1
|
Braaß H, Wolf S, Feldheim J, Chu Y, Tinnermann A, Finsterbusch J, Büchel C, Gerloff C, Schulz R. Altered Functional Connectivity Between Cortical Premotor Areas and the Spinal Cord in Chronic Stroke. Stroke 2025; 56:1159-1168. [PMID: 40110598 PMCID: PMC12036787 DOI: 10.1161/strokeaha.124.048384] [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: 07/05/2024] [Revised: 01/14/2025] [Accepted: 02/12/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Neuroscience research has contributed significantly to understanding alterations in brain structure and function after ischemic stroke. Technical limitations have excluded the spinal cord from imaging-based research. Available data are restricted to a few microstructural analyses, and functional connectivity data are absent. The present study attempted to close this knowledge gap and assess alterations in corticospinal coupling in chronic stroke and their relation to motor deficits. METHODS In this cross-sectional study, patients with chronic stroke and healthy controls underwent corticospinal functional magnetic resonance imaging while performing a simple force generation task at the University Medical Center Hamburg-Eppendorf between September 2021 and June 2023. Task-related activation was localized in the ipsilesional ventral premotor cortex, the supplementary motor area, and the cervical spinal cord. Psycho-physiological interactions and linear modeling were used to infer functional connectivity between cortical motor regions and the cervical spinal cord and their associations with clinical scores. RESULTS Thirteen well-recovered patients with stroke (1 woman, 12 men; mean age, 62.6 years; mean time after stroke: 47.6 months) and 13 healthy controls (5 women, 8 men; mean age, 64.5 years) were included. The main finding was that ventral premotor cortex and supplementary motor area showed topographically distinct alterations in their connectivity with the spinal cord. Specifically, we found a reduced coupling between the supplementary motor area and the ipsilateral ventral spinal cord and an enhanced coupling between the ventral premotor cortex and ventral and intermediate central spinal zones. Lower supplementary motor area and higher ventral premotor cortex-related spinal cord couplings were correlated with residual deficits. CONCLUSIONS This work provides first-in-human functional insights into stroke-related alterations in the functional connectivity between cortical premotor areas and the spinal cord, suggesting that different premotor areas and spinal neuronal assemblies might be involved in coupling changes. It adds a novel, promising approach to better understanding stroke recovery and developing innovative models to comprehend treatment strategies with spinal cord stimulation.
Collapse
Affiliation(s)
- Hanna Braaß
- Department of Neurology (H.B., S.W., J. Feldheim, C.G., R.S.), University Medical Center Hamburg-Eppendorf, Germany
- Department of Systems Neuroscience (H.B., Y.C., A.T., J. Finsterbusch, C.B.), University Medical Center Hamburg-Eppendorf, Germany
| | - Silke Wolf
- Department of Neurology (H.B., S.W., J. Feldheim, C.G., R.S.), University Medical Center Hamburg-Eppendorf, Germany
| | - Jan Feldheim
- Department of Neurology (H.B., S.W., J. Feldheim, C.G., R.S.), University Medical Center Hamburg-Eppendorf, Germany
| | - Ying Chu
- Department of Systems Neuroscience (H.B., Y.C., A.T., J. Finsterbusch, C.B.), University Medical Center Hamburg-Eppendorf, Germany
| | - Alexandra Tinnermann
- Department of Systems Neuroscience (H.B., Y.C., A.T., J. Finsterbusch, C.B.), University Medical Center Hamburg-Eppendorf, Germany
| | - Jürgen Finsterbusch
- Department of Systems Neuroscience (H.B., Y.C., A.T., J. Finsterbusch, C.B.), University Medical Center Hamburg-Eppendorf, Germany
| | - Christian Büchel
- Department of Systems Neuroscience (H.B., Y.C., A.T., J. Finsterbusch, C.B.), University Medical Center Hamburg-Eppendorf, Germany
| | - Christian Gerloff
- Department of Neurology (H.B., S.W., J. Feldheim, C.G., R.S.), University Medical Center Hamburg-Eppendorf, Germany
| | - Robert Schulz
- Department of Neurology (H.B., S.W., J. Feldheim, C.G., R.S.), University Medical Center Hamburg-Eppendorf, Germany
| |
Collapse
|
2
|
Huang Z, Li J, Wo J, Li C, Wu Z, Deng X, Liang Y, Li F, Chen B, Jia B, Wang L, Wang Y, Sun G, Li Z, Zhu H, Guest JD, So K, Fu Q, Zhou L. Intranasal Delivery of Brain-Derived Neurotrophic Factor (BDNF)-Loaded Small Extracellular Vesicles for Treating Acute Spinal Cord Injury in Rats and Monkeys. J Extracell Vesicles 2025; 14:e70066. [PMID: 40194993 PMCID: PMC11975507 DOI: 10.1002/jev2.70066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 03/10/2025] [Indexed: 04/09/2025] Open
Abstract
Besides surgical decompression, neuroprotection and neuroinflammation reduction are critical for acute spinal cord injury (SCI). In this study, we prepared small extracellular vesicles (sEVs) from immortalised mesenchymal stem cells overexpressing brain-derived neurotrophic factor (BDNF) and evaluated whether intranasal administration of BDNF-sEVs is a therapeutic option for acute SCI. In cultured neurons, BDNF loading enhanced neurite outgrowth promoted by sEVs. After intranasal administration, mCherry-labelled sEVs were transported to the injured spinal cords of rats and monkeys and mainly taken up by neurons. In acute SCI rats, intranasal administration of sEVs and BDNF-sEVs reduced glial responses and proinflammatory cytokine production, enhanced neuronal survival and angiogenesis in the lesion, promoted injured axon rewiring, delayed lumbar spinal motoneuron atrophy below the lesion, and improved functional performance. The rats receiving BDNF-sEV treatment showed improved neural repair and functional recovery compared to those with sEV treatment. Intranasal administration of BDNF-sEVs, but not of sEVs, increased BDNF levels and phosphorylation of downstream signals in the rat-injured spinal cord samples, indicating activation of the BDNF/TrkB signalling pathway. In acute SCI monkeys, intranasal administration of BDNF-sEVs was further confirmed to inhibit glial reactivities and proinflammatory cytokine release, increasing BDNF levels in the cerebrospinal fluid, enhancing neural network rewiring of injured spinal cords and neuronal activities of the brain, and improving functional performances in behavioural tests and electrophysiological recordings. In conclusion, BDNF-sEVs play a combinatory therapeutic role of sEVs and BDNF, and intranasal administration of BDNF-sEVs is a potential option for the clinical treatment of acute SCI.
