1
|
He J, Liu Q, Guo J, Wu D, Guo Y. Circulatory factors in stroke protection and recovery. Brain Res 2025; 1855:149594. [PMID: 40122323 DOI: 10.1016/j.brainres.2025.149594] [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: 10/10/2024] [Revised: 01/14/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Over the past decade, the management of acute ischemic stroke has undergone a paradigm shift, especially a longer time-window and a wider indication for endovascular treatments. However, many patients still have long-term dysfunction despite the best medical care at present. Based on findings from innovative proteomic and transcriptomic technologies, researchers have identified an array of novel or previously underappreciated circulatory factors that play pivotal roles in mediating post-injuries brain communication. Thus, the previous concept of the brain as a privileged compartment isolated from the rest of the body has been replaced by the novel consensus that brain bidirectionally interacts with the other organs after brain diseases. In this review, we make a summary of several axes that connect the brain with the rest of the body after stroke. More importantly, we summarize several circulatory factors that play pivotal roles in fostering post-stroke functional recovery in the chronic stage. Special attention is given to the instrumental role of circulatory signals, positing them as significant contributors to the complex process of brain function recovery and as translational therapeutic targets for ischemic stroke in future studies.
Collapse
Affiliation(s)
- Jiachen He
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China; Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin 150081 Heilongjiang, China
| | - Qi Liu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China
| | - Jiaqi Guo
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China
| | - Di Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China; Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 10053, China.
| | - Yansu Guo
- Beijing Geriatric Healthcare Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
2
|
Sun X, Li L, Huang L, Li Y, Wang L, Wei Q. Harnessing spinal circuit reorganization for targeted functional recovery after spinal cord injury. Neurobiol Dis 2025; 207:106854. [PMID: 40010611 DOI: 10.1016/j.nbd.2025.106854] [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: 01/10/2025] [Revised: 02/13/2025] [Accepted: 02/23/2025] [Indexed: 02/28/2025] Open
Abstract
Spinal cord injury (SCI) disrupts the communication between the brain and spinal cord, resulting in the loss of motor function below the injury site. However, spontaneous structural and functional plasticity occurs in neural circuits after SCI, with unaffected synaptic inputs forming new connections and detour pathways to support recovery. The review discusses various mechanisms of circuit reorganization post-SCI, including supraspinal pathways, spinal interneurons, and spinal central pattern generators. Functional recovery may rely on maintaining a balance between excitatory and inhibitory neural activity, as well as enhancing proprioceptive input, which plays a key role in limb stability. The review emphasizes the importance of endogenous neuronal regeneration, neuromodulation therapies (such as electrical stimulation) and proprioception in SCI treatment. Future research should integrate advanced technologies such as gene targeting, imaging, and single-cell mapping to better understand the mechanisms underpinning SCI recovery, aiming to identify key neuronal subpopulations for targeted reconstruction and enhanced functional recovery. By harnessing spinal circuit reorganization, these efforts hold the potential to pave the way for more precise and effective strategies for functional recovery after SCI.
Collapse
Affiliation(s)
- Xin Sun
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Lijuan Li
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Liyi Huang
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Yangan Li
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Lu Wang
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Quan Wei
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China.
| |
Collapse
|
3
|
Hernandez-Navarro A, Ros-Alsina A, Yurtseven M, Wright M, Kumru H. Non-invasive cerebral and spinal cord stimulation for motor and gait recovery in incomplete spinal cord injury: systematic review and meta-analysis. J Neuroeng Rehabil 2025; 22:53. [PMID: 40050875 PMCID: PMC11887137 DOI: 10.1186/s12984-025-01557-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/15/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Spinal cord injury (SCI) leads to gait impairment and loss of motor function and can be traumatic or non-traumatic in nature. Recently there has been important progress in the field of non-invasive central nervous stimulation, which can target the brain or spinal cord. In this review we aim to compare the effect of non-invasive cerebral and spinal cord stimulation on gait recovery and motor strength of lower limbs in subjects with SCI. METHODS We conducted a search (from September 2022 until March 2024) using the PubMed, Cochrane, and PEDro databases, including all studies published since the year 2000. The protocol of the review followed PRISMA guidelines and only RCTs scoring above 5 on the PEDro scale were selected. RESULTS A total of 12 RCTs with 341 participants were included. When all studies were pooled together, non-invasive central nervous system stimulation had significant effects on Lower Extremity Motor Scale (LEMS) score and gait speed. However, data was less apparent when subgrouped by type and level of stimulation. Repetitive transcranial magnetic stimulation (rTMS) showed large effect on LEMS, however transcranial direct current stimulation (tDCS) displayed a small effect on motor strength and gait speed. No meta-analysis could be performed for non-invasive spinal cord stimulation due to a lack of studies. CONCLUSIONS When all non-invasive stimulation techniques were pooled together, significant effects on motor strength and gait function were observed. However, subgroup analyses based on stimulation types and levels revealed a significant reduction in these effects, particularly when categorized by stimulation type (rTMS and tDCS). Furthermore, a meta-analysis could not be conducted for non-invasive spinal cord stimulation due to a lack of studies (only one study each on tsDCS and tSCS). Therefore, more randomized controlled trials are needed to evaluate neuromodulation interventions in spinal cord injury, particularly at the spinal cord level. Registration This systematic review with meta-analysis was registered in PROSPERO under the ID 512864.
Collapse
Affiliation(s)
- Agustin Hernandez-Navarro
- Fundación Institut Guttmann, Hospital de Neurorehabilitació Institut Guttmann, Institut Universitari de Neurorrehabilitació Adscrit a la UAB, Camí Can Ruti S/N, 08916, Badalona, Spain.
- Universitat Autònoma de Barcelona, 08193, Barcelona, Spain.
| | - Aina Ros-Alsina
- Fundación Institut Guttmann, Hospital de Neurorehabilitació Institut Guttmann, Institut Universitari de Neurorrehabilitació Adscrit a la UAB, Camí Can Ruti S/N, 08916, Badalona, Spain
| | - Muhammed Yurtseven
- Department of Physiotherapy, Vocational School of Health Services, Istanbul Gelisim University, Istanbul, 34310, Turkey
| | - Mark Wright
- Fundación Institut Guttmann, Hospital de Neurorehabilitació Institut Guttmann, Institut Universitari de Neurorrehabilitació Adscrit a la UAB, Camí Can Ruti S/N, 08916, Badalona, Spain
- Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
| | - Hatice Kumru
- Fundación Institut Guttmann, Hospital de Neurorehabilitació Institut Guttmann, Institut Universitari de Neurorrehabilitació Adscrit a la UAB, Camí Can Ruti S/N, 08916, Badalona, Spain.
- Universitat Autònoma de Barcelona, 08193, Barcelona, Spain.
- Fundació Institut d'Investigació en Ciéncies de la Salut Germans Trias i Pujol, 08916, Badalona, Spain.
| |
Collapse
|
4
|
Liu Y, Li X, Xu H, Sun K, Gong HJ, Luo C. Spinal cord stimulation induces Neurotrophin-3 to improve diabetic foot disease. Med Mol Morphol 2025; 58:43-52. [PMID: 39550735 PMCID: PMC11829938 DOI: 10.1007/s00795-024-00410-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/30/2024] [Indexed: 11/18/2024]
Abstract
Low-extremity ischemic disease is a common complication in diabetic patients, leading to reduced quality of life and potential amputation. This study investigated the therapeutic effect of spinal cord stimulation (SCS) on patients with diabetic foot disease and a rat model of diabetic foot injury. SCS was applied to patients with diabetic foot disease, with clinical assessments performed before and after therapy. Blood levels of NGF, BDNF, and NT-3 were determined by ELISA. A rat model of diabetic foot injury was established to validate NT-3's role in SCS therapy. SCS therapy improved the condition of patients with diabetic ischemic foot disease and promoted wound healing in the rat model. NT-3 levels significantly increased after SCS therapy in both patients and rats. Recombinant NT-3 administration improved wound healing and re-vascularization in the rat model, while NT-3 neutralization abrogated SCS's therapeutic effect. SCS improves the condition of patients with diabetic ischemic foot disease by inducing NT-3 production. Both SCS and NT-3 supplementation show therapeutic potential for ameliorating diabetic foot disease.
Collapse
Affiliation(s)
- Yi Liu
- Department of Neurosurgery, National Regional Trauma Center, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, China
| | - XuanPeng Li
- Department of Neurosurgery, National Regional Trauma Center, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, China
| | - HaiWen Xu
- Department of Neurosurgery, National Regional Trauma Center, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, China
| | - Ke Sun
- Department of Neurosurgery, National Regional Trauma Center, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, China
| | - Hui Jun Gong
- Department of Neurosurgery, National Regional Trauma Center, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, China.
| | - Cheng Luo
- The Second Department of Neurosurgery, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, China.
| |
Collapse
|
5
|
Boudreault-Morales GE, Marquez-Chin C, Liu X, Zariffa J. The effect of depth data and upper limb impairment on lightweight monocular RGB human pose estimation models. Biomed Eng Online 2025; 24:12. [PMID: 39920692 PMCID: PMC11804014 DOI: 10.1186/s12938-025-01347-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Markerless vision-based human pose estimation (HPE) is a promising avenue towards scalable data collection in rehabilitation. Deploying this technology will require self-contained systems able to process data efficiently and accurately. The aims of this work are to (1) Determine how depth data affects lightweight monocular red-green-blue (RGB) HPE performance (accuracy and speed), to inform sensor selection and (2) Validate HPE models using data from individuals with physical impairments. METHODS Two HPE models were investigated: Dite-HRNet and MobileHumanPose (capable of 2D and 3D HPE, respectively). The models were modified to include depth data as an input using three different fusion techniques: an early fusion method, a simple intermediate fusion method (using concatenation), and a complex intermediate fusion method (using specific fusion blocks, additional convolutional layers, and concatenation). All fusion techniques used RGB-D data, in contrast to the original models which only used RGB data. The models were trained, validated and tested using the CMU Panoptic and Human3.6 M data sets as well as a custom data set. The custom data set includes RGB-D and optical motion capture data of 15 uninjured and 12 post-stroke individuals, while they performed movements involving their upper limbs. HPE model performances were monitored through accuracy and computational efficiency. Evaluation metrics include Mean per Joint Position Error (MPJPE), Floating Point Operations (FLOPs) and frame rates (frames per second). RESULTS The early fusion architecture consistently delivered the lowest MPJPE in both 2D and 3D HPE cases while achieving similar FLOPs and frame rates to its RGB counterpart. These results were consistent regardless of the data used for training and testing the HPE models. Comparisons between the uninjured and stroke groups did not reveal a significant effect (all p values > 0.36) of motor impairment on the accuracy of any model. CONCLUSIONS Including depth data using an early fusion architecture improves the accuracy-efficiency trade-off of the HPE model. HPE accuracy is not affected by the presence of physical impairments. These results suggest that using depth data with RGB data is beneficial to HPE, and that models trained with data collected from uninjured individuals can generalize to persons with physical impairments.
Collapse
Affiliation(s)
- Gloria-Edith Boudreault-Morales
- KITE Research Institute, Toronto Rehabilitation Institute - University Health Network, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Cesar Marquez-Chin
- KITE Research Institute, Toronto Rehabilitation Institute - University Health Network, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Xilin Liu
- KITE Research Institute, Toronto Rehabilitation Institute - University Health Network, Toronto, ON, Canada
- Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada
| | - José Zariffa
- KITE Research Institute, Toronto Rehabilitation Institute - University Health Network, Toronto, ON, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.
- Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada.
- Rehabilitation Sciences Institute, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
6
|
He J, Zhang Y, Guo Y, Guo J, Chen X, Xu S, Xu X, Wu C, Liu C, Chen J, Ding Y, Fisher M, Jiang M, Liu G, Ji X, Wu D. Blood-derived factors to brain communication in brain diseases. Sci Bull (Beijing) 2024; 69:3618-3632. [PMID: 39353815 DOI: 10.1016/j.scib.2024.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 10/04/2024]
Abstract
Brain diseases, mainly including acute brain injuries, neurodegenerative diseases, and mental disorders, have posed a significant threat to human health worldwide. Due to the limited regenerative capability and the existence of the blood-brain barrier, the brain was previously thought to be separated from the rest of the body. Currently, various cross-talks between the central nervous system and peripheral organs have been widely described, including the brain-gut axis, the brain-liver axis, the brain-skeletal muscle axis, and the brain-bone axis. Moreover, several lines of evidence indicate that leveraging systemic biology intervention approaches, including but not limited to lifestyle interventions, exercise, diet, blood administration, and peripheral immune responses, have demonstrated a significant influence on the progress and prognosis of brain diseases. The advancement of innovative proteomic and transcriptomic technologies has enriched our understanding of the nuanced interplay between peripheral organs and brain diseases. An array of novel or previously underappreciated blood-derived factors have been identified to play pivotal roles in mediating these communications. In this review, we provide a comprehensive summary of blood-to-brain communication following brain diseases. Special attention is given to the instrumental role of blood-derived signals, positing them as significant contributors to the complex process of brain diseases. The insights presented here aim to bridge the current knowledge gaps and inspire novel therapeutic strategies for brain diseases.
Collapse
Affiliation(s)
- Jiachen He
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China; Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin 150081, China
| | - Yanming Zhang
- Department of Rehabilitation, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yansu Guo
- Beijing Geriatric Healthcare Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jiaqi Guo
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China
| | - Xi Chen
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China
| | - Shuaili Xu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China
| | - Xiaohan Xu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China
| | - Chuanjie Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Chengeng Liu
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Jian Chen
- Department of Neurosurgery, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China
| | - Yuchuan Ding
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit MI 46801, USA
| | - Marc Fisher
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02115, USA
| | - Miaowen Jiang
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China.
| | - Guiyou Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China; Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, Wuhu 241002, China; Brain Hospital, Shengli Oilfield Central Hospital, Dongying 257034, China.
| | - Xunming Ji
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China.
| | - Di Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
7
|
Pu B, Chen Y, Bi Q, Shen J, Wang L, Han Y. Prediction of serum neuritin and neuron-specific enolase for prognosis in patients with traumatic brain injury combined with spinal cord injury. J Med Biochem 2024; 43:870-878. [PMID: 39876915 PMCID: PMC11771968 DOI: 10.5937/jomb0-45469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/18/2024] [Indexed: 01/31/2025] Open
Abstract
Background Serum neuritin and neuron-specific enolase (NSE) have predictive value for the prognosis of patients with combined traumatic brain injury (TBI) and spinal cord injury (SCI). Studying their predictive effects has positive value for disease control and treatment. Methods Sixty patients with combined TBI and SCI were recruited and rolled into three groups according to prognosis: Group I (n=42, favourable prognosis), Group II (n=11, poor prognosis), and Group III (n=7, death). Clinical indicators were compared between the groups, and the predictive value of different indicators for prognosis was analyzed. Results The proportion of patients with combined injuries to other organs and hypotension, as well as levels of platelets (PLT), D-dimer (D-D), antithrombin III (AT-III), S100 protein (S100), NSE, and serum neurofilament levels were significantly higher in Groups II and III compared to Group I. Conversely, the Glasgow Coma Scale (GCS) scores were significantly lower in Group I (P<0.05). Multivariable logistic regression analysis revealed that other organ injuries, GCS score, PLT, D-D, and AT-III significantly influenced the prognosis of TBI combined with SCI patients (P<0.05), while hypotension, NSE, serum neurofilament levels, S100, and accompanying organ injuries were highly correlated with the prognosis of TBI combined with SCI patients (P<0.001). The predictive sensitivity, specificity, accuracy, positive predictive value, and negative predictive value of NSE combined with serum neurofilament in predicting the prognosis of TBI combined with SCI patients were significantly higher than the singular predictive efficacy of NSE or serum neurofilament alone (P<0.05). Conclusions To evaluate the prognosis of TBI combined with SCI patients, consideration should be given to factors such as other organ injuries, hypotension, consciousness assessment, and levels of various biomarkers. Furthermore, combined testing of serum neurofilament and NSE can more accurately predict the prognosis of TBI combined with SCI patients.
