department of Rehabilitation Medicine, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
- First Department of Rehabilitation Medicine, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330006, China
| | - Xiaoyang Dong
- Department of Rehabilitation Medicine, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Qi Xiong
- Department of Rehabilitation Medicine, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Zhen Feng
- Department of Rehabilitation Medicine, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| |
Collapse
|
6
|
Winter Y, Sandner K, Bassetti CLA, Glaser M, Ciolac D, Ziebart A, Karakoyun A, Saryyeva A, Krauss JK, Ringel F, Groppa S. Vagus nerve stimulation for the treatment of narcolepsy. Brain Stimul 2024; 17:83-88. [PMID: 38184192 DOI: 10.1016/j.brs.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/10/2023] [Accepted: 01/02/2024] [Indexed: 01/08/2024] Open
Abstract
BACKGROUND AND OBJECTIVE No study on neurostimulation in narcolepsy is available until now. Arousal- and wake-promoting effects of vagus nerve stimulation (VNS) have been demonstrated in animal experiments and are well-known as side effects of VNS therapy in epilepsy and depression. The objective was to evaluate the therapeutic effect of VNS on daily sleepiness and cataplexies in narcolepsy. METHODS In our open-label prospective comparative study, we included narcolepsy patients who were treated with VNS because of depression or epilepsy and compared them to controls without narcolepsy treated with VNS for depression or epilepsy (18 patients in each group, aged 31.5 ± 8.2 years). We evaluated daily sleepiness (Epworth Sleepiness Scale, ESS) and the number of cataplexies per week before the implantation of VNS and at three and six month follow-ups. RESULTS Compared to baseline (ESS: 15.9 ± 2.5) patients with narcolepsy showed a significant improvement on ESS after three months (11.2 ± 3.3, p < 0.05) and six months (9.6 ± 2.8, p < 0.001) and a trend to reduction of cataplexies. No significant ESS-improvement was observed in patients without narcolepsy (14.9 ± 3.9, 13.6 ± 3.7, 13.2 ± 3.5, p = 0.2 at baseline, three and six months, correspondingly). Side effects did not differ between the study groups. CONCLUSION In this first evaluation of VNS in narcolepsy, we found a significant improvement of daily sleepiness due to this type of neurostimulation. VNS could be a promising non-medical treatment in narcolepsy.
Collapse
Affiliation(s)
- Yaroslav Winter
- Mainz Comprehensive Epilepsy and Sleep Medicine Center, Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Department of Neurology, Philipps-University Marburg, Germany.
| | - Katharina Sandner
- Mainz Comprehensive Epilepsy and Sleep Medicine Center, Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Claudio L A Bassetti
- Department of Neurology, Inselspital, University Hospital of Bern, University of Bern, Switzerland
| | - Martin Glaser
- Department of Neurosurgery, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Dumitru Ciolac
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Andreas Ziebart
- Department of Neurosurgery, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ali Karakoyun
- Department of Neurosurgery, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Assel Saryyeva
- Department of Neurosurgery, Hannover Medical School, MHH, Hannover, Germany
| | - Joachim K Krauss
- Department of Neurosurgery, Hannover Medical School, MHH, Hannover, Germany
| | - Florian Ringel
- Department of Neurosurgery, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sergiu Groppa
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
7
|
Zhou Y, Sun Y, He P, Xiong Q, Kang J, Tang Y, Feng Z, Dong X. The efficacy and safety of transcutaneous auricular vagus nerve stimulation for patients with minimally conscious state: a sham-controlled randomized double-blind clinical trial. Front Neurosci 2023; 17:1323079. [PMID: 38156271 PMCID: PMC10752952 DOI: 10.3389/fnins.2023.1323079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023] Open
Abstract
Background Transcutaneous auricular vagus nerve stimulation (taVNS) has emerged as a potentially effective neuromodulation technique for addressing neurological disorders, including disorders of consciousness. Expanding upon our prior clinical study, which demonstrated the superior effectiveness of a 4-week taVNS treatment in patients with minimally conscious state (MCS) compared to those in a vegetative state/unresponsive wakefulness state, the aim of this investigation was to evaluate the safety and therapeutic efficacy of taVNS in individuals with MCS through a sham-controlled randomized double-blind clinical trial. Methods A cohort of 50 adult patients (male = 33, female = 17) diagnosed with a MCS were randomly assigned to either the active taVNS (N = 25) or sham taVNS (N = 25) groups. The treatment period lasted for 4 weeks, followed by an 8-week follow-up period. The Coma Recovery Scale-Revised (CRS-R) and Glasgow Coma Scale (GCS) were administered at baseline and weekly during the initial 4 weeks. Additionally, the Disability Rating Scale (DRS) was used to assess the patients' functional abilities via telephone at week 12. Furthermore, various neurophysiological measures, including electroencephalogram (EEG), upper-limb somatosensory evoked potentials (USEP), brainstem auditory evoked potentials (BAEP), and P300 event-related potentials (P300), were employed to monitor changes in brain activity and neural conduction pathways. Results The scores for the active taVNS group in the CRS-R and GCS showed greater improvement over time compared to the sham taVNS group (CRS-R: 1-week, Z = -1.248, p = 0.212; 2-week, Z = -1.090, p = 0.276; 3-week, Z = -2.017, p = 0.044; 4-week, Z = -2.267, p = 0.023. GCS: 1-week, Z = -1.325, p = 0.185; 2-week, Z = -1.245, p = 0.213; 3-week, Z = -1.848, p = 0.065; 4-week, Z = -1.990, p = 0.047). Additionally, the EEG, USEP, BAEP, and P300 also demonstrated significant improvement in the active taVNS group compared to the sham taVNS group at week 4 (EEG, Z = -2.086, p = 0.037; USEP, Z = -2.014, p = 0.044; BAEP, Z = -2.298, p = 0.022; P300 amplitude, Z = -1.974, p = 0.049; P300 latency, t = 2.275, p = 0.027). Subgroup analysis revealed that patients with MCS derived greater benefits from receiving taVNS treatment earlier (CRS-R, Disease duration ≤ 1-month, mean difference = 8.50, 95% CI = [2.22, 14.78], p = 0.027; GCS, Disease duration ≤ 1-month, mean difference = 3.58, 95% CI = [0.14, 7.03], p = 0.044). By week 12, the active taVNS group exhibited lower Disability Rating Scale (DRS) scores compared to the sham taVNS group (Z = -2.105, p = 0.035), indicating a more favorable prognosis for MCS patients who underwent taVNS. Furthermore, no significant adverse events related to taVNS were observed during treatment. Conclusion The findings of this study suggest that taVNS may serve as a potentially effective and safe intervention for facilitating the restoration of consciousness in individuals diagnosed with MCS. This therapeutic approach appears to enhance cerebral functioning and optimize neural conduction pathways. Clinical trial registration http://www.chictr.org.cn, Identifier ChiCTR2200066629.
Collapse
Affiliation(s)
- Yifan Zhou
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Rehabilitation Medicine Clinical Research Center of Jiangxi Province, Nanchang, Jiangxi, China
| | - Yejing Sun
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Rehabilitation Medicine Clinical Research Center of Jiangxi Province, Nanchang, Jiangxi, China
| | - Pei He
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Rehabilitation Medicine Clinical Research Center of Jiangxi Province, Nanchang, Jiangxi, China
| | - Qi Xiong
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Rehabilitation Medicine Clinical Research Center of Jiangxi Province, Nanchang, Jiangxi, China
| | - Junwei Kang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Rehabilitation Medicine Clinical Research Center of Jiangxi Province, Nanchang, Jiangxi, China
| | - Yunliang Tang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Rehabilitation Medicine Clinical Research Center of Jiangxi Province, Nanchang, Jiangxi, China
| | - Zhen Feng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Rehabilitation Medicine Clinical Research Center of Jiangxi Province, Nanchang, Jiangxi, China
| | - Xiaoyang Dong
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Rehabilitation Medicine Clinical Research Center of Jiangxi Province, Nanchang, Jiangxi, China
| |
Collapse
|
8
|
Wang L, Gao F, Wang Z, Liang F, Dai Y, Wang M, Wu J, Chen Y, Yan Q, Wang L. Transcutaneous auricular vagus nerve stimulation in the treatment of disorders of consciousness: mechanisms and applications. Front Neurosci 2023; 17:1286267. [PMID: 37920298 PMCID: PMC10618368 DOI: 10.3389/fnins.2023.1286267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/05/2023] [Indexed: 11/04/2023] Open
Abstract
This review provides an in-depth exploration of the mechanisms and applications of transcutaneous auricular vagus nerve stimulation (taVNS) in treating disorders of consciousness (DOC). Beginning with an exploration of the vagus nerve's role in modulating brain function and consciousness, we then delve into the neuroprotective potential of taVNS demonstrated in animal models. The subsequent sections assess the therapeutic impact of taVNS on human DOC, discussing the safety, tolerability, and various factors influencing the treatment response. Finally, the review identifies the current challenges in taVNS research and outlines future directions, emphasizing the need for large-scale trials, optimization of treatment parameters, and comprehensive investigation of taVNS's long-term effects and underlying mechanisms. This comprehensive overview positions taVNS as a promising and safe modality for DOC treatment, with a focus on understanding its intricate neurophysiological influence and optimizing its application in clinical settings.
Collapse
Affiliation(s)
- Likai Wang
- Department of Rehabilitation Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Fei Gao
- Department of Rehabilitation Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Zhan Wang
- Department of Rehabilitation Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Feng Liang
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Yongli Dai
- Department of Rehabilitation Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Mengchun Wang
- Department of Rehabilitation Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Jingyi Wu
- Department of Rehabilitation Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Yaning Chen
- Department of Rehabilitation Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Qinjie Yan
- Department of Rehabilitation Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Litong Wang
- Department of Rehabilitation Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
9
|
Dong X, Tang Y, Zhou Y, Feng Z. Stimulation of vagus nerve for patients with disorders of consciousness: a systematic review. Front Neurosci 2023; 17:1257378. [PMID: 37781261 PMCID: PMC10540190 DOI: 10.3389/fnins.2023.1257378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/06/2023] [Indexed: 10/03/2023] Open
Abstract
Purpose The purpose of this study is to evaluate the efficacy and safety of stimulating the vagus nerve in patients with disorders of consciousness (DOCs). Methods A comprehensive systematic review was conducted, encompassing the search of databases such as PubMed, CENTRAL, EMBASE and PEDro from their inception until July 2023. Additionally, manual searches and exploration of grey literature were performed. The literature review was conducted independently by two reviewers for search strategy, selection of studies, data extraction, and judgment of evidence quality according to the American Academy of Cerebral Palsy and Developmental Medicine (AACPDM) Study Quality Scale. Results A total of 1,269 articles were retrieved, and 10 studies met the inclusion criteria. Among these, there were three case reports, five case series, and only two randomized controlled trials (RCTs). Preliminary studies have suggested that stimulation of vagus nerve can enhance the levels of DOCs in both vegetative state/unresponsive wakefulness state (VS/UWS) and minimally conscious state (MCS). However, due to a lack of high-quality RCTs research and evidence-based medical evidence, no definitive conclusion can be drawn regarding the intervention's effectiveness on consciousness level. Additionally, there were no significant adverse effects observed following stimulation of vagus nerve. Conclusion A definitive conclusion cannot be drawn from this systematic review as there was a limited number of eligible studies and low-quality evidence. The findings of this systematic review can serve as a roadmap for future research on the use of stimulation of vagus nerve to facilitate recovery from DOCs.
Collapse
Affiliation(s)
| | | | | | - Zhen Feng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
10
|
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
|
11
|
Maruyama T, Ueta Y. Internal and external modulation factors of the orexin system (REVIEW). Peptides 2023; 165:171009. [PMID: 37054895 DOI: 10.1016/j.peptides.2023.171009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/15/2023]
Abstract
Orexin-A and -B (identical to hypocretin-1 and -2) are neuropeptides synthesized in the lateral hypothalamus and perifornical area, and orexin neurons project their axon terminals broadly throughout the entire central nervous system (CNS). The activity of orexins is mediated by two specific G protein-coupled receptors (GPCRs), termed orexin type1 receptor (OX1R) and orexin type2 receptor (OX2R). The orexin system plays a relevant role in various physiological functions, including arousal, feeding, reward, and thermogenesis, and is key to human health. Orexin neurons receive various signals related to environmental, physiological, and emotional stimuli. Previous studies have reported that several neurotransmitters and neuromodulators influence the activation or inhibition of orexin neuron activity. In this review, we summarize the modulating factors of orexin neurons in the sleep/wake rhythm and feeding behavior, particularly in the context of the modulation of appetite, body fluids, and circadian signaling. We also describe the effects of life activity, behavior, and diet on the orexin system. Some studies have observed phenomena that have been verified in animal experiments, revealing the detailed mechanism and neural pathway, while their applications to humans is expected in future research.
Collapse
Affiliation(s)
- Takashi Maruyama
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Japan.
| | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Japan
| |
Collapse
|
12
|
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
|
13
|
Zhou YF, Kang JW, Xiong Q, Feng Z, Dong XY. Transauricular vagus nerve stimulation for patients with disorders of consciousness: A randomized controlled clinical trial. Front Neurol 2023; 14:1133893. [PMID: 36937511 PMCID: PMC10017768 DOI: 10.3389/fneur.2023.1133893] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction Disorders of consciousness (DoCs) are a frequent complication of brain injury disease, and effective treatments are currently lacking. Transauricular vagus nerve stimulation (tVNS) has been proposed as a promising therapeutic method for neurological disorders such as epilepsy and depression. In our previous study, we demonstrated that vagus nerve stimulation promoted recovery in rats with DoCs caused by traumatic brain injury. However, the clinical effect of vagus nerve stimulation on consciousness disorders is unclear. We aimed to investigate the therapeutic efficacy and safety of tVNS in patients with DoCs. Methods We conducted a randomized, double-blinded, sham-controlled trial. Patients (N = 60) with DoCs, including minimally conscious state (MCS) and vegetative state/unresponsive wakefulness syndrome, were enrolled and randomized to groups receiving either active or sham tVNS. A frequency of 20 Hz and pulse wave of 200 us was used in the active-tVNS protocol, which was performed in the auricular branch of the vagus nerve in the left outer ear. The sham-tVNS protocol was the same as the active-tVNS protocol although without current input. Both groups of patients also received conventional treatments. Consciousness was evaluated according to the Coma Recovery Scale-Revised before and after the 4-week intervention. We also recorded the type and number of behavioral responses. Safety was primarily assessed according to the incidence of treatment-emergent adverse events. Each patient's heart rate and blood pressure were monitored during all treatment sessions. Results Ultimately, 57 patients completed the study: 28 patients underwent active tVNS and 29 patients underwent sham tVNS. No significant differences were observed in Coma Recovery Scale-Revised scores between the active- and sham-tVNS groups before the tVNS sessions. Compared with patients in the sham-tVNS group (9.28 ± 4.38), patients with DoCs treated with active tVNS showed improved consciousness (10.93 ± 4.99), although not statistically significant. Further analysis revealed obvious differences between patients with MCS receiving active and sham tVNS, but no significant difference in patients with vegetative state/unresponsive wakefulness syndrome in both groups. All side effects were considered common medical conditions with no obvious correlation to tVNS. Conclusion These preliminary data provide early evidence that tVNS may be an effective and safe approach for promoting the recovery of consciousness, especially in patients with MCS. Clinical trial registration https://www.chictr.org.cn/edit.aspx?pid=175938&htm=4, identifier: ChiCTR2200066629.
Collapse
|
14
|
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
|
15
|
Chen H, Feng Z, Min L, Deng W, Tan M, Hong J, Gong Q, Zhang D, Liu H, Hou J. Vagus Nerve Stimulation Reduces Neuroinflammation Through Microglia Polarization Regulation to Improve Functional Recovery After Spinal Cord Injury. Front Neurosci 2022; 16:813472. [PMID: 35464311 PMCID: PMC9022634 DOI: 10.3389/fnins.2022.813472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/11/2022] [Indexed: 01/02/2023] Open
Abstract
BackgroundSpinal cord injury (SCI) is a devastating disease that lacks effective treatment. Interestingly, recent studies indicated that vagus nerve stimulation (VNS), neuromodulation that is widely used in a variety of central nervous system (CNS) diseases, improved motor function recovery after SCI. But the exact underlying mechanism of how VNS ameliorates SCI is unclear. This study aimed to confirm the efficacy and further explore the potential therapeutic mechanism of VNS in SCI.MethodA T10 spinal cord compression model was established in adult female Sprague-Dawley rats. Then the stimulation electrode was placed in the left cervical vagus nerve (forming Sham-VNS, VNS, and VNS-MLA groups). Basso-Beattie-Bresnahan (BBB) behavioral scores and Motor evoked potentials (MEPs) analysis were used to detect motor function. A combination of histological and molecular methods was used to clarify the relevant mechanism.ResultsCompared with the Sham-VNS group, the VNS group exhibited better functional recovery, reduced scar formation (both glial and fibrotic scars), tissue damage, and dark neurons, but these beneficial effects of VNS were diminished after alpha 7 nicotinic acetylcholine receptor (α7nAchR) blockade. Specifically, VNS inhibited the pro-inflammatory factors TNF-α, IL-1β, and IL-6 and increased the expression of the anti-inflammatory factors IL-10. Furthermore, we found that VNS promotes the shift of M1-polarized Iba-1+/CD86+ microglia to M2-polarized Iba-1+/CD206+ microglia via upregulating α7nAchR to alleviate neuroinflammation after SCI.ConclusionOur results demonstrated that VNS promotes microglial M2 polarization through upregulating α7nAChR to reduce neuroinflammation, thus improving motor function recovery after SCI. These findings indicate VNS might be a promising neuromodulation strategy for SCI.
Collapse
|
16
|
Raymond JS, Rehn S, Hoyos CM, Bowen MT. The influence of oxytocin-based interventions on sleep-wake and sleep-related behaviour and neurobiology: A systematic review of preclinical and clinical studies. Neurosci Biobehav Rev 2021; 131:1005-1026. [PMID: 34673110 DOI: 10.1016/j.neubiorev.2021.10.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 12/26/2022]
Abstract
The oxytocin (OXT) system has garnered considerable interest due to its influence on diverse behaviours. However, scant research has considered the influence of oxytocin on sleep-wake and sleep-related behaviour and neurobiology. Consequently, the objective of this systematic review was to assess the extant preclinical and clinical evidence for the influence of oxytocin-based interventions on sleep-wake outcomes. The primary search was conducted on 22/7/2020 using six electronic databases; 30 studies (19 preclinical, 11 clinical) were included based on inclusion criteria. Studies were evaluated for risk of bias using the SYRCLE tool and the Cochrane risk of bias tools for preclinical and clinical studies, respectively. Results indicated manipulation of the OXT system can influence sleep-wake outcomes. Preclinical evidence suggests a wake-promoting influence of OXT system activation whereas the clinical evidence suggests little or no sleep-promoting influence of OXT. OXT dose was identified as a likely modulatory factor of OXT-induced effects on sleep-wake behaviour. Future studies are necessary to validate and strengthen these tentative conclusions about the influence of OXT on sleep-wake behaviour.
Collapse
Affiliation(s)
- Joel S Raymond
- The University of Sydney, Faculty of Science, School of Psychology, Camperdown, NSW, Australia; The University of Sydney, Brain and Mind Centre, Camperdown, NSW, Australia
| | - Simone Rehn
- The University of Sydney, Faculty of Science, School of Psychology, Camperdown, NSW, Australia
| | - Camilla M Hoyos
- The University of Sydney, Faculty of Science, School of Psychology, Camperdown, NSW, Australia; The University of Sydney, Brain and Mind Centre, Camperdown, NSW, Australia; The University of Sydney, Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Camperdown, NSW, Australia
| | - Michael T Bowen
- The University of Sydney, Faculty of Science, School of Psychology, Camperdown, NSW, Australia; The University of Sydney, Brain and Mind Centre, Camperdown, NSW, Australia.
| |
Collapse
|
17
|
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
|
18
|
Carta G, Seregni A, Casamassima A, Galli M, Geuna S, Pagliaro P, Zago M. Validation and Reliability of a Novel Vagus Nerve Neurodynamic Test and Its Effects on Heart Rate in Healthy Subjects: Little Differences Between Sexes. Front Neurosci 2021; 15:698470. [PMID: 34552462 PMCID: PMC8450330 DOI: 10.3389/fnins.2021.698470] [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: 06/10/2021] [Accepted: 07/21/2021] [Indexed: 11/24/2022] Open
Abstract
Background The vagus nerve (VN), also called the pneumogastric nerve, connects the brainstem to organs contained in the chest and abdomen. Physiologically, VN stimulation can rapidly affect cardiac activity and heart rate (HR). VN neuropathy can increase the risk of arrhythmias and sudden death. Therefore, a selective test of VN function may be very useful. Since peripheral neurodynamic tests (NDT) are reliable for the assessment of neuropathies in somatic nerves, we aimed to validate a novel NDT to assess VN activity, namely, the VN-NTD. Methods In this cross-sectional double-blind, sex-balanced study, 30 participants (15 females) completed a checklist of autonomic dysfunction symptoms. During the VN-NDT administration, HR and symptoms (i.e., mechanical allodynia) were monitored in parallel to a real-time ultrasonography imaging (USI) and motion capture analysis of the neck. The VN-NDT impact on HR and its accuracy for autonomic symptoms reported in the last 7 days were tested. Results The VN-NDT induced a significant HR reduction of about 12 and 8 bpm in males and females [t(1, 119) = 2.425; p < 0.017; ηp2 = 0.047, 95% confidence interval (CI): 0.93–9.18], respectively. No adverse events were observed during VN-NDT. A substantial interexaminer agreement between the evaluators in symptoms induction by VN-NDT was detected [F(1, 119) = 0.540; p = 0.464; ηp2 = 0.005, low effect]. Notably, mechanical allodynia accuracy for gastrointestinal dysfunctions was excellent (p < 0.05; 95% CI: 0.52–0.73; p < 0.001; 95% CI: 0.81–0.96). Conclusions The novel VN-NDT is a valid and accurate test capable of detecting VN activation with high sensitivity. Data provided are suitable for both sexes as a hallmark of HR variation due to VN normal response. The proposed VN-NDT may be reliable as daily routine neurological examination tests for the evaluation of neuropathic signs related to neuroinflammation of the VN. Clinical Trial Registration www.ClinicalTrials.gov, identifier NCT04192877.
Collapse
Affiliation(s)
- Giacomo Carta
- Human Anatomy, Department of Biological and Clinical Sciences, University of Turin, Turin, Italy.,Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Turin, Italy.,Department of Rehabilitation, Sesto Hospital, ASST Nord Milano, Milan, Italy
| | - Agnese Seregni
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| | - Andrea Casamassima
- General Surgery Department, S. Maria delle Stelle Hospital, ASST Melegnano e Martesana Melzo, Milan, Italy
| | - Manuela Galli
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| | - Stefano Geuna
- Human Anatomy, Department of Biological and Clinical Sciences, University of Turin, Turin, Italy.,Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Turin, Italy
| | - Pasquale Pagliaro
- Human Physiology, Department of Biological and Clinical Sciences, University of Turin, Turin, Italy
| | - Matteo Zago
- Department of Mechanics, Politecnico di Milano, Lecco, Italy
| |
Collapse
|
19
|
Wakefulness-Promoting Effects of Lateral Hypothalamic Area-Deep Brain Stimulation in Traumatic Brain Injury-Induced Comatose Rats: Upregulation of α1-Adrenoceptor Subtypes and Downregulation of Gamma-Aminobutyric Acid β Receptor Expression Via the Orexins Pathway. World Neurosurg 2021; 152:e321-e331. [PMID: 34062300 DOI: 10.1016/j.wneu.2021.05.089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 05/21/2021] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Previous studies have shown that deep brain stimulation (DBS) can improve the level of consciousness of comatose patients with traumatic brain injuries (TBIs). However, the most suitable targets for DBS are unknown, and the mechanisms underlying recovery remain to be determined. The aim of the present study was to assess the effects of lateral hypothalamic area-DBS (LHA-DBS) in comatose rats with TBIs. METHODS A total of 55 Sprague-Dawley rats were randomly assigned to 5 groups: the control group, TBI group, stimulated (TBI+LHA-DBS) group, antagonist (TBI+SB334867+LHA-DBS) group, and antagonist control (TBI+saline+LHA-DBS) group. The rats in the control group had undergone a sham operation and anesthesia, without coma induction. Coma was induced using a free-fall drop method. The rats in the stimulated group received bilateral LHA stimulation (frequency, 200 Hz; voltage, 2-4 V; pulse width, 0.1 ms) for 1 hour, with 5-minute intervals between subsequent stimulations, which were applied alternately to the left and right sides of the lateral hypothalamus. The comatose rats in the antagonist group received an intracerebroventricular injection with an orexins receptor type 1 (OX1R) antagonist (SB334867) and then received LHA-DBS. A I-VI consciousness scale and electroencephalography were used to assess the level of consciousness in each group of rats after LHA-DBS. Western blotting and immunofluorescence were used to detect OX1R expression in the LHA and α1-adrenoceptor (α1-AR) subtype and gamma-aminobutyric acid β receptor (GABABR) expression in the prefrontal cortex. RESULTS In the TBI, stimulated, antagonist, and antagonist control groups, 5, 10, 6, and 9 rats were awakened. The electroencephalographic readings indicated that the proportion of δ waves was lower in the stimulated group than in the TBI and antagonist groups (P < 0.05). Western blotting and immunofluorescence analysis showed that OX1R expression was greater in the stimulated group than in the TBI group (P < 0.05). The expression of α1-AR was also greater in the stimulated group than in the TBI and antagonist groups (P < 0.05). In contrast, the GABABR levels in the stimulated group were lower than those in the TBI and antagonist groups (P < 0.05). A statistically significant difference was found between the antagonist and antagonist control groups. CONCLUSIONS Taken together, these results suggest that LHA-DBS promotes the recovery of consciousness in comatose rats with TBIs. Upregulation of α1-AR expression and downregulation of GABABR expression in the prefrontal cortex via the orexins and OX1R pathways might be involved in the wakefulness-promoting effects of LHA-DBS.
Collapse
|
20
|
Ye W, Yan Y, Tang Y, Dong X, Chen G, Kang J, Huang L, Xiong Q, Feng Z. Orexin-A Attenuates Inflammatory Responses in Lipopolysaccharide-Induced Neural Stem Cells by Regulating NF-KB and Phosphorylation of MAPK/P38/Erk Pathways. J Inflamm Res 2021; 14:2007-2017. [PMID: 34040413 PMCID: PMC8140926 DOI: 10.2147/jir.s308078] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/14/2021] [Indexed: 11/23/2022] Open
Abstract
Background Neuronal damage is the main cause of neurological diseases. Neural stem cells (NSCs) have the functions of cell repair and replacement of neurons, secretion of neurotrophic factors, and immune regulation of the neural microenvironment. Objective Previous study found that Orexin-A had a protective effect on neurons in the central nervous system, but it is lacking in making great efforts on the function of Orexin-A on NSCs. This study aimed to investigate the anti-inflammatory responses and signaling mechanisms of Orexin-A on lipopolysaccharide (LPS)-induced NSCs. Methods Quantitative real-time polymerase chain reaction was used to detect the mRNA level. Signaling pathway-related protein expression was detected by Western blot. The proliferation and migration of NSCs were investigated by Cell Counting Kit-8 (CCK-8) detection kit and transwell assay. Besides, the staining of hematoxylin and eosin (HE) was performed to study the morphology of cell. Results Orexin-A decreased the pro-inflammatory cytokines of IL-1β, TNF-α, and IL-6 induced by LPS by regulating nuclear factor-k-gene binding (NF-kB) and phosphorylation of P38/Erk-mitogen-activated protein kinases (MAPKs) pathways, but not p-JNK signaling. Conclusion Our findings indicate that Orexin-A can alleviate the inflammatory response of NSC. It can provide beneficial help in neural stem cell therapy applications.
Collapse
Affiliation(s)
- Wen Ye
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, People's Republic of China
| | - Yan Yan
- Department of Nephrology, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, People's Republic of China
| | - Yunliang Tang
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, People's Republic of China
| | - Xiaoyang Dong
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, People's Republic of China
| | - Gengfa Chen
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, People's Republic of China
| | - Junwei Kang
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, People's Republic of China
| | - Lianghua Huang
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, People's Republic of China
| | - Qi Xiong
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, People's Republic of China
| | - Zhen Feng
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, People's Republic of China
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Despite the development of several medications for the acute and preventive treatment of migraine, there are still many patients in whom lack of efficacy, tolerability, interactions or contraindications make other options necessary. CGRP-based drugs have opened the door to a new era of migraine-targeted treatments. Beyond CGRP, there are other promising targets covered here. RECENT FINDINGS For the acute treatment of migraine, 5-HT1F receptor agonists, ditans, are now available. Unlike triptans, 5-HT1B/1D receptor agonists, cardiovascular disease is not a contraindication for the use of ditans. The first study on a monoclonal antibody targeting PAC1 receptor was negative, although this may not be the end for the pituitary adenylate cyclase-activating polypeptide (PACAP) pathway as a target. SUMMARY Following positive phase-III clinical trials, lasmiditan is the first ditan to be FDA-approved. PACAP has experimental evidence suggesting a role in migraine pathophysiology. As for CGRP, the presence of PACAP in key migraine structures along with positive provocative tests for both PACAP-38 and PACAP-27 indicate this pathway may still be a pharmacological target. Glutamate-based targets have long been considered in migraine. Two clinical trials with memantine, an NMDA-R antagonist, for the preventive treatment of migraine have now been published. The hypothalamus has also been implicated in migraine pathophysiology: the potential role of orexins in migraine is discussed. Acid-sensing ion channels, as well as amylin-blocking drugs, may also become migraine treatments in the future: more research is warranted.
Collapse
|
22
|
Intranasal Orexin After Cardiac Arrest Leads to Increased Electroencephalographic Gamma Activity and Enhanced Neurologic Recovery in Rats. Crit Care Explor 2021; 3:e0349. [PMID: 33634267 PMCID: PMC7901796 DOI: 10.1097/cce.0000000000000349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Objectives Prolonged cardiac arrest is known to cause global ischemic brain injury and functional impairment. Upon resuscitation, electroencephalographic recordings of brain activity begin to resume and can potentially be used to monitor neurologic recovery. We have previously shown that intrathecal orexin shows promise as a restorative drug and arousal agent in rodents. Our goal is to determine the electrophysiology effects of orexin in a rodent model of asphyxial cardiac arrest, focusing on the electroencephalographic activity in the gamma and super-gamma bands (indicative of return of higher brain function). Design Experimental animal study. Setting University-based animal research laboratory. Subjects Adult male Wistar rats. Interventions In an established model of asphyxial cardiac arrest (n = 24), we treated half of Wistar rats with orexin administered intranasally by atomizer 30 minutes post return of spontaneous circulation in one of two dose levels (10 and 50 µM); the rest were treated with saline as control. Continuous electroencephalographic recording was obtained and quantitatively analyzed for the gamma fraction. Gamma and high-frequency super-gamma band measures were compared against clinical recovery according to Neuro-Deficit Score. Measurements and Main Results Compared with the control cohort, the high-dose orexin cohort showed significantly better Neuro-Deficit Score 4 hours after return of spontaneous circulation (55.17 vs 47.58; p < 0.02) and significantly higher mean gamma fraction (0.251 vs 0.177; p < 0.02) in cerebral regions surveyed by rostral electrodes for the first 170 minutes after administration of orexin. Conclusions Our findings support early and continuous monitoring of electroencephalography-based gamma activity as a marker of better functional recovery after intranasal administration of orexin as measured by Neuro-Deficit Score in an established animal model of asphyxial cardiac arrest.
Collapse
|
23
|
Long-term outcomes of two patients with progressive myoclonic epilepsy treated with vagus nerve stimulation therapy. Heliyon 2020; 6:e05324. [PMID: 33134590 PMCID: PMC7586113 DOI: 10.1016/j.heliyon.2020.e05324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/17/2020] [Accepted: 10/19/2020] [Indexed: 11/23/2022] Open
|
24
|
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
|
25
|
Tang H, Zhu Q, Li W, Qin S, Gong Y, Wang H, Shioda S, Li S, Huang J, Liu B, Fang Y, Liu Y, Wang S, Guo Y, Xia Q, Guo Y, Xu Z. Neurophysiology and Treatment of Disorders of Consciousness Induced by Traumatic Brain Injury: Orexin Signaling as a Potential Therapeutic Target. Curr Pharm Des 2020; 25:4208-4220. [PMID: 31663471 DOI: 10.2174/1381612825666191029101830] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) can cause disorders of consciousness (DOC) by impairing the neuronal circuits of the ascending reticular activating system (ARAS) structures, including the hypothalamus, which are responsible for the maintenance of the wakefulness and awareness. However, the effectiveness of drugs targeting ARAS activation is still inadequate, and novel therapeutic modalities are urgently needed. METHODS The goal of this work is to describe the neural loops of wakefulness, and explain how these elements participate in DOC, with emphasis on the identification of potential new therapeutic options for DOC induced by TBI. RESULTS Hypothalamus has been identified as a sleep/wake center, and its anterior and posterior regions have diverse roles in the regulation of the sleep/wake function. In particular, the posterior hypothalamus (PH) possesses several types of neurons, including the orexin neurons in the lateral hypothalamus (LH) with widespread projections to other wakefulness-related regions of the brain. Orexins have been known to affect feeding and appetite, and recently their profound effect on sleep disorders and DOC has been identified. Orexin antagonists are used for the treatment of insomnia, and orexin agonists can be used for narcolepsy. Additionally, several studies demonstrated that the agonists of orexin might be effective in the treatment of DOC, providing novel therapeutic opportunities in this field. CONCLUSION The hypothalamic-centered orexin has been adopted as the point of entry into the system of consciousness control, and modulators of orexin signaling opened several therapeutic opportunities for the treatment of DOC.
Collapse
Affiliation(s)
- Huiling Tang
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qiumei Zhu
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wei Li
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Siru Qin
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yinan Gong
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hong Wang
- Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Seiji Shioda
- Peptide Drug Innovation, Global Research Center for Innovative Life Science, Hoshi University School of Pharmacy and Pharmaceutical Sciences, 2-4-41 Ebara, Shinagawa, Tokyo 142-8501, Japan
| | - Shanshan Li
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jin Huang
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Baohu Liu
- Department of Rehabilitation, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuxin Fang
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yangyang Liu
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shenjun Wang
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yongming Guo
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qing Xia
- Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Guo
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhifang Xu
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
26
|
Hakon J, Moghiseh M, Poulsen I, Øland CML, Hansen CP, Sabers A. Transcutaneous Vagus Nerve Stimulation in Patients With Severe Traumatic Brain Injury: A Feasibility Trial. Neuromodulation 2020; 23:859-864. [PMID: 32227429 DOI: 10.1111/ner.13148] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Preclinical studies have shown that surgically implanted vagus nerve stimulation (VNS) promotes recovery of consciousness and cognitive function following experimental traumatic brain injury (TBI). The aim of this study is to report the feasibility and safety of a noninvasive transcutaneous vagus nerve stimulation (tVNS) in patients with persistent impairment of consciousness following severe TBI. MATERIALS AND METHODS The feasibility of tVNS was evaluated in five patients presenting with diffuse axonal injury and reduced dominant EEG activity one month following severe TBI. tVNS was applied to the left cymba conchae of the external ear using a skin electrode four hours daily for eight weeks. Possible effects of tVNS on physiological parameters and general side effects were recorded. In addition, we report the rate of recovery using coma recovery scale revised (CRS-R). RESULTS The tVNS regime of four hours daily for eight weeks was feasible and well tolerated with little side effects and no clinically relevant effects on physiological parameters. Three patients showed improvements (>3 points) in the CRS-R following eight weeks tVNS. CONCLUSION We demonstrated that tVNS is a feasible and safe VNS strategy for patients following severe TBI. Controlled studies are needed to clarify whether tVNS has a potential to promote recovery of consciousness following severe TBI.
Collapse
Affiliation(s)
- Jakob Hakon
- Research Unit on Brain Injury Rehabilitation Copenhagen (RUBRIC), Department of Neurorehabilitation, TBI Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Neurosurgery, Rigshospitalet, Copenhagen, Denmark
| | - Melika Moghiseh
- Research Unit on Brain Injury Rehabilitation Copenhagen (RUBRIC), Department of Neurorehabilitation, TBI Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Ingrid Poulsen
- Research Unit on Brain Injury Rehabilitation Copenhagen (RUBRIC), Department of Neurorehabilitation, TBI Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Research Unit Nursing and Health Care, Health, Aarhus University, Aarhus, Denmark
| | - Christoffer M L Øland
- Research Unit on Brain Injury Rehabilitation Copenhagen (RUBRIC), Department of Neurorehabilitation, TBI Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Christian P Hansen
- Research Unit on Brain Injury Rehabilitation Copenhagen (RUBRIC), Department of Neurorehabilitation, TBI Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Anne Sabers
- The Epilepsy Clinic, Department of Neurology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
27
|
Sattin D, Leonardi M, Picozzi M. The autonomic nervous system and the brainstem: A fundamental role or the background actors for consciousness generation? Hypothesis, evidence, and future directions for rehabilitation and theoretical approaches. Brain Behav 2020; 10:e01474. [PMID: 31782916 PMCID: PMC6955833 DOI: 10.1002/brb3.1474] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/08/2019] [Accepted: 08/15/2019] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION One of the hardest challenges of the third century is to develop theories that could joint different results for a global explanation of human consciousness. Some important theories have been proposed, trying to explain the emergence of consciousness as the result of different progressive changes in the elaboration of information during brain processing, giving particular attention to the thalamocortical system. METHODS In this article, a summary review of results that highlighted as cerebral cortex could not be so fundamental for consciousness generation is proposed. In detail, three topics were analyzed: (a) studies using experimental approach (manipulating stimuli or brain areas), such as decorticated animals or subliminal presentation of stimuli; (b) studies using anatomo-clinical method (conscious inferenced from observed behaviors); and (c) data from neurostimulation of subcortical areas or of the autonomic nervous system. RESULTS We sketch two speculative hypothesis relative, firstly, to the possible independence from cortical areas of the on/off mechanism for consciousness generation and, secondly, to the possible role of information variability generated by the bottom-up exchange of information among neural systems as a switch for consciousness. CONCLUSIONS A broad range of evidence regarding the functional role of the brainstem and autonomic nervous system is reviewed for its bearing on a future hypothesis regarding the generation of consciousness experience.
Collapse
Affiliation(s)
- Davide Sattin
- Neurology, Public Health, Disability Unit and Coma Research CentreFondazione IRCCS Istituto Neurologico C.BestaMilanItaly
- Experimental Medicine and Medical Humanities‐PhD ProgramBiotechnology and Life Sciences Department and Center for Clinical EthicsInsubria UniversityVareseItaly
| | - Matilde Leonardi
- Neurology, Public Health, Disability Unit and Coma Research CentreFondazione IRCCS Istituto Neurologico C.BestaMilanItaly
| | - Mario Picozzi
- Biotechnology and Life Sciences Department and Center for Clinical EthicsInsubria UniversityVareseItaly
| |
Collapse
|
28
|
Xiang XJ, Sun LZ, Xu CB, Xie Y, Pan MY, Ran J, Hu Y, Nong BX, Shen Q, Huang H, Huang SH, Yu YZ. The clinical effect of vagus nerve stimulation in the treatment of patients with a minimally conscious state. JOURNAL OF NEURORESTORATOLOGY 2020. [DOI: 10.26599/jnr.2020.9040016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Objective: Vagus nerve stimulation (VNS) has recently been used in neurorehabilitation and the recovery of consciousness based on its effects on cortical plasticity. The aim of this study was to examine the therapeutic effects of VNS on patients with a minimally conscious state (MCS). Methods: All patients included in the study were assessed more than 5 months after injury and were receiving regular rehabilitation at our hospital from August 2018 to October 2019. Ten patients diagnosed with MCS by Coma Recovery Scale-Revised (CRS-R) test who underwent VNS surgery were enrolled. The scores on CRS-R evaluation at baseline (before VNS implantation) and 1, 3, and 6 months after VNS treatment were recorded. The stimulation parameters were chosen according to a previous study. All clinical rehabilitation protocols remained unchanged during the study. Furthermore, safety was assessed by analyzing treatment-emergent adverse events (TEAEs). Results: No significant improvement in the total CRS-R scores at the end of the 1-month follow-up was observed (p > 0.05). After 3 months of stimulation, a significant difference (p = 0.0078) was observed in the total CRS-R scores compared with the baseline. After 6 months of VNS treatment, CRS-R assessments showed a continuous significant improvement (p = 0.0039); one patient emerged from the MCS and recovered functional communication and object use. Interestingly, one item of CRS-R scores on visual domain was sensitive to VNS treatment (p = 0.0039). Furthermore, no serious adverse event occurred throughout the study. Conclusion: This exploratory study provides preliminary evidence suggesting that VNS is a safe and effective tool for consciousness recovery in patients with MCS.
Collapse
|
29
|
He LX, Wan L, Xiang W, Li JM, Pan AH, Lu DH. Synaptic development of layer V pyramidal neurons in the prenatal human prefrontal neocortex: a Neurolucida-aided Golgi study. Neural Regen Res 2020; 15:1490-1495. [PMID: 31997813 PMCID: PMC7059576 DOI: 10.4103/1673-5374.274345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The prefrontal neocortex is involved in many high cognitive functions in humans. Deficits in neuronal and neurocircuitry development in this part of the cerebrum have been associated with various neuropsychiatric disorders in adolescents and adults. There are currently little available data regarding prenatal dendrite and spine formation on projecting neurons in the human prefrontal neocortex. Previous studies have demonstrated that Golgi silver staining can identify neurons in the frontal lobe and visual cortex in human embryos. In the present study, five fetal brains, at 19, 20, 26, 35, and 38 gestational weeks, were obtained via the body donation program at Xiangya School of Medicine, Central South University, China. Golgi-stained pyramidal neurons in layer V of Brodmann area 46 in fetuses were quantitatively analyzed using the Neurolucida morphometry system. Results revealed that somal size, total dendritic length, and branching points of these neurons increased from 26 to 38 gestational weeks. There was also a large increase in dendritic spines from 35 to 38 gestational weeks. These findings indicate that, in the human prefrontal neocortex, dendritic growth in layer V pyramidal neurons occurs rapidly during the third trimester of gestation. The use of human fetal brain tissue was approved by the Animal Ethics Committee of Xiangya School of Medicine, Central South University, China (approval No. 2011-045) on April 5, 2011.
Collapse
Affiliation(s)
- Li-Xin He
- Xiangtan Medicine and Health Vocational College, Xiangtan, Hunan Province, China
| | - Lily Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Wei Xiang
- Changde Vocational Technical College, Changde, Hunan Province, China
| | - Jian-Ming Li
- Department of Anatomy, Changsha Medical University, Changsha, Hunan Province, China
| | - An-Hua Pan
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Da-Hua Lu
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| |
Collapse
|