Collapse
Affiliation(s)
- Zhonghai Huang
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionThe Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital)Jinan UniversityHeyuanChina
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Jing Li
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
- Department of Human Anatomy, The College of Basic Medical SciencesJinan UniversityGuangzhouChina
| | - Jin Wo
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionThe Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital)Jinan UniversityHeyuanChina
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Cheng‐Lin Li
- Otorhinolaryngology Hospital of The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zi‐Cong Wu
- Otorhinolaryngology Hospital of The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Xiao‐Hui Deng
- Otorhinolaryngology Hospital of The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Extracellular Vesicle Research and Clinical Translational CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yaying Liang
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
| | - Fuxiang Li
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
| | - Boli Chen
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
| | - Bin Jia
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT‐MRI CenterThe First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Ying Wang
- Medical Imaging CenterThe First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Guodong Sun
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionThe Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital)Jinan UniversityHeyuanChina
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Zhizhong Li
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionThe Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital)Jinan UniversityHeyuanChina
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Hui Zhu
- Kunming International Spine and Spinal Cord Injury Treatment CenterKunming Tongren HospitalKunmingChina
| | - James D Guest
- Neurological Surgery and the Miami Project to Cure ParalysisMiller School of MedicineMiamiFloridaUSA
| | - Kwok‐Fai So
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
- Department of Human Anatomy, The College of Basic Medical SciencesJinan UniversityGuangzhouChina
- Neuroscience and Neurorehabilitation InstituteUniversity of Health and Rehabilitation SciencesQingdaoShandongChina
- Co‐Innovation Center of NeuroregenerationNantong UniversityJiangsuChina
- Center for Exercise and Brain ScienceSchool of PsychologyShanghai University of SportShanghaiChina
| | - Qing‐Ling Fu
- Otorhinolaryngology Hospital of The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Extracellular Vesicle Research and Clinical Translational CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Libing Zhou
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionThe Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital)Jinan UniversityHeyuanChina
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouChina
- Neuroscience and Neurorehabilitation InstituteUniversity of Health and Rehabilitation SciencesQingdaoShandongChina
- Co‐Innovation Center of NeuroregenerationNantong UniversityJiangsuChina
- Center for Exercise and Brain ScienceSchool of PsychologyShanghai University of SportShanghaiChina
| |
Collapse
|
3
|
Inoue T, Ueno M. The diversity and plasticity of descending motor pathways rewired after stroke and trauma in rodents. Front Neural Circuits 2025; 19:1566562. [PMID: 40191711 PMCID: PMC11968733 DOI: 10.3389/fncir.2025.1566562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 03/10/2025] [Indexed: 04/09/2025] Open
Abstract
Descending neural pathways to the spinal cord plays vital roles in motor control. They are often damaged by brain injuries such as stroke and trauma, which lead to severe motor impairments. Due to the limited capacity for regeneration of neural circuits in the adult central nervous system, currently no essential treatments are available for complete recovery. Notably, accumulating evidence shows that residual circuits of the descending pathways are dynamically reorganized after injury and contribute to motor recovery. Furthermore, recent technological advances in cell-type classification and manipulation have highlighted the structural and functional diversity of these pathways. Here, we focus on three major descending pathways, namely, the corticospinal tract from the cerebral cortex, the rubrospinal tract from the red nucleus, and the reticulospinal tract from the reticular formation, and summarize the current knowledge of their structures and functions, especially in rodent models (mice and rats). We then review and discuss the process and patterns of reorganization induced in these pathways following injury, which compensate for lost connections for recovery. Understanding the basic structural and functional properties of each descending pathway and the principles of the induction and outcome of the rewired circuits will provide therapeutic insights to enhance interactive rewiring of the multiple descending pathways for motor recovery.
Collapse
Affiliation(s)
- Takahiro Inoue
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
4
|
Urbin MA. Adaptation in the spinal cord after stroke: Implications for restoring cortical control over the final common pathway. J Physiol 2025; 603:685-721. [PMID: 38787922 DOI: 10.1113/jp285563] [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: 01/24/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Control of voluntary movement is predicated on integration between circuits in the brain and spinal cord. Although damage is often restricted to supraspinal or spinal circuits in cases of neurological injury, both spinal motor neurons and axons linking these cells to the cortical origins of descending motor commands begin showing changes soon after the brain is injured by stroke. The concept of 'transneuronal degeneration' is not new and has been documented in histological, imaging and electrophysiological studies dating back over a century. Taken together, evidence from these studies comports more with a system attempting to survive rather than one passively surrendering to degeneration. There tends to be at least some preservation of fibres at the brainstem origin and along the spinal course of the descending white matter tracts, even in severe cases. Myelin-associated proteins are observed in the spinal cord years after stroke onset. Spinal motor neurons remain morphometrically unaltered. Skeletal muscle fibres once innervated by neurons that lose their source of trophic input receive collaterals from adjacent neurons, causing spinal motor units to consolidate and increase in size. Although some level of excitability within the distributed brain network mediating voluntary movement is needed to facilitate recovery, minimal structural connectivity between cortical and spinal motor neurons can support meaningful distal limb function. Restoring access to the final common pathway via the descending input that remains in the spinal cord therefore represents a viable target for directed plasticity, particularly in light of recent advances in rehabilitation medicine.
Collapse
Affiliation(s)
- Michael A Urbin
- Human Engineering Research Laboratories, VA RR&D Center of Excellence, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
5
|
Saijilafu, Ye LC, Li H, Li H, Lin X, Hu K, Huang Z, Chimedtseren C, Fang L, Saijilahu, Xu RJ. A bibliometric analysis of the top 100 most cited articles on corticospinal tract regeneration from 2004 to 2024. Front Neurosci 2025; 18:1509850. [PMID: 39935762 PMCID: PMC11811756 DOI: 10.3389/fnins.2024.1509850] [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: 10/11/2024] [Accepted: 12/23/2024] [Indexed: 02/13/2025] Open
Abstract
Objective Here, bibliometric and visual analytical techniques were employed to assess the key features of the 100 most cited publications concerning corticospinal tract (CST) regeneration. Methods Research was conducted within the Web of Science Core Collection to pinpoint the 100 most cited publications on CST regeneration. From these, comprehensive data encompassing titles, authorship, key terms, publication venues, release timelines, geographic origins, and institutional affiliations were extracted, followed by an in-depth bibliometric examination. Results The 100 most cited publications were all published between 2004 and 2024. These seminal papers amassed an aggregate of 18,321 citations, with individual citation counts ranging from 83 to 871 and a median of 136 citations per paper. Schwab M. E., stood out as the most prominent contributor, with significant authorship in 9 of the 100 papers. The United States dominated the geographical distribution, accounting for 49 of the articles. With 17 publications, the University of California System led the institutional rankings. A thorough keyword analysis revealed pivotal themes in the field, encompassing the optic nerve, gene expression, CST integrity and regeneration, diffusion tensor imaging, myelin-associated glycoproteins, inhibitors of neurite outgrowth, and methods of electrical and intracortical microstimulation. Conclusion This investigation provides a bibliometric analysis of CST regeneration, underscoring the significant contribution of the United States to this field. Our findings unveiled the dynamics and trends within the field of CST regeneration, providing a scientific foundation for advancing clinical applications. Building on this analysis, the clinical application of CST regeneration should be optimized through interdisciplinary collaboration, enabling the exploration and validation of a variety of therapeutic approaches, including the use of neurotrophic factors, stem cell therapies, biomaterials, and electrical stimulation. Concurrently, additional clinical trials are necessary to test the safety and efficacy of these therapeutic methods and develop assessment tools for monitoring the recovery of patients. Furthermore, rehabilitation strategies should be refined, and professional education and training should be provided to enhance the understanding of CST regeneration treatments among both medical professionals and patients. The implementation of these strategies promises to enhance therapeutic outcomes and the quality of life of patients with spinal cord injury (SCI).
Collapse
Affiliation(s)
- Saijilafu
- Hangzhou Lin’an Traditional Chinese Medicine Hospital, Affiliated Hospital, Hangzhou City University, Hangzhou, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Ling-Chen Ye
- Department of Orthopaedics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Huanyi Li
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Haokun Li
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Xinyi Lin
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Kehui Hu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Zekai Huang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | | | - Linjun Fang
- Hangzhou Lin’an Traditional Chinese Medicine Hospital, Affiliated Hospital, Hangzhou City University, Hangzhou, China
| | - Saijilahu
- Tongliao Centers for Disease Control and Prevention, Tongliao, China
| | - Ren-Jie Xu
- Department of Orthopaedics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| |
Collapse
|
6
|
Li Z, Jiang A, Fang J, Jiang Y, He W, Yan L, Qiu S, Qin B, Zhu Q, Wang H. METTL5-mediated 18S rRNA m 6A modification promotes corticospinal tract sprouting after unilateral traumatic brain injury. Exp Neurol 2024; 383:115000. [PMID: 39406306 DOI: 10.1016/j.expneurol.2024.115000] [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: 07/15/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
The key to improving function of an impaired limb after unilateral brain injury is promotion of corticospinal tract (CST) sprouting across the midline into the denervated hemicord. Previous studies have unveiled specific genes that regulate CST sprouting. CST sprouting may also be regulated by RNA modification. We examined METTL5, the methyltransferase for 18S rRNA m6A modification, as a regulator of CST sprouting in mice. Overexpression of METTL5 in contralesional corticospinal neurons promoted CST sprouting after unilateral traumatic brain injury. Mechanically, METTL5-mediated 18S rRNA m6A modification promoted the translation efficiency (TE) of various genes. Notably, the upregulation of TE in the gene Cfl1, which encodes cofilin, led to an increase in its expression. Additionally, the upregulation of TE in the gene Inpp5k led to the activation of cofilin. Active cofilin stimulates actin polymerization and facilitates protrusion and bundling of microtubules, thus promoting axon outgrowth. These findings offer valuable insights for developing novel strategies to promote CST sprouting.
Collapse
Affiliation(s)
- Zhenpeng Li
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - An Jiang
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Jintao Fang
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Yifei Jiang
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Wenting He
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Liwei Yan
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Shuai Qiu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Bengang Qin
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Qingtang Zhu
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Honggang Wang
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China.
| |
Collapse
|
7
|
Poinsatte K, Kenwood M, Betz D, Nawaby A, Ajay AD, Xu W, Plautz EJ, Kong X, Ramirez DMO, Goldberg MP. SpinalTRAQ: A novel volumetric cervical spinal cord atlas identifies the corticospinal tract synaptic projectome in healthy and post-stroke mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609434. [PMID: 39416130 PMCID: PMC11482800 DOI: 10.1101/2024.08.23.609434] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Descending corticospinal tract (CST) connections to the neurons of the cervical spinal cord are vital for performance of forelimb-specific fine motor skills. In rodents, CST axons are almost entirely crossed at the level of the medullary decussation. While specific contralateral axon projections have been well-characterized using anatomic and molecular approaches, the field currently lacks a cohesive imaging modality allowing rapid quantitative assessment of the entire, bilateral cervical cord projectome at the level of individual laminae and cervical levels. This is potentially important as the CST is known to undergo marked structural remodeling in development, injury, and disease. We developed SpinalTRAQ (Spinal cord Tomographic Registration and Automated Quantification), a novel volumetric cervical spinal cord atlas and machine learning-driven microscopy acquisition and analysis pipeline that uses serial two-photon tomography- images to generate unbiased, region-specific quantification of the fluorescent pixels of anterograde AAV-labeled CST pre-synaptic terminals. In adult mice, the CST synaptic projectome densely innervates the contralateral hemicord, particularly in laminae 5 and 7, with sparse, monosynaptic input to motoneurons in lamina 9. Motor pools supplying axial musculature in the upper cervical cord are bilaterally innervated. The remainder of the ipsilateral cord has sparse labeling in a distinct distribution compared to the contralateral side. Following a focal stroke of the motor cortex, there is a complete loss of descending corticospinal axons from the injured side. Consistent with prior reports of axon collateralization, the CST spinal projectome increases at four weeks post-stroke and continues to elevate by six weeks post stroke. At six weeks post-stroke, we observed striking synapse formation in the denervated hemicord from the uninjured CST in a homotopic distribution. Additionally, CST synaptic reinnervation increases in the denervated lamina 9 in nearly all motoneuron pools, exhibiting novel patterns of connectivity. Detailed level- and lamina-specific quantification of the bilateral cervical spinal cord synaptic projectome reveals previously undescribed patterns of CST connectivity in health and injury-related plasticity.
Collapse
Affiliation(s)
- Katherine Poinsatte
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Matthew Kenwood
- Department of Neurology, Long School of Medicine, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
- Graduate School of Biomedical Science (Neuroscience), University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Dene Betz
- Department of Neurology, Long School of Medicine, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
- Graduate School of Biomedical Science (Neuroscience), University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Ariana Nawaby
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Apoorva D Ajay
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Wei Xu
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Erik J Plautz
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xiangmei Kong
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Denise M O Ramirez
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Mark P Goldberg
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology, Long School of Medicine, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Lead Contact
| |
Collapse
|
8
|
Huang L, Yi L, Huang H, Zhan S, Chen R, Yue Z. Corticospinal tract: a new hope for the treatment of post-stroke spasticity. Acta Neurol Belg 2024; 124:25-36. [PMID: 37704780 PMCID: PMC10874326 DOI: 10.1007/s13760-023-02377-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/30/2023] [Indexed: 09/15/2023]
Abstract
Stroke is the third leading cause of death and disability worldwide. Post-stroke spasticity (PSS) is the most common complication of stroke but represents only one of the many manifestations of upper motor neuron syndrome. As an upper motor neuron, the corticospinal tract (CST) is the only direct descending motor pathway that innervates the spinal motor neurons and is closely related to the recovery of limb function in patients with PSS. Therefore, promoting axonal remodeling in the CST may help identify new therapeutic strategies for PSS. In this review, we outline the pathological mechanisms of PSS, specifically their relationship with CST, and therapeutic strategies for axonal regeneration of the CST after stroke. We found it to be closely associated with astroglial scarring produced by astrocyte activation and its secretion of neurotrophic factors, mainly after the onset of cerebral ischemia. We hope that this review offers insight into the relationship between CST and PSS and provides a basis for further studies.
Collapse
Affiliation(s)
- Linxing Huang
- College of Acupuncture, Massage and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Lizhen Yi
- College of Acupuncture, Massage and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Huiyuan Huang
- College of Acupuncture, Massage and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Sheng Zhan
- College of Acupuncture, Massage and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ruixue Chen
- College of Acupuncture, Massage and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zenghui Yue
- College of Acupuncture, Massage and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
9
|
Dumitru ML. Brain asymmetry is globally different in males and females: exploring cortical volume, area, thickness, and mean curvature. Cereb Cortex 2023; 33:11623-11633. [PMID: 37851852 PMCID: PMC10724869 DOI: 10.1093/cercor/bhad396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/20/2023] Open
Abstract
Brain asymmetry is a cornerstone in the development of higher-level cognition, but it is unclear whether and how it differs in males and females. Asymmetry has been investigated using the laterality index, which compares homologous regions as pairwise weighted differences between the left and the right hemisphere. However, if asymmetry differences between males and females are global instead of pairwise, involving proportions between multiple brain areas, novel methodological tools are needed to evaluate them. Here, we used the Amsterdam Open MRI collection to investigate sexual dimorphism in brain asymmetry by comparing laterality index with the distance index, which is a global measure of differences within and across hemispheres, and with the subtraction index, which compares pairwise raw values in the left and right hemisphere. Machine learning models, robustness tests, and group analyses of cortical volume, area, thickness, and mean curvature revealed that, of the three indices, distance index was the most successful biomarker of sexual dimorphism. These findings suggest that left-right asymmetry in males and females involves global coherence rather than pairwise contrasts. Further studies are needed to investigate the biological basis of local and global asymmetry based on growth patterns under genetic, hormonal, and environmental factors.
Collapse
Affiliation(s)
- Magda L Dumitru
- Department of Biological Sciences, University of Bergen, Postboks 7803, 5020 Bergen, Norway
- Department of Biological and Medical Psychology, University of Bergen, Postboks 7807, 5020 Bergen, Norway
| |
Collapse
|
10
|
Van Steenbergen V, Burattini L, Trumpp M, Fourneau J, Aljović A, Chahin M, Oh H, D’Ambra M, Bareyre FM. Coordinated neurostimulation promotes circuit rewiring and unlocks recovery after spinal cord injury. J Exp Med 2023; 220:e20220615. [PMID: 36571760 PMCID: PMC9794600 DOI: 10.1084/jem.20220615] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 10/26/2022] [Accepted: 12/15/2022] [Indexed: 12/27/2022] Open
Abstract
Functional recovery after incomplete spinal cord injury depends on the effective rewiring of neuronal circuits. Here, we show that selective chemogenetic activation of either corticospinal projection neurons or intraspinal relay neurons alone led to anatomically restricted plasticity and little functional recovery. In contrast, coordinated stimulation of both supraspinal centers and spinal relay stations resulted in marked and circuit-specific enhancement of neuronal rewiring, shortened EMG latencies, and improved locomotor recovery.
Collapse
Affiliation(s)
- Valérie Van Steenbergen
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Laura Burattini
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Michelle Trumpp
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Julie Fourneau
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Almir Aljović
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Planegg-Martinsried, Germany
| | - Maryam Chahin
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Planegg-Martinsried, Germany
| | - Hanseul Oh
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Planegg-Martinsried, Germany
| | - Marta D’Ambra
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Florence M. Bareyre
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), LMU Munich, Munich, Germany
| |
Collapse
|
11
|
Wang J, Cai Y, Sun J, Feng H, Zhu X, Chen Q, Gao F, Ni Q, Mao L, Yang M, Sun B. Administration of intramuscular AAV-BDNF and intranasal AAV-TrkB promotes neurological recovery via enhancing corticospinal synaptic connections in stroke rats. Exp Neurol 2023; 359:114236. [PMID: 36183811 DOI: 10.1016/j.expneurol.2022.114236] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/14/2022] [Accepted: 09/25/2022] [Indexed: 12/30/2022]
Abstract
Stroke causes long-term disability in survivors. BDNF/TrkB plays an important role in synaptic plasticity and synaptic transmission in the central nervous system (CNS), promoting neurological recovery. In this study, we performed non-invasive treatment methods focused on intramuscular injection into stroke-injured forelimb muscles, or intranasal administration using adeno-associated virus (AAV) vectors carrying genes encoding BDNF or TrkB. In a permanent rat middle cerebral artery occlusion (MCAO) model, we assessed the effects of combination therapy with AAV-BDNF and AAV-TrkB on motor functional recovery and synaptic plasticity of the corticospinal connections. Our results showed that BDNF or TrkB gene transduced in the spinal anterior horn neurons and cerebral cortical neurons. Compared to AAV vector treatment alone, behavioral and electrophysiological results showed that the combination therapy significantly improved upper limb motor functional recovery and neurotransmission efficiency after stroke. BDA tracing, immunofluorescence staining, qRT-PCR, and transmission electron microscopy of synaptic ultrastructure results revealed that the combination therapy not only potently increased the expression of Synapsin I, PSD-95, and GAP-43, but also promoted the axonal remodeling and restoration of abnormal synaptic structures. These findings provide a new strategy for enhancing neural plasticity and a potential means to treat stroke clinically.
Collapse
Affiliation(s)
- Jing Wang
- Medical College of Qingdao University, Qingdao 266021, Shandong, China; Institute for Neurological Research, The Second Affiliated Hospital; School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Yichen Cai
- Institute for Neurological Research, The Second Affiliated Hospital; School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Jingyi Sun
- Department of Spinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Hua Feng
- Department of Otolaryngology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, China
| | - Xiaoyu Zhu
- Institute for Neurological Research, The Second Affiliated Hospital; School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Qian Chen
- Institute for Neurological Research, The Second Affiliated Hospital; School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Feng Gao
- Institute for Neurological Research, The Second Affiliated Hospital; School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Qingbin Ni
- Postdoctoral Workstation, Taian Central Hospital, Taian 271000, Shandong, China
| | - Leilei Mao
- Institute for Neurological Research, The Second Affiliated Hospital; School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China.
| | - Mingfeng Yang
- Institute for Neurological Research, The Second Affiliated Hospital; School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China.
| | - Baoliang Sun
- Medical College of Qingdao University, Qingdao 266021, Shandong, China; Institute for Neurological Research, The Second Affiliated Hospital; School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China.
| |
Collapse
|
12
|
Matsubayashi J, Kawaguchi Y, Kawakami Y, Takei K. Brain-derived neurotrophic factor (BDNF) induces antagonistic action to Nogo signaling by the upregulation of lateral olfactory tract usher substance (LOTUS) expression. J Neurochem 2023; 164:29-43. [PMID: 36448220 DOI: 10.1111/jnc.15732] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022]
Abstract
Neurons in the central nervous system (CNS) have limited capacity for axonal regeneration after trauma and neurological disorders due to an endogenous nonpermissive environment for axon regrowth in the CNS. Lateral olfactory tract usher substance (LOTUS) contributes to axonal tract formation in the developing brain and axonal regeneration in the adult brain as an endogenous Nogo receptor-1 (NgR1) antagonist. However, how LOTUS expression is regulated remains unclarified. This study examined molecular mechanism of regulation in LOTUS expression and found that brain-derived neurotrophic factor (BDNF) increased LOTUS expression in cultured hippocampal neurons. Exogenous application of BDNF increased LOTUS expression at both mRNA and protein levels in a dose-dependent manner. We also found that pharmacological inhibition with K252a and gene knockdown by siRNA of tropomyosin-related kinase B (TrkB), BDNF receptor suppressed BDNF-induced increase in LOTUS expression. Further pharmacological analysis of the TrkB signaling pathway revealed that BDNF increased LOTUS expression through mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K) cascades, but not phospholipase C-γ (PLCγ) cascade. Additionally, treatment with c-AMP response element binding protein (CREB) inhibitor partially suppressed BDNF-induced LOTUS expression. Finally, neurite outgrowth assay in cultured hippocampal neurons revealed that BDNF treatment-induced antagonism for NgR1 by up-regulating LOTUS expression. These findings suggest that BDNF may acts as a positive regulator of LOTUS expression through the TrkB signaling, thereby inducing an antagonistic action for NgR1 function by up-regulating LOTUS expression. Also, BDNF may synergistically affect axon regrowth through the upregulation of LOTUS expression.
Collapse
Affiliation(s)
- Junpei Matsubayashi
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan
| | - Yuki Kawaguchi
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan
| | - Yutaka Kawakami
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan.,Department of Anesthesiology, National Center for Neurology and Psychiatry, Kodaira, Japan
| | - Kohtaro Takei
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan
| |
Collapse
|
13
|
Nakamura Y, Kurabe M, Matsumoto M, Sato T, Miytashita S, Hoshina K, Kamiya Y, Tainaka K, Matsuzawa H, Ohno N, Ueno M. Cerebrospinal fluid-contacting neuron tracing reveals structural and functional connectivity for locomotion in the mouse spinal cord. eLife 2023; 12:83108. [PMID: 36805807 PMCID: PMC9943067 DOI: 10.7554/elife.83108] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 02/03/2023] [Indexed: 02/22/2023] Open
Abstract
Cerebrospinal fluid-contacting neurons (CSF-cNs) are enigmatic mechano- or chemosensory cells lying along the central canal of the spinal cord. Recent studies in zebrafish larvae and lampreys have shown that CSF-cNs control postures and movements via spinal connections. However, the structures, connectivity, and functions in mammals remain largely unknown. Here we developed a method to genetically target mouse CSF-cNs that highlighted structural connections and functions. We first found that intracerebroventricular injection of adeno-associated virus with a neuron-specific promoter and Pkd2l1-Cre mice specifically labeled CSF-cNs. Single-cell labeling of 71 CSF-cNs revealed rostral axon extensions of over 1800 μm in unmyelinated bundles in the ventral funiculus and terminated on CSF-cNs to form a recurrent circuitry, which was further determined by serial electron microscopy and electrophysiology. CSF-cNs were also found to connect with axial motor neurons and premotor interneurons around the central canal and within the axon bundles. Chemogenetic CSF-cNs inactivation reduced speed and step frequency during treadmill locomotion. Our data revealed the basic structures and connections of mouse CSF-cNs to control spinal motor circuits for proper locomotion. The versatile methods developed in this study will contribute to further understanding of CSF-cN functions in mammals.
Collapse
Affiliation(s)
- Yuka Nakamura
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata UniversityNiigataJapan
| | - Miyuki Kurabe
- Division of Anesthesiology, Niigata University Graduate School of Medical and Dental SciencesNiigataJapan
| | - Mami Matsumoto
- Section of Electron Microscopy, Supportive Center for Brain Research, National Institute for Physiological SciencesOkazakiJapan,Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Tokiharu Sato
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata UniversityNiigataJapan
| | - Satoshi Miytashita
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata UniversityNiigataJapan
| | - Kana Hoshina
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata UniversityNiigataJapan
| | - Yoshinori Kamiya
- Division of Anesthesiology, Niigata University Graduate School of Medical and Dental SciencesNiigataJapan
| | - Kazuki Tainaka
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata UniversityNiigataJapan
| | - Hitoshi Matsuzawa
- Center for Advanced Medicine and Clinical Research, Kashiwaba Neurosurgical HospitalSapporoJapan,Center for Integrated Human Brain Science, Niigata UniversityNiigataJapan
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, Jichi Medical University, School of MedicineShimotsukeJapan,Division of Ultrastructural Research, National Institute for Physiological SciencesOkazakiJapan
| | - Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata UniversityNiigataJapan
| |
Collapse
|
14
|
Chovsepian A, Empl L, Bareyre FM. Plasticity of callosal neurons in the contralesional cortex following traumatic brain injury. Neural Regen Res 2022; 18:1257-1258. [PMID: 36453402 PMCID: PMC9838154 DOI: 10.4103/1673-5374.360167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Affiliation(s)
- Alexandra Chovsepian
- Institute of Clinical Neuroimmunology, Biomedical Building and Clinic of the Ludwig-Maximilians Universität München, Munich, Germany
| | - Laura Empl
- Institute of Clinical Neuroimmunology, Biomedical Building and Clinic of the Ludwig-Maximilians Universität München, Munich, Germany
| | - Florence M. Bareyre
- Institute of Clinical Neuroimmunology, Biomedical Building and Clinic of the Ludwig-Maximilians Universität München, Munich, Germany,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany,Correspondence to: Florence M. Bareyre, .
| |
Collapse
|
15
|
Hemati-Gourabi M, Cao T, Romprey MK, Chen M. Capacity of astrocytes to promote axon growth in the injured mammalian central nervous system. Front Neurosci 2022; 16:955598. [PMID: 36203815 PMCID: PMC9530187 DOI: 10.3389/fnins.2022.955598] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/15/2022] [Indexed: 01/02/2023] Open
Abstract
Understanding the regulation of axon growth after injury to the adult central nervous system (CNS) is crucial to improve neural repair. Following acute focal CNS injury, astrocytes are one cellular component of the scar tissue at the primary lesion that is traditionally associated with inhibition of axon regeneration. Advances in genetic models and experimental approaches have broadened knowledge of the capacity of astrocytes to facilitate injury-induced axon growth. This review summarizes findings that support a positive role of astrocytes in axon regeneration and axon sprouting in the mature mammalian CNS, along with potential underlying mechanisms. It is important to recognize that astrocytic functions, including modulation of axon growth, are context-dependent. Evidence suggests that the local injury environment, neuron-intrinsic regenerative potential, and astrocytes’ reactive states determine the astrocytic capacity to support axon growth. An integrated understanding of these factors will optimize therapeutic potential of astrocyte-targeted strategies for neural repair.
Collapse
Affiliation(s)
| | - Tuoxin Cao
- Spinal Cord and Brain Injury Research Center, Lexington, KY, United States
| | - Megan K. Romprey
- Spinal Cord and Brain Injury Research Center, Lexington, KY, United States
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Meifan Chen
- Spinal Cord and Brain Injury Research Center, Lexington, KY, United States
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- *Correspondence: Meifan Chen,
| |
Collapse
|
16
|
Chen M, Ingle L, Plautz EJ, Kong X, Tang R, Ghosh N, Romprey MK, Fenske WK, Goldberg MP. LZK-dependent stimulation of astrocyte reactivity promotes corticospinal axon sprouting. Front Cell Neurosci 2022; 16:969261. [PMID: 36187291 PMCID: PMC9520579 DOI: 10.3389/fncel.2022.969261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Injury to the adult mammalian central nervous system induces compensatory plasticity of spared axons-referred to as collateral axon sprouting-that can facilitate neural recovery. The contribution of reactive astrocytes to axon sprouting remains elusive. Here, we sought to investigate the role of axon degeneration-reactive astrocytes in the regulation of collateral axon sprouting that occurs in the mouse spinal cord after unilateral photothrombotic stroke of the primary motor cortex. We identified astrocytic leucine zipper-bearing kinase (LZK) as a positive regulator of astrocyte reactivity to corticospinal axon degeneration. Remarkably, genetic stimulation of astrocyte reactivity, via LZK overexpression in adult astrocytes, enhanced corticospinal axon sprouting. LZK promoted the production of astrocyte-derived ciliary neurotrophic factor (CNTF) that likely enhanced axon growth in mice with astrocytic LZK overexpression after injury. Our finding that LZK-dependent stimulation of astrocyte reactivity promotes corticospinal axon sprouting highlights the potential of engineering astrocytes to support injury-induced axon plasticity for neural repair.
Collapse
Affiliation(s)
- Meifan Chen
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY, United States
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Laura Ingle
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Erik J. Plautz
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Xiangmei Kong
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Rui Tang
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Neil Ghosh
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Megan K. Romprey
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY, United States
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - William K. Fenske
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Mark P. Goldberg
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Neurology, University of Texas Health Science Center San Antonio, San Antonio, TX, United States
| |
Collapse
|
17
|
Selective plasticity of callosal neurons in the adult contralesional cortex following murine traumatic brain injury. Nat Commun 2022; 13:2659. [PMID: 35551446 PMCID: PMC9098892 DOI: 10.1038/s41467-022-29992-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/11/2022] [Indexed: 11/17/2022] Open
Abstract
Traumatic brain injury (TBI) results in deficits that are often followed by recovery. The contralesional cortex can contribute to this process but how distinct contralesional neurons and circuits respond to injury remains to be determined. To unravel adaptations in the contralesional cortex, we used chronic in vivo two-photon imaging. We observed a general decrease in spine density with concomitant changes in spine dynamics over time. With retrograde co-labeling techniques, we showed that callosal neurons are uniquely affected by and responsive to TBI. To elucidate circuit connectivity, we used monosynaptic rabies tracing, clearing techniques and histology. We demonstrate that contralesional callosal neurons adapt their input circuitry by strengthening ipsilateral connections from pre-connected areas. Finally, functional in vivo two-photon imaging demonstrates that the restoration of pre-synaptic circuitry parallels the restoration of callosal activity patterns. Taken together our study thus delineates how callosal neurons structurally and functionally adapt following a contralateral murine TBI. Which contralesional circuits adapt after traumatic brain injury (TBI) is unclear. Here the authors used in vivo imaging, retrograde labeling, rabies tracing, clearing and functional imaging to demonstrate that callosal neurons selectively adapt after TBI in mice.
Collapse
|
18
|
Saikia JM, Chavez-Martinez CL, Kim ND, Allibhoy S, Kim HJ, Simonyan L, Smadi S, Tsai KM, Romaus-Sanjurjo D, Jin Y, Zheng B. A Critical Role for DLK and LZK in Axonal Repair in the Mammalian Spinal Cord. J Neurosci 2022; 42:3716-3732. [PMID: 35361703 PMCID: PMC9087816 DOI: 10.1523/jneurosci.2495-21.2022] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/23/2022] [Accepted: 03/24/2022] [Indexed: 11/21/2022] Open
Abstract
The limited ability for axonal repair after spinal cord injury underlies long-term functional impairment. Dual leucine-zipper kinase [DLK; MAP kinase kinase kinase 12; MAP3K12] is an evolutionarily conserved MAP3K implicated in neuronal injury signaling from Caenorhabditis elegans to mammals. However, whether DLK or its close homolog leucine zipper kinase (LZK; MAP3K13) regulates axonal repair in the mammalian spinal cord remains unknown. Here, we assess the role of endogenous DLK and LZK in the regeneration and compensatory sprouting of corticospinal tract (CST) axons in mice of both sexes with genetic analyses in a regeneration competent background provided by PTEN deletion. We found that inducible neuronal deletion of both DLK and LZK, but not either kinase alone, abolishes PTEN deletion-induced regeneration and sprouting of CST axons, and reduces naturally-occurring axon sprouting after injury. Thus, DLK/LZK-mediated injury signaling operates not only in injured neurons to regulate regeneration, but also unexpectedly in uninjured neurons to regulate sprouting. Deleting DLK and LZK does not interfere with PTEN/mTOR signaling, indicating that injury signaling and regenerative competence are independently controlled. Together with our previous study implicating LZK in astrocytic reactivity and scar formation, these data illustrate the multicellular function of this pair of MAP3Ks in both neurons and glia in the injury response of the mammalian spinal cord.SIGNIFICANCE STATEMENT Functional recovery after spinal cord injury is limited because of a lack of axonal repair in the mammalian CNS. Dual leucine-zipper kinase (DLK) and leucine zipper kinase (LZK) are two closely related protein kinases that have emerged as regulators of neuronal responses to injury. However, their role in axonal repair in the mammalian spinal cord has not been described. Here, we show that DLK and LZK together play critical roles in axonal repair in the mammalian spinal cord, validating them as potential targets to promote repair and recovery after spinal cord injury. In addition to regulating axonal regeneration from injured neurons, both kinases also regulate compensatory axonal growth from uninjured neurons, indicating a more pervasive role in CNS repair than originally anticipated.
Collapse
Affiliation(s)
- Junmi M Saikia
- Department of Neurosciences, School of Medicine
- Neurosciences Graduate Program
| | | | - Noah D Kim
- Department of Neurosciences, School of Medicine
| | | | - Hugo J Kim
- Department of Neurosciences, School of Medicine
| | | | | | | | | | - Yishi Jin
- Department of Neurosciences, School of Medicine
- Department of Neurobiology, School of Biological Sciences
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, California 92093
| | - Binhai Zheng
- Department of Neurosciences, School of Medicine
- VA San Diego Healthcare System Research Service, San Diego, California 92161
| |
Collapse
|
19
|
Aljovic A, Zhao S, Chahin M, de la Rosa C, Van Steenbergen V, Kerschensteiner M, Bareyre FM. A deep learning-based toolbox for Automated Limb Motion Analysis (ALMA) in murine models of neurological disorders. Commun Biol 2022; 5:131. [PMID: 35169263 PMCID: PMC8847458 DOI: 10.1038/s42003-022-03077-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 01/21/2022] [Indexed: 11/24/2022] Open
Abstract
In neuroscience research, the refined analysis of rodent locomotion is complex and cumbersome, and access to the technique is limited because of the necessity for expensive equipment. In this study, we implemented a new deep learning-based open-source toolbox for Automated Limb Motion Analysis (ALMA) that requires only basic behavioral equipment and an inexpensive camera. The ALMA toolbox enables the consistent and comprehensive analyses of locomotor kinematics and paw placement and can be applied to neurological conditions affecting the brain and spinal cord. We demonstrated that the ALMA toolbox can (1) robustly track the evolution of locomotor deficits after spinal cord injury, (2) sensitively detect locomotor abnormalities after traumatic brain injury, and (3) correctly predict disease onset in a multiple sclerosis model. We, therefore, established a broadly applicable automated and standardized approach that requires minimal financial and time commitments to facilitate the comprehensive analysis of locomotion in rodent disease models. Presenting ALMA toolbox, an open source Python repository for automatic analysis of mouse locomotion using bodypart coordinates from markerless pose estimation tools. ALMA is capable of analyzing both healthy and CNS-injured mice. ALMA is also capable of predicting onset of disease in a multiple sclerosis model.
Collapse
Affiliation(s)
- Almir Aljovic
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universitaet Munich, 82152, Planegg-Martinsried, Germany
| | - Shuqing Zhao
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universitaet Munich, 82152, Planegg-Martinsried, Germany
| | - Maryam Chahin
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universitaet Munich, 82152, Planegg-Martinsried, Germany
| | - Clara de la Rosa
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universitaet Munich, 82152, Planegg-Martinsried, Germany
| | - Valerie Van Steenbergen
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany.,Munich Cluster of Systems Neurology (SyNergy), 81377, Munich, Germany
| | - Florence M Bareyre
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany. .,Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany. .,Munich Cluster of Systems Neurology (SyNergy), 81377, Munich, Germany.
| |
Collapse
|
20
|
Buzoianu-Anguiano V, Torres-Llacsa M, Doncel-Pérez E. Role of Aldynoglia Cells in Neuroinflammatory and Neuroimmune Responses after Spinal Cord Injury. Cells 2021; 10:2783. [PMID: 34685763 PMCID: PMC8534338 DOI: 10.3390/cells10102783] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/07/2021] [Accepted: 10/12/2021] [Indexed: 11/16/2022] Open
Abstract
Aldynoglia are growth-promoting cells with a morphology similar to radial glia and share properties and markers with astrocytes and Schwann cells. They are distributed in several locations throughout the adult central nervous system, where the cells of the aldynoglia interact and respond to the signals of the immune cells. After spinal cord injury (SCI), the functions of resident aldynoglia, identified as ependymocytes, tanycytes, and ependymal stem cells (EpSCs) of the spinal cord are crucial for the regeneration of spinal neural tissue. These glial cells facilitate axonal regrowth and remyelination of injured axons. Here, we review the influence of M1 or M2 macrophage/microglia subpopulations on the fate of EpSCs during neuroinflammation and immune responses in the acute, subacute, and chronic phases after SCI.
Collapse
Affiliation(s)
| | - Mabel Torres-Llacsa
- Servicio de Radiología, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain;
| | - Ernesto Doncel-Pérez
- Grupo de Química Neuro-Regenerativa, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain;
| |
Collapse
|
21
|
Sato T, Nakamura Y, Takeda A, Ueno M. Lesion Area in the Cerebral Cortex Determines the Patterns of Axon Rewiring of Motor and Sensory Corticospinal Tracts After Stroke. Front Neurosci 2021; 15:737034. [PMID: 34707476 PMCID: PMC8542932 DOI: 10.3389/fnins.2021.737034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/21/2021] [Indexed: 11/18/2022] Open
Abstract
The corticospinal tract (CST) is an essential neural pathway for reorganization that recovers motor functions after brain injuries such as stroke. CST comprises multiple pathways derived from different sensorimotor areas of the cerebral cortex; however, the patterns of reorganization in such complex pathways postinjury are largely unknown. Here we comprehensively examined the rewiring patterns of the CST pathways of multiple cerebral origins in a mouse stroke model that varied in size and location in the sensorimotor cortex. We found that spared contralesional motor and sensory CST axons crossed the midline and sprouted into the denervated side of the cervical spinal cord after stroke in a large cortical area. In contrast, the contralesional CST fibers did not sprout in a small stroke, whereas the ipsilesional axons from the spared motor area grew on the denervated side. We further showed that motor and sensory CST axons did not innervate the projecting areas mutually when either one was injured. The present results reveal the basic principles that generate the patterns of CST rewiring, which depend on stroke location and CST subtype. Our data indicate the importance of targeting different neural substrates to restore function among the types of injury.
Collapse
Affiliation(s)
| | | | | | - Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
22
|
Moreno-Lopez Y, Bichara C, Delbecq G, Isope P, Cordero-Erausquin M. The corticospinal tract primarily modulates sensory inputs in the mouse lumbar cord. eLife 2021; 10:65304. [PMID: 34497004 PMCID: PMC8439650 DOI: 10.7554/elife.65304] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 07/27/2021] [Indexed: 01/01/2023] Open
Abstract
It is generally assumed that the main function of the corticospinal tract (CST) is to convey motor commands to bulbar or spinal motoneurons. Yet the CST has also been shown to modulate sensory signals at their entry point in the spinal cord through primary afferent depolarization (PAD). By sequentially investigating different routes of corticofugal pathways through electrophysiological recordings and an intersectional viral strategy, we here demonstrate that motor and sensory modulation commands in mice belong to segregated paths within the CST. Sensory modulation is executed exclusively by the CST via a population of lumbar interneurons located in the deep dorsal horn. In contrast, the cortex conveys the motor command via a relay in the upper spinal cord or supraspinal motor centers. At lumbar level, the main role of the CST is thus the modulation of sensory inputs, which is an essential component of the selective tuning of sensory feedback used to ensure well-coordinated and skilled movement.
Collapse
Affiliation(s)
- Yunuen Moreno-Lopez
- Institut des Neurosciences Cellulaires et Intégrées, CNRS - Université de Strasbourg, Strasbourg, France
| | - Charlotte Bichara
- Institut des Neurosciences Cellulaires et Intégrées, CNRS - Université de Strasbourg, Strasbourg, France
| | - Gilles Delbecq
- Institut des Neurosciences Cellulaires et Intégrées, CNRS - Université de Strasbourg, Strasbourg, France
| | - Philippe Isope
- Institut des Neurosciences Cellulaires et Intégrées, CNRