1
|
Di Giovanni S. The neuronal-like behaviour of macrophages. Trends Cell Biol 2025:S0962-8924(25)00091-1. [PMID: 40335360 DOI: 10.1016/j.tcb.2025.04.003] [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: 02/13/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 05/09/2025]
Abstract
Tissue-resident macrophages (TRMs) contribute to both organ physiology and pathology by communicating with the nervous system and tissue-resident cells. They have been increasingly reported to be extremely plastic and adaptable, as shown by their broad range of molecular signatures and functional roles well beyond their classical duties as phagocytes. It has recently been found that macrophages can reside in proximity to muscle spindles, which are sensory neurons sensing muscle length. Surprisingly muscle spindle macrophages express neuron-like gene expression signatures and support muscle contraction by releasing glutamate. This behaviour is more in keeping with neurons rather than immune cells. Here, the potential additional roles for muscle spindle macrophages in muscle contraction and locomotion at homeostasis and in disease states will be discussed. It will also be debated whether the use of neurotransmitters by myeloid cells as shared currency to optimise cell to cell communication, to favour neural circuit development, homeostasis and repair is an exception or could rather be the rule.
Collapse
Affiliation(s)
- Simone Di Giovanni
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK.
| |
Collapse
|
2
|
Stachowski NJ, Wheel JH, Singh S, Atoche SJ, Yao L, Garcia-Ramirez DL, Giszter SF, Dougherty KJ. Activity of spinal RORβ neurons is related to functional improvements following combination treatment after complete SCI. Proc Natl Acad Sci U S A 2025; 122:e2406333122. [PMID: 40198697 PMCID: PMC12012501 DOI: 10.1073/pnas.2406333122] [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: 03/29/2024] [Accepted: 02/05/2025] [Indexed: 04/10/2025] Open
Abstract
Various strategies targeting spinal locomotor circuitry have been associated with functional improvements after spinal cord injury (SCI). However, the neuronal populations mediating beneficial effects remain largely unknown. Using a combination therapy in a mouse model of complete SCI, we show that virally delivered brain-derived neurotrophic factor (BDNF) (AAV-BDNF) activates hindlimb stepping and causes hyperreflexia, whereas submotor threshold epidural stimulation (ES) reduces BDNF-induced hyperreflexia. Given their role in gating proprioceptive afferents and as a potential convergence point of BDNF and ES, we hypothesized that an enhanced excitability of inhibitory RORβ neurons would be associated with locomotor improvements. Ex vivo spinal slice recordings from mice with a range of locomotor and hyperreflexia scores revealed that the excitability of RORβ neurons was related to functional outcome post-SCI. Mice with poor locomotor function after SCI had less excitable RORβ neurons, but the excitability of RORβ neurons was similar between the uninjured and "best stepping" SCI groups. Further, chemogenetic activation of RORβ neurons reduced BDNF-induced hyperreflexia and improved stepping, similar to ES. Our findings identify inhibitory RORβ neurons as a target population to limit hyperreflexia and enhance locomotor function after SCI.
Collapse
Affiliation(s)
- Nicholas J. Stachowski
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA19129
| | - Jaimena H. Wheel
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA19129
| | - Shayna Singh
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA19129
| | - Sebastian J. Atoche
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA19129
| | - Lihua Yao
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA19129
| | - D. Leonardo Garcia-Ramirez
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA19129
| | - Simon F. Giszter
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA19129
| | - Kimberly J. Dougherty
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA19129
| |
Collapse
|
3
|
Eneanya C, Smith GM. The Sensory Input from the External Cuneate Nucleus and Central Cervical Nucleus to the Cerebellum Refines Forelimb Movements. Cells 2025; 14:589. [PMID: 40277914 PMCID: PMC12025734 DOI: 10.3390/cells14080589] [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: 03/20/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Goal-directed reaching movements are extremely accurate to the point that the location, placement, and speed of the limbs are specific from trial to trial. These movements require descending motor commands and feedback modulation from ascending sensory information. The descending motor commands and ascending sensory information work in conjunction to ensure that the movement is accurate and precise through an error-corrected process that resides in the cerebellum. Disruptions to this information may cause errors in the precision of forelimb motor targeting. According to the previous literature, the external cuneate nucleus (ECN) and central cervical nucleus (CeCv) are responsible for conveying unconscious sensory information from the forelimbs, shoulders, and neck muscles to the cerebellum. Here, we examined the significance of the ECN and CeCv, separately, in forelimb function. In conjunction with inhibitory DREADDs (hM4Di), we observed an obstruction in single pellet reaching and grasping when ECN activity was repressed, both unilaterally and bilaterally, in normal rats. We also observed reduced reach in the grooming assessment bilaterally. We discovered that the CeCv terminates in the medial cerebellar nucleus (MCN), within the deep cerebellar nuclei (DCN), which, to the best of our knowledge, was previously not clearly defined. Together, this information provides evidence that the requirement of ascending sensory information is important in forelimb function.
Collapse
Affiliation(s)
| | - George M. Smith
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA;
| |
Collapse
|
4
|
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
|
5
|
Passini FS, Bornstein B, Rubin S, Kuperman Y, Krief S, Masschelein E, Mehlman T, Brandis A, Addadi Y, Shalom SHO, Richter EA, Yardeni T, Tirosh A, De Bock K, Zelzer E. Piezo2 in sensory neurons regulates systemic and adipose tissue metabolism. Cell Metab 2025; 37:987-1000.e6. [PMID: 39919739 DOI: 10.1016/j.cmet.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 10/23/2024] [Accepted: 12/31/2024] [Indexed: 02/09/2025]
Abstract
Systemic metabolism ensures energy homeostasis through inter-organ crosstalk regulating thermogenic adipose tissue. Unlike the well-described inductive role of the sympathetic system, the inhibitory signal ensuring energy preservation remains poorly understood. Here, we show that, via the mechanosensor Piezo2, sensory neurons regulate morphological and physiological properties of brown and beige fat and prevent systemic hypermetabolism. Targeting runt-related transcription factor 3 (Runx3)/parvalbumin (PV) sensory neurons in independent genetic mouse models resulted in a systemic metabolic phenotype characterized by reduced body fat and increased insulin sensitivity and glucose tolerance. Deletion of Piezo2 in PV sensory neurons reproduced the phenotype, protected against high-fat-diet-induced obesity, and caused adipose tissue browning and beiging, likely driven by elevated norepinephrine levels. Finding that brown and beige fat are innervated by Runx3/PV sensory neurons expressing Piezo2 suggests a model in which mechanical signals, sensed by Piezo2 in sensory neurons, protect energy storage and prevent a systemic hypermetabolic phenotype.
Collapse
Affiliation(s)
- Fabian S Passini
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| | - Bavat Bornstein
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Sarah Rubin
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Yael Kuperman
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Sharon Krief
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Evi Masschelein
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Tevie Mehlman
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander Brandis
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Yoseph Addadi
- MICC Cell Observatory, Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Shira Huri-Ohev Shalom
- Bert Strassburger Metabolic Center for Preventive Medicine, Sheba Medical Center, Tel-Hashomer, Israel
| | - Erik A Richter
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Tal Yardeni
- Bert Strassburger Metabolic Center for Preventive Medicine, Sheba Medical Center, Tel-Hashomer, Israel
| | - Amir Tirosh
- The Dalia and David Arabov Endocrinology and Diabetes Research Center, Institute of Endocrinology, Sheba Medical Center, Tel-Hashomer, Israel; Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Katrien De Bock
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
6
|
Glover JC. Hodological patterning as an organizing principle in vertebrate motor circuitry. Front Neuroanat 2025; 18:1510944. [PMID: 39844798 PMCID: PMC11750774 DOI: 10.3389/fnana.2024.1510944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Hodological patterning refers to developmental mechanisms that link the location of neurons in the brain or spinal cord to specific axonal trajectories that direct connectivity to synaptic targets either within the central nervous system or in the periphery. In vertebrate motor circuits, hodological patterning has been demonstrated at different levels, from the final motor output of somatic and preganglionic autonomic neurons targeting peripheral motoneurons and ganglion cells, to premotor inputs from spinal and brainstem neuron populations targeting the somatic motoneurons and preganglionic autonomic neurons, to cortical neurons that delegate movement commands to the brainstem and spinal neurons. In many cases molecular profiling reveals potential underlying mechanisms whereby selective gene expression creates the link between location and axon trajectory. At the cortical level, somatotopic organization suggests a potential underlying hodological patterning, but this has not been proven. This review describes examples of hodological patterning in motor circuits and covers current knowledge about how this patterning arises.
Collapse
Affiliation(s)
- Joel C. Glover
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
7
|
Godesberg V, Bockemühl T, Büschges A. Natural variability and individuality of walking behavior in Drosophila. J Exp Biol 2024; 227:jeb247878. [PMID: 39422060 DOI: 10.1242/jeb.247878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Insects use walking behavior in a large number of contexts, such as exploration, foraging, escape and pursuit, or migration. A lot is known about how nervous systems produce this behavior in general and also how certain parameters vary with regard to walking direction or speed, for instance. An aspect that has not received much attention is whether and how walking behavior varies across individuals of a particular species. To address this, we created a large corpus of kinematic walking data of many individuals of the fruit fly Drosophila. We only selected instances of straight walking in a narrow range of walking speeds to minimize the influence of high-level parameters, such as turning and walking speed, aiming to uncover more subtle aspects of variability. Using high-speed videography and automated annotation, we captured the positions of the six leg tips for thousands of steps and used principal components analysis to characterize the postural space individuals used during walking. Our analysis shows that the largest part of walking kinematics can be described by five principal components (PCs). Separation of these five PCs into a 2D and a 3D subspace divided the description of walking behavior into invariant features shared across individuals and features that relate to the specifics of individuals; the latter features can be regarded as idiosyncrasies. We also demonstrate that this approach can detect the effects of experimental interventions in an unbiased manner and that general aspects of individuality, such as the individual walking posture, can be described.
Collapse
Affiliation(s)
- Vincent Godesberg
- Department of Animal Physiology, Institute of Zoology, University of Cologne, 50674 Cologne, Germany
| | - Till Bockemühl
- Department of Animal Physiology, Institute of Zoology, University of Cologne, 50674 Cologne, Germany
| | - Ansgar Büschges
- Department of Animal Physiology, Institute of Zoology, University of Cologne, 50674 Cologne, Germany
| |
Collapse
|
8
|
Rybak IA, Shevtsova NA, Markin SN, Prilutsky BI, Frigon A. Operation regimes of spinal circuits controlling locomotion and the role of supraspinal drives and sensory feedback. eLife 2024; 13:RP98841. [PMID: 39401073 PMCID: PMC11473106 DOI: 10.7554/elife.98841] [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] [Indexed: 10/15/2024] Open
Abstract
Locomotion in mammals is directly controlled by the spinal neuronal network, operating under the control of supraspinal signals and somatosensory feedback that interact with each other. However, the functional architecture of the spinal locomotor network, its operation regimes, and the role of supraspinal and sensory feedback in different locomotor behaviors, including at different speeds, remain unclear. We developed a computational model of spinal locomotor circuits receiving supraspinal drives and limb sensory feedback that could reproduce multiple experimental data obtained in intact and spinal-transected cats during tied-belt and split-belt treadmill locomotion. We provide evidence that the spinal locomotor network operates in different regimes depending on locomotor speed. In an intact system, at slow speeds (<0.4 m/s), the spinal network operates in a non-oscillating state-machine regime and requires sensory feedback or external inputs for phase transitions. Removing sensory feedback related to limb extension prevents locomotor oscillations at slow speeds. With increasing speed and supraspinal drives, the spinal network switches to a flexor-driven oscillatory regime and then to a classical half-center regime. Following spinal transection, the model predicts that the spinal network can only operate in the state-machine regime. Our results suggest that the spinal network operates in different regimes for slow exploratory and fast escape locomotor behaviors, making use of different control mechanisms.
Collapse
Affiliation(s)
- Ilya A Rybak
- Department of Neurobiology and Anatomy, College of Medicine, Drexel UniversityPhiladelphiaUnited States
| | - Natalia A Shevtsova
- Department of Neurobiology and Anatomy, College of Medicine, Drexel UniversityPhiladelphiaUnited States
| | - Sergey N Markin
- Department of Neurobiology and Anatomy, College of Medicine, Drexel UniversityPhiladelphiaUnited States
| | - Boris I Prilutsky
- School of Biological Sciences, Georgia Institute of TechnologyAtlantaUnited States
| | - Alain Frigon
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Centre de Recherche du CHUS, Université de SherbrookeSherbrookeCanada
| |
Collapse
|
9
|
Deng L, Song N, Wang J, Wang X, Chen Y, Wu S. Effect of Intermittent Theta Burst Stimulation Dual-Target Stimulation on Lower Limb Function in Patients with Incomplete Spinal Cord Injury: A Randomized, Single-Blind, Sham-Controlled Study. World Neurosurg 2024; 190:e46-e59. [PMID: 38960308 DOI: 10.1016/j.wneu.2024.06.141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
OBJECTIVE To explore the influence of intermittent theta burst stimulation (iTBS) dual-target stimulation on lower limb function in patients with incomplete spinal cord injury (iSCI). METHODS A randomized, single -blind, sham-controlled trial was used in this study. Thirty iSCI patients with lower limb dysfunction meeting the inclusion criteria were randomly divided into a sham group and an iTBS group, with 15 cases in each group. The iTBS group received conventional rehabilitation therapy combined with iTBS dual-target stimulation on the central cerebral sulcus and the nerve root of the spinal cord injury segment. The sham group was treated with conventional rehabilitation therapy combined with iTBS dual-target sham stimulation therapy. Comprehensive functional assessment was performed on all patients before treatment, on the day 3 and day 21 of treatment. The main evaluation indicators were as follows: amplitude and latency of motor-evoked potential (MEP) in the anterior tibial muscles of both lower limbs, latency of sensory-evoked potential (SEP) of both lower limbs, knee flexor strength and knee extensor strength, lower extremity motor score (LEMS), lower extremity sensory score, spinal cord independence measure (SCIM) score, and gait parameters (stride speed, stride frequency, stride length, and ground reaction force). RESULTS On day 21 of treatment, in the iTBS group, the MEP amplitude of the anterior tibial muscles increased, the latency of MEP shortened, knee flexor strength and knee extensor strength increased, and the LEMS and SCIM score of both lower limbs increased. In addition, there were statistically significant differences in the muscle strength of the knee flexion muscle, knee extensor muscle, MEP amplitude, LEMS, and SCIM between the 2 groups (P < 0.05). Among the 10 patients who could walk with an assisted walker, the step length and step frequency of the iTBS group were increased compared with the sham group after treatment (P < 0.01), and the ground reaction force was increased (P < 0.05). There was no significant difference in the lower extremity sensory score of the lower limbs between the 2 groups (P > 0.05). CONCLUSIONS ITBS dual-target stimulation can significantly improve the motor function of both lower limbs in patients with iSCI but does not significantly improve the sensory function of both lower limbs. Therefore, this treatment mode may participate in the reconstruction and repair of some nerve circuits in patients with iSCI. In addition, iTBS dual-target stimulation can improve the ability of iSCI patients to perform daily living.
Collapse
Affiliation(s)
- Luoyi Deng
- Department of Rehabilitation Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China
| | - Ning Song
- Department of Rehabilitation Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China;; School of Clinical Medicine, Guizhou Medical University, Guiyang, PR China
| | - Jia Wang
- Department of Rehabilitation Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China
| | - Xianbin Wang
- Department of Rehabilitation Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China
| | - Yan Chen
- Department of Rehabilitation Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China
| | - Shuang Wu
- Department of Rehabilitation Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China;.
| |
Collapse
|
10
|
Zhu X, Zhang C, Hu Y, Wang Y, Xiao S, Zhu Y, Sun H, Sun J, Xu C, Xu Y, Chen Y, He X, Liu B, Liu J, Du J, Liang Y, Liu B, Li X, Jiang Y, Shen Z, Shao X, Fang J. Modulation of Comorbid Chronic Neuropathic Pain and Anxiety-Like Behaviors by Glutamatergic Neurons in the Ventrolateral Periaqueductal Gray and the Analgesic and Anxiolytic Effects of Electroacupuncture. eNeuro 2024; 11:ENEURO.0454-23.2024. [PMID: 39084906 PMCID: PMC11360982 DOI: 10.1523/eneuro.0454-23.2024] [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: 10/31/2023] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024] Open
Abstract
Comorbid chronic neuropathic pain and anxiety is a common disease that represents a major clinical challenge. The underlying mechanisms of chronic neuropathic pain and anxiety are not entirely understood, which limits the exploration of effective treatment methods. Glutamatergic neurons in the ventrolateral periaqueductal gray (vlPAG) have been implicated in regulating pain, but the potential roles of the vlPAG in neuropathic pain-induced anxiety have not been investigated. Herein, whole-cell recording and immunofluorescence showed that the excitability of CamkIIα neurons in the vlPAG (vlPAGCamkIIα+ neurons) was decreased in mice with spared nerve injury (SNI), while electroacupuncture (EA) activated these neurons. We also showed that chemogenetic inhibition of vlPAGCamkIIα+ neurons resulted in allodynia and anxiety-like behaviors in naive mice. Furthermore, chemogenetic activation of vlPAGCamkIIα+ neurons reduced anxiety-like behaviors and allodynia in mice with SNI, and EA had a similar effect in alleviating these symptoms. Nevertheless, EA combined with chemogenetic activation failed to further relieve allodynia and anxiety-like behaviors. Artificial inhibition of vlPAGCamkIIα+ neurons abolished the analgesic and anxiolytic effects of EA. Overall, our study reveals a novel mechanism of neuropathic pain-induced anxiety and shows that EA may relieve comorbid chronic neuropathic pain and anxiety by activating vlPAGCamkIIα+ neurons.
Collapse
Affiliation(s)
- Xixiao Zhu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chi Zhang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yuxin Hu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yifang Wang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Siqi Xiao
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yichen Zhu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Haiju Sun
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jing Sun
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chi Xu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yunyun Xu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yuerong Chen
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaofen He
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Boyu Liu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jinggen Liu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Junying Du
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yi Liang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Boyi Liu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaoyu Li
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yongliang Jiang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zui Shen
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaomei Shao
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jianqiao Fang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
11
|
Yoshioka N, Kurose M, Sano H, Tran DM, Chiken S, Tainaka K, Yamamura K, Kobayashi K, Nambu A, Takebayashi H. Sensory-motor circuit is a therapeutic target for dystonia musculorum mice, a model of hereditary sensory and autonomic neuropathy 6. SCIENCE ADVANCES 2024; 10:eadj9335. [PMID: 39058787 PMCID: PMC11277474 DOI: 10.1126/sciadv.adj9335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 06/25/2024] [Indexed: 07/28/2024]
Abstract
Mutations in Dystonin (DST), which encodes cytoskeletal linker proteins, cause hereditary sensory and autonomic neuropathy 6 (HSAN-VI) in humans and the dystonia musculorum (dt) phenotype in mice; however, the neuronal circuit underlying the HSAN-VI and dt phenotype is unresolved. dt mice exhibit dystonic movements accompanied by the simultaneous contraction of agonist and antagonist muscles and postnatal lethality. Here, we identified the sensory-motor circuit as a major causative neural circuit using a gene trap system that enables neural circuit-selective inactivation and restoration of Dst by Cre-mediated recombination. Sensory neuron-selective Dst deletion led to motor impairment, degeneration of proprioceptive sensory neurons, and disruption of the sensory-motor circuit. Restoration of Dst expression in sensory neurons using Cre driver mice or a single postnatal injection of Cre-expressing adeno-associated virus ameliorated sensory degeneration and improved abnormal movements. These findings demonstrate that the sensory-motor circuit is involved in the movement disorders in dt mice and that the sensory circuit is a therapeutic target for HSAN-VI.
Collapse
Affiliation(s)
- Nozomu Yoshioka
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Transdisciplinary Research Programs, Niigata University, Niigata, Japan
| | - Masayuki Kurose
- Department of Physiology, School of Dentistry, Iwate Medical University, Yahaba, Japan
- Division of Oral Physiology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hiromi Sano
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences, SOKENDAI, Okazaki, Japan
- Division of Behavioral Neuropharmacology, International Center for Brain Science, Fujita Health University, Toyoake, Japan
| | - Dang Minh Tran
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Satomi Chiken
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences, SOKENDAI, Okazaki, Japan
| | - Kazuki Tainaka
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kensuke Yamamura
- Division of Oral Physiology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki, Japan
| | - Atsushi Nambu
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences, SOKENDAI, Okazaki, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Center for Coordination of Research Facilities, Niigata University, Niigata, Japan
| |
Collapse
|
12
|
Rybak IA, Shevtsova NA, Markin SN, Prilutsky BI, Frigon A. Operation regimes of spinal circuits controlling locomotion and role of supraspinal drives and sensory feedback. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586122. [PMID: 38585778 PMCID: PMC10996463 DOI: 10.1101/2024.03.21.586122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Locomotion in mammals is directly controlled by the spinal neuronal network, operating under the control of supraspinal signals and somatosensory feedback that interact with each other. However, the functional architecture of the spinal locomotor network, its operation regimes, and the role of supraspinal and sensory feedback in different locomotor behaviors, including at different speeds, remain unclear. We developed a computational model of spinal locomotor circuits receiving supraspinal drives and limb sensory feedback that could reproduce multiple experimental data obtained in intact and spinal-transected cats during tied-belt and split-belt treadmill locomotion. We provide evidence that the spinal locomotor network operates in different regimes depending on locomotor speed. In an intact system, at slow speeds (< 0.4 m/s), the spinal network operates in a non-oscillating state-machine regime and requires sensory feedback or external inputs for phase transitions. Removing sensory feedback related to limb extension prevents locomotor oscillations at slow speeds. With increasing speed and supraspinal drives, the spinal network switches to a flexor-driven oscillatory regime and then to a classical half-center regime. Following spinal transection, the model predicts that the spinal network can only operate in the state-machine regime. Our results suggest that the spinal network operates in different regimes for slow exploratory and fast escape locomotor behaviors, making use of different control mechanisms.
Collapse
|
13
|
Lavaud S, Bichara C, D'Andola M, Yeh SH, Takeoka A. Two inhibitory neuronal classes govern acquisition and recall of spinal sensorimotor adaptation. Science 2024; 384:194-201. [PMID: 38603479 DOI: 10.1126/science.adf6801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 03/06/2024] [Indexed: 04/13/2024]
Abstract
Spinal circuits are central to movement adaptation, yet the mechanisms within the spinal cord responsible for acquiring and retaining behavior upon experience remain unclear. Using a simple conditioning paradigm, we found that dorsal inhibitory neurons are indispensable for adapting protective limb-withdrawal behavior by regulating the transmission of a specific set of somatosensory information to enhance the saliency of conditioning cues associated with limb position. By contrast, maintaining previously acquired motor adaptation required the ventral inhibitory Renshaw cells. Manipulating Renshaw cells does not affect the adaptation itself but flexibly alters the expression of adaptive behavior. These findings identify a circuit basis involving two distinct populations of spinal inhibitory neurons, which enables lasting sensorimotor adaptation independently from the brain.
Collapse
Affiliation(s)
- Simon Lavaud
- VIB-Neuroelectronics Research Flanders (NERF), 3001 Leuven, Belgium
- KU Leuven, Department of Neuroscience and Leuven Brain Institute, 3000 Leuven, Belgium
| | - Charlotte Bichara
- VIB-Neuroelectronics Research Flanders (NERF), 3001 Leuven, Belgium
- KU Leuven, Department of Neuroscience and Leuven Brain Institute, 3000 Leuven, Belgium
| | - Mattia D'Andola
- VIB-Neuroelectronics Research Flanders (NERF), 3001 Leuven, Belgium
- KU Leuven, Department of Neuroscience and Leuven Brain Institute, 3000 Leuven, Belgium
| | - Shu-Hao Yeh
- VIB-Neuroelectronics Research Flanders (NERF), 3001 Leuven, Belgium
- KU Leuven, Department of Neuroscience and Leuven Brain Institute, 3000 Leuven, Belgium
| | - Aya Takeoka
- VIB-Neuroelectronics Research Flanders (NERF), 3001 Leuven, Belgium
- KU Leuven, Department of Neuroscience and Leuven Brain Institute, 3000 Leuven, Belgium
- IMEC, 3001 Leuven, Belgium
- RIKEN Center for Brain Science, Laboratory for Motor Circuit Plasticity, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| |
Collapse
|
14
|
Dominguez-Bajo A, Clotman F. Potential Roles of Specific Subclasses of Premotor Interneurons in Spinal Cord Function Recovery after Traumatic Spinal Cord Injury in Adults. Cells 2024; 13:652. [PMID: 38667267 PMCID: PMC11048910 DOI: 10.3390/cells13080652] [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: 03/01/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
The differential expression of transcription factors during embryonic development has been selected as the main feature to define the specific subclasses of spinal interneurons. However, recent studies based on single-cell RNA sequencing and transcriptomic experiments suggest that this approach might not be appropriate in the adult spinal cord, where interneurons show overlapping expression profiles, especially in the ventral region. This constitutes a major challenge for the identification and direct targeting of specific populations that could be involved in locomotor recovery after a traumatic spinal cord injury in adults. Current experimental therapies, including electrical stimulation, training, pharmacological treatments, or cell implantation, that have resulted in improvements in locomotor behavior rely on the modulation of the activity and connectivity of interneurons located in the surroundings of the lesion core for the formation of detour circuits. However, very few publications clarify the specific identity of these cells. In this work, we review the studies where premotor interneurons were able to create new intraspinal circuits after different kinds of traumatic spinal cord injury, highlighting the difficulties encountered by researchers, to classify these populations.
Collapse
Affiliation(s)
- Ana Dominguez-Bajo
- Université catholique de Louvain, Louvain Institute of Biomolecular Science and Technology (LIBST), Animal Molecular and Cellular Biology Group (AMCB), Place Croix du Sud 4–5, 1348 Louvain la Neuve, Belgium
| | - Frédéric Clotman
- Université catholique de Louvain, Louvain Institute of Biomolecular Science and Technology (LIBST), Animal Molecular and Cellular Biology Group (AMCB), Place Croix du Sud 4–5, 1348 Louvain la Neuve, Belgium
| |
Collapse
|
15
|
Tian T, Li H, Zhang S, Yang M. Characterization of sensory and motor dysfunction and morphological alterations in late stages of type 2 diabetic mice. Front Endocrinol (Lausanne) 2024; 15:1374689. [PMID: 38532899 PMCID: PMC10964478 DOI: 10.3389/fendo.2024.1374689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/28/2024] [Indexed: 03/28/2024] Open
Abstract
Diabetic neuropathy is the most common complication of diabetes and lacks effective treatments. Although sensory dysfunction during the early stages of diabetes has been extensively studied in various animal models, the functional and morphological alterations in sensory and motor systems during late stages of diabetes remain largely unexplored. In the current work, we examined the influence of diabetes on sensory and motor function as well as morphological changes in late stages of diabetes. The obese diabetic Leprdb/db mice (db/db) were used for behavioral assessments and subsequent morphological examinations. The db/db mice exhibited severe sensory and motor behavioral defects at the age of 32 weeks, including significantly higher mechanical withdrawal threshold and thermal latency of hindpaws compared with age-matched nondiabetic control animals. The impaired response to noxious stimuli was mainly associated with the remarkable loss of epidermal sensory fibers, particularly CGRP-positive nociceptive fibers. Unexpectedly, the area of CGRP-positive terminals in the spinal dorsal horn was dramatically increased in diabetic mice, which was presumably associated with microglial activation. In addition, the db/db mice showed significantly more foot slips and took longer time during the beam-walking examination compared with controls. Meanwhile, the running duration in the rotarod test was markedly reduced in db/db mice. The observed sensorimotor deficits and motor dysfunction were largely attributed to abnormal sensory feedback and muscle atrophy as well as attenuated neuromuscular transmission in aged diabetic mice. Morphological analysis of neuromuscular junctions (NMJs) demonstrated partial denervation of NMJs and obvious fragmentation of acetylcholine receptors (AChRs). Intrafusal muscle atrophy and abnormal muscle spindle innervation were also detected in db/db mice. Additionally, the number of VGLUT1-positive excitatory boutons on motor neurons was profoundly increased in aged diabetic mice as compared to controls. Nevertheless, inhibitory synaptic inputs onto motor neurons were similar between the two groups. This excitation-inhibition imbalance in synaptic transmission might be implicated in the disturbed locomotion. Collectively, these results suggest that severe sensory and motor deficits are present in late stages of diabetes. This study contributes to our understanding of mechanisms underlying neurological dysfunction during diabetes progression and helps to identify novel therapeutic interventions for patients with diabetic neuropathy.
Collapse
Affiliation(s)
- Ting Tian
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
- Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Haofeng Li
- Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Sensen Zhang
- Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Maojun Yang
- Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
- Cryo-EM Facility Center, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
16
|
Grau JW, Hudson KE, Johnston DT, Partipilo SR. Updating perspectives on spinal cord function: motor coordination, timing, relational processing, and memory below the brain. Front Syst Neurosci 2024; 18:1184597. [PMID: 38444825 PMCID: PMC10912355 DOI: 10.3389/fnsys.2024.1184597] [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: 03/12/2023] [Accepted: 01/29/2024] [Indexed: 03/07/2024] Open
Abstract
Those studying neural systems within the brain have historically assumed that lower-level processes in the spinal cord act in a mechanical manner, to relay afferent signals and execute motor commands. From this view, abstracting temporal and environmental relations is the province of the brain. Here we review work conducted over the last 50 years that challenges this perspective, demonstrating that mechanisms within the spinal cord can organize coordinated behavior (stepping), induce a lasting change in how pain (nociceptive) signals are processed, abstract stimulus-stimulus (Pavlovian) and response-outcome (instrumental) relations, and infer whether stimuli occur in a random or regular manner. The mechanisms that underlie these processes depend upon signal pathways (e.g., NMDA receptor mediated plasticity) analogous to those implicated in brain-dependent learning and memory. New data show that spinal cord injury (SCI) can enable plasticity within the spinal cord by reducing the inhibitory effect of GABA. It is suggested that the signals relayed to the brain may contain information about environmental relations and that spinal cord systems can coordinate action in response to descending signals from the brain. We further suggest that the study of stimulus processing, learning, memory, and cognitive-like processing in the spinal cord can inform our views of brain function, providing an attractive model system. Most importantly, the work has revealed new avenues of treatment for those that have suffered a SCI.
Collapse
Affiliation(s)
- James W. Grau
- Lab of Dr. James Grau, Department of Psychological and Brain Sciences, Cellular and Behavioral Neuroscience, Texas A&M University, College Station, TX, United States
| | | | | | | |
Collapse
|
17
|
Chalif JI, Chavarro VS, Mensah E, Johnston B, Fields DP, Chalif EJ, Chiang M, Sutton O, Yong R, Trumbower R, Lu Y. Epidural Spinal Cord Stimulation for Spinal Cord Injury in Humans: A Systematic Review. J Clin Med 2024; 13:1090. [PMID: 38398403 PMCID: PMC10889415 DOI: 10.3390/jcm13041090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
(1) Background: Spinal cord injury (SCI) represents a major health challenge, often leading to significant and permanent sensorimotor and autonomic dysfunctions. This study reviews the evolving role of epidural spinal cord stimulation (eSCS) in treating chronic SCI, focusing on its efficacy and safety. The objective was to analyze how eSCS contributes to the recovery of neurological functions in SCI patients. (2) Methods: We utilized the PRISMA guidelines and performed a comprehensive search across MEDLINE/PubMed, Embase, Web of Science, and IEEE Xplore databases up until September 2023. We identified studies relevant to eSCS in SCI and extracted assessments of locomotor, cardiovascular, pulmonary, and genitourinary functions. (3) Results: A total of 64 studies encompassing 306 patients were identified. Studies investigated various stimulation devices, parameters, and rehabilitation methods. Results indicated significant improvements in motor function: 44% of patients achieved assisted or independent stepping or standing; 87% showed enhanced muscle activity; 65% experienced faster walking speeds; and 80% improved in overground walking. Additionally, eSCS led to better autonomic function, evidenced by improvements in bladder and sexual functions, airway pressures, and bowel movements. Notable adverse effects included device migration, infections, and post-implant autonomic dysreflexia, although these were infrequent. (4) Conclusion: Epidural spinal cord stimulation is emerging as an effective and generally safe treatment for chronic SCI, particularly when combined with intensive physical rehabilitation. Future research on standardized stimulation parameters and well-defined therapy regimens will optimize benefits for specific patient populations.
Collapse
Affiliation(s)
- J. I. Chalif
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (J.I.C.); (V.S.C.); (B.J.)
- Harvard Medical School, Boston, MA 02115, USA; (M.C.); (R.Y.); (R.T.)
| | - V. S. Chavarro
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (J.I.C.); (V.S.C.); (B.J.)
- Harvard Medical School, Boston, MA 02115, USA; (M.C.); (R.Y.); (R.T.)
- Department of Physical Medicine and Rehabilitation, Spaulding Hospital Cambridge, Cambridge, MA 02115, USA
| | - E. Mensah
- Chan School of Public Health, Harvard University, Boston, MA 02115, USA;
| | - B. Johnston
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (J.I.C.); (V.S.C.); (B.J.)
- Harvard Medical School, Boston, MA 02115, USA; (M.C.); (R.Y.); (R.T.)
| | - D. P. Fields
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - E. J. Chalif
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (J.I.C.); (V.S.C.); (B.J.)
- Harvard Medical School, Boston, MA 02115, USA; (M.C.); (R.Y.); (R.T.)
| | - M. Chiang
- Harvard Medical School, Boston, MA 02115, USA; (M.C.); (R.Y.); (R.T.)
- Department of Physical Medicine and Rehabilitation, Spaulding Hospital Cambridge, Cambridge, MA 02115, USA
- Department of Anesthesiology Perioperative and Pain Management, Brigham and Women’s Hospital, Boston, MA 02115, USA;
| | - O. Sutton
- Department of Anesthesiology Perioperative and Pain Management, Brigham and Women’s Hospital, Boston, MA 02115, USA;
| | - R. Yong
- Harvard Medical School, Boston, MA 02115, USA; (M.C.); (R.Y.); (R.T.)
- Department of Anesthesiology Perioperative and Pain Management, Brigham and Women’s Hospital, Boston, MA 02115, USA;
| | - R. Trumbower
- Harvard Medical School, Boston, MA 02115, USA; (M.C.); (R.Y.); (R.T.)
- Department of Physical Medicine and Rehabilitation, Spaulding Hospital Cambridge, Cambridge, MA 02115, USA
| | - Y. Lu
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (J.I.C.); (V.S.C.); (B.J.)
- Harvard Medical School, Boston, MA 02115, USA; (M.C.); (R.Y.); (R.T.)
| |
Collapse
|
18
|
Yang HB, Li Y, Li XH, Yan QM, Han XZ, Cao J, Sang HP, Li JL. The compensatory increase of Gli-similar 3 inhibited neuronal apoptosis through regulating Mps one binder kinase activator 1b (MOB1b): a possible strategy for the functional recovery after spinal cord injury. Exp Anim 2024; 73:61-72. [PMID: 37574269 PMCID: PMC10877155 DOI: 10.1538/expanim.23-0041] [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: 03/16/2023] [Accepted: 08/02/2023] [Indexed: 08/15/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating disease characterized by neuronal apoptosis. Gli-similar 3 (GLIS3), a transcriptional factor, was involved in cell apoptosis and associated with the transcription of downstream target genes related to neuronal function. However, the function of GLIS3 in SCI remains unknown. Therefore, we used the mouse model of SCI to explore the role of GLIS3 in SCI. The results showed that GLIS3 expression was significantly increased in spinal cord tissues of SCI mice, and GLIS3 overexpression promoted the functional recovery, reserved histological changes, and inhibited neuronal apoptosis after SCI. Through online tools, the potential target genes of GLIS3 were analyzed and we found that Mps one binder kinase activator 1b (Mob1b) had a strong association with SCI among these genes. MOB1b is a core component of Hippo signaling pathway, which was reported to inhibit cell apoptosis. MOB1b expression was significantly increased in mice at 7 days post-SCI and GLIS3 overexpression further increased its expression. Dual-luciferase reporter assay revealed that GLIS3 bound to the promoter of Mob1b and promoted its transcription. In conclusion, our findings reveal that the compensatory increase of GLIS3 promotes functional recovery after SCI through inhibiting neuronal apoptosis by transcriptionally regulating MOB1b. Our study provides a novel target for functional recovery after SCI.
Collapse
Affiliation(s)
- Hong-Bo Yang
- Department of Orthopedics, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan District, Chifeng, Inner Mongolia, 024000, P.R. China
- Institute of Orthopedic Diseases, Chifeng University, No.1 Yingbin Road, Hongshan District, Chifeng, 024000, P.R. China
| | - Ying Li
- Department of Neurology, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan District, Chifeng, 024000, P.R. China
| | - Xiu-Hai Li
- Department of Orthopedics, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan District, Chifeng, Inner Mongolia, 024000, P.R. China
| | - Qing-Ming Yan
- Department of Orthopedics, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan District, Chifeng, Inner Mongolia, 024000, P.R. China
| | - Xian-Zhang Han
- Department of Orthopedics, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan District, Chifeng, Inner Mongolia, 024000, P.R. China
| | - Jian Cao
- Department of Orthopedics, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan District, Chifeng, Inner Mongolia, 024000, P.R. China
- Institute of Orthopedic Diseases, Chifeng University, No.1 Yingbin Road, Hongshan District, Chifeng, 024000, P.R. China
| | - Hong-Peng Sang
- Department of Orthopedics, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan District, Chifeng, Inner Mongolia, 024000, P.R. China
- Institute of Orthopedic Diseases, Chifeng University, No.1 Yingbin Road, Hongshan District, Chifeng, 024000, P.R. China
| | - Jin-Long Li
- Department of Orthopedics, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan District, Chifeng, Inner Mongolia, 024000, P.R. China
- Institute of Orthopedic Diseases, Chifeng University, No.1 Yingbin Road, Hongshan District, Chifeng, 024000, P.R. China
| |
Collapse
|
19
|
Ghai S, Ghai I, Narciss S. Influence of taping on force sense accuracy: a systematic review with between and within group meta-analysis. BMC Sports Sci Med Rehabil 2023; 15:138. [PMID: 37864268 PMCID: PMC10588111 DOI: 10.1186/s13102-023-00740-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/20/2023] [Indexed: 10/22/2023]
Abstract
Taping is a common technique used to address proprioceptive deficits in both healthy and patient population groups. Although there is increasing interest in taping to address proprioceptive deficits, little is known about its effects on the kinetic aspects of proprioception as measured by force sense accuracy. To address this gap in the literature, the present systematic review and meta-analysis was conducted to evaluate the impact of taping on force sense accuracy. A search for relevant literature was conducted following PRISMA guidelines across seven databases and one register. Eleven studies with 279 participants were included in the review out of 7362 records. In the between-group analyses, we found a significant improvement in absolute (p < 0.01) and relative (p = 0.01) force sense accuracy with taping compared to no comparator. Likewise, a significant improvement in absolute (p = 0.01) force sense accuracy was also observed with taping compared to placebo tape. In the within group analysis, this reduction in the absolute (p = 0.11) force sense accuracy was not significant. Additional exploratory subgroup analyses revealed between group improvement in force sense accuracy in both healthy individuals and individuals affected by medial epicondylitis. The findings of this meta-analysis should be interpreted with caution due to the limited number of studies and a lack of blinded randomized controlled trials, which may impact the generalizability of the results. More high-quality research is needed to confirm the overall effect of taping on force sense accuracy.
Collapse
Affiliation(s)
- Shashank Ghai
- Department of Political, Historical, Religious and Cultural Studies, Karlstad University, Karlstad, Sweden.
- Centre for Societal Risk Research, Karlstad University, Karlstad, Sweden.
- Psychology of Learning and Instruction, Department of Psychology, School of Science, Technische Universität Dresden, Dresden, Germany.
- Centre for Tactile Internet With Human-in-the-Loop (CeTI), Technische Universität Dresden, Dresden, Germany.
| | - Ishan Ghai
- School of Life Sciences, Jacobs University Bremen, Bremen, Germany
| | - Susanne Narciss
- Psychology of Learning and Instruction, Department of Psychology, School of Science, Technische Universität Dresden, Dresden, Germany
- Centre for Tactile Internet With Human-in-the-Loop (CeTI), Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
20
|
Danner SM, Shepard CT, Hainline C, Shevtsova NA, Rybak IA, Magnuson DSK. Spinal control of locomotion before and after spinal cord injury. Exp Neurol 2023; 368:114496. [PMID: 37499972 PMCID: PMC10529867 DOI: 10.1016/j.expneurol.2023.114496] [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: 03/25/2023] [Revised: 07/10/2023] [Accepted: 07/23/2023] [Indexed: 07/29/2023]
Abstract
Thoracic spinal cord injury affects long propriospinal neurons that interconnect the cervical and lumbar enlargements. These neurons are crucial for coordinating forelimb and hindlimb locomotor movements in a speed-dependent manner. However, recovery from spinal cord injury is usually studied over a very limited range of speeds that may not fully expose circuitry dysfunction. To overcome this limitation, we investigated overground locomotion in rats trained to move over an extended distance with a wide range of speeds both pre-injury and after recovery from thoracic hemisection or contusion injuries. In this experimental context, intact rats expressed a speed-dependent continuum of alternating (walk and trot) and non-alternating (canter, gallop, half-bound gallop, and bound) gaits. After a lateral hemisection injury, rats recovered the ability to locomote over a wide range of speeds but lost the ability to use the highest-speed gaits (half-bound gallop and bound) and predominantly used the limb contralateral to the injury as lead during canter and gallop. A moderate contusion injury caused a greater reduction in maximal speed, loss of all non-alternating gaits, and emergence of novel alternating gaits. These changes resulted from weak fore-hind coupling together with appropriate control of left-right alternation. After hemisection, animals expressed a subset of intact gaits with appropriate interlimb coordination even on the side of the injury, where the long propriospinal connections were severed. These observations highlight how investigating locomotion over the full range of speeds can reveal otherwise hidden aspects of spinal locomotor control and post-injury recovery.
Collapse
Affiliation(s)
- Simon M Danner
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, USA.
| | - Courtney T Shepard
- Interdisciplinary Program in Translational Neuroscience, University of Louisville School of Medicine, Health Sciences Campus, Louisville, KY, USA
| | - Casey Hainline
- Speed School of Engineering, University of Louisville School of Medicine, Health Sciences Campus, Louisville, KY, USA
| | - Natalia A Shevtsova
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Ilya A Rybak
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - David S K Magnuson
- Department of Neurological Surgery, University of Louisville School of Medicine, Health Sciences Campus, Louisville, KY, USA; Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Health Sciences Campus, Louisville, KY, USA
| |
Collapse
|
21
|
Lin S, Hari K, Black S, Khatmi A, Fouad K, Gorassini MA, Li Y, Lucas-Osma AM, Fenrich KK, Bennett DJ. Locomotor-related propriospinal V3 neurons produce primary afferent depolarization and modulate sensory transmission to motoneurons. J Neurophysiol 2023; 130:799-823. [PMID: 37609680 PMCID: PMC10650670 DOI: 10.1152/jn.00482.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/24/2023] Open
Abstract
When a muscle is stretched, sensory feedback not only causes reflexes but also leads to a depolarization of sensory afferents throughout the spinal cord (primary afferent depolarization, PAD), readying the whole limb for further disturbances. This sensory-evoked PAD is thought to be mediated by a trisynaptic circuit, where sensory input activates first-order excitatory neurons that activate GABAergic neurons that in turn activate GABAA receptors on afferents to cause PAD, though the identity of these first-order neurons is unclear. Here, we show that these first-order neurons include propriospinal V3 neurons, as they receive extensive sensory input and in turn innervate GABAergic neurons that cause PAD, because optogenetic activation or inhibition of V3 neurons in mice mimics or inhibits sensory-evoked PAD, respectively. Furthermore, persistent inward sodium currents intrinsic to V3 neurons prolong their activity, explaining the prolonged duration of PAD. Also, local optogenetic activation of V3 neurons at one segment causes PAD in other segments, due to the long propriospinal tracts of these neurons, helping to explain the radiating nature of PAD. This in turn facilitates monosynaptic reflex transmission to motoneurons across the spinal cord. In addition, V3 neurons directly innervate proprioceptive afferents (including Ia), causing a glutamate receptor-mediated PAD (glutamate PAD). Finally, increasing the spinal cord excitability with either GABAA receptor blockers or chronic spinal cord injury causes an increase in the glutamate PAD. Overall, we show the V3 neuron has a prominent role in modulating sensory transmission, in addition to its previously described role in locomotion.NEW & NOTEWORTHY Locomotor-related propriospinal neurons depolarize sensory axons throughout the spinal cord by either direct glutamatergic axoaxonic contacts or indirect innervation of GABAergic neurons that themselves form axoaxonic contacts on sensory axons. This depolarization (PAD) increases sensory transmission to motoneurons throughout the spinal cord, readying the sensorimotor system for external disturbances. The glutamate-mediated PAD is particularly adaptable, increasing with either an acute block of GABA receptors or chronic spinal cord injury, suggesting a role in motor recovery.
Collapse
Affiliation(s)
- Shihao Lin
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Krishnapriya Hari
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Sophie Black
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Aysan Khatmi
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Karim Fouad
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Monica A Gorassini
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Yaqing Li
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Ana M Lucas-Osma
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Keith K Fenrich
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - David J Bennett
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
22
|
O'Keeffe C, Gill C, Etzelmueller M, Taylor C, Hablani S, Reilly RB, Fleming N. Multimodal analysis of the biomechanical impact of knee angle on the Sit-to-Stand transition. Gait Posture 2023; 105:125-131. [PMID: 37542885 DOI: 10.1016/j.gaitpost.2023.07.283] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/07/2023]
Abstract
BACKGROUND The Sit-to-Stand (STS) transition is one of the most used activities of daily living and vital for independence. Neurological, or physical injuries impairing functional mobility or sensory feedback often require rehabilitative programs or therapeutic interventions. Understanding the biomechanical elements of daily movements and the interaction between these elements may help inform rehabilitation protocols and optimize targeted interventions, such as stimulation protocols. RESEARCH QUESTION What are the effects of different initial knee angle, arm facilitation and proprioceptive input on leg muscle activation patterns and balance during and after a sit-to-stand? METHODS EMG of four lower limb muscles were recorded in 20 healthy participants as well centre-of-pressure sway amplitude and velocity, as participants stood from a seated position. Initial knee angles were set to various levels of extension (80°, 90°, 100°) and surface stability and arm facilitation were altered using a foam mat or crossing arms. Data were analysed across 3 phases of the STS transition. RESULTS More extended knee angles resulted in greater mediolateral sway during each phase (p < .01) and had a detrimental effect on anterior-posterior sway in phases 1 and 3. EMG data suggested more extended initial knee angles also increased EMG activity of the Tibialis Anterior (p < .001) and Bicep Femoris (p < .02) within Phases 1 and 2 to assist lift and stabilisation. SIGNIFICANCE Findings of this study outline phase-based muscle involvement as well as the compounding effects of reduced proprioceptive input and knee angle, on difficulty of the STS transition. Such results emphasising the need to take sensory and mobility issues into consideration when designing rehabilitative programs or stimulation control systems.
Collapse
Affiliation(s)
- Clodagh O'Keeffe
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland; School of Medicine, Trinity College Dublin, Dublin, Ireland.
| | - Conor Gill
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland; School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Mark Etzelmueller
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland; School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Clare Taylor
- School of Medicine, Trinity College Dublin, Dublin, Ireland; Departments of Anatomy and Physiology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Surbhi Hablani
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland; School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Richard B Reilly
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland; School of Medicine, Trinity College Dublin, Dublin, Ireland; School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Neil Fleming
- School of Medicine, Trinity College Dublin, Dublin, Ireland; Departments of Anatomy and Physiology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
23
|
Bohic M, Upadhyay A, Eisdorfer JT, Keating J, Simon RC, Briones BA, Azadegan C, Nacht HD, Oputa O, Martinez AM, Bethell BN, Gradwell MA, Romanienko P, Ramer MS, Stuber GD, Abraira VE. A new Hoxb8FlpO mouse line for intersectional approaches to dissect developmentally defined adult sensorimotor circuits. Front Mol Neurosci 2023; 16:1176823. [PMID: 37603775 PMCID: PMC10437123 DOI: 10.3389/fnmol.2023.1176823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/04/2023] [Indexed: 08/23/2023] Open
Abstract
Improvements in the speed and cost of expression profiling of neuronal tissues offer an unprecedented opportunity to define ever finer subgroups of neurons for functional studies. In the spinal cord, single cell RNA sequencing studies support decades of work on spinal cord lineage studies, offering a unique opportunity to probe adult function based on developmental lineage. While Cre/Flp recombinase intersectional strategies remain a powerful tool to manipulate spinal neurons, the field lacks genetic tools and strategies to restrict manipulations to the adult mouse spinal cord at the speed at which new tools develop. This study establishes a new workflow for intersectional mouse-viral strategies to dissect adult spinal function based on developmental lineages in a modular fashion. To restrict manipulations to the spinal cord, we generate a brain-sparing Hoxb8FlpO mouse line restricting Flp recombinase expression to caudal tissue. Recapitulating endogenous Hoxb8 gene expression, Flp-dependent reporter expression is present in the caudal embryo starting day 9.5. This expression restricts Flp activity in the adult to the caudal brainstem and below. Hoxb8FlpO heterozygous and homozygous mice do not develop any of the sensory or locomotor phenotypes evident in Hoxb8 heterozygous or mutant animals, suggesting normal developmental function of the Hoxb8 gene and protein in Hoxb8FlpO mice. Compared to the variability of brain recombination in available caudal Cre and Flp lines, Hoxb8FlpO activity is not present in the brain above the caudal brainstem, independent of mouse genetic background. Lastly, we combine the Hoxb8FlpO mouse line with dorsal horn developmental lineage Cre mouse lines to express GFP in developmentally determined dorsal horn populations. Using GFP-dependent Cre recombinase viruses and Cre recombinase-dependent inhibitory chemogenetics, we target developmentally defined lineages in the adult. We show how developmental knock-out versus transient adult silencing of the same ROR𝛃 lineage neurons affects adult sensorimotor behavior. In summary, this new mouse line and viral approach provides a blueprint to dissect adult somatosensory circuit function using Cre/Flp genetic tools to target spinal cord interneurons based on genetic lineage.
Collapse
Affiliation(s)
- Manon Bohic
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Aman Upadhyay
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- Neuroscience PhD Program at Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Jaclyn T. Eisdorfer
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Jessica Keating
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- School of Medicine, Oregon Health and Science University, Portland, OR, United States
- M.D./PhD Program in Neuroscience, School of Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Rhiana C. Simon
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Brandy A. Briones
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Chloe Azadegan
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Hannah D. Nacht
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Olisemeka Oputa
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Alana M. Martinez
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Bridget N. Bethell
- International Collaboration on Repair Discoveries and Department of Zoology, The University of British Columbia, Vancouver, BC, Canada
| | - Mark A. Gradwell
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Peter Romanienko
- Genome Editing Shared Resource, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Matt S. Ramer
- International Collaboration on Repair Discoveries and Department of Zoology, The University of British Columbia, Vancouver, BC, Canada
| | - Garret D. Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Victoria E. Abraira
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| |
Collapse
|
24
|
Goltash S, Stevens SJ, Topcu E, Bui TV. Changes in synaptic inputs to dI3 INs and MNs after complete transection in adult mice. Front Neural Circuits 2023; 17:1176310. [PMID: 37476398 PMCID: PMC10354275 DOI: 10.3389/fncir.2023.1176310] [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: 02/28/2023] [Accepted: 06/21/2023] [Indexed: 07/22/2023] Open
Abstract
Introduction Spinal cord injury (SCI) is a debilitating condition that disrupts the communication between the brain and the spinal cord. Several studies have sought to determine how to revive dormant spinal circuits caudal to the lesion to restore movements in paralyzed patients. So far, recovery levels in human patients have been modest at best. In contrast, animal models of SCI exhibit more recovery of lost function. Previous work from our lab has identified dI3 interneurons as a spinal neuron population central to the recovery of locomotor function in spinalized mice. We seek to determine the changes in the circuitry of dI3 interneurons and motoneurons following SCI in adult mice. Methods After a complete transection of the spinal cord at T9-T11 level in transgenic Isl1:YFP mice and subsequent treadmill training at various time points of recovery following surgery, we examined changes in three key circuits involving dI3 interneurons and motoneurons: (1) Sensory inputs from proprioceptive and cutaneous afferents, (2) Presynaptic inhibition of sensory inputs, and (3) Central excitatory glutamatergic synapses from spinal neurons onto dI3 INs and motoneurons. Furthermore, we examined the possible role of treadmill training on changes in synaptic connectivity to dI3 interneurons and motoneurons. Results Our data suggests that VGLUT1+ inputs to dI3 interneurons decrease transiently or only at later stages after injury, whereas levels of VGLUT1+ remain the same for motoneurons after injury. Levels of VGLUT2+ inputs to dI3 INs and MNs may show transient increases but fall below levels seen in sham-operated mice after a period of time. Levels of presynaptic inhibition to VGLUT1+ inputs to dI3 INs and MNs can rise shortly after SCI, but those increases do not persist. However, levels of presynaptic inhibition to VGLUT1+ inputs never fell below levels observed in sham-operated mice. For some synaptic inputs studied, levels were higher in spinal cord-injured animals that received treadmill training, but these increases were observed only at some time points. Discussion These results suggest remodeling of spinal circuits involving spinal interneurons that have previously been implicated in the recovery of locomotor function after spinal cord injury in mice.
Collapse
|
25
|
Pradat PF, Hayon D, Blancho S, Neveu P, Khamaysa M, Guerout N. Advances in Spinal Cord Neuromodulation: The Integration of Neuroengineering, Computational Approaches, and Innovative Conceptual Frameworks. J Pers Med 2023; 13:993. [PMID: 37373982 DOI: 10.3390/jpm13060993] [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: 03/27/2023] [Revised: 06/07/2023] [Accepted: 06/11/2023] [Indexed: 06/29/2023] Open
Abstract
Spinal cord stimulation (SCS) is an approved treatment for intractable pain and has recently emerged as a promising area of research for restoring function after spinal cord lesion. This review will focus on the historical evolution of this transition and the path that remains to be taken for these methods to be rigorously evaluated for application in clinical practice. New developments in SCS are being driven by advances in the understanding of spinal cord lesions at the molecular, cellular, and neuronal levels, as well as the understanding of compensatory mechanisms. Advances in neuroengineering and the computational neurosciences have enabled the development of new conceptual SCS strategies, such as spatiotemporal neuromodulation, which allows spatially selective stimulation at precise time points during anticipated movement. It has also become increasingly clear that these methods are only effective when combined with intensive rehabilitation techniques, such as new task-oriented methods and robotic aids. The emergence of innovative approaches to spinal cord neuromodulation has sparked significant enthusiasm among patients and in the media. Non-invasive methods are perceived to offer improved safety, patient acceptance, and cost-effectiveness. There is an immediate need for well-designed clinical trials involving consumer or advocacy groups to evaluate and compare the effectiveness of various treatment modalities, assess safety considerations, and establish outcome priorities.
Collapse
Affiliation(s)
- Pierre-François Pradat
- Laboratoire d'Imagerie Biomédicale, Sorbonne Université, CNRS, INSERM, 75013 Paris, France
- APHP, Département de Neurologie, Hôpital Pitié-Salpêtrière, Centre Référent SLA, 75013 Paris, France
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute Ulster University, C-TRIC, Altnagelvin Hospital, Derry/Londonderry BT47 6SB, UK
- Institut Pour la Recherche Sur la Moelle Epiniere et l'Encéphale (IRME), 25 Rue Duranton, 75015 Paris, France
| | - David Hayon
- Clinique Saint-Roch, Service d'Anesthésie, 56 Rue de Lille, 59223 Roncq, France
| | - Sophie Blancho
- Institut Pour la Recherche Sur la Moelle Epiniere et l'Encéphale (IRME), 25 Rue Duranton, 75015 Paris, France
| | - Pauline Neveu
- Saints Pères Paris Institute for the Neurosciences, Université Paris Cité, CNRS UMR8003, 75006 Paris, France
| | - Mohammed Khamaysa
- Laboratoire d'Imagerie Biomédicale, Sorbonne Université, CNRS, INSERM, 75013 Paris, France
| | - Nicolas Guerout
- Saints Pères Paris Institute for the Neurosciences, Université Paris Cité, CNRS UMR8003, 75006 Paris, France
| |
Collapse
|
26
|
Danner SM, Shepard CT, Hainline C, Shevtsova NA, Rybak IA, Magnuson DS. Spinal control of locomotion before and after spinal cord injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.22.533794. [PMID: 36993490 PMCID: PMC10055332 DOI: 10.1101/2023.03.22.533794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Thoracic spinal cord injury affects long propriospinal neurons that interconnect the cervical and lumbar enlargements. These neurons are crucial for coordinating forelimb and hindlimb locomotor movements in a speed-dependent manner. However, recovery from spinal cord injury is usually studied over a very limited range of speeds that may not fully expose circuitry dysfunction. To overcome this limitation, we investigated overground locomotion in rats trained to move over an extended distance with a wide range of speeds both pre-injury and after recovery from thoracic hemisection or contusion injuries. In this experimental context, intact rats expressed a speed-dependent continuum of alternating (walk and trot) and non-alternating (canter, gallop, half-bound gallop, and bound) gaits. After a lateral hemisection injury, rats recovered the ability to locomote over a wide range of speeds but lost the ability to use the highest-speed gaits (half-bound gallop and bound) and predominantly used the limb contralateral to the injury as lead during canter and gallop. A moderate contusion injury caused a greater reduction in maximal speed, loss of all non-alternating gaits, and emergence of novel alternating gaits. These changes resulted from weak fore-hind coupling together with appropriate control of left-right alternation. After hemisection, animals expressed a subset of intact gaits with appropriate interlimb coordination even on the side of the injury, where the long propriospinal connections were severed. These observations highlight how investigating locomotion over the full range of speeds can reveal otherwise hidden aspects of spinal locomotor control and post-injury recovery.
Collapse
Affiliation(s)
- Simon M. Danner
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Courtney T. Shepard
- Interdisciplinary Program in Translational Neuroscience, University of Louisville School of Medicine, Health Sciences Campus, Louisville, Kentucky, USA
| | - Casey Hainline
- Speed School of Engineering, University of Louisville School of Medicine, Health Sciences Campus, Louisville, Kentucky, USA
| | - Natalia A. Shevtsova
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Ilya A. Rybak
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - David S.K. Magnuson
- Department of Neurological Surgery, University of Louisville School of Medicine, Health Sciences Campus, Louisville, Kentucky, USA
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Health Sciences Campus, Louisville, Kentucky, USA
| |
Collapse
|
27
|
Griffin JM, Hingorani Jai Prakash S, Bockemühl T, Benner JM, Schaffran B, Moreno-Manzano V, Büschges A, Bradke F. Rehabilitation enhances epothilone-induced locomotor recovery after spinal cord injury. Brain Commun 2023; 5:fcad005. [PMID: 36744011 PMCID: PMC9893225 DOI: 10.1093/braincomms/fcad005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/21/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023] Open
Abstract
Microtubule stabilization through epothilones is a promising preclinical therapy for functional recovery following spinal cord injury that stimulates axon regeneration, reduces growth-inhibitory molecule deposition and promotes functional improvements. Rehabilitation therapy is the only clinically validated approach to promote functional improvements following spinal cord injury. However, whether microtubule stabilization can augment the beneficial effects of rehabilitation therapy or act in concert with it to further promote repair remains unknown. Here, we investigated the pharmacokinetic, histological and functional efficacies of epothilone D, epothilone B and ixabepilone alone or in combination with rehabilitation following a moderate contusive spinal cord injury. Pharmacokinetic analysis revealed that ixabepilone only weakly crossed the blood-brain barrier and was subsequently excluded from further investigations. In contrast, epothilones B and D rapidly distributed to CNS compartments displaying similar profiles after either subcutaneous or intraperitoneal injections. Following injury and subcutaneous administration of epothilone B or D, rats were subjected to 7 weeks of sequential bipedal and quadrupedal training. For all outcome measures, epothilone B was efficacious compared with epothilone D. Specifically, epothilone B decreased fibrotic scaring which was associated with a retention of fibronectin localized to perivascular cells in sections distal to the lesion. This corresponded to a decreased number of cells present within the intralesional space, resulting in less axons within the lesion. Instead, epothilone B increased serotonergic fibre regeneration and vesicular glutamate transporter 1 expression caudal to the lesion, which was not affected by rehabilitation. Multiparametric behavioural analyses consisting of open-field locomotor scoring, horizontal ladder, catwalk gait analysis and hindlimb kinematics revealed that rehabilitation and epothilone B both improved several aspects of locomotion. Specifically, rehabilitation improved open-field locomotor and ladder scores, as well as improving the gait parameters of limb coupling, limb support, stride length and limb speed; epothilone B improved these same gait parameters but also hindlimb kinematic profiles. Functional improvements by epothilone B and rehabilitation acted complementarily on gait parameters leading to an enhanced recovery in the combination group. As a result, principal component analysis of gait showed the greatest improvement in the epothilone B plus rehabilitation group. Thus, these results support the combination of epothilone B with rehabilitation in a clinical setting.
Collapse
Affiliation(s)
- Jarred M Griffin
- Correspondence may also be addressed to: Jarred Griffin The German Center for Neurodegenerative Diseases (DZNE) Venusberg-Campus 1/99, Bonn 53127, Germany E-mail:
| | - Sonia Hingorani Jai Prakash
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe (CIPF), Valencia 46012, Spain
| | - Till Bockemühl
- Department of Animal Physiology, Institute of Zoology, University of Cologne, Cologne 50674, Germany
| | - Jessica M Benner
- Laboratory for Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn 53127, Germany
| | - Barbara Schaffran
- Laboratory for Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn 53127, Germany
| | - Victoria Moreno-Manzano
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe (CIPF), Valencia 46012, Spain
| | - Ansgar Büschges
- Department of Animal Physiology, Institute of Zoology, University of Cologne, Cologne 50674, Germany
| | - Frank Bradke
- Correspondence to: Frank Bradke The German Center for Neurodegenerative Diseases (DZNE) Venusberg-Campus 1/99, Bonn 53127, Germany E-mail:
| |
Collapse
|
28
|
Aljović A, Jacobi A, Marcantoni M, Kagerer F, Loy K, Kendirli A, Bräutigam J, Fabbio L, Van Steenbergen V, Pleśniar K, Kerschensteiner M, Bareyre FM. Synaptogenic gene therapy with FGF22 improves circuit plasticity and functional recovery following spinal cord injury. EMBO Mol Med 2023; 15:e16111. [PMID: 36601738 PMCID: PMC9906383 DOI: 10.15252/emmm.202216111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 01/06/2023] Open
Abstract
Functional recovery following incomplete spinal cord injury (SCI) depends on the rewiring of motor circuits during which supraspinal connections form new contacts onto spinal relay neurons. We have recently identified a critical role of the presynaptic organizer FGF22 for the formation of new synapses in the remodeling spinal cord. Here, we now explore whether and how targeted overexpression of FGF22 can be used to mitigate the severe functional consequences of SCI. By targeting FGF22 expression to either long propriospinal neurons, excitatory interneurons, or a broader population of interneurons, we establish that FGF22 can enhance neuronal rewiring both in a circuit-specific and comprehensive way. We can further demonstrate that the latter approach can restore functional recovery when applied either on the day of the lesion or within 24 h. Our study thus establishes viral gene transfer of FGF22 as a new synaptogenic treatment for SCI and defines a critical therapeutic window for its application.
Collapse
Affiliation(s)
- Almir Aljović
- Institute of Clinical Neuroimmunology, University HospitalLMU MunichMunichGermany,Biomedical Center Munich (BMC), Faculty of MedicineLMU MunichPlaneggGermany,Graduate School of Systemic NeurosciencesLMU MunichPlaneggGermany
| | - Anne Jacobi
- Institute of Clinical Neuroimmunology, University HospitalLMU MunichMunichGermany,Biomedical Center Munich (BMC), Faculty of MedicineLMU MunichPlaneggGermany,Present address:
F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of NeurologyHarvard Medical SchoolBostonMAUSA
| | - Maite Marcantoni
- Institute of Clinical Neuroimmunology, University HospitalLMU MunichMunichGermany,Biomedical Center Munich (BMC), Faculty of MedicineLMU MunichPlaneggGermany
| | - Fritz Kagerer
- Institute of Clinical Neuroimmunology, University HospitalLMU MunichMunichGermany,Biomedical Center Munich (BMC), Faculty of MedicineLMU MunichPlaneggGermany,Elite Graduate Program M.Sc. Biomedical NeuroscienceTUMMunichGermany
| | - Kristina Loy
- Institute of Clinical Neuroimmunology, University HospitalLMU MunichMunichGermany,Biomedical Center Munich (BMC), Faculty of MedicineLMU MunichPlaneggGermany
| | - Arek Kendirli
- Institute of Clinical Neuroimmunology, University HospitalLMU MunichMunichGermany,Biomedical Center Munich (BMC), Faculty of MedicineLMU MunichPlaneggGermany,Graduate School of Systemic NeurosciencesLMU MunichPlaneggGermany
| | - Jonas Bräutigam
- Institute of Clinical Neuroimmunology, University HospitalLMU MunichMunichGermany,Biomedical Center Munich (BMC), Faculty of MedicineLMU MunichPlaneggGermany
| | - Luca Fabbio
- Institute of Clinical Neuroimmunology, University HospitalLMU MunichMunichGermany,Biomedical Center Munich (BMC), Faculty of MedicineLMU MunichPlaneggGermany
| | - Valérie Van Steenbergen
- Institute of Clinical Neuroimmunology, University HospitalLMU MunichMunichGermany,Biomedical Center Munich (BMC), Faculty of MedicineLMU MunichPlaneggGermany
| | - Katarzyna Pleśniar
- Institute of Clinical Neuroimmunology, University HospitalLMU MunichMunichGermany,Biomedical Center Munich (BMC), Faculty of MedicineLMU MunichPlaneggGermany
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University HospitalLMU MunichMunichGermany,Biomedical Center Munich (BMC), Faculty of MedicineLMU MunichPlaneggGermany,Munich Cluster of Systems Neurology (SyNergy)MunichGermany
| | - Florence M Bareyre
- Institute of Clinical Neuroimmunology, University HospitalLMU MunichMunichGermany,Biomedical Center Munich (BMC), Faculty of MedicineLMU MunichPlaneggGermany,Munich Cluster of Systems Neurology (SyNergy)MunichGermany
| |
Collapse
|
29
|
Santuz A, Akay T. Muscle spindles and their role in maintaining robust locomotion. J Physiol 2023; 601:275-285. [PMID: 36510697 PMCID: PMC10483674 DOI: 10.1113/jp282563] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Muscle spindles, one of the two main classes of proprioceptors together with Golgi tendon organs, are sensory structures that keep the central nervous system updated about the position and movement of body parts. Although they were discovered more than 150 years ago, their function during movement is not yet fully understood. Here, we summarize the morphology and known functions of muscle spindles, with a particular focus on locomotion. Although certain properties such as the sensitivity to dynamic and static muscle stretch are long known, recent advances in molecular biology have allowed the characterization of the molecular mechanisms for signal transduction in muscle spindles. Building upon classic literature showing that a lack of sensory feedback is deleterious to locomotion, we bring to the discussion more recent findings that support a pivotal role of muscle spindles in maintaining murine and human locomotor robustness, defined as the ability to cope with perturbations. Yet, more research is needed to expand the existing mechanistic understanding of how muscle spindles contribute to the production of robust, functional locomotion in real world settings. Future investigations should focus on combining different animal models to identify, in health and disease, those peripheral, spinal and brain proprioceptive structures involved in the fine tuning of motor control when locomotion happens in challenging conditions.
Collapse
Affiliation(s)
- Alessandro Santuz
- Atlantic Mobility Action Project, Brain Repair Centre, Department of Medical Neuroscience, Life Sciences Research Institute, Dalhousie University, Halifax, NS, Canada
| | - Turgay Akay
- Atlantic Mobility Action Project, Brain Repair Centre, Department of Medical Neuroscience, Life Sciences Research Institute, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
30
|
Shin JC, Jeon HR, Kim D, Min WK, Lee JS, Cho SI, Oh DS, Yoo J. Effects of end-effector robot-assisted gait training on gait ability, muscle strength, and balance in patients with spinal cord injury. NeuroRehabilitation 2023; 53:335-346. [PMID: 37638457 DOI: 10.3233/nre-230085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
BACKGROUND There is no randomized controlled study about the effects of end-effector robot-assisted gait training (RAGT) in patients with spinal cord injury (SCI). OBJECTIVE To examine the effects of end-effector RAGT on gait and balance abilities in SCI. METHODS Thirty-one patients were randomly assigned to the RAGT (Morning Walk®, Curexo, Seoul, South Korea) or conventional therapy (CT) group. Patients were assessed using the 10-meter walk test (10MWT), 6-minute walk test (6mWT), lower extremity motor score (LEMS) and proprioception, Berg Balance Scale (BBS), Walking Index for Spinal Cord Injury-II (WISCI-II), and mobility category of Spinal Cord Independence Measure-III. RESULTS All clinical outcome measures significantly improved in both groups. The BBS and WISCI-II were significantly improved in the RAGT group compared to the CT group. In the RAGT group, pre-LEMS and pre-WISCI-II of the 10MWT improved group and pre-BBS of the 6mWT improved group were higher than those of the 10MWT non-improved and 6mWT non-improved group, respectively. CONCLUSION End-effector RAGT and CT in patients with incomplete SCI could lead to improvements in gait ability, lower extremity muscle strength, balance, proprioception, and mobility. Additionally, end-effector RAGT could improve balance and gait abilities substantially better than CT.
Collapse
Affiliation(s)
- Ji Cheol Shin
- Research Institute of Rehabilitation Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Ha Ra Jeon
- Department of Physical Medicine and Rehabilitation, National Health Insurance Service Ilsan Hospital, Goyang, South Korea
| | - Dahn Kim
- Research Institute of Rehabilitation Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Won Kyu Min
- Rehabilitation Center, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - June Sung Lee
- Rehabilitation Center, National Health Insurance Service Ilsan Hospital, Goyang, South Korea
| | - Sung Il Cho
- Rehabilitation Center, Inje University Ilsan Paik Hospital, Goyang, South Korea
| | - Da Som Oh
- Inje Industry, Academic Cooperation Foundation, Goyang, South Korea
| | - Jeehyun Yoo
- Department of Rehabilitation Medicine, Inje University Ilsan Paik Hospital, Goyang, South Korea
| |
Collapse
|
31
|
Wang Z, Duan H, Hao F, Hao P, Zhao W, Gao Y, Gu Y, Song J, Li X, Yang Z. Circuit reconstruction of newborn neurons after spinal cord injury in adult rats via an NT3-chitosan scaffold. Prog Neurobiol 2023; 220:102375. [PMID: 36410665 DOI: 10.1016/j.pneurobio.2022.102375] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 11/07/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022]
Abstract
An implanted neurotrophin-3 (NT3)-chitosan scaffold can recruit endogenous neural stem cells to migrate to a lesion region and differentiate into mature neurons after adult spinal cord injury (SCI). However, the identities of these newborn neurons and whether they can form functional synapses and circuits to promote recovery after paraplegia remain unknown. By using combined advanced technologies, we revealed here that the newborn neurons of several subtypes received synaptic input from the corticospinal tract (CST), rubrospinal tract (RST), and supraspinal tracts. They formed a functional neural circuit at the injured spinal region, further driving the local circuits beneath the lesion. Our results showed that the NT3-chitosan scaffold facilitated the maturation of spinal neurons and the reestablishment of the spinal neural circuit in the lesion region 12 weeks after SCI. Transsynaptic virus experiments revealed that these newborn spinal neurons received synaptic connections from the CST and RST and drove the neural circuit beneath the lesion via newly formed synapses. These re-established circuits successfully recovered the formation and function of the neuromuscular junction (NMJ) beneath the lesion spinal segments. These findings suggest that the NT3-chitosan scaffold promotes the formation of relay neural circuits to accommodate various types of brain descending inputs and facilitate functional recovery after paraplegia.
Collapse
Affiliation(s)
- Zijue Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Hongmei Duan
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Fei Hao
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Peng Hao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Wen Zhao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yudan Gao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yiming Gu
- Physical Education Department, Capital University of Economics and Business, Beijing 100070, China
| | - Jianren Song
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China; Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai 200092, China.
| | - Xiaoguang Li
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Department of Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| | - Zhaoyang Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
32
|
Yu H, Li Q, Sandoval A, Gibbs HC, English A, Dunn T, Moth J, Elahi H, Chen B. Pipeline for fluorescent imaging and volumetric analysis of neurons in cleared mouse spinal cords. STAR Protoc 2022; 3:101759. [PMID: 36227743 PMCID: PMC9576748 DOI: 10.1016/j.xpro.2022.101759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/12/2022] [Accepted: 09/15/2022] [Indexed: 11/05/2022] Open
Abstract
Precisely measuring the number and somatic volume of neurons in the central nervous system at single-cell resolution is technically challenging. Here, we combine multiple techniques to address this challenge in optically cleared mouse spinal cords. We describe in vivo neuron labeling approaches, tissue-clearing technology, light sheet fluorescence microscopy, and machine learning-guided imaging analysis. This combination provides a precise determination of the cell number and somatic volume of any neuron population in the spinal cords.
Collapse
Affiliation(s)
- Hao Yu
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77550, USA.
| | - Qiang Li
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Alfredo Sandoval
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Holly C Gibbs
- Microscopy and Imaging Center, Texas A&M University, College Station, TX 77843, USA; Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Amber English
- Application Support Specialist, Bitplane Inc., 300 Baker Avenue Extension No. 150, Concord, MA 01742, USA
| | - Tiffany Dunn
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - John Moth
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Hajira Elahi
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Bo Chen
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77550, USA.
| |
Collapse
|
33
|
Ronzano R, Skarlatou S, Barriga BK, Bannatyne BA, Bhumbra GS, Foster JD, Moore JD, Lancelin C, Pocratsky AM, Özyurt MG, Smith CC, Todd AJ, Maxwell DJ, Murray AJ, Pfaff SL, Brownstone RM, Zampieri N, Beato M. Spinal premotor interneurons controlling antagonistic muscles are spatially intermingled. eLife 2022; 11:e81976. [PMID: 36512397 PMCID: PMC9844990 DOI: 10.7554/elife.81976] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022] Open
Abstract
Elaborate behaviours are produced by tightly controlled flexor-extensor motor neuron activation patterns. Motor neurons are regulated by a network of interneurons within the spinal cord, but the computational processes involved in motor control are not fully understood. The neuroanatomical arrangement of motor and premotor neurons into topographic patterns related to their controlled muscles is thought to facilitate how information is processed by spinal circuits. Rabies retrograde monosynaptic tracing has been used to label premotor interneurons innervating specific motor neuron pools, with previous studies reporting topographic mediolateral positional biases in flexor and extensor premotor interneurons. To more precisely define how premotor interneurons contacting specific motor pools are organized, we used multiple complementary viral-tracing approaches in mice to minimize systematic biases associated with each method. Contrary to expectations, we found that premotor interneurons contacting motor pools controlling flexion and extension of the ankle are highly intermingled rather than segregated into specific domains like motor neurons. Thus, premotor spinal neurons controlling different muscles process motor instructions in the absence of clear spatial patterns among the flexor-extensor circuit components.
Collapse
Affiliation(s)
- Remi Ronzano
- Department of Neuromuscular Diseases, University College LondonLondonUnited Kingdom
| | | | - Bianca K Barriga
- Gene Expression Laboratory, Salk Institute for Biological StudiesLa JollaUnited States
- Biological Sciences Graduate Program, University of California, San DiegoSan DiegoUnited States
| | - B Anne Bannatyne
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUnited Kingdom
| | - Gardave Singh Bhumbra
- Department of Neuroscience Physiology and Pharmacology, University College LondonLondonUnited Kingdom
| | - Joshua D Foster
- Department of Neuroscience Physiology and Pharmacology, University College LondonLondonUnited Kingdom
| | - Jeffrey D Moore
- Howard Hughes Medical Institute and Department of Molecular and Cellular Biology, Center for Brain Science, Harvard UniversityCambridgeUnited States
| | - Camille Lancelin
- Department of Neuromuscular Diseases, University College LondonLondonUnited Kingdom
| | - Amanda M Pocratsky
- Department of Neuromuscular Diseases, University College LondonLondonUnited Kingdom
| | | | - Calvin Chad Smith
- Department of Neuromuscular Diseases, University College LondonLondonUnited Kingdom
| | - Andrew J Todd
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUnited Kingdom
| | - David J Maxwell
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUnited Kingdom
| | - Andrew J Murray
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College LondonLondonUnited Kingdom
| | - Samuel L Pfaff
- Gene Expression Laboratory, Salk Institute for Biological StudiesLa JollaUnited States
| | - Robert M Brownstone
- Department of Neuromuscular Diseases, University College LondonLondonUnited Kingdom
| | | | - Marco Beato
- Department of Neuroscience Physiology and Pharmacology, University College LondonLondonUnited Kingdom
| |
Collapse
|
34
|
Santuz A, Laflamme OD, Akay T. The brain integrates proprioceptive information to ensure robust locomotion. J Physiol 2022; 600:5267-5294. [PMID: 36271747 DOI: 10.1113/jp283181] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/10/2022] [Indexed: 01/05/2023] Open
Abstract
Robust locomotion relies on information from proprioceptors: sensory organs that communicate the position of body parts to the spinal cord and brain. Proprioceptive circuits in the spinal cord are known to coarsely regulate locomotion in the presence of perturbations. Yet, the regulatory importance of the brain in maintaining robust locomotion remains less clear. Here, through mouse genetic studies and in vivo electrophysiology, we examined the role of the brain in integrating proprioceptive information during perturbed locomotion. The systemic removal of proprioceptors left the mice in a constantly perturbed state, similar to that observed during mechanically perturbed locomotion in wild-type mice and characterised by longer and less accurate synergistic activation patterns. By contrast, after surgically interrupting the ascending proprioceptive projection to the brain through the dorsal column of the spinal cord, wild-type mice showed normal walking behaviour, yet lost the ability to respond to external perturbations. Our findings provide direct evidence of a pivotal role for ascending proprioceptive information in achieving robust, safe locomotion. KEY POINTS: Whether brain integration of proprioceptive feedback is crucial for coping with perturbed locomotion is not clear. We showed a crucial role of the brain for responding to external perturbations and ensure robust locomotion. We used mouse genetics to remove proprioceptors and a spinal lesion model to interrupt the flow of proprioceptive information to the brain through the dorsal column in wild-type animals. Using a custom-built treadmill, we administered sudden and random mechanical perturbations to mice during walking. External perturbations affected locomotion in wild-type mice similar to the absence of proprioceptors in genetically modified mice. Proprioceptive feedback from muscle spindles and Golgi tendon organs contributed to locomotor robustness. Wild-type mice lost the ability to respond to external perturbations after interruption of the ascending proprioceptive projection to the brainstem.
Collapse
Affiliation(s)
- Alessandro Santuz
- Atlantic Mobility Action Project, Brain Repair Centre, Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - Olivier D Laflamme
- Atlantic Mobility Action Project, Brain Repair Centre, Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - Turgay Akay
- Atlantic Mobility Action Project, Brain Repair Centre, Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
35
|
Lafitte MN, Kadone H, Kubota S, Shimizu Y, Tan CK, Koda M, Hada Y, Sankai Y, Suzuki K, Yamazaki M. Alteration of muscle activity during voluntary rehabilitation training with single-joint Hybrid Assistive Limb (HAL) in patients with shoulder elevation dysfunction from cervical origin. Front Neurosci 2022; 16:817659. [PMID: 36440285 PMCID: PMC9682184 DOI: 10.3389/fnins.2022.817659] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 10/17/2022] [Indexed: 08/27/2023] Open
Abstract
Shoulder elevation, defined here as arm raising, being essential for activities of daily living, dysfunctions represent a substantial burden in patients' lives. Owing to the complexity of the shoulder joint, the tightly coordinated muscular activity is a fundamental component, and neuromuscular impairments have devastating effects. A single-joint shoulder type version of the Hybrid Assistive Limb (HAL) allowing motion assistance based on the intention of the user via myoelectric activation has recently been developed, and its safety was demonstrated for shoulder rehabilitation. Yet, little is known about the physiological effects of the device. This study aims to monitor the changes in muscle activity and motion during shoulder HAL rehabilitation in several patients suffering from shoulder elevation dysfunction from cervical radicular origin. 8 patients (6 males, 2 females, mean age 62.4 ± 9.3 years old) with weakness of the deltoid muscle resulting from a damage to the C5 nerve root underwent HAL-assisted rehabilitation. We combined surface electromyography and three-dimensional motion capture to record muscular activity and kinematics. All participants showed functional recovery, with improvements in their Manual Muscle Testing (MMT) scores and range of motion (ROM). During training, HAL decreased the activity of deltoid and trapezius, significantly more for the latter, as well as the coactivation of both muscles. We also report a reduction of the characteristic shrugging compensatory motion which is an obstacle to functional recovery. This reduction was notably demonstrated by a stronger reliance on the deltoid rather than the trapezius, indicating a muscle coordination tending toward a pattern similar to healthy individuals. Altogether, the results of the evaluation of motion and muscular changes hint toward a functional recovery in acute, and chronic shoulder impairments from cervical radicular origin following shoulder HAL rehabilitation training and provide information on the physiological effect of the device.
Collapse
Affiliation(s)
- Margaux Noémie Lafitte
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Japan
- Artificial Intelligence Laboratory, University of Tsukuba, Tsukuba, Japan
| | - Hideki Kadone
- Center for Cybernics Research, University of Tsukuba, Tsukuba, Japan
| | - Shigeki Kubota
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yukiyo Shimizu
- Department of Rehabilitation Medicine, University of Tsukuba Hospital, Tsukuba, Japan
| | - Chun Kwang Tan
- Artificial Intelligence Laboratory, University of Tsukuba, Tsukuba, Japan
| | - Masao Koda
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yasushi Hada
- Department of Rehabilitation Medicine, University of Tsukuba Hospital, Tsukuba, Japan
| | - Yoshiyuki Sankai
- Center for Cybernics Research, University of Tsukuba, Tsukuba, Japan
| | - Kenji Suzuki
- Artificial Intelligence Laboratory, University of Tsukuba, Tsukuba, Japan
- Center for Cybernics Research, University of Tsukuba, Tsukuba, Japan
| | - Masashi Yamazaki
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
36
|
Huang CX, Wang Z, Cheng J, Zhu Z, Guan NN, Song J. De novo establishment of circuit modules restores locomotion after spinal cord injury in adult zebrafish. Cell Rep 2022; 41:111535. [DOI: 10.1016/j.celrep.2022.111535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/12/2022] [Accepted: 09/29/2022] [Indexed: 11/03/2022] Open
|
37
|
Spinal Cord Circuits: Models and Reality. NEUROPHYSIOLOGY+ 2022. [DOI: 10.1007/s11062-022-09927-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
38
|
Eisdorfer JT, Sobotka-Briner H, Schramfield S, Moukarzel G, Chen J, Campion TJ, Smit R, Rauscher BC, Lemay MA, Smith GM, Spence AJ. Chemogenetic modulation of sensory afferents induces locomotor changes and plasticity after spinal cord injury. Front Mol Neurosci 2022; 15:872634. [PMID: 36090254 PMCID: PMC9461563 DOI: 10.3389/fnmol.2022.872634] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 07/26/2022] [Indexed: 12/12/2022] Open
Abstract
Neuromodulatory therapies for spinal cord injury (SCI) such as electrical epidural stimulation (EES) are increasingly effective at improving patient outcomes. These improvements are thought to be due, at least in part, to plasticity in neuronal circuits. Precisely which circuits are influenced and which afferent classes are most effective in stimulating change remain important open questions. Genetic tools, such as Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), support targeted and reversible neuromodulation as well as histological characterization of manipulated neurons. We therefore transduced and activated lumbar large diameter peripheral afferents with excitatory (hM3Dq) DREADDs, in a manner analogous to EES, in a rat hemisection model, to begin to trace plasticity and observe concomitant locomotor changes. Chronic DREADDs activation, coupled with thrice weekly treadmill training, was observed to increase afferent fluorescent labeling within motor pools and Clarke's column when compared to control animals. This plasticity may underlie kinematic differences that we observed across stages of recovery, including an increased and less variable hindquarters height in DREADDs animals, shorter step durations, a more flexed ankle joint early in recovery, a less variable ankle joint angle in swing phase, but a more variable hip joint angle. Withdrawal of DREADDs agonist, clozapine-N-oxide (CNO) left these kinematic differences largely unaffected; suggesting that DREADDs activation is not necessary for them later in recovery. However, we observed an intermittent “buckling” phenomenon in DREADDs animals without CNO activation, that did not occur with CNO re-administration. Future studies could use more refined genetic targeted of specific afferent classes, and utilize muscle recordings to find where afferent modulation is most influential in altering motor output.
Collapse
Affiliation(s)
- Jaclyn T. Eisdorfer
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, United States
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Hannah Sobotka-Briner
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, United States
| | - Susan Schramfield
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, United States
| | - George Moukarzel
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, United States
| | - Jie Chen
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Thomas J. Campion
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Rupert Smit
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Bradley C. Rauscher
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, United States
| | - Michel A. Lemay
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, United States
| | - George M. Smith
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Andrew J. Spence
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, United States
- *Correspondence: Andrew J. Spence
| |
Collapse
|
39
|
Neurotransmitter phenotype switching by spinal excitatory interneurons regulates locomotor recovery after spinal cord injury. Nat Neurosci 2022; 25:617-629. [PMID: 35524138 PMCID: PMC9076533 DOI: 10.1038/s41593-022-01067-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 03/29/2022] [Indexed: 11/08/2022]
Abstract
Severe spinal cord injury in adults leads to irreversible paralysis below the lesion. However, adult rodents that received a complete thoracic lesion just after birth demonstrate proficient hindlimb locomotion without input from the brain. How the spinal cord achieves such striking plasticity remains unknown. In this study, we found that adult spinal cord injury prompts neurotransmitter switching of spatially defined excitatory interneurons to an inhibitory phenotype, promoting inhibition at synapses contacting motor neurons. In contrast, neonatal spinal cord injury maintains the excitatory phenotype of glutamatergic interneurons and causes synaptic sprouting to facilitate excitation. Furthermore, genetic manipulation to mimic the inhibitory phenotype observed in excitatory interneurons after adult spinal cord injury abrogates autonomous locomotor functionality in neonatally injured mice. In comparison, attenuating this inhibitory phenotype improves locomotor capacity after adult injury. Together, these data demonstrate that neurotransmitter phenotype of defined excitatory interneurons steers locomotor recovery after spinal cord injury.
Collapse
|
40
|
Hippocampal Mitochondrial Abnormalities Induced the Dendritic Complexity Reduction and Cognitive Decline in a Rat Model of Spinal Cord Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9253916. [PMID: 35571236 PMCID: PMC9095360 DOI: 10.1155/2022/9253916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/19/2022] [Accepted: 04/12/2022] [Indexed: 11/18/2022]
Abstract
Spinal cord injury (SCI) is a progressive neurodegenerative disease in addition to a traumatic event. Cognitive dysfunction following SCI has been widely reported in patients and animal models. However, the neuroanatomical changes affecting cognitive function after SCI, as well as the mechanisms behind these changes, have so far remained elusive. Herein, we found that SCI accelerates oxidative stress damage of hippocampal neuronal mitochondria. Then, for the first time, we presented a three-dimensional morphological atlas of rat hippocampal neurons generated using a fluorescence Micro-Optical Sectioning Tomography system, a method that accurately identifies the spatial localization of neurons and trace neurites. We showed that the number of dendritic branches and dendritic length was decreased in late stage of SCI. Western blot and transmission electron microscopy analyses also showed a decrease in synaptic communication. In addition, a battery of behavioral tests in these animals revealed hippocampal based cognitive dysfunction, which could be attributed to changes in the dendritic complexity of hippocampal neurons. Taken together, these results suggested that mitochondrial abnormalities in hippocampal neurons induced the dendritic complexity reduction and cognitive decline following SCI. Our study highlights the neuroanatomical basis and importance of mitochondria in brain degeneration following SCI, which might contribute to propose new therapeutic strategies.
Collapse
|
41
|
Kjell J, Svensson M. Advancing Peripheral Nerve Graft Transplantation for Incomplete Spinal Cord Injury Repair. Front Cell Neurosci 2022; 16:885245. [PMID: 35573831 PMCID: PMC9097274 DOI: 10.3389/fncel.2022.885245] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
Peripheral nerves have a propensity for axon growth and regeneration that the central nervous system lacks (CNS). However, CNS axons can also grow long distances if introduced to a graft harvested from a peripheral nerve (PNGs), which is the rationale for using PNGs as repair strategy for injuries of the spinal cord. From a clinical perspective, PNGs provide interesting possibilities with potential to repair the injured spinal cord. First, there are numerous options to harvest autologous grafts associated with low risk for the patient. Second, a PNG allow axons to grow considerable distances and can, by the surgical procedure, be navigated to specific target sites in the CNS. Furthermore, a PNG provides all necessary biological substrates for myelination of elongating axons. A PNG can thus be suited to bridge axons long distances across an injury site and restore long tracts in incomplete SCI. Experimentally, locomotor functions have been improved transplanting a PNG after incomplete injury. However, we still know little with regard to the formation of new circuitries and functional outcome in association to when, where, and how grafts are inserted into the injured spinal cord, especially for sensory functions. In this perspective, we discuss the advantages of PNG from a clinical and surgical perspective, the need for adding/repairing long tracts, how PNGs are best applied for incomplete injuries, and the unexplored areas we believe are in need of answers.
Collapse
Affiliation(s)
- Jacob Kjell
- Department of Clinical Neuroscience, Karolinska Institutet, Solna, Sweden
| | - Mikael Svensson
- Department of Clinical Neuroscience, Karolinska Institutet, Solna, Sweden
- Neurosurgery, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
42
|
Fortino TA, Randelman ML, Hall AA, Singh J, Bloom DC, Engel E, Hoh DJ, Hou S, Zholudeva LV, Lane MA. Transneuronal tracing to map connectivity in injured and transplanted spinal networks. Exp Neurol 2022; 351:113990. [DOI: 10.1016/j.expneurol.2022.113990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/09/2021] [Accepted: 01/20/2022] [Indexed: 11/24/2022]
|
43
|
Abstract
When animals walk overground, mechanical stimuli activate various receptors located in muscles, joints, and skin. Afferents from these mechanoreceptors project to neuronal networks controlling locomotion in the spinal cord and brain. The dynamic interactions between the control systems at different levels of the neuraxis ensure that locomotion adjusts to its environment and meets task demands. In this article, we describe and discuss the essential contribution of somatosensory feedback to locomotion. We start with a discussion of how biomechanical properties of the body affect somatosensory feedback. We follow with the different types of mechanoreceptors and somatosensory afferents and their activity during locomotion. We then describe central projections to locomotor networks and the modulation of somatosensory feedback during locomotion and its mechanisms. We then discuss experimental approaches and animal models used to investigate the control of locomotion by somatosensory feedback before providing an overview of the different functional roles of somatosensory feedback for locomotion. Lastly, we briefly describe the role of somatosensory feedback in the recovery of locomotion after neurological injury. We highlight the fact that somatosensory feedback is an essential component of a highly integrated system for locomotor control. © 2021 American Physiological Society. Compr Physiol 11:1-71, 2021.
Collapse
Affiliation(s)
- Alain Frigon
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Quebec, Canada
| | - Turgay Akay
- Department of Medical Neuroscience, Atlantic Mobility Action Project, Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Boris I Prilutsky
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
44
|
Moreno-López Y, Hollis ER. Sensory Circuit Remodeling and Movement Recovery After Spinal Cord Injury. Front Neurosci 2021; 15:787690. [PMID: 34955735 PMCID: PMC8692650 DOI: 10.3389/fnins.2021.787690] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/10/2021] [Indexed: 12/18/2022] Open
Abstract
Restoring sensory circuit function after spinal cord injury (SCI) is essential for recovery of movement, yet current interventions predominantly target motor pathways. Integrated cortical sensorimotor networks, disrupted by SCI, are critical for perceiving, shaping, and executing movement. Corticocortical connections between primary sensory (S1) and motor (M1) cortices are critical loci of functional plasticity in response to learning and injury. Following SCI, in the motor cortex, corticocortical circuits undergo dynamic remodeling; however, it remains unknown how rehabilitation shapes the plasticity of S1-M1 networks or how these changes may impact recovery of movement.
Collapse
Affiliation(s)
| | - Edmund R Hollis
- Burke Neurological Institute, White Plains, NY, United States.,Weill Cornell Medicine, Feil Family Brain & Mind Research Institute, New York, NY, United States
| |
Collapse
|
45
|
Hu S, Wu G, Wu B, Du Z, Zhang Y. Rehabilitative training paired with peripheral stimulation promotes motor recovery after ischemic cerebral stroke. Exp Neurol 2021; 349:113960. [PMID: 34953896 DOI: 10.1016/j.expneurol.2021.113960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 12/12/2021] [Accepted: 12/18/2021] [Indexed: 12/22/2022]
Abstract
Spontaneous recovery of ischemic stroke is very limited and often results in the loss of motor and sensory function. Till now, rehabilitative training is the most widely accepted therapy to improve long-term outcome. However, its effectiveness is often suboptimal, largely due to a sharp decline of neuroplasticity in adults. In this study, we hypothesized that a combination of proprioceptive stimulation and rehabilitative training will promote neuroplasticity and functional recovery post injury. To test this hypothesis, we first established a photothrombotic stroke model that lesions the hindlimb sensorimotor cortex. Next, we demonstrated that injecting Cre-dependent AAV-retro viruses into the dorsal column of PV-Cre mice achieves specific and efficient targeting of proprioceptors. With chemogenetics, this method enables chronic activation of proprioceptors. We then assessed effects of combinatorial treatment on motor and sensory functional recovery. Our results showed that pairing proprioceptive stimulation with rehabilitative training significantly promoted skilled motor, but not tactile sensory functional recovery. This further led to significant improvement when compared to rehabilitation training or proprioceptor stimulation alone. Mechanistically, combinatorial treatment promoted cortical layer V neuronal mTOR activity and sprouting of corticospinal axon into the area where proprioceptive afferents terminate in the denervated side of the spinal cord. Serving as a proof of principle, our study thus provided novel insights into the application of combining proprioceptive stimulation and rehabilitative training to improve functional recovery of ischemic stroke and other traumatic brain or spinal cord injuries.
Collapse
Affiliation(s)
- Shukun Hu
- Department of Neurosurgery, Affiliated Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Gang Wu
- Department of Neurosurgery, Affiliated Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Biwu Wu
- Department of Neurosurgery, Affiliated Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Zhouying Du
- Department of Neurosurgery, Affiliated Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Yi Zhang
- Department of Neurosurgery, Affiliated Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
| |
Collapse
|
46
|
Faw TD, Lakhani B, Schmalbrock P, Knopp MV, Lohse KR, Kramer JLK, Liu H, Nguyen HT, Phillips EG, Bratasz A, Fisher LC, Deibert RJ, Boyd LA, McTigue DM, Basso DM. Eccentric rehabilitation induces white matter plasticity and sensorimotor recovery in chronic spinal cord injury. Exp Neurol 2021; 346:113853. [PMID: 34464653 PMCID: PMC10084731 DOI: 10.1016/j.expneurol.2021.113853] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/04/2021] [Accepted: 08/26/2021] [Indexed: 12/12/2022]
Abstract
Experience-dependent white matter plasticity offers new potential for rehabilitation-induced recovery after neurotrauma. This first-in-human translational experiment combined myelin water imaging in humans and genetic fate-mapping of oligodendrocyte lineage cells in mice to investigate whether downhill locomotor rehabilitation that emphasizes eccentric muscle actions promotes white matter plasticity and recovery in chronic, incomplete spinal cord injury (SCI). In humans, of 20 individuals with SCI that enrolled, four passed the imaging screen and had myelin water imaging before and after a 12-week (3 times/week) downhill locomotor treadmill training program (SCI + DH). One individual was excluded for imaging artifacts. Uninjured control participants (n = 7) had two myelin water imaging sessions within the same day. Changes in myelin water fraction (MWF), a histopathologically-validated myelin biomarker, were analyzed in a priori motor learning and non-motor learning brain regions and the cervical spinal cord using statistical approaches appropriate for small sample sizes. PDGFRα-CreERT2:mT/mG mice, that express green fluorescent protein on oligodendrocyte precursor cells and subsequent newly-differentiated oligodendrocytes upon tamoxifen-induced recombination, were either naive (n = 6) or received a moderate (75 kilodyne), contusive SCI at T9 and were randomized to downhill training (n = 6) or unexercised groups (n = 6). We initiated recombination 29 days post-injury, seven days prior to downhill training. Mice underwent two weeks of daily downhill training on the same 10% decline grade used in humans. Between-group comparison of functional (motor and sensory) and histological (oligodendrogenesis, oligodendroglial/axon interaction, paranodal structure) outcomes occurred post-training. In humans with SCI, downhill training increased MWF in brain motor learning regions (postcentral, precuneus) and mixed motor and sensory tracts of the ventral cervical spinal cord compared to control participants (P < 0.05). In mice with thoracic SCI, downhill training induced oligodendrogenesis in cervical dorsal and lateral white matter, increased axon-oligodendroglial interactions, and normalized paranodal structure in dorsal column sensory tracts (P < 0.05). Downhill training improved sensorimotor recovery in mice by normalizing hip and knee motor control and reducing hyperalgesia, both of which were associated with new oligodendrocytes in the cervical dorsal columns (P < 0.05). Our findings indicate that eccentric-focused, downhill rehabilitation promotes white matter plasticity and improved function in chronic SCI, likely via oligodendrogenesis in nervous system regions activated by the training paradigm. Together, these data reveal an exciting role for eccentric training in white matter plasticity and sensorimotor recovery after SCI.
Collapse
Affiliation(s)
- Timothy D Faw
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; Department of Orthopaedic Surgery, Duke University, Durham, NC 27710, USA
| | - Bimal Lakhani
- Department of Physical Therapy, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Petra Schmalbrock
- Department of Radiology, The Ohio State University, Columbus, OH 43210, USA
| | - Michael V Knopp
- Department of Radiology, The Ohio State University, Columbus, OH 43210, USA
| | - Keith R Lohse
- Department of Health, Kinesiology, and Recreation, University of Utah, Salt Lake City, UT 84112, USA; Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT 84108, USA
| | - John L K Kramer
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Hanwen Liu
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC V5Z 1M9, Canada; Department of Physics and Astronomy, University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Huyen T Nguyen
- Department of Radiology, The Ohio State University, Columbus, OH 43210, USA
| | - Eileen G Phillips
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Anna Bratasz
- Small Animal Imaging Shared Resources, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Lesley C Fisher
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Rochelle J Deibert
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Lara A Boyd
- Department of Physical Therapy, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Dana M McTigue
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - D Michele Basso
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
47
|
Hachmann JT, Yousak A, Wallner JJ, Gad PN, Edgerton VR, Gorgey AS. Epidural spinal cord stimulation as an intervention for motor recovery after motor complete spinal cord injury. J Neurophysiol 2021; 126:1843-1859. [PMID: 34669485 DOI: 10.1152/jn.00020.2021] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 10/12/2021] [Indexed: 12/19/2022] Open
Abstract
Spinal cord injury (SCI) commonly results in permanent loss of motor, sensory, and autonomic function. Recent clinical studies have shown that epidural spinal cord stimulation may provide a beneficial adjunct for restoring lower extremity and other neurological functions. Herein, we review the recent clinical advances of lumbosacral epidural stimulation for restoration of sensorimotor function in individuals with motor complete SCI and we discuss the putative neural pathways involved in this promising neurorehabilitative approach. We focus on three main sections: review recent clinical results for locomotor restoration in complete SCI; discuss the contemporary understanding of electrical neuromodulation and signal transduction pathways involved in spinal locomotor networks; and review current challenges of motor system modulation and future directions toward integrative neurorestoration. The current understanding is that initial depolarization occurs at the level of large diameter dorsal root proprioceptive afferents that when integrated with interneuronal and latent residual supraspinal translesional connections can recruit locomotor centers and augment downstream motor units. Spinal epidural stimulation can initiate excitability changes in spinal networks and supraspinal networks. Different stimulation parameters can facilitate standing or stepping, and it may also have potential for augmenting myriad other sensorimotor and autonomic functions. More comprehensive investigation of the mechanisms that mediate the transformation of dysfunctional spinal networks to higher functional states with a greater focus on integrated systems-based control system may reveal the key mechanisms underlying neurological augmentation and motor restoration after severe paralysis.
Collapse
Affiliation(s)
- Jan T Hachmann
- Department of Neurological Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Andrew Yousak
- Spinal Cord Injury and Disorders Center, Hunter Holmes McGuire VAMC, Richmond, Virginia
| | - Josephine J Wallner
- Spinal Cord Injury and Disorders Center, Hunter Holmes McGuire VAMC, Richmond, Virginia
| | - Parag N Gad
- Department of Neurobiology, University of California, Los Angeles, California
| | - V Reggie Edgerton
- Department of Neurobiology, University of California, Los Angeles, California
- Fundación Institut Guttmann, Institut Universitari de Neurorehabilitació Badalona, Barcelona, Spain
| | - Ashraf S Gorgey
- Spinal Cord Injury and Disorders Center, Hunter Holmes McGuire VAMC, Richmond, Virginia
- Physical Medicine and Rehabilitation, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
48
|
Zhang H, Liu Y, Zhou K, Wei W, Liu Y. Restoring Sensorimotor Function Through Neuromodulation After Spinal Cord Injury: Progress and Remaining Challenges. Front Neurosci 2021; 15:749465. [PMID: 34720867 PMCID: PMC8551759 DOI: 10.3389/fnins.2021.749465] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/13/2021] [Indexed: 12/27/2022] Open
Abstract
Spinal cord injury (SCI) is a major disability that results in motor and sensory impairment and extensive complications for the affected individuals which not only affect the quality of life of the patients but also result in a heavy burden for their families and the health care system. Although there are few clinically effective treatments for SCI, research over the past few decades has resulted in several novel treatment strategies which are related to neuromodulation. Neuromodulation-the use of neuromodulators, electrical stimulation or optogenetics to modulate neuronal activity-can substantially promote the recovery of sensorimotor function after SCI. Recent studies have shown that neuromodulation, in combination with other technologies, can allow paralyzed patients to carry out intentional, controlled movement, and promote sensory recovery. Although such treatments hold promise for completely overcoming SCI, the mechanisms by which neuromodulation has this effect have been difficult to determine. Here we review recent progress relative to electrical neuromodulation and optogenetics neuromodulation. We also examine potential mechanisms by which these methods may restore sensorimotor function. We then highlight the strengths of these approaches and remaining challenges with respect to its application.
Collapse
Affiliation(s)
- Hui Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yaping Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Kai Zhou
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Wei Wei
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yaobo Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| |
Collapse
|
49
|
Jankowska E, Hammar I. The plasticity of nerve fibers: the prolonged effects of polarization of afferent fibers. J Neurophysiol 2021; 126:1568-1591. [PMID: 34525323 DOI: 10.1152/jn.00718.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The review surveys various aspects of the plasticity of nerve fibers, in particular the prolonged increase in their excitability evoked by polarization, focusing on a long-lasting increase in the excitability of myelinated afferent fibers traversing the dorsal columns of the spinal cord. We review the evidence that increased axonal excitability 1) follows epidurally applied direct current (DC) as well as relatively short (5 or 10 ms) current pulses and synaptically evoked intrinsic field potentials; 2) critically depends on the polarization of branching regions of afferent fibers at the sites where they bifurcate and give off axon collaterals entering the spinal gray matter in conjunction with actions of extrasynaptic GABAA membrane receptors; and 3) shares the feature of being activity-independent with the short-lasting effects of polarization of peripheral nerve fibers. A comparison between the polarization evoked sustained increase in the excitability of dorsal column fibers and spinal motoneurons (plateau potentials) indicates the possibility that they are mediated by partly similar membrane channels (including noninactivating type L Cav++ 1.3 but not Na+ channels) and partly different mechanisms. We finally consider under which conditions transspinally applied DC (tsDCS) might reproduce the effects of epidural polarization on dorsal column fibers and the possible advantages of increased excitability of afferent fibers for the rehabilitation of motor and sensory functions after spinal cord injuries.NEW & NOTEWORTHY This review supplements previous reviews of properties of nerve fibers by surveying recent experimental evidence for their long-term plasticity. It also extends recent descriptions of spinal effects of DC by reviewing effects of polarization of afferent nerve fibers within the dorsal columns, the mechanisms most likely underlying the long-lasting increase in their excitability and possible clinical implications.
Collapse
Affiliation(s)
- Elzbieta Jankowska
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ingela Hammar
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
50
|
Shin JC, Jeon HR, Kim D, Cho SI, Min WK, Lee JS, Oh DS, Yoo J. Effects on the Motor Function, Proprioception, Balance, and Gait Ability of the End-Effector Robot-Assisted Gait Training for Spinal Cord Injury Patients. Brain Sci 2021; 11:brainsci11101281. [PMID: 34679346 PMCID: PMC8534051 DOI: 10.3390/brainsci11101281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 11/16/2022] Open
Abstract
The primary aim of this study was to reveal the effects of end-effector robot-assisted gait training (RAGT) on motor function, proprioception, balance, and gait ability in patients with incomplete spinal cord injury (SCI). The secondary aim was to determine the correlation between clinical outcomes. This study was a prospective and multi-center study. A total of 13 incomplete SCI patients who met inclusion criteria received 30 min of RAGT with Morning Walk® (Curexo, Seoul, South Korea), and 1 h of conventional physiotherapy 5 times per week for 4 weeks. Clinical outcome measures were 10 m walk test (10MWT), 6 min walk test (6mWT), lower extremity motor score (LEMS), proprioception, Berg Balance Scale (BBS), and Walking Index for Spinal Cord Injury (WISCI)-II. All participants were assessed within 48 h before and after the intervention. All clinical outcomes were statistically improved after RAGT. Subgroup analysis according to the initial proprioception, WISCI-II in the normal group showed a statistically significant improvement compared to the abnormal group. Initial BBS and WISCI-II had a positive correlation with most of the final clinical outcomes. The final BBS had a strong positive correlation with the final 10MWT, 6mWT, and WISCI-II. Initial proprioception had a positive correlation with the final WISCI-II. The final proprioception also had a moderate positive correlation with 6mWT and BBS. This study's results suggest that the end-effector RAGT could promote proprioception, balance ability and walking ability. Postural control ability and proprioception also had a positive relationship with gait ability.
Collapse
Affiliation(s)
- Ji Cheol Shin
- Department and Research Institute of Rehabilitation Medicine, Severance Hospital, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Korea; (J.C.S.); (D.K.)
| | - Ha Ra Jeon
- Department of Physical Medicine and Rehabilitation, National Health Insurance Service Ilsan Hospital, Ilsan-ro 100, Ilsandong-gu, Goyang-si 10444, Korea;
| | - Dahn Kim
- Department and Research Institute of Rehabilitation Medicine, Severance Hospital, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Korea; (J.C.S.); (D.K.)
| | - Sung Il Cho
- Rehabilitation Center, Inje University Ilsan Paik Hospital, Juhwa-ro 170, Ilsanseo-gu, Goyang-si 10380, Korea;
| | - Won Kyu Min
- Department of Rehabilitation Center, Severance Hospital, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Korea;
| | - June Sung Lee
- Department of Rehabilitation Center, National Health Insurance Service Ilsan Hospital, Ilsan-ro 100, Ilsandong-gu, Goyang-si 10444, Korea;
| | - Da Som Oh
- Inje Industry, Academic Cooperation Foundation, Inje-ro 197, Gimhae-si 50834, Korea;
| | - Jeehyun Yoo
- Department of Rehabilitation Medicine, Inje University Ilsan Paik Hospital, Juhwa-ro 170, Ilsanseo-gu, Goyang-si 10380, Korea
- Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Korea
- Correspondence: ; Tel.: +82-31-910-7440
| |
Collapse
|