Collapse
Affiliation(s)
- Bingbing Pu
- Pudong Hospital, Department of Rehabilitation, Shanghai, China
| | - Yu Chen
- Pudong Hospital, Department of Rehabilitation, Shanghai, China
| | - Qingguo Bi
- Zhoupu Hospital, Department of Critical Care, Shanghai, China
| | - Jian Shen
- Pudong Hospital, Department of Medical Equipment, Shanghai, China
| | - Lihui Wang
- Pudong Hospital, Department of Rehabilitation, Shanghai, China
| | - Ye Han
- People's Hospital of Xuyi County, Department of Pathology, Huaian, Jiangsu Province, China
| |
Collapse
|
8
|
Eliason M, Kalbande PP, Saleem GT. Is non-invasive neuromodulation a viable technique to improve neuroplasticity in individuals with acquired brain injury? A review. Front Hum Neurosci 2024; 18:1341707. [PMID: 39296918 PMCID: PMC11408216 DOI: 10.3389/fnhum.2024.1341707] [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: 11/20/2023] [Accepted: 07/22/2024] [Indexed: 09/21/2024] Open
Abstract
Objective This study aimed to explore and evaluate the efficacy of non-invasive brain stimulation (NIBS) as a standalone or coupled intervention and understand its mechanisms to produce positive alterations in neuroplasticity and behavioral outcomes after acquired brain injury (ABI). Data sources Cochrane Library, Web of Science, PubMed, and Google Scholar databases were searched from January 2013 to January 2024. Study selection Using the PICO framework, transcranial magnetic stimulation (TMS) and transcranial direct current stimulation (tDCS) randomized controlled trials (RCTs), retrospective, pilot, open-label, and observational large group and single-participant case studies were included. Two authors reviewed articles according to pre-established inclusion criteria. Data extraction Data related to participant and intervention characteristics, mechanisms of change, methods, and outcomes were extracted by two authors. The two authors performed quality assessments using SORT. Results Twenty-two studies involving 657 participants diagnosed with ABIs were included. Two studies reported that NIBS was ineffective in producing positive alterations or behavioral outcomes. Twenty studies reported at least one, or a combination of, positively altered neuroplasticity and improved neuropsychological, neuropsychiatric, motor, or somatic symptoms. Twenty-eight current articles between 2020 and 2024 have been studied to elucidate potential mechanisms of change related to NIBS and other mediating or confounding variables. Discussion tDCS and TMS may be efficacious as standalone interventions or coupled with neurorehabilitation therapies to positively alter maladaptive brain physiology and improve behavioral symptomology resulting from ABI. Based on postintervention and follow-up results, evidence suggests NIBS may offer a direct or mediatory contribution to improving behavioral outcomes post-ABI. Conclusion More research is needed to better understand the extent of rTMS and tDCS application in affecting changes in symptoms after ABI.
Collapse
Affiliation(s)
- Michelle Eliason
- Rehabilitation Science Department, University at Buffalo, Buffalo, NY, United States
| | | | - Ghazala T Saleem
- Rehabilitation Science Department, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
9
|
Oishi R, Takeda I, Ode Y, Okada Y, Kato D, Nakashima H, Imagama S, Wake H. Neuromodulation with transcranial direct current stimulation contributes to motor function recovery via microglia in spinal cord injury. Sci Rep 2024; 14:18031. [PMID: 39098975 PMCID: PMC11298548 DOI: 10.1038/s41598-024-69127-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 07/31/2024] [Indexed: 08/06/2024] Open
Abstract
Spinal cord injury (SCI) is damage or trauma to the spinal cord, which often results in loss of function, sensation, or mobility below the injury site. Transcranial direct current stimulation (tDCS) is a non-invasive and affordable brain stimulation technique used to modulate neuronal circuits, which changes the morphology and activity of microglia in the cerebral cortex. However, whether similar morphological changes can be observed in the spinal cord remains unclear. Therefore, we evaluated neuronal population activity in layer 5 (L5) of M1 following SCI and investigated whether changes in the activities of L5 neurons affect microglia-axon interactions using C57BL/6J mice. We discovered that L5 of the primary motor cortex (corticospinal neurons) exhibited reduced synchronized activity after SCI that correlates with microglial morphology, which was recovered using tDCS. This indicates that tDCS promotes changes in the morphological properties and recovery of microglia after SCI. Combining immunotherapy with tDCS may be effective in treating SCI.
Collapse
Affiliation(s)
- Ryotaro Oishi
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Ikuko Takeda
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, Myodaiji, Okazaki, 444-8585, Japan
| | - Yukihito Ode
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Yuya Okada
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Daisuke Kato
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Hiroaki Nakashima
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Shiro Imagama
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Hiroaki Wake
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan.
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, Myodaiji, Okazaki, 444-8585, Japan.
- Center for Optical Scattering Image Science, Kobe University, Kobe, Japan.
- Department of Physiological Sciences, Graduate University for Advanced Studies, SOKENDAI, Shonan, Hayama, Kanagawa, 240-0193, Japan.
- Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan.
| |
Collapse
|
10
|
García-Alén L, Ros-Alsina A, Sistach-Bosch L, Wright M, Kumru H. Noninvasive Electromagnetic Neuromodulation of the Central and Peripheral Nervous System for Upper-Limb Motor Strength and Functionality in Individuals with Cervical Spinal Cord Injury: A Systematic Review and Meta-Analysis. SENSORS (BASEL, SWITZERLAND) 2024; 24:4695. [PMID: 39066092 PMCID: PMC11280769 DOI: 10.3390/s24144695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024]
Abstract
(1) Background: Restoring arm and hand function is one of the priorities of people with cervical spinal cord injury (cSCI). Noninvasive electromagnetic neuromodulation is a current approach that aims to improve upper-limb function in individuals with SCI. The aim of this study is to review updated information on the different applications of noninvasive electromagnetic neuromodulation techniques that focus on restoring upper-limb functionality and motor function in people with cSCI. (2) Methods: The Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines were used to structure the search protocol. A systematic review of the literature was performed in three databases: the Cochrane Library, PubMed, and Physiotherapy Evidence Database (PEDro). (3) Results: Twenty-five studies were included: four were on transcranial magnetic stimulation (TMS), four on transcranial direct current stimulation (tDCS), two on transcutaneous spinal cord stimulation (tSCS), ten on functional electrical stimulation (FES), four on transcutaneous electrical nerve stimulation (TENS), and one on neuromuscular stimulation (NMS). The meta-analysis could not be completed due to a lack of common motor or functional evaluations. Finally, we realized a narrative review of the results, which reported that noninvasive electromagnetic neuromodulation combined with rehabilitation at the cerebral or spinal cord level significantly improved upper-limb functionality and motor function in cSCI subjects. Results were significant compared with the control group when tSCS, FES, TENS, and NMS was applied. (4) Conclusions: To perform a meta-analysis and contribute to more evidence, randomized controlled trials with standardized outcome measures for the upper extremities in cSCI are needed, even though significant improvement was reported in each non-invasive electromagnetic neuromodulation study.
Collapse
Affiliation(s)
- Loreto García-Alén
- Fundación Institut Guttmann, Institut Universitari de Neurorrehabilitació Adscrit a la UAB, 08916 Badalona, Spain; (A.R.-A.); (L.S.-B.); (M.W.)
- Universitat Autónoma de Barcelona, 08193 Barcelona, Spain
| | - Aina Ros-Alsina
- Fundación Institut Guttmann, Institut Universitari de Neurorrehabilitació Adscrit a la UAB, 08916 Badalona, Spain; (A.R.-A.); (L.S.-B.); (M.W.)
| | - Laura Sistach-Bosch
- Fundación Institut Guttmann, Institut Universitari de Neurorrehabilitació Adscrit a la UAB, 08916 Badalona, Spain; (A.R.-A.); (L.S.-B.); (M.W.)
| | - Mark Wright
- Fundación Institut Guttmann, Institut Universitari de Neurorrehabilitació Adscrit a la UAB, 08916 Badalona, Spain; (A.R.-A.); (L.S.-B.); (M.W.)
| | - Hatice Kumru
- Fundación Institut Guttmann, Institut Universitari de Neurorrehabilitació Adscrit a la UAB, 08916 Badalona, Spain; (A.R.-A.); (L.S.-B.); (M.W.)
- Universitat Autónoma de Barcelona, 08193 Barcelona, Spain
- Fundació Institut d’Investigació en Ciéncies de la Salut Germans Trias i Pujol, 08916 Badalona, Spain
| |
Collapse
|
11
|
Vorobyev AN, Burmistrova AV, Puzin KM, Varyukhina MD, Radutnaya ML, Yakovlev AA, Chmutin GE, Musa G, Chmutin EG, Grechko AV, Reyes Soto G, Catillo-Rangel C, Nurmukhametov R, Ramirez MDJE, Montemurro N. Clinical Outcome After Epidural Spinal Cord Stimulation in Patients With Severe Traumatic Brain Injury. Cureus 2024; 16:e65753. [PMID: 39211655 PMCID: PMC11361623 DOI: 10.7759/cureus.65753] [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] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION Epidural spinal cord stimulation is a minimally invasive procedure with a growing list of indications. It has a good safety profile and analgesic effect, reduces the severity of spasticity, and activates various brain regions. The purpose of this study is to evaluate the clinical outcome of epidural spinal cord stimulation in patients with spastic syndrome and chronic disorders of consciousness resulting from severe traumatic brain injury (sTBI). METHODS Between 2021 and 2023, an epidural spinal cord stimulation test was performed in 34 patients with central paresis, severe hypertonia, and chronically altered consciousness following sTBI. The severity of spastic syndrome was assessed using a modified Ashworth scale. All patients underwent implantation of a cylindrical eight-contact test epidural electrode at C3-C5 cervical level, followed by neurostimulation and selection of individual modes. Tonic stimulation at a frequency of 60 Hz, "burst" mode, or a combination of the two was used. RESULTS Epidural spinal cord stimulation was administered for an average of 4 ± 1.5 days, with tonic stimulation mode applied in 15 (44.1%) patients, "burst" mode in 10 (29.4%), and a combination of two in nine (26.5%) patients. A reduction in spasticity with clinical improvement was observed in 21 patients (61.8%). The Ashworth scale scores for distal and proximal upper extremities decreased from 3 points to 2.5 points and from 3 points to 2 points, respectively. This was significant in the right upper limbs (p = 0.0152 distally and p = 0.0164 proximally). Significant improvements were also seen in the lower extremities. Active movements in paretic limbs increased or appeared in 12 patients (35.3%), while a heightened level of consciousness was observed in six patients (17.6%). Permanent neurostimulator implantation was performed in 12 patients (35.3%), with no reported surgical complications. CONCLUSION Epidural spinal cord stimulation shows promise as an invasive rehabilitation method for patients with sTBI sequelae. Its use reduced the severity of spastic syndrome in over half of patients and increased active movements in paretic limbs in over a third. Notably, neuromodulation at the cervical level yielded pronounced effects on the upper extremities, both proximally and distally. Findings regarding consciousness level improvement are particularly intriguing but warrant further validation through randomized trials.
Collapse
Affiliation(s)
- Alexey N Vorobyev
- Reanimatology and Rehabilitation, Federal Scientific and Clinical Center for Reanimatology and Rehabilitation, Moscow, RUS
| | - Aleksandra V Burmistrova
- Reanimatology and Rehabilitation, Federal Scientific and Clinical Center for Reanimatology and Rehabilitation, Moscow, RUS
| | - Kiril M Puzin
- Reanimatology and Rehabilitation, Federal Scientific and Clinical Center for Reanimatology and Rehabilitation, Moscow, RUS
| | - Maria D Varyukhina
- Reanimatology and Rehabilitation, Federal Scientific and Clinical Center for Reanimatology and Rehabilitation, Moscow, RUS
| | - Margarita L Radutnaya
- Reanimatology and Rehabilitation, Federal Scientific and Clinical Center for Reanimatology and Rehabilitation, Moscow, RUS
| | - Alexey A Yakovlev
- Reanimatology and Rehabilitation, Federal Scientific and Clinical Center for Reanimatology and Rehabilitation, Moscow, RUS
| | - Gennady E Chmutin
- Neurological Surgery, Peoples' Friendship University of Russia, Moscow, RUS
| | - Gerald Musa
- Neurosurgery, Livingstone Central Hospital, Livingstone, ZMB
- Neurosurgery, Peoples' Friendship University of Russia, Moscow, RUS
| | - Egor G Chmutin
- Nervous Diseases and Neurosurgery, Peoples' Friendship University of Russia, Moscow, RUS
| | - Andrey V Grechko
- Reanimatology and Rehabilitation, Federal Scientific and Clinical Center for Reanimatology and Rehabilitation, Moscow, RUS
| | | | | | | | | | - Nicola Montemurro
- Neurosurgery, Azienda Ospedaliera Universitaria Pisana (AOUP) University of Pisa, Pisa, ITA
| |
Collapse
|
12
|
Sun F, Liu W, Li X, Wang X, Ou Y, Li X, Shi M. Median nerve electrical stimulation improves traumatic brain injury by reducing TACR1 to inhibit nuclear factor-κB and CCL7 activation in microglia. Histol Histopathol 2024; 39:889-902. [PMID: 38098319 DOI: 10.14670/hh-18-686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
The existing report elucidates that median nerve electrical stimulation (MNS) plays a role in treating traumatic brain injury (TBI). Herein, we explored the mechanism of MNS in TBI. A TBI-induced coma model (skull was hit by a cylindrical impact hammer) was established in adult Sprague-Dawley rats. Microglia were isolated from newborn Sprague-Dawley rats and was injured by lipopolysaccharide (LPS; 10 ng/mL). Consciousness was assessed by sensory and motor functions. Brain tissue morphology was detected using hematoxylin-eosin staining assay. Ionized calcium binding adapter molecule 1, NeuN and tachykinin receptor 1 (TACR1) level were detected by immunohistochemical assay. Levels of pro-inflammatory and anti-inflammatory factors were measured by enzyme linked immune sorbent assay (ELISA). Levels of TACR1, C-C motif chemokine 7 (CCL7), phosphorylation (p)-P65 and P65 were assessed by quantitative real time polymerase chain reaction (qRT-PCR) and western blot. M1 markers (inducible nitric oxide synthase and CD86) and M2 markers (arginase-1 (Arg1) and chitinase 3-like 3 (YM1)) of microglia as well as the transfection efficiency of short hairpin TACR1 (shTACR1) were assessed by qRT-PCR. Immunofluorescence and flow cytometry assay were used to detect microglia morphology and neuron apoptosis. MNS reduced neuron injury and microglia activation in the TBI-induced rat coma model. MNS reversed the effects of TBI on levels of inflammation-related factors, M1/M2 microglia markers, TACR1, p-P65/P65 and CCL7 in rats. shTACR1 reversed the effects of LPS on inflammation-related factors, M1/M2 microglia markers, microglia activation, neuron apoptosis, p-P65/P65 value and CCL7 level. Our results revealed that MNS improved TBI by reducing TACR1 to inhibit nuclear factor-κB (NF-κB) and CCL7 activation in microglia.
Collapse
Affiliation(s)
- Fan Sun
- Cardiopulmonary Intensive Care Rehabilitation Department, the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, PR China
| | - Wenbing Liu
- Cardiopulmonary Intensive Care Rehabilitation Department, the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, PR China
| | - Xiaodong Li
- Cardiopulmonary Intensive Care Rehabilitation Department, the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, PR China
| | - Xiaowei Wang
- Cardiopulmonary Intensive Care Rehabilitation Department, the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, PR China
| | - Yali Ou
- Cardiopulmonary Intensive Care Rehabilitation Department, the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, PR China
| | - Xuesong Li
- Cardiopulmonary Intensive Care Rehabilitation Department, Zhejiang Rehabilitation Medical Center, Hangzhou, Zhejiang Province, PR China
| | - Min Shi
- Neurology Department, the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, PR China.
| |
Collapse
|
13
|
Shimizu Y, Ntege EH, Takahara E, Matsuura N, Matsuura R, Kamizato K, Inoue Y, Sowa Y, Sunami H. Adipose-derived stem cell therapy for spinal cord injuries: Advances, challenges, and future directions. Regen Ther 2024; 26:508-519. [PMID: 39161365 PMCID: PMC11331855 DOI: 10.1016/j.reth.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 07/18/2024] [Indexed: 08/21/2024] Open
Abstract
Spinal cord injury (SCI) has limited treatment options for regaining function. Adipose-derived stem cells (ADSCs) show promise owing to their ability to differentiate into multiple cell types, promote nerve cell survival, and modulate inflammation. This review explores ADSC therapy for SCI, focusing on its potential for improving function, preclinical and early clinical trial progress, challenges, and future directions. Preclinical studies have demonstrated ADSC transplantation's effectiveness in promoting functional recovery, reducing cavity formation, and enhancing nerve regrowth and myelin repair. To improve ADSC efficacy, strategies including genetic modification and combination with rehabilitation are being explored. Early clinical trials have shown safety and feasibility, with some suggesting motor and sensory function improvements. Challenges remain for clinical translation, including optimizing cell survival and delivery, determining dosing, addressing tumor formation risks, and establishing standardized protocols. Future research should focus on overcoming these challenges and exploring the potential for combining ADSC therapy with other treatments, including rehabilitation and medication.
Collapse
Affiliation(s)
- Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Edward Hosea Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Eisaku Takahara
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Naoki Matsuura
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Rikako Matsuura
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Kota Kamizato
- Department of Anesthesiology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Yoshikazu Inoue
- Department of Plastic and Reconstructive Surgery, School of Medicine, Fujita Health University, 1-98, Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Yoshihiro Sowa
- Department of Plastic Surgery, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, 329-0498, Tochigi, Japan
| | - Hiroshi Sunami
- Center for Advanced Medical Research, School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| |
Collapse
|
14
|
Kumru H, Ros-Alsina A, García Alén L, Vidal J, Gerasimenko Y, Hernandez A, Wrigth M. Improvement in Motor and Walking Capacity during Multisegmental Transcutaneous Spinal Stimulation in Individuals with Incomplete Spinal Cord Injury. Int J Mol Sci 2024; 25:4480. [PMID: 38674065 PMCID: PMC11050444 DOI: 10.3390/ijms25084480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Transcutaneous multisegmental spinal cord stimulation (tSCS) has shown superior efficacy in modulating spinal locomotor circuits compared to single-site stimulation in individuals with spinal cord injury (SCI). Building on these findings, we hypothesized that administering a single session of tSCS at multiple spinal segments may yield greater enhancements in muscle strength and gait function during stimulation compared to tSCS at only one or two segments. In our study, tSCS was applied at single segments (C5, L1, and Coc1), two segments (C5-L1, C5-Coc1, and L1-Coc1), or multisegments (C5-L1-Coc1) in a randomized order. We evaluated the 6-m walking test (6MWT) and maximum voluntary contraction (MVC) and assessed the Hmax/Mmax ratio during stimulation in ten individuals with incomplete motor SCI. Our findings indicate that multisegmental tSCS improved walking time and reduced spinal cord excitability, as measured by the Hmax/Mmax ratio, similar to some single or two-site tSCS interventions. However, only multisegmental tSCS resulted in increased tibialis anterior (TA) muscle strength. These results suggest that multisegmental tSCS holds promise for enhancing walking capacity, increasing muscle strength, and altering spinal cord excitability in individuals with incomplete SCI.
Collapse
Affiliation(s)
- Hatice Kumru
- Fundación Institut Guttmann, Institut Universitari de NeurorehabilitacióAdscrit a la UAB, 08916 Badalona, Spain; (A.R.-A.); (L.G.A.); (J.V.); (A.H.); (M.W.)
- Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, 08916 Badalona, Spain
| | - Aina Ros-Alsina
- Fundación Institut Guttmann, Institut Universitari de NeurorehabilitacióAdscrit a la UAB, 08916 Badalona, Spain; (A.R.-A.); (L.G.A.); (J.V.); (A.H.); (M.W.)
| | - Loreto García Alén
- Fundación Institut Guttmann, Institut Universitari de NeurorehabilitacióAdscrit a la UAB, 08916 Badalona, Spain; (A.R.-A.); (L.G.A.); (J.V.); (A.H.); (M.W.)
- Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Joan Vidal
- Fundación Institut Guttmann, Institut Universitari de NeurorehabilitacióAdscrit a la UAB, 08916 Badalona, Spain; (A.R.-A.); (L.G.A.); (J.V.); (A.H.); (M.W.)
- Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, 08916 Badalona, Spain
| | - Yury Gerasimenko
- Pavlov Institute of Physiology, St. Petersburg 199034, Russia;
- Department of Physiology and Biophysics, University of Louisville, Louisville, KY 40292, USA
| | - Agusti Hernandez
- Fundación Institut Guttmann, Institut Universitari de NeurorehabilitacióAdscrit a la UAB, 08916 Badalona, Spain; (A.R.-A.); (L.G.A.); (J.V.); (A.H.); (M.W.)
- Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Mark Wrigth
- Fundación Institut Guttmann, Institut Universitari de NeurorehabilitacióAdscrit a la UAB, 08916 Badalona, Spain; (A.R.-A.); (L.G.A.); (J.V.); (A.H.); (M.W.)
- Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| |
Collapse
|
15
|
Scheuber MI, Guidolin C, Martins S, Sartori AM, Hofer AS, Schwab ME. Electrical stimulation of the cuneiform nucleus enhances the effects of rehabilitative training on locomotor recovery after incomplete spinal cord injury. Front Neurosci 2024; 18:1352742. [PMID: 38595973 PMCID: PMC11002271 DOI: 10.3389/fnins.2024.1352742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/13/2024] [Indexed: 04/11/2024] Open
Abstract
Most human spinal cord injuries are anatomically incomplete, leaving some fibers still connecting the brain with the sublesional spinal cord. Spared descending fibers of the brainstem motor control system can be activated by deep brain stimulation (DBS) of the cuneiform nucleus (CnF), a subnucleus of the mesencephalic locomotor region (MLR). The MLR is an evolutionarily highly conserved structure which initiates and controls locomotion in all vertebrates. Acute electrical stimulation experiments in female adult rats with incomplete spinal cord injury conducted in our lab showed that CnF-DBS was able to re-establish a high degree of locomotion five weeks after injury, even in animals with initially very severe functional deficits and white matter lesions up to 80-95%. Here, we analyzed whether CnF-DBS can be used to support medium-intensity locomotor training and long-term recovery in rats with large but incomplete spinal cord injuries. Rats underwent rehabilitative training sessions three times per week in an enriched environment, either with or without CnF-DBS supported hindlimb stepping. After 4 weeks, animals that trained under CnF-DBS showed a higher level of locomotor performance than rats that trained comparable distances under non-stimulated conditions. The MLR does not project to the spinal cord directly; one of its main output targets is the gigantocellular reticular nucleus in the medulla oblongata. Long-term electrical stimulation of spared reticulospinal fibers after incomplete spinal cord injury via the CnF could enhance reticulospinal anatomical rearrangement and in this way lead to persistent improvement of motor function. By analyzing the spared, BDA-labeled giganto-spinal fibers we found that their gray matter arborization density after discontinuation of CnF-DBS enhanced training was lower in the lumbar L2 and L5 spinal cord in stimulated as compared to unstimulated animals, suggesting improved pruning with stimulation-enhanced training. An on-going clinical study in chronic paraplegic patients investigates the effects of CnF-DBS on locomotor capacity.
Collapse
Affiliation(s)
- Myriam I. Scheuber
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
- ETH Phenomics Center, ETH Zurich, Zurich, Switzerland
| | - Carolina Guidolin
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
- ETH Phenomics Center, ETH Zurich, Zurich, Switzerland
| | - Suzi Martins
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
- ETH Phenomics Center, ETH Zurich, Zurich, Switzerland
| | - Andrea M. Sartori
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
- ETH Phenomics Center, ETH Zurich, Zurich, Switzerland
| | - Anna-Sophie Hofer
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
- ETH Phenomics Center, ETH Zurich, Zurich, Switzerland
- Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Martin E. Schwab
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
- ETH Phenomics Center, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
16
|
Liu J, Chen Z, Wu R, Yu H, Yang S, Xu J, Wu C, Guo Y, Hua N, Zeng X, Ma Y, Li G, Zhang L, Chen Y, Zeng Y, Ding Y, Lai B. Effects of tail nerve electrical stimulation on the activation and plasticity of the lumbar locomotor circuits and the prevention of skeletal muscle atrophy after spinal cord transection in rats. CNS Neurosci Ther 2024; 30:e14445. [PMID: 37752787 PMCID: PMC10916423 DOI: 10.1111/cns.14445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 09/28/2023] Open
Abstract
INTRODUCTION Severe spinal cord injury results in the loss of neurons in the relatively intact spinal cord below the injury area and skeletal muscle atrophy in the paralyzed limbs. These pathological processes are significant obstacles for motor function reconstruction. OBJECTIVE We performed tail nerve electrical stimulation (TNES) to activate the motor neural circuits below the injury site of the spinal cord to elucidate the regulatory mechanisms of the excitatory afferent neurons in promoting the reconstruction of locomotor function. METHODS Eight days after T10 spinal cord transection in rats, TNES was performed for 7 weeks. Behavioral scores were assessed weekly. Electrophysiological tests and double retrograde tracings were performed at week 8. RESULTS After 7 weeks of TNES treatment, there was restoration in innervation, the number of stem cells, and mitochondrial metabolism in the rats' hindlimb muscles. Double retrograde tracings of the tail nerve and sciatic nerve further confirmed the presence of synaptic connections between the tail nerve and central pattern generator (CPG) neurons in the lumbar spinal cord, as well as motor neurons innervating the hindlimb muscles. CONCLUSION The mechanisms of TNES induced by the stimulation of primary afferent nerve fibers involves efficient activation of the motor neural circuits in the lumbosacral segment, alterations of synaptic plasticity, and the improvement of muscle and nerve regeneration, which provides the structural and functional foundation for the future use of cutting-edge biological treatment strategies to restore voluntary movement of paralyzed hindlimbs.
Collapse
Affiliation(s)
- Jia‐Lin Liu
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
| | - Zheng‐Hong Chen
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
- Rehabilitation Medicine DepartmentThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Rong‐Jie Wu
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
- Shantou University Medical CollegeShantouGuangdongChina
- Department of OrthopedicsGuangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouGuangdongChina
| | - Hai‐Yang Yu
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
- Department of OrthopedicsGuangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouGuangdongChina
| | - Shang‐Bin Yang
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
- Department of Histology and EmbryologyZhongshan School of Medicine, Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Jing Xu
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
- Department of Histology and EmbryologyZhongshan School of Medicine, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Brain Function and DiseaseZhongshan School of Medicine, Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Chuang‐Ran Wu
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
- Department of OrthopedicsGuangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouGuangdongChina
| | - Yi‐Nan Guo
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
- Department of Histology and EmbryologyZhongshan School of Medicine, Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Nan Hua
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
| | - Xiang Zeng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
- Department of Histology and EmbryologyZhongshan School of Medicine, Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yuan‐Huan Ma
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
- Guangzhou First People's Hospital, Guangzhou Institute of Clinical Medicine, South China University of TechnologyGuangzhouGuangdongChina
| | - Ge Li
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart DiseaseGuangdong Provincial People's Hospital(Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouGuangdongChina
| | - Ling Zhang
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
- Rehabilitation Medicine DepartmentThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yuan‐Feng Chen
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
- Department of OrthopedicsGuangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouGuangdongChina
| | - Yuan‐Shan Zeng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
- Department of Histology and EmbryologyZhongshan School of Medicine, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Brain Function and DiseaseZhongshan School of Medicine, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Co‐innovation Center of NeuroregenerationNantong UniversityNantongJiangsuChina
| | - Ying Ding
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
- Department of Histology and EmbryologyZhongshan School of Medicine, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Brain Function and DiseaseZhongshan School of Medicine, Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Bi‐Qin Lai
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University), Ministry of EducationGuangzhouGuangdongChina
- Department of Histology and EmbryologyZhongshan School of Medicine, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Brain Function and DiseaseZhongshan School of Medicine, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Co‐innovation Center of NeuroregenerationNantong UniversityNantongJiangsuChina
| |
Collapse
|
17
|
Bonnet M, Ertlen C, Seblani M, Brezun JM, Coyle T, Cereda C, Zuccotti G, Colli M, Desouches C, Decherchi P, Carelli S, Marqueste T. Activated Human Adipose Tissue Transplantation Promotes Sensorimotor Recovery after Acute Spinal Cord Contusion in Rats. Cells 2024; 13:182. [PMID: 38247873 PMCID: PMC10814727 DOI: 10.3390/cells13020182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024] Open
Abstract
Traumatic spinal cord injuries (SCIs) often result in sensory, motor, and vegetative function loss below the injury site. Although preclinical results have been promising, significant solutions for SCI patients have not been achieved through translating repair strategies to clinical trials. In this study, we investigated the effective potential of mechanically activated lipoaspirated adipose tissue when transplanted into the epicenter of a thoracic spinal contusion. Male Sprague Dawley rats were divided into three experimental groups: SHAM (uninjured and untreated), NaCl (spinal cord contusion with NaCl application), and AF (spinal cord contusion with transplanted activated human fat). Pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) were measured to assess endogenous inflammation levels 14 days after injury. Sensorimotor recovery was monitored weekly for 12 weeks, and gait and electrophysiological analyses were performed at the end of this observational period. The results indicated that AF reduced endogenous inflammation post-SCI and there was a significant improvement in sensorimotor recovery. Moreover, activated adipose tissue also reinstated the segmental sensorimotor loop and the communication between supra- and sub-lesional spinal cord regions. This investigation highlights the efficacy of activated adipose tissue grafting in acute SCI, suggesting it is a promising therapeutic approach for spinal cord repair after traumatic contusion in humans.
Collapse
Affiliation(s)
- Maxime Bonnet
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Céline Ertlen
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Mostafa Seblani
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Jean-Michel Brezun
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Thelma Coyle
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Cristina Cereda
- Center of Functional Genomics and Rare Diseases, Department of Paediatrics, Buzzi Children’s Hospital, Via Ludovico Castelvetro 32, 20154 Milano, Italy
| | - Gianvincenzo Zuccotti
- Pediatric Clinical Research Center «Romeo ed Enrica Invernizzi», Department of Biomedical and Clinical Sciences, University of Milano (UNIMI), Via G.B. Grassi 74, 20157 Milan, Italy;
- Department of Paediatrics, Buzzi Children’s Hospital, Via Ludovico Castelvetro 32, 20154 Milano, Italy
| | - Mattia Colli
- Podgora7 Clinic, Via Podgora 7, 20122 Milano, Italy
| | - Christophe Desouches
- Clinique Phénicia—CD Esthétique, 5 Boulevard Notre Dame, F-13006 Marseille, France
| | - Patrick Decherchi
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Stephana Carelli
- Center of Functional Genomics and Rare Diseases, Department of Paediatrics, Buzzi Children’s Hospital, Via Ludovico Castelvetro 32, 20154 Milano, Italy
- Pediatric Clinical Research Center «Romeo ed Enrica Invernizzi», Department of Biomedical and Clinical Sciences, University of Milano (UNIMI), Via G.B. Grassi 74, 20157 Milan, Italy;
| | - Tanguy Marqueste
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| |
Collapse
|
18
|
Balbinot G. Neuromodulation to guide circuit reorganization with regenerative therapies in upper extremity rehabilitation following cervical spinal cord injury. FRONTIERS IN REHABILITATION SCIENCES 2024; 4:1320211. [PMID: 38234989 PMCID: PMC10791849 DOI: 10.3389/fresc.2023.1320211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/12/2023] [Indexed: 01/19/2024]
Abstract
Spinal cord injury (SCI) is a profoundly debilitating condition with no effective treatment to date. The complex response of the central nervous system (CNS) to injury and its limited regeneration capacity pose bold challenges for restoring function. Cervical SCIs are the most prevalent and regaining hand function is a top priority for individuals living with cervical SCI. A promising avenue for addressing this challenge arises from the emerging field of regenerative rehabilitation, which combines regenerative biology with physical medicine approaches. The hypothesis for optimizing gains in upper extremity function centers on the integration of targeted neurorehabilitation with novel cell- and stem cell-based therapies. However, the precise roles and synergistic effects of these components remain poorly understood, given the intricate nature of SCI and the diversity of regenerative approaches. This perspective article sheds light on the current state of regenerative rehabilitation for cervical SCI. Notably, preclinical research has yet to fully incorporate rehabilitation protocols that mimic current clinical practices, which often rely on neuromodulation strategies to activate spared circuits below the injury level. Therefore, it becomes imperative to comprehensively investigate the combined effects of neuromodulation and regenerative medicine strategies in animal models before translating these therapies to individuals with SCI. In cases of severe upper extremity paralysis, the advent of neuromodulation strategies, such as corticospinal tract (CST) and spinal cord stimulation, holds promise as the next frontier in enhancing the effectiveness of cell- and stem cell-based therapies. Future preclinical studies should explore this convergence of neuromodulation and regenerative approaches to unlock new possibilities for upper extremity treatment after SCI.
Collapse
Affiliation(s)
- Gustavo Balbinot
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Center for Advancing Neurotechnological Innovation to Application, University of Toronto, Toronto, ON, Canada
- KITE Research Institute – Toronto Rehabilitation Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
19
|
Xu J, Wu S, Huo L, Zhang Q, Liu L, Ye Z, Cao J, Ma H, Shang C, Ma C. Trigeminal nerve stimulation restores hippocampal dopamine deficiency to promote cognitive recovery in traumatic brain injury. Prog Neurobiol 2023:102477. [PMID: 37270025 DOI: 10.1016/j.pneurobio.2023.102477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/20/2023] [Accepted: 05/30/2023] [Indexed: 06/05/2023]
Abstract
Cognitive impairment (CI) is a common neurological disease resulting from traumatic brain injury (TBI). Trigeminal nerve stimulation (TNS) is an emerging, non-invasive, and effective neuromodulation therapy especially for patients suffering from brain function disorders. However, the treatment and recovery mechanisms of TNS remain poorly understood. By using combined advanced technologies, we revealed here that the neuroprotective potential of TNS to improve CI caused by TBI. The study results found that 40Hz TNS treatment has the ability to improve CI in TBI mice and communicates with central nervous system via the trigeminal ganglion (TG). Transsynaptic virus experiments revealed that TG is connected to the hippocampus (HPC) through the corticotropin-releasing hormone (CRH) neurons of paraventricular hypothalamic nucleus (PVN) and the dopamine transporter (DAT) neurons of substantia nigra pars compacta/ventral tegmental area (SNc/VTA). Mechanistically, the data showed that TNS can increase the release of dopamine in the HPC by activating the following neural circuit: TG→CRH+ PVN→DAT+ SNc/VTA → HPC. Bulk RNA sequencing confirmed changes in the expression of dopamine-related genes in the HPC. This work preliminarily explains the efficacy and mechanism of TNS and adds to the increasing evidence demonstrating that nerve stimulation is an effective method to treat neurological diseases. DATA AVAILABILITY: The data that support the findings of this study are available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Jing Xu
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510030, China
| | - Shaoling Wu
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510030, China
| | - Lifang Huo
- Guangzhou Laboratory, Guangzhou, 510005, China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Qian Zhang
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510030, China
| | - Lijiaqi Liu
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510030, China
| | - Zhimin Ye
- Guangzhou Laboratory, Guangzhou, 510005, China
| | - Jie Cao
- Guangzhou Laboratory, Guangzhou, 510005, China
| | - Haiyun Ma
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510030, China
| | - Congping Shang
- Guangzhou Laboratory, Guangzhou, 510005, China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China; School of Basic Medical Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510005, China.
| | - Chao Ma
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510030, China.
| |
Collapse
|
20
|
Lin YP, Ku CH, Chang CC, Chang ST. Effects of intravascular photobiomodulation on cognitive impairment and crossed cerebellar diaschisis in patients with traumatic brain injury: a longitudinal study. Lasers Med Sci 2023; 38:108. [PMID: 37076743 PMCID: PMC10115718 DOI: 10.1007/s10103-023-03764-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 04/04/2023] [Indexed: 04/21/2023]
Abstract
The association between intravascular photobiomodulation (iPBM) and crossed cerebellar diaschisis (CCD) and cognitive dysfunction in patients with traumatic brain injury (TBI) remains unknown. We postulate that iPBM might enable greater neurologic improvements. The objective of this study was to evaluate the clinical impact of iPBM on the prognosis of patients with TBI. In this longitudinal study, patients who were diagnosed with TBI were recruited. CCD was identified from brain perfusion images when the uptake difference of both cerebella was > 20%. Thus, two groups were identified: CCD( +) and CCD( -). All patients received general traditional physical therapy and three courses of iPBM (helium-neon laser illuminator, 632.8 nm). Treatment assemblies were conducted on weekdays for 2 consecutive weeks as a solitary treatment course. Three courses of iPBM were performed over 2-3 months, with 1-3 weeks of rest between each course. The outcomes were measured using the Rancho Los Amigos Levels of Cognitive Functioning (LCF) tool. The chi-square test was used to compare categorical variables. Generalized estimating equations were used to verify the associations of various effects between the two groups. p < 0.05 indicated a statistically significant difference. Thirty patients were included and classified into the CCD( +) and CCD( -) groups (n = 15, each group). Statistics showed that before iPBM, CCD in the CCD( +) group was 2.74 (exp 1.0081) times higher than that of CCD( -) group (p = 0.1632). After iPBM, the CCD was 0.64 (exp-0.4436) times lower in the CCD( +) group than in the CCD( -) group (p < 0.0001). Cognitive assessment revealed that, before iPBM, the CCD( +) group had a non-significantly 0.1030 lower LCF score than that of CCD( -) group (p = 0.1632). Similarly, the CCD( +) group had a non-significantly 0.0013 higher score than that of CCD( -) after iPBM treatment (p = 0.7041), indicating no significant differences between the CCD( +) or CCD( -) following iPBM and general physical therapy. CCD was less likely to appear in iPBM-treated patients. Additionally, iPBM was not associated with LCF score. Administration of iPBM could be applied in TBI patients to reduce the occurrence of CCD. The study failed to show differences in cognitive function after iPBM, which still serves as an alternative non-pharmacological intervention.
Collapse
Affiliation(s)
- Yen-Po Lin
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Medical Education and Research, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Chih-Hung Ku
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-Chiang Chang
- Department of Physical Medicine and Rehabilitation, School of Medicine, Tri-Service General Hospital, National Defense Medical Center, Neihu District, # 161, Section 6, Minquan East Road, Taipei, 114201, Taiwan
| | - Shin-Tsu Chang
- Department of Physical Medicine and Rehabilitation, School of Medicine, Tri-Service General Hospital, National Defense Medical Center, Neihu District, # 161, Section 6, Minquan East Road, Taipei, 114201, Taiwan.
- Department of Physical Medicine and Rehabilitation, Kaohsiung Veterans General Hospital, Zuoying Dist., # 386, Dazhong 1st Rd., 813414, Kaohsiung, Taiwan.
| |
Collapse
|
21
|
Diaz MJ, Root KT, Beneke A, Penev Y, Lucke-Wold B. Neurostimulation for Traumatic Brain Injury: Emerging Innovation. OBM NEUROBIOLOGY 2023; 7:161. [PMID: 36938307 PMCID: PMC10019379 DOI: 10.21926/obm.neurobiol.2301161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Traumatic brain injury (TBI) is a significant source of brain deficit and death among neurosurgical patients, with limited prospects for functional recovery in the cases of moderate-to-severe injury. Until now, the relevant body of literature on TBI intervention has focused on first-line, invasive treatment options (namely craniectomy and hematoma evacuation) with underwhelming focus on non-invasive therapies following surgical stabilization. Recent advances in our understanding of the impaired brain have encouraged deeper investigation of neurostimulation strategies, owed largely to its demonstrated livening of damaged neural circuitry and capacity to stabilize erratic network activity. The objective of the present study is to provide a scoping review of new knowledge in neurostimulation published in the PubMed, Scopus, and Google Scholar databases from inception to November 2022. We critically assess and appraise the available data on primary neurostimulation delivery techniques, with marked emphasis on restorative opportunities for accessory neurostimulation in the interdisciplinary care of moderate-to-severe TBI (msTBI) patients. These data identify two primary future directions: 1) to relate obtained gain-of-function outcomes to hemodynamic and histological changes and 2) to develop a clearer understanding of neurostimulation efficacy, when combined with pharmacologic interventions or other modulatory techniques, for complex brain insult.
Collapse
Affiliation(s)
| | | | - Alice Beneke
- College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yordan Penev
- College of Medicine, University of Florida, Gainesville, FL, USA
| | | |
Collapse
|
22
|
Skiadopoulos A, Famodimu GO, Solomon SK, Agarwal P, Harel NY, Knikou M. Priming locomotor training with transspinal stimulation in people with spinal cord injury: study protocol of a randomized clinical trial. Trials 2023; 24:145. [PMID: 36841773 PMCID: PMC9960224 DOI: 10.1186/s13063-023-07193-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 02/20/2023] [Indexed: 02/27/2023] Open
Abstract
BACKGROUND The seemingly simple tasks of standing and walking require continuous integration of complex spinal reflex circuits between descending motor commands and ascending sensory inputs. Spinal cord injury greatly impairs standing and walking ability, but both improve with locomotor training. However, even after multiple locomotor training sessions, abnormal muscle activity and coordination persist. Thus, locomotor training alone cannot fully optimize the neuronal plasticity required to strengthen the synapses connecting the brain, spinal cord, and local circuits and potentiate neuronal activity based on need. Transcutaneous spinal cord (transspinal) stimulation alters motoneuron excitability over multiple segments by bringing motoneurons closer to threshold, a prerequisite for effectively promoting spinal locomotor network neuromodulation and strengthening neural connectivity of the injured human spinal cord. Importantly, whether concurrent treatment with transspinal stimulation and locomotor training maximizes motor recovery after spinal cord injury is unknown. METHODS Forty-five individuals with chronic spinal cord injury are receiving 40 sessions of robotic gait training primed with 30 Hz transspinal stimulation at the Thoracic 10 vertebral level. Participants are randomized to receive 30 min of active or sham transspinal stimulation during standing or active transspinal stimulation while supine followed by 30 min of robotic gait training. Over the course of locomotor training, the body weight support, treadmill speed, and leg guidance force are adjusted as needed for each participant based on absence of knee buckling during the stance phase and toe dragging during the swing phase. At baseline and after completion of all therapeutic sessions, neurophysiological recordings registering corticospinal and spinal neural excitability changes along with clinical assessment measures of standing and walking, and autonomic function via questionnaires regarding bowel, bladder, and sexual function are taken. DISCUSSION The results of this mechanistic randomized clinical trial will demonstrate that tonic transspinal stimulation strengthens corticomotoneuronal connectivity and dynamic neuromodulation through posture-dependent corticospinal and spinal neuroplasticity. We anticipate that this mechanistic clinical trial will greatly impact clinical practice because, in real-world clinical settings, noninvasive transspinal stimulation can be more easily and widely implemented than invasive epidural stimulation. Additionally, by applying multiple interventions to accelerate motor recovery, we are employing a treatment regimen that reflects a true clinical approach. TRIAL REGISTRATION ClinicalTrials.gov NCT04807764 . Registered on March 19, 2021.
Collapse
Affiliation(s)
- Andreas Skiadopoulos
- grid.254498.60000 0001 2198 5185Klab4Recovery Research Program, The City University of New York, College of Staten Island, Staten Island, NY USA ,grid.254498.60000 0001 2198 5185Department of Physical Therapy, College of Staten Island, The City University of New York, Staten Island, NY USA
| | - Grace O. Famodimu
- Spinal Cord Damage Research Center, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY USA
| | - Shammah K. Solomon
- grid.254498.60000 0001 2198 5185Klab4Recovery Research Program, The City University of New York, College of Staten Island, Staten Island, NY USA ,grid.254498.60000 0001 2198 5185Department of Physical Therapy, College of Staten Island, The City University of New York, Staten Island, NY USA
| | - Parul Agarwal
- grid.59734.3c0000 0001 0670 2351Population Health Science & Policy, Institute for Health Care Delivery Science, Icahn School of Medicine at Mount Sinai, Manhattan, NY USA
| | - Noam Y. Harel
- Spinal Cord Damage Research Center, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY USA ,grid.59734.3c0000 0001 0670 2351Population Health Science & Policy, Institute for Health Care Delivery Science, Icahn School of Medicine at Mount Sinai, Manhattan, NY USA
| | - Maria Knikou
- Klab4Recovery Research Program, The City University of New York, College of Staten Island, Staten Island, NY, USA. .,Department of Physical Therapy, College of Staten Island, The City University of New York, Staten Island, NY, USA. .,PhD Program in Biology and Collaborative Neuroscience Program, Graduate Center of The City University of New York and College of Staten Island, Manhattan & Staten Island, NY, USA.
| |
Collapse
|
23
|
Skiadopoulos A, Famodimu GO, Solomon SK, Agrawal P, Harel NY, Knikou M. Priming locomotor training with transspinal stimulation in people with spinal cord injury: study protocol of a randomized clinical trial. RESEARCH SQUARE 2023:rs.3.rs-2527617. [PMID: 36824823 PMCID: PMC9949167 DOI: 10.21203/rs.3.rs-2527617/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Background The seemingly simple tasks of standing and walking require continuous integration of complex spinal reflex circuits between descending motor commands and ascending sensory inputs. Spinal cord injury greatly impairs standing and walking ability, but both improve with locomotor training. However, even after multiple locomotor training sessions, abnormal muscle activity and coordination persist. Thus, locomotor training alone cannot fully optimize the neuronal plasticity required to strengthen the synapses connecting the brain, spinal cord, and local circuits and potentiate neuronal activity based on need. Transcutaneous spinal cord (transspinal) stimulation alters motoneuron excitability over multiple segments by bringing motoneurons closer to threshold, a prerequisite for effectively promoting spinal locomotor network neuromodulation and strengthening neural connectivity of the injured human spinal cord. Importantly, whether concurrent treatment with transspinal stimulation and locomotor training maximizes motor recovery after spinal cord injury is unknown. Methods Forty-five individuals with chronic spinal cord injury are receiving 40 sessions of robotic gait training primed with 30 Hz transspinal stimulation at the Thoracic 10 vertebral level. Participants are randomized to receive 30-minutes of active or sham transspinal stimulation during standing or active transspinal stimulation while supine followed by 30-minutes of robotic gait training. Over the course of locomotor training, the body weight support, treadmill speed, and leg guidance force are adjusted as needed for each participant based on absence of knee buckling during the stance phase and toe dragging during the swing phase. At baseline and after completion of all therapeutic sessions, neurophysiological recordings registering corticospinal and spinal neural excitability changes along with clinical assessment measures of standing and walking, and autonomic function via questionnaires regarding bowel, bladder and sexual function are taken. Discussion The results of this mechanistic randomized clinical trial will demonstrate that tonic transspinal stimulation strengthens corticomotoneuronal connectivity and dynamic neuromodulation through posture-dependent corticospinal and spinal neuroplasticity. We anticipate that this mechanistic clinical trial will greatly impact clinical practice because in real-world clinical settings, noninvasive transspinal stimulation can be more easily and widely implemented than invasive epidural stimulation. Additionally, by applying multiple interventions to accelerate motor recovery, we are employing a treatment regimen that reflects a true clinical approach. Trial registration ClinicalTrials.gov: NCT04807764; Registered on March 19, 2021.
Collapse
Affiliation(s)
| | | | | | - Parul Agrawal
- Icahn School of Medicine at Mount Sinai Department of Population Health Science and Policy
| | - Noam Y Harel
- James J Peters VAMC: James J Peters VA Medical Center
| | - Maria Knikou
- College of Staten Island School of Health Sciences
| |
Collapse
|
24
|
Neuromodulation with transcutaneous spinal stimulation reveals different groups of motor profiles during robot-guided stepping in humans with incomplete spinal cord injury. Exp Brain Res 2023; 241:365-382. [PMID: 36534141 PMCID: PMC10278039 DOI: 10.1007/s00221-022-06521-5] [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: 08/16/2022] [Accepted: 12/03/2022] [Indexed: 12/23/2022]
Abstract
Neuromodulation via spinal stimulation has been investigated for improving motor function and reducing spasticity after spinal cord injury (SCI) in humans. Despite the reported heterogeneity of outcomes, few investigations have attempted to discern commonalities among individual responses to neuromodulation, especially the impact of stimulation frequencies. Here, we examined how exposure to continuous lumbosacral transcutaneous spinal stimulation (TSS) across a range of frequencies affects robotic torques and EMG patterns during stepping in a robotic gait orthosis on a motorized treadmill. We studied nine chronic motor-incomplete SCI individuals (8/1 AIS-C/D, 8 men) during robot-guided stepping with body-weight support without and with TSS applied at random frequencies between 1 and up to 100 Hz at a constant, individually selected stimulation intensity below the common motor threshold for posterior root reflexes. The hip and knee robotic torques needed to maintain the predefined stepping trajectory and EMG in eight bilateral leg muscles were recorded. We calculated the standardized mean difference between the stimulation conditions grouped into frequency bins and the no stimulation condition to determine changes in the normalized torques and the average EMG envelopes. We found heterogeneous changes in robotic torques across individuals. Agglomerative clustering of robotic torques identified four groups wherein the patterns of changes differed in magnitude and direction depending mainly on the stimulation frequency and stance/swing phase. On one end of the spectrum, the changes in robotic torques were greater with increasing stimulation frequencies (four participants), which coincided with a decrease in EMG, mainly due to the reduction of clonogenic motor output in the lower leg muscles. On the other end, we found an inverted u-shape change in torque over the mid-frequency range along with an increase in EMG, reflecting the augmentation of gait-related physiological (two participants) or pathophysiological (one participant) output. We conclude that TSS during robot-guided stepping reveals different frequency-dependent motor profiles among individuals with chronic motor incomplete SCI. This suggests the need for a better understanding and characterization of motor control profiles in SCI when applying TSS as a therapeutic intervention for improving gait.
Collapse
|
25
|
Valencia-Hernández I, González-Piña R, García-Díaz G, Ramos-Languren L, Parra-Cid C, Lomelí J, Montes S, Ríos C, Bueno-Nava A. Alpha 2-adrenergic receptor activation reinstates motor deficits in rats recovering from cortical injury. Neural Regen Res 2023; 18:875-880. [DOI: 10.4103/1673-5374.353501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
26
|
Tsai MJ, Fay LY, Liou DY, Chen Y, Chen YT, Lee MJ, Tu TH, Huang WC, Cheng H. Multifaceted Benefits of GDF11 Treatment in Spinal Cord Injury: In Vitro and In Vivo Studies. Int J Mol Sci 2022; 24:ijms24010421. [PMID: 36613862 PMCID: PMC9820576 DOI: 10.3390/ijms24010421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Traumatic spinal cord injury (SCI) initiates a series of cellular and molecular events that include both primary and secondary injury cascades. This secondary cascade provides opportunities for the delivery of therapeutic intervention. Growth differentiation factor 11 (GDF11), a member of the transforming growth factor-β (TGF-β) superfamily, regulates various biological processes in mammals. The effects of GDF11 in the nervous system were not fully elucidated. Here, we perform extensive in vitro and in vivo studies to unravel the effects of GDF11 on spinal cord after injury. In vitro culture studies showed that GDF11 increased the survival of both neuronal and oligodendroglial cells but decreased microglial cells. In stressed cultures, GDF11 effectively inhibited LPS stimulation and also protected neurons from ischemic damage. Intravenous GDF11 administration to rat after eliciting SCI significantly improved hindlimb functional restoration of SCI rats. Reduced neuronal connectivity was evident at 6 weeks post-injury and these deficits were markedly attenuated by GDF11 treatment. Furthermore, SCI-associated oligodendroglial alteration were more preserved by GDF11 treatment. Taken together, GDF11 infusion via intravenous route to SCI rats is beneficial, facilitating its therapeutic application in the future.
Collapse
Affiliation(s)
- May-Jywan Tsai
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Li-Yu Fay
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Division of Neural Regeneration and Repair, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Dann-Ying Liou
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Yi Chen
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Ya-Tzu Chen
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Meng-Jen Lee
- Department of Applied Chemistry, Chaoyang University of Technology, Taichung 41349, Taiwan
| | - Tsung-Hsi Tu
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Division of Neural Regeneration and Repair, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Wen-Cheng Huang
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Division of Neural Regeneration and Repair, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Henrich Cheng
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Division of Neural Regeneration and Repair, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Correspondence: ; Tel.: +886-2-28757718
| |
Collapse
|
27
|
Wang S, Qian W, Chen S, Xian S, Jin M, Liu Y, Zhang H, Qin H, Zhang X, Zhu J, Yue X, Shi C, Yan P, Huang R, Huang Z. Bibliometric analysis of research on gene expression in spinal cord injury. Front Mol Neurosci 2022; 15:1023692. [PMID: 36385766 PMCID: PMC9661966 DOI: 10.3389/fnmol.2022.1023692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 10/10/2022] [Indexed: 11/29/2022] Open
Abstract
Background Spinal cord injury (SCI) is a severe disease with motor and sensory function being destroyed, which leads to a poor prognosis and a serious financial burden. It is urgent to figure out the molecular and pathological mechanisms of SCI to develop feasible therapeutic strategies. This article aims to review documents focused on gene expression in SCI and summarize research hotspots and the development process in this field. Methods Publications of SCI-related studies from 2000 to 2022 were retrieved from the Web of Science Core Collection database. Biblioshiny was used to evaluate the research performance, core authors, journals and contributed countries, together with trend topics, hotspots in the field, and keyword co-occurrence analysis. Visualized images were obtained to help comprehension. Results Among 351 documents, it was found that the number of annual publications increased in general. The most productive country was China, followed by the United States with the highest influence and the most international cooperation. Plos One was the journal of the maximum publications, while Journal of Neuroscience was the most influential one. According to keyword co-occurrence and trend topics analysis, these articles mainly focused on molecular and pathological mechanisms as well as novel therapies for SCI. Neuropathic pain, axonal regeneration and messenger RNA are significant and promising research areas. Conclusion As the first bibliometric study focused on gene expression in SCI, we demonstrated the evolution of the field and provided future research directions like mechanisms and treatments of SCI with great innovativeness and clinical value. Further studies are recommended to develop more viable therapeutic methods for SCI.
Collapse
Affiliation(s)
- Siqiao Wang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Tongji University School of Medicine, Shanghai, China
| | - Weijin Qian
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaofeng Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shuyuan Xian
- Tongji University School of Medicine, Shanghai, China
| | - Minghao Jin
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifan Liu
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Zhang
- Department of Orthopedics, Naval Medical Center of PLA, Second Military Medical University Shanghai, Shanghai, China
| | - Hengwei Qin
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinkun Zhang
- Tongji University School of Medicine, Shanghai, China
| | - Jiwen Zhu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xi Yue
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chaofeng Shi
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Penghui Yan
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Zongqiang Huang, ; Runzhi Huang, ; Penghui Yan,
| | - Runzhi Huang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
- *Correspondence: Zongqiang Huang, ; Runzhi Huang, ; Penghui Yan,
| | - Zongqiang Huang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Zongqiang Huang, ; Runzhi Huang, ; Penghui Yan,
| |
Collapse
|
28
|
Hofer AS, Scheuber MI, Sartori AM, Good N, Stalder SA, Hammer N, Fricke K, Schalbetter SM, Engmann AK, Weber RZ, Rust R, Schneider MP, Russi N, Favre G, Schwab ME. Stimulation of the cuneiform nucleus enables training and boosts recovery after spinal cord injury. Brain 2022; 145:3681-3697. [PMID: 35583160 PMCID: PMC9586551 DOI: 10.1093/brain/awac184] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 04/07/2022] [Accepted: 05/04/2022] [Indexed: 11/15/2022] Open
Abstract
Severe spinal cord injuries result in permanent paraparesis in spite of the frequent sparing of small portions of white matter. Spared fibre tracts are often incapable of maintaining and modulating the activity of lower spinal motor centres. Effects of rehabilitative training thus remain limited. Here, we activated spared descending brainstem fibres by electrical deep brain stimulation of the cuneiform nucleus of the mesencephalic locomotor region, the main control centre for locomotion in the brainstem, in adult female Lewis rats. We show that deep brain stimulation of the cuneiform nucleus enhances the weak remaining motor drive in highly paraparetic rats with severe, incomplete spinal cord injuries and enables high-intensity locomotor training. Stimulation of the cuneiform nucleus during rehabilitative aquatraining after subchronic (n = 8 stimulated versus n = 7 unstimulated versus n = 7 untrained rats) and chronic (n = 14 stimulated versus n = 9 unstimulated versus n = 9 untrained rats) spinal cord injury re-established substantial locomotion and improved long-term recovery of motor function. We additionally identified a safety window of stimulation parameters ensuring context-specific locomotor control in intact rats (n = 18) and illustrate the importance of timing of treatment initiation after spinal cord injury (n = 14). This study highlights stimulation of the cuneiform nucleus as a highly promising therapeutic strategy to enhance motor recovery after subchronic and chronic incomplete spinal cord injury with direct clinical applicability.
Collapse
Affiliation(s)
- Anna-Sophie Hofer
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Myriam I Scheuber
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Andrea M Sartori
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Nicolas Good
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Stephanie A Stalder
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Nicole Hammer
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Kai Fricke
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Sina M Schalbetter
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Anne K Engmann
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Rebecca Z Weber
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Ruslan Rust
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Marc P Schneider
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Natalie Russi
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Giacomin Favre
- Department of Economics, University of Zurich, 8032 Zurich, Switzerland
| | - Martin E Schwab
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
29
|
Dietz VA, Roberts N, Knox K, Moore S, Pitonak M, Barr C, Centeno J, Leininger S, New KC, Nowell P, Rodreick M, Geoffroy CG, Stampas A, Dulin JN. Fighting for recovery on multiple fronts: The past, present, and future of clinical trials for spinal cord injury. Front Cell Neurosci 2022; 16:977679. [PMID: 36212690 PMCID: PMC9533868 DOI: 10.3389/fncel.2022.977679] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
Through many decades of preclinical research, great progress has been achieved in understanding the complex nature of spinal cord injury (SCI). Preclinical research efforts have guided and shaped clinical trials, which are growing in number by the year. Currently, 1,149 clinical trials focused on improving outcomes after SCI are registered in the U.S. National Library of Medicine at ClinicalTrials.gov. We conducted a systematic analysis of these SCI clinical trials, using publicly accessible data downloaded from ClinicalTrials.gov. After extracting all available data for these trials, we categorized each trial according to the types of interventions being tested and the types of outcomes assessed. We then evaluated clinical trial characteristics, both globally and by year, in order to understand the areas of growth and change over time. With regard to clinical trial attributes, we found that most trials have low enrollment, only test single interventions, and have limited numbers of primary outcomes. Some gaps in reporting are apparent; for instance, over 75% of clinical trials with "Completed" status do not have results posted, and the Phase of some trials is incorrectly classified as "Not applicable" despite testing a drug or biological compound. When analyzing trials based on types of interventions assessed, we identified the largest representation in trials testing rehab/training/exercise, neuromodulation, and behavioral modifications. Most highly represented primary outcomes include motor function of the upper and lower extremities, safety, and pain. The most highly represented secondary outcomes include quality of life and pain. Over the past 15 years, we identified increased representation of neuromodulation and rehabilitation trials, and decreased representation of drug trials. Overall, the number of new clinical trials initiated each year continues to grow, signifying a hopeful future for the clinical treatment of SCI. Together, our work provides a comprehensive glimpse into the past, present, and future of SCI clinical trials, and suggests areas for improvement in clinical trial reporting.
Collapse
Affiliation(s)
- Valerie A. Dietz
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Nolan Roberts
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Katelyn Knox
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Sherilynne Moore
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Michael Pitonak
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Chris Barr
- Unite 2 Fight Paralysis, Minneapolis, MN, United States
| | - Jesus Centeno
- Unite 2 Fight Paralysis, Minneapolis, MN, United States
| | | | - Kent C. New
- Unite 2 Fight Paralysis, Minneapolis, MN, United States
| | - Peter Nowell
- Unite 2 Fight Paralysis, Minneapolis, MN, United States
| | | | - Cedric G. Geoffroy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University, College Station, TX, United States
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| | - Argyrios Stampas
- Department of Physical Medicine and Rehabilitation, UTHealth Houston McGovern Medical School, Houston, TX, United States
| | - Jennifer N. Dulin
- Department of Biology, Texas A&M University, College Station, TX, United States
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| |
Collapse
|
30
|
Wireless charging-mediated angiogenesis and nerve repair by adaptable microporous hydrogels from conductive building blocks. Nat Commun 2022; 13:5172. [PMID: 36056007 PMCID: PMC9440098 DOI: 10.1038/s41467-022-32912-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/22/2022] [Indexed: 02/04/2023] Open
Abstract
Traumatic brain injury causes inflammation and glial scarring that impede brain tissue repair, so stimulating angiogenesis and recovery of brain function remain challenging. Here we present an adaptable conductive microporous hydrogel consisting of gold nanoyarn balls-coated injectable building blocks possessing interconnected pores to improve angiogenesis and recovery of brain function in traumatic brain injury. We show that following minimally invasive implantation, the adaptable hydrogel is able to fill defects with complex shapes and regulate the traumatic brain injury environment in a mouse model. We find that placement of this injectable hydrogel at peri-trauma regions enhances mature brain-derived neurotrophic factor by 180% and improves angiogenesis by 250% in vivo within 2 weeks after electromagnetized stimulation, and that these effects facilitate neuron survival and motor function recovery by 50%. We use blood oxygenation level-dependent functional neuroimaging to reveal the successful restoration of functional brain connectivity in the corticostriatal and corticolimbic circuits.
Collapse
|
31
|
Chen M, Chen Z, Xiao X, Zhou L, Fu R, Jiang X, Pang M, Xia J. Corticospinal circuit neuroplasticity may involve silent synapses: Implications for functional recovery facilitated by neuromodulation after spinal cord injury. IBRO Neurosci Rep 2022; 14:185-194. [PMID: 36824667 PMCID: PMC9941655 DOI: 10.1016/j.ibneur.2022.08.005] [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: 08/15/2022] [Indexed: 10/15/2022] Open
Abstract
Spinal cord injury (SCI) leads to devastating physical consequences, such as severe sensorimotor dysfunction even lifetime disability, by damaging the corticospinal system. The conventional opinion that SCI is intractable due to the poor regeneration of neurons in the adult central nervous system (CNS) needs to be revisited as the CNS is capable of considerable plasticity, which underlie recovery from neural injury. Substantial spontaneous neuroplasticity has been demonstrated in the corticospinal motor circuitry following SCI. Some of these plastic changes appear to be beneficial while others are detrimental toward locomotor function recovery after SCI. The beneficial corticospinal plasticity in the spared corticospinal circuits can be harnessed therapeutically by multiple contemporary neuromodulatory approaches, especially the electrical stimulation-based modalities, in an activity-dependent manner to improve functional outcomes in post-SCI rehabilitation. Silent synapse generation and unsilencing contribute to profound neuroplasticity that is implicated in a variety of neurological disorders, thus they may be involved in the corticospinal motor circuit neuroplasticity following SCI. Exploring the underlying mechanisms of silent synapse-mediated neuroplasticity in the corticospinal motor circuitry that may be exploited by neuromodulation will inform a novel direction for optimizing therapeutic repair strategies and rehabilitative interventions in SCI patients.
Collapse
Key Words
- AMPARs, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors
- BDNF, brain-derived neurotrophic factor
- BMIs, brain-machine interfaces
- CPG, central pattern generator
- CST, corticospinal tract
- Corticospinal motor circuitry
- DBS, deep brain stimulation
- ESS, epidural spinal stimulation
- MEPs, motor-evoked potentials
- NHPs, non-human primates
- NMDARs, N-methyl-d-aspartate receptors
- Neuromodulation
- Neuroplasticity
- PSNs, propriospinal neurons
- Rehabilitation
- SCI, spinal cord injury
- STDP, spike timing-dependent plasticity
- Silent synapses
- Spinal cord injury
- TBS, theta burst stimulation
- TMS, transcranial magnetic stimulation
- TrkB, tropomyosin-related kinase B
- cTBS, continuous TBS
- iTBS, intermittent TBS
- mTOR, mammalian target of rapamycin
- rTMS, repetitive TMS
- tDCS, transcranial direct current stimulation
- tcSCS, transcutaneous spinal cord stimulation
Collapse
Affiliation(s)
- Mingcong Chen
- Department of Orthopedics and Traumatology, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, China
| | - Zuxin Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS); Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong 518055, China
| | - Xiao Xiao
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence; MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200433, China
| | - Libing Zhou
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, Guangdong 510632, China
| | - Rao Fu
- Department of Anatomy, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong 518100, China
| | - Xian Jiang
- Institute of Neurological and Psychiatric Disorder, Shenzhen Bay laboratory, Shenzhen, Guangdong 518000, China
| | - Mao Pang
- Department of Spine Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, Guangdong 510630, China
| | - Jianxun Xia
- Department of Basic Medical Sciences, Yunkang School of Medicine and Health, Nanfang College, Guangzhou, Guangdong 510970, China,Corresponding author.
| |
Collapse
|
32
|
Hong KS, Khan MNA, Ghafoor U. Non-invasive transcranial electrical brain stimulation guided by functional near-infrared spectroscopy for targeted neuromodulation: A review. J Neural Eng 2022; 19. [PMID: 35905708 DOI: 10.1088/1741-2552/ac857d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/29/2022] [Indexed: 11/12/2022]
Abstract
One of the primary goals in cognitive neuroscience is to understand the neural mechanisms on which cognition is based. Researchers are trying to find how cognitive mechanisms are related to oscillations generated due to brain activity. The research focused on this topic has been considerably aided by developing non-invasive brain stimulation techniques. The dynamics of brain networks and the resultant behavior can be affected by non-invasive brain stimulation techniques, which make their use a focus of interest in many experiments and clinical fields. One essential non-invasive brain stimulation technique is transcranial electrical stimulation (tES), subdivided into transcranial direct and alternating current stimulation. tES has recently become more well-known because of the effective results achieved in treating chronic conditions. In addition, there has been exceptional progress in the interpretation and feasibility of tES techniques. Summarizing the beneficial effects of tES, this article provides an updated depiction of what has been accomplished to date, brief history, and the open questions that need to be addressed in the future. An essential issue in the field of tES is stimulation duration. This review briefly covers the stimulation durations that have been utilized in the field while monitoring the brain using functional-near infrared spectroscopy-based brain imaging.
Collapse
Affiliation(s)
- Keum-Shik Hong
- Department of Cogno-mechatronics Engineering, Pusan National University, 2 Busandaehak-ro, Geumgeong-gu, Busan, Busan, 609735, Korea (the Republic of)
| | - M N Afzal Khan
- Pusan National University, Department of Mechanical Engineering, Busan, 46241, Korea (the Republic of)
| | - Usman Ghafoor
- School of Mechanical Engineering, Pusan National University College of Engineering, room 204, Busan, 46241, Korea (the Republic of)
| |
Collapse
|
33
|
Novel Noninvasive Spinal Neuromodulation Strategy Facilitates Recovery of Stepping after Motor Complete Paraplegia. J Clin Med 2022; 11:jcm11133670. [PMID: 35806954 PMCID: PMC9267673 DOI: 10.3390/jcm11133670] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/03/2022] [Accepted: 06/16/2022] [Indexed: 02/04/2023] Open
Abstract
It has been suggested that neuroplasticity-promoting neuromodulation can restore sensory-motor pathways after spinal cord injury (SCI), reactivating the dormant locomotor neuronal circuitry. We introduce a neuro-rehabilitative approach that leverages locomotor training with multi-segmental spinal cord transcutaneous electrical stimulation (scTS). We hypothesized that scTS neuromodulates spinal networks, complementing the neuroplastic effects of locomotor training, result in a functional progression toward recovery of locomotion. We conducted a case-study to test this approach on a 27-year-old male classified as AIS A with chronic SCI. The training regimen included task-driven non-weight-bearing training (1 month) followed by weight-bearing training (2 months). Training was paired with multi-level continuous and phase-dependent scTS targeting function-specific motor pools. Results suggest a convergence of cross-lesional networks, improving kinematics during voluntary non-weight-bearing locomotor-like stepping. After weight-bearing training, coordination during stepping improved, suggesting an important role of afferent feedback in further improvement of voluntary control and reorganization of the sensory-motor brain-spinal connectome.
Collapse
|
34
|
Pierre K, Clark A, Felisma P, Weisman S, Lucke-Wold B. Neurologic Injury and Dementia: Update on Current Physiotherapeutic Intervention. ARCHIVES OF EMERGENCY MEDICINE AND CRITICAL CARE 2022; 6:1050. [PMID: 36468938 PMCID: PMC9717692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Neurologic injury and dementia can lead to devastating outcomes for patients with extended course of disease. Secondary and tertiary injury can progress and lead to continued deficits and rapid neurodegeneration. In this review, we highlight alternative strategies that can target recovery for these patients and prevent further neurologic decline. We discuss the benefit of music therapy and acupuncture. We then look at transcranlal magnetic stimulation and transcranial direct current stimulation. Finally, we look at the role of yoga and virtual reality. While several of these modalities are in their infancy, some have been used for generations. We argue for higher quality evidence to confirm effectiveness and clinical utility.
Collapse
Affiliation(s)
- Kevin Pierre
- Department of Neurosurgery, University of Florida, USA
| | - Alec Clark
- College of Medicine, University of Central Florida, USA
| | | | | | | |
Collapse
|
35
|
Estrada V, Oldenburg E, Popa O, Muller HW. Mapping the long rocky road to effective spinal cord injury therapy - A meta-review of pre-clinical and clinical research. J Neurotrauma 2022; 39:591-612. [PMID: 35196894 DOI: 10.1089/neu.2021.0298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Spinal cord injury (SCI) is a rare condition, which even after decades of research, to date still presents an incurable condition with a complex symptomatology. SCI can result in paralysis, pain, loss of sensation, bladder and sexual dysfunction, and muscle degeneration to name but a few. The large number of publications makes it difficult to keep track of current progress in the field and of the many treatment options, which have been suggested and are being proposed with increasing frequency. Scientific databases with user-oriented search options will offer possible solutions, but they are still mostly in the development phase. In this meta-analysis, we summarize and narrow down SCI therapeutic approaches applied in pre-clinical and clinical research. Statistical analyses of treatment clusters - assorted after counting annual publication numbers in PubMed and ClinicalTrials.gov databases - were performed to allow the comparison of research foci and of their translation efficacy into clinical therapy. Using the example of SCI research, our findings demonstrate the challenges that come with the accelerating research progress - an issue, which many research fields are faced with today. The analyses point out similarities and differences in the prioritization of SCI research in pre-clinical versus clinical therapy strategies. Moreover, the results demonstrate the rapidly growing importance of modern (bio-)engineering technologies.
Collapse
Affiliation(s)
- Veronica Estrada
- Heinrich Heine University Düsseldorf, 9170, Neurology, Molecular Neurobiology Laboratory, Düsseldorf, Germany;
| | - Ellen Oldenburg
- Heinrich Heine University Düsseldorf, 9170, Institute of Quantitative and Theoretical Biology, Düsseldorf, Germany;
| | - Ovidiu Popa
- Heinrich Heine University Düsseldorf, 9170, Institute of Quantitative and Theoretical Biology, Düsseldorf, Germany;
| | - Hans W Muller
- Heinrich Heine University Düsseldorf, 9170, Neurology, Düsseldorf, Germany;
| |
Collapse
|
36
|
Kiyotake EA, Martin MD, Detamore MS. Regenerative rehabilitation with conductive biomaterials for spinal cord injury. Acta Biomater 2022; 139:43-64. [PMID: 33326879 DOI: 10.1016/j.actbio.2020.12.021] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/24/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
Abstract
The individual approaches of regenerative medicine efforts alone and rehabilitation efforts alone have not yet fully restored function after severe spinal cord injury (SCI). Regenerative rehabilitation may be leveraged to promote regeneration of the spinal cord tissue, and promote reorganization of the regenerated neural pathways and intact spinal circuits for better functional recovery for SCI. Conductive biomaterials may be a linchpin that empowers the synergy between regenerative medicine and rehabilitation approaches, as electrical stimulation applied to the spinal cord could facilitate neural reorganization. In this review, we discuss current regenerative medicine approaches in clinical trials and the rehabilitation, or neuromodulation, approaches for SCI, along with their respective translational limitations. Furthermore, we review the translational potential, in a surgical context, of conductive biomaterials (e.g., conductive polymers, carbon-based materials, metallic nanoparticle-based materials) as they pertain to SCI. While pre-formed scaffolds may be difficult to translate to human contusion SCIs, injectable composites that contain blended conductive components and can form within the injury may be more translational. However, given that there are currently no in vivo SCI studies that evaluated conductive materials combined with rehabilitation approaches, we discuss several limitations of conductive biomaterials, including demonstrating safety and efficacy, that will need to be addressed in the future for conductive biomaterials to become SCI therapeutics. Even so, the use of conductive biomaterials creates a synergistic opportunity to merge the fields of regenerative medicine and rehabilitation and redefine what regenerative rehabilitation means for the spinal cord. STATEMENT OF SIGNIFICANCE: For spinal cord injury (SCI), the individual approaches of regenerative medicine and rehabilitation are insufficient to fully restore functional recovery; however, the goal of regenerative rehabilitation is to combine these two disparate fields to maximize the functional outcomes. Concepts similar to regenerative rehabilitation for SCI have been discussed in several reviews, but for the first time, this review considers how conductive biomaterials may synergize the two approaches. We cover current regenerative medicine and rehabilitation approaches for SCI, and the translational advantages and disadvantages, in a surgical context, of conductive biomaterials used in biomedical applications that may be additionally applied to SCI. Furthermore, we identify the current limitations and translational challenges for conductive biomaterials before they may become therapeutics for SCI.
Collapse
|
37
|
Remyelination trial failures: Repercussions of ignoring neurorehabilitation and exercise in repair. Mult Scler Relat Disord 2022; 58:103539. [DOI: 10.1016/j.msard.2022.103539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/13/2022] [Accepted: 01/16/2022] [Indexed: 11/18/2022]
|
38
|
Lin J, Shi J, Min X, Chen S, Zhao Y, Zhang Y, Cheng L. The GDF11 Promotes Nerve Regeneration After Sciatic Nerve Injury in Adult Rats by Promoting Axon Growth and Inhibiting Neuronal Apoptosis. Front Bioeng Biotechnol 2022; 9:803052. [PMID: 35059389 PMCID: PMC8764262 DOI: 10.3389/fbioe.2021.803052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/15/2021] [Indexed: 11/20/2022] Open
Abstract
Introduction: Sciatic nerve injury is a common injury of the nervous system. Stem cell-based therapies, drug-based therapies and rehabilitation physiotherapy therapies are currently available, but their limited therapeutic efficacy limits their use. Here, we aimed to explore a novel lentiviral-based gene therapeutic strategy and to elaborate its mechanism. Materials and Methods: Recombinant GDF11 protein was used for the in vitro treatment of dorsal root ganglion (DRG) cells. Lentivirus was used to construct a vector system for the in vivo expression of GDF11. The nerve conduction function was detected using action-evoked potentials at different time periods, and the regulatory effect of nerves on target organs was detected by weighing the gastrocnemius muscle. Immunofluorescence of NF200 and S100 was used to show the regeneration of the sciatic nerve, and myelin and Nissl staining were performed to observe the pathological features of the tissue. Western was used to validate signaling pathways. The expression of related genes was observed by qPCR and Western blotting, and cell apoptosis was detected by flow cytometry. Result: GDF11 promotes the axonal growth of DRG cells and inhibits DGR cell apoptosis in vitro. GDF11 acts by activating the Smad pathway. GDF11 promotes the recovery of damaged sciatic nerve function in rats, the regeneration of damaged sciatic nerves in rats, and myelin regeneration of damaged sciatic nerves in rats. GDF11 also exerts a protective effect on neuronal cells in rats. Conclusion: Based on the present study, we conclude that GDF11 promotes axonal growth and inhibits DRG cell apoptosis in vitro through the Smad pathway, and lentivirus-mediated GDF11 overexpression in vivo can promote the recovery of sciatic nerves after transection by promoting axonal growth and inhibiting neuronal apoptosis in the spinal cord.
Collapse
Affiliation(s)
- Junhao Lin
- Department of Orthopaedic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Shi
- Department of Orthopaedic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Cheeloo College of Medicine, Shandong University, Jinan, China.,NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiang Min
- Department of Health Management Center, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Si Chen
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan, China
| | - Yunpeng Zhao
- Department of Orthopaedic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuanqiang Zhang
- Department of Orthopaedic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lei Cheng
- Department of Orthopaedic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
39
|
Abstract
The sudden loss of movement after spinal cord injury (SCI) is life-changing and is a major impetus to study spinal cord motor system plasticity and devise novel repair strategies. This review focuses on the motor cortex and the corticospinal tract, which are key to producing voluntary movements. The motor cortex projects directly to the spinal cord, via the corticospinal tract, and indirectly, via relays in the brain stem. With loss of the corticospinal tract after SCI, the indirect paths may bypass the injury and play an important role in voluntary control. In health and after injury, the spinal cord is a key site for activity-dependent neuroplasticity of the corticospinal system. Three kinds of activity-dependent plasticity have been identified: (1) corticospinal tract axon sprouting after electrical stimulation of the motor cortex; (2) synaptic competition between corticospinal tract and proprioceptive afferent fiber terminations; and (3) long-term potentiation (LTP) at the corticospinal tract-spinal interneuron synapse. SCI damages descending motor pathway connections and, in turn, triggers a loss of down-stream activity-dependent processes. This activity loss produces spinal interneuron degeneration and several activity-dependent maladaptive changes that underly hyperreflexia, spasticity, and spasms. Animal studies show that phasic electrical and tonic direct current stimulation can be used to supplement activity after SCI to reduce the activity-dependent degenerative and maladaptive changes. Importantly, when applied chronically neuromodulation recruits spinal neuroplasticity to improve function after SCI by promoting activity-dependent corticospinal axon sprouting and synapse formation. This helps establish new functional connections and strengthens spared connections. Combining neuromodulation to promote repair and motor rehabilitation to train circuits can most effectively promote motor recovery.
Collapse
Affiliation(s)
- John H Martin
- Department of Molecular, Cellular, and Biomedical Sciences, Center for Discovery and Innovation, City University of New York School of Medicine, New York, NY, United States; Neuroscience Program, Graduate Center of the City University of New York, New York, NY, United States.
| |
Collapse
|
40
|
Denison T, Morrell MJ. Neuromodulation in 2035: The Neurology Future Forecasting Series. Neurology 2022; 98:65-72. [PMID: 35263267 PMCID: PMC8762584 DOI: 10.1212/wnl.0000000000013061] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 11/08/2021] [Indexed: 11/15/2022] Open
Abstract
Neuromodulation devices are approved in the United States for the treatment of movement disorders, epilepsy, pain, and depression, and are used off-label for other neurologic indications. By 2035, advances in our understanding of neuroanatomical networks and in the mechanism of action of stimulation, coupled with developments in material science, miniaturization, energy storage, and delivery, will expand the use of neuromodulation devices. Neuromodulation approaches are flexible and modifiable. Stimulation can be targeted to a dysfunctional brain focus, region, or network, and can be delivered as a single treatment, continuously, according to a duty cycle, or in response to physiologic changes. Programming can be titrated and modified based on the clinical response or a physiologic biomarker. In addition to keeping pace with clinical and technological developments, neurologists in 2035 will need to navigate complex ethical and economic considerations to ensure access to neuromodulation technology for a rapidly expanding population of patients. This article provides an overview of systems in use today and those that are anticipated and highlights the opportunities and challenges for the future, some of which are technical, but most of which will be addressed by learning about brain networks, and from rapidly growing experience with neuromodulation devices.
Collapse
Affiliation(s)
- Tim Denison
- From the Department of Engineering Science (T.D.), University of Oxford, UK; Department of Neurology and Neurological Sciences (M.J.M), Stanford University, CA; and NeuroPace (M.J.M), Mountain View, CA
| | - Martha J Morrell
- From the Department of Engineering Science (T.D.), University of Oxford, UK; Department of Neurology and Neurological Sciences (M.J.M), Stanford University, CA; and NeuroPace (M.J.M), Mountain View, CA.
| |
Collapse
|
41
|
Yang Q, Zhang S, Xu Z, Liu L, Fan S, Wu S, Ma C. The Effectiveness of Trigeminal Nerve Stimulation on Traumatic Brain Injury. Neuromodulation 2022; 25:1330-1337. [PMID: 35088758 DOI: 10.1016/j.neurom.2021.10.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 09/10/2021] [Accepted: 10/12/2021] [Indexed: 11/18/2022]
Abstract
OBJECTIVES Trigeminal nerve stimulation (TNS) is a promising strategy in treating diseases of the nervous system. In this study, the effects of TNS on traumatic brain injury (TBI) were investigated in a mouse model. MATERIALS AND METHODS TBI was induced using a weight-drop device, and TNS treatment was delivered in the first hour after the TBI. Twenty-four hours later, the mice's behavior, brain edema, and expression of inflammatory factors were tested. Functional magnetic resonance imaging also was used to explore the possible effects of TNS on brain activity. RESULTS TNS alleviates TBI-induced neurological dysfunction in animal behavior tests, besides protecting the blood-brain barrier and reducing the level of brain edema. TNS also effectively reduces the level of tumor necrosis factor-α and interleukin 6 and downregulates the cleaved caspase-3 signaling pathway. A series of brain areas was found to be possibly regulated by TNS, thus affecting the neural functions of animals. CONCLUSION This study elucidates the role of TNS as an effective treatment for TBI by inhibiting the occurrence of a secondary brain injury.
Collapse
Affiliation(s)
- Qian Yang
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Subo Zhang
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhen Xu
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lijiaqi Liu
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shengnuo Fan
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shaoling Wu
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chao Ma
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
42
|
Souza TR, Schallig W, Veerkamp K, Magalhães FA, Okai-Nóbrega LA, Fonseca ST, van der Krogt MM. Muscle actions on crossed and non-crossed joints during upright standing and gait: A comprehensive description based on induced acceleration analysis. J Biomech 2021; 130:110874. [PMID: 34847446 DOI: 10.1016/j.jbiomech.2021.110874] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/18/2021] [Accepted: 11/21/2021] [Indexed: 11/16/2022]
Abstract
The multibody nature of the musculoskeletal system makes each applied force potentially accelerate all body segments. Hence, muscles' actions on the kinematics of crossed and non-crossed joints should be estimated based on multibody dynamics. The objective of this study was to systematically investigate the actions of main lower limb muscles on the sagittal-plane angular kinematics of the hip, knee, and ankle joints, during upright standing and gait. Subject-specific simulations were performed to compute the muscle-tendon forces based on three-dimensional kinematic data collected from 10 able-bodied subjects during walking at preferred speed and during relaxed standing posture. A subject-scaled model consisting of the lower limb segments, 19 degrees of freedom and 92 Hill-type muscle-tendon units was used. Muscle-induced joint angular accelerations were estimated by Induced Acceleration Analysis in OpenSim. A comprehensive description of the estimated joint accelerations induced by lower limb muscles was presented, for upright standing and for the whole gait cycle. The observed muscle actions on crossed and non-crossed joints were phase- and task-specific. The main flexors and extensors for each joint were reported. Particular biarticular muscles presented actions opposite to their anatomical classification for specific joints. Antagonist muscle actions were revealed, such as the hitherto unknown opposite actions of the soleus and gastrocnemius at the ankle, and of the iliopsoas and soleus at the knee and ankle, during upright standing. Agonist actions among remote muscles were also identified. The presented muscle actions and their roles in joint kinematics of bipedal standing and walking contribute to understanding task-specific coordination.
Collapse
Affiliation(s)
- Thales R Souza
- Graduate Program in Rehabilitation Sciences, Universidade Federal de Minas Gerais (UFMG), Brazil; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Rehabilitation Medicine, Amsterdam Movement Sciences, Amsterdam, The Netherlands.
| | - Wouter Schallig
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Rehabilitation Medicine, Amsterdam Movement Sciences, Amsterdam, The Netherlands; Amsterdam UMC, University of Amsterdam, Radiology and Nuclear Medicine, Amsterdam Movement Sciences, Meibergdreef 9, Amsterdam, The Netherlands
| | - Kirsten Veerkamp
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Rehabilitation Medicine, Amsterdam Movement Sciences, Amsterdam, The Netherlands; School of Allied Health Sciences, Griffith University, Gold Coast, Australia; Griffith Centre for Biomedical & Rehabilitation Engineering (GCORE), Menzies Health Institute Queensland, Gold Coast, Australia
| | - Fabrício A Magalhães
- Graduate Program in Rehabilitation Sciences, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Liria A Okai-Nóbrega
- Graduate Program in Rehabilitation Sciences, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Sérgio T Fonseca
- Graduate Program in Rehabilitation Sciences, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Marjolein M van der Krogt
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Rehabilitation Medicine, Amsterdam Movement Sciences, Amsterdam, The Netherlands; Amsterdam UMC, University of Amsterdam, Radiology and Nuclear Medicine, Amsterdam Movement Sciences, Meibergdreef 9, Amsterdam, The Netherlands
| |
Collapse
|
43
|
Stieglitz LH, Hofer AS, Bolliger M, Oertel MF, Filli L, Willi R, Cathomen A, Meyer C, Schubert M, Hubli M, Kessler TM, Baumann CR, Imbach L, Krüsi I, Prusse A, Schwab ME, Regli L, Curt A. Deep brain stimulation for locomotion in incomplete human spinal cord injury (DBS-SCI): protocol of a prospective one-armed multi-centre study. BMJ Open 2021; 11:e047670. [PMID: 34593490 PMCID: PMC8487195 DOI: 10.1136/bmjopen-2020-047670] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 09/09/2021] [Indexed: 01/11/2023] Open
Abstract
INTRODUCTION Spinal cord injury (SCI) is a devastating condition with immediate impact on the individual's health and quality of life. Major functional recovery reaches a plateau 3-4 months after injury despite intensive rehabilitative training. To enhance training efficacy and improve long-term outcomes, the combination of rehabilitation with electrical modulation of the spinal cord and brain has recently aroused scientific interest with encouraging results. The mesencephalic locomotor region (MLR), an evolutionarily conserved brainstem locomotor command and control centre, is considered a promising target for deep brain stimulation (DBS) in patients with SCI. Experiments showed that MLR-DBS can induce locomotion in rats with spinal white matter destructions of >85%. METHODS AND ANALYSIS In this prospective one-armed multi-centre study, we investigate the safety, feasibility, and therapeutic efficacy of MLR-DBS to enable and enhance locomotor training in severely affected, subchronic and chronic American Spinal Injury Association Impairment Scale C patients in order to improve functional recovery. Patients undergo an intensive training programme with MLR-DBS while being regularly followed up until 6 months post-implantation. The acquired data of each timepoint are compared with baseline while the primary endpoint is performance in the 6-minute walking test. The clinical trial protocol was written in accordance with the Standard Protocol Items: Recommendations for Interventional Trials checklist. ETHICS AND DISSEMINATION This first in-man study investigates the therapeutic potential of MLR-DBS in SCI patients. One patient has already been implanted with electrodes and underwent MLR stimulation during locomotion. Based on the preliminary results which promise safety and feasibility, recruitment of further patients is currently ongoing. Ethical approval has been obtained from the Ethical Committee of the Canton of Zurich (case number BASEC 2016-01104) and Swissmedic (10000316). Results will be published in peer-reviewed journals and presented at conferences. TRIAL REGISTRATION NUMBER NCT03053791.
Collapse
Affiliation(s)
| | - Anna-Sophie Hofer
- Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
| | - Marc Bolliger
- Spinal Cord Injury Center, Balgrist University Hospital, Zurich, Switzerland
| | - Markus F Oertel
- Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Linard Filli
- Spinal Cord Injury Center, Balgrist University Hospital, Zurich, Switzerland
| | - Romina Willi
- Spinal Cord Injury Center, Balgrist University Hospital, Zurich, Switzerland
| | - Adrian Cathomen
- Spinal Cord Injury Center, Balgrist University Hospital, Zurich, Switzerland
| | - Christian Meyer
- Spinal Cord Injury Center, Balgrist University Hospital, Zurich, Switzerland
| | - Martin Schubert
- Spinal Cord Injury Center, Balgrist University Hospital, Zurich, Switzerland
| | - Michèle Hubli
- Spinal Cord Injury Center, Balgrist University Hospital, Zurich, Switzerland
| | - Thomas M Kessler
- Department of Neuro-Urology, Balgrist University Hospital, Zurich, Switzerland
| | | | - Lukas Imbach
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Iris Krüsi
- Spinal Cord Injury Center, Balgrist University Hospital, Zurich, Switzerland
| | - Andrea Prusse
- Spinal Cord Injury Center, Balgrist University Hospital, Zurich, Switzerland
| | - Martin E Schwab
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
| | - Luca Regli
- Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Armin Curt
- Spinal Cord Injury Center, Balgrist University Hospital, Zurich, Switzerland
| |
Collapse
|
44
|
Corticospinal Motor Circuit Plasticity After Spinal Cord Injury: Harnessing Neuroplasticity to Improve Functional Outcomes. Mol Neurobiol 2021; 58:5494-5516. [PMID: 34341881 DOI: 10.1007/s12035-021-02484-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 07/07/2021] [Indexed: 10/20/2022]
Abstract
Spinal cord injury (SCI) is a devastating condition that affects approximately 294,000 people in the USA and several millions worldwide. The corticospinal motor circuitry plays a major role in controlling skilled movements and in planning and coordinating movements in mammals and can be damaged by SCI. While axonal regeneration of injured fibers over long distances is scarce in the adult CNS, substantial spontaneous neural reorganization and plasticity in the spared corticospinal motor circuitry has been shown in experimental SCI models, associated with functional recovery. Beneficially harnessing this neuroplasticity of the corticospinal motor circuitry represents a highly promising therapeutic approach for improving locomotor outcomes after SCI. Several different strategies have been used to date for this purpose including neuromodulation (spinal cord/brain stimulation strategies and brain-machine interfaces), rehabilitative training (targeting activity-dependent plasticity), stem cells and biological scaffolds, neuroregenerative/neuroprotective pharmacotherapies, and light-based therapies like photodynamic therapy (PDT) and photobiomodulation (PMBT). This review provides an overview of the spontaneous reorganization and neuroplasticity in the corticospinal motor circuitry after SCI and summarizes the various therapeutic approaches used to beneficially harness this neuroplasticity for functional recovery after SCI in preclinical animal model and clinical human patients' studies.
Collapse
|
45
|
Kondiles BR, Wei H, Chaboub LS, Horner PJ, Wu JQ, Perlmutter SI. Transcriptome of rat subcortical white matter and spinal cord after spinal injury and cortical stimulation. Sci Data 2021; 8:175. [PMID: 34267212 PMCID: PMC8282877 DOI: 10.1038/s41597-021-00953-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 05/18/2021] [Indexed: 12/05/2022] Open
Abstract
Spinal cord injury disrupts ascending and descending neural signals causing sensory and motor dysfunction. Neuromodulation with electrical stimulation is used in both clinical and research settings to induce neural plasticity and improve functional recovery following spinal trauma. However, the mechanisms by which electrical stimulation affects recovery remain unclear. In this study we examined the effects of cortical electrical stimulation following injury on transcription at several levels of the central nervous system. We performed a unilateral, incomplete cervical spinal contusion injury in rats and delivered stimulation for one week to the contralesional motor cortex to activate the corticospinal tract and other pathways. RNA was purified from bilateral subcortical white matter and 3 levels of the spinal cord. Here we provide the complete data set in the hope that it will be useful for researchers studying electrical stimulation as a therapy to improve recovery from the deficits associated with spinal cord injury.
Collapse
Affiliation(s)
- Bethany R Kondiles
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Haichao Wei
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX, USA
| | - Lesley S Chaboub
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Philip J Horner
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Jia Qian Wu
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX, USA.
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| | - Steve I Perlmutter
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
46
|
Zhao Z, Cea C, Gelinas JN, Khodagholy D. Responsive manipulation of neural circuit pathology by fully implantable, front-end multiplexed embedded neuroelectronics. Proc Natl Acad Sci U S A 2021; 118:e2022659118. [PMID: 33972429 PMCID: PMC8157942 DOI: 10.1073/pnas.2022659118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Responsive neurostimulation is increasingly required to probe neural circuit function and treat neuropsychiatric disorders. We introduce a multiplex-then-amplify (MTA) scheme that, in contrast to current approaches (which necessitate an equal number of amplifiers as number of channels), only requires one amplifier per multiplexer, significantly reducing the number of components and the size of electronics in multichannel acquisition systems. It also enables simultaneous stimulation of arbitrary waveforms on multiple independent channels. We validated the function of MTA by developing a fully implantable, responsive embedded system that merges the ability to acquire individual neural action potentials using conformable conducting polymer-based electrodes with real-time onboard processing, low-latency arbitrary waveform stimulation, and local data storage within a miniaturized physical footprint. We verified established responsive neurostimulation protocols and developed a network intervention to suppress pathological coupling between the hippocampus and cortex during interictal epileptiform discharges. The MTA design enables effective, self-contained, chronic neural network manipulation with translational relevance to the treatment of neuropsychiatric disease.
Collapse
Affiliation(s)
- Zifang Zhao
- Department of Electrical Engineering, Columbia University, New York, NY 10027
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY 10032
| | - Claudia Cea
- Department of Electrical Engineering, Columbia University, New York, NY 10027
| | - Jennifer N Gelinas
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY 10032;
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032
| | - Dion Khodagholy
- Department of Electrical Engineering, Columbia University, New York, NY 10027;
| |
Collapse
|
47
|
Morse LR, Field-Fote EC, Contreras-Vidal J, Noble-Haeusslein LJ, Rodreick M, Shields RK, Sofroniew M, Wudlick R, Zanca JM. Meeting Proceedings for SCI 2020: Launching a Decade of Disruption in Spinal Cord Injury Research. J Neurotrauma 2021; 38:1251-1266. [PMID: 33353467 PMCID: PMC11984770 DOI: 10.1089/neu.2020.7174] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The spinal cord injury (SCI) research community has experienced great advances in discovery research, technology development, and promising clinical interventions in the past decade. To build upon these advances and maximize the benefit to persons with SCI, the National Institutes of Health (NIH) hosted a conference February 12-13, 2019 titled "SCI 2020: Launching a Decade of Disruption in Spinal Cord Injury Research." The purpose of the conference was to bring together a broad range of stakeholders, including researchers, clinicians and healthcare professionals, persons with SCI, industry partners, regulators, and funding agency representatives to break down existing communication silos. Invited speakers were asked to summarize the state of the science, assess areas of technological and community readiness, and build collaborations that could change the trajectory of research and clinical options for people with SCI. In this report, we summarize the state of the science in each of five key domains and identify the gaps in the scientific literature that need to be addressed to move the field forward.
Collapse
Affiliation(s)
- Leslie R. Morse
- Department of Rehabilitation Medicine, University of Minnesota School of Medicine, Minneapolis, Minnesota, USA
| | - Edelle C. Field-Fote
- Shepherd Center, Atlanta, Georgia, USA
- Division of Physical Therapy, Department of Rehabilitation Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jose Contreras-Vidal
- Laboratory for Non-Invasive Brain Machine Interfaces, NSF IUCRC BRAIN, Cullen College of Engineering, University of Houston, Houston, Texas, USA
| | - Linda J. Noble-Haeusslein
- Departments of Neurology and Psychology and the Institute of Neuroscience, University of Texas at Austin, Austin, Texas, USA
| | | | - Richard K. Shields
- Department of Physical Therapy and Rehabilitation Science, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Michael Sofroniew
- Department of Neurobiology, University of California, Los Angeles, California, USA
| | - Robert Wudlick
- Department of Rehabilitation Medicine, University of Minnesota School of Medicine, Minneapolis, Minnesota, USA
| | - Jeanne M. Zanca
- Spinal Cord Injury Research, Kessler Foundation, West Orange, New Jersey, USA
- Physical Medicine and Rehabilitation, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
48
|
Collagen-based scaffolds: An auspicious tool to support repair, recovery, and regeneration post spinal cord injury. Int J Pharm 2021; 601:120559. [PMID: 33831486 DOI: 10.1016/j.ijpharm.2021.120559] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/24/2021] [Accepted: 03/27/2021] [Indexed: 02/07/2023]
Abstract
Spinal cord injury (SCI) is a perplexing traumatic disease that habitually gives ride to permanent disability, motor, and sensory impairment. Despite the existence of several therapeutic approaches for the injured motor or sensory neurons, they can't promote axonal regeneration. Whether prepared by conventional or rapid prototyping techniques, scaffolds can be applied to refurbish the continuity of the injured site, by creating a suitable environment for tissue repair, axonal regeneration, and vascularization. Collagen is a multi-sourced protein, found in animals skin, tendons, cartilage, bones, and human placenta, in addition to marine biomass. Collagen is highly abundant in the extracellular matrix and is known for its biocompatibility, biodegradability, porous structure, good permeability, low immunogenicity and thus is extensively applied in the pharmaceutical, cosmetic, and food industries as well as the tissue engineering field. Collagen in scaffolds is usually functionalized with different ligands and factors such as, stem cells, embryonic or human cells to augment its binding specificity and activity. The review summarizes the significance of collagen-based scaffolds and their influence on regeneration, repair and recovery of spinal cord injuries.
Collapse
|
49
|
Park E, Lyon JG, Alvarado-Velez M, Betancur MI, Mokarram N, Shin JH, Bellamkonda RV. Enriching neural stem cell and anti-inflammatory glial phenotypes with electrical stimulation after traumatic brain injury in male rats. J Neurosci Res 2021; 99:1864-1884. [PMID: 33772860 PMCID: PMC8360147 DOI: 10.1002/jnr.24834] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/08/2021] [Indexed: 12/26/2022]
Abstract
Traumatic brain injury (TBI) by an external physical impact results in compromised brain function via undesired neuronal death. Following the injury, resident and peripheral immune cells, astrocytes, and neural stem cells (NSCs) cooperatively contribute to the recovery of the neuronal function after TBI. However, excessive pro‐inflammatory responses of immune cells, and the disappearance of endogenous NSCs at the injury site during the acute phase of TBI, can exacerbate TBI progression leading to incomplete healing. Therefore, positive outcomes may depend on early interventions to control the injury‐associated cellular milieu in the early phase of injury. Here, we explore electrical stimulation (ES) of the injury site in a rodent model (male Sprague–Dawley rats) to investigate its overall effect on the constituent brain cell phenotype and composition during the acute phase of TBI. Our data showed that a brief ES for 1 hr on day 2 of TBI promoted anti‐inflammatory phenotypes of microglia as assessed by CD206 expression and increased the population of NSCs and Nestin+ astrocytes at 7 days post‐TBI. Also, ES effectively increased the number of viable neurons when compared to the unstimulated control group. Given the salience of microglia and neural stem cells for healing after TBI, our results strongly support the potential benefit of the therapeutic use of ES during the acute phase of TBI to regulate neuroinflammation and to enhance neuroregeneration.
Collapse
Affiliation(s)
- Eunyoung Park
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Johnathan G Lyon
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Melissa Alvarado-Velez
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Martha I Betancur
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Nassir Mokarram
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Jennifer H Shin
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Ravi V Bellamkonda
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
50
|
Al’joboori Y, Massey SJ, Knight SL, Donaldson NDN, Duffell LD. The Effects of Adding Transcutaneous Spinal Cord Stimulation (tSCS) to Sit-To-Stand Training in People with Spinal Cord Injury: A Pilot Study. J Clin Med 2020; 9:jcm9092765. [PMID: 32858977 PMCID: PMC7565331 DOI: 10.3390/jcm9092765] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 02/08/2023] Open
Abstract
Spinal cord stimulation may enable recovery of volitional motor control in people with chronic Spinal Cord Injury (SCI). In this study we explored the effects of adding SCS, applied transcutaneously (tSCS) at vertebral levels T10/11, to a sit-to-stand training intervention in people with motor complete and incomplete SCI. Nine people with chronic SCI (six motor complete; three motor incomplete) participated in an 8-week intervention, incorporating three training sessions per week. Participants received either tSCS combined with sit-to-stand training (STIM) or sit-to-stand training alone (NON-STIM). Outcome measures were carried out before and after the intervention. Seven participants completed the intervention (STIM N = 5; NON-STIM N = 2). Post training, improvements in International Standards for Neurological Classification of Spinal Cord Injury (ISNCSCI) motor scores were noted in three STIM participants (range 1.0–7.0), with no change in NON-STIM participants. Recovery of volitional lower limb muscle activity and/or movement (with tSCS off) was noted in three STIM participants. Unassisted standing was not achieved in any participant, although standing with minimal assistance was achieved in one STIM participant. This pilot study has shown that the recruitment of participants, intervention and outcome measures were all feasible in this study design. However, some modifications are recommended for a larger trial.
Collapse
Affiliation(s)
- Yazi Al’joboori
- Department of Medical Physics & Biomedical Engineering, UCL, London WC1E 6BT, UK; (N.d.N.D.); (L.D.D.)
- Aspire CREATe, UCL, Stanmore HA7 4LP, UK;
- Correspondence: ; Tel.: +44-020-3108-4083
| | | | - Sarah L. Knight
- London Spinal Cord Injury Centre, Royal National Orthopaedic Hospital, Stanmore HA7 4LP, UK;
| | - Nick de N. Donaldson
- Department of Medical Physics & Biomedical Engineering, UCL, London WC1E 6BT, UK; (N.d.N.D.); (L.D.D.)
| | - Lynsey D. Duffell
- Department of Medical Physics & Biomedical Engineering, UCL, London WC1E 6BT, UK; (N.d.N.D.); (L.D.D.)
- Aspire CREATe, UCL, Stanmore HA7 4LP, UK;
| |
Collapse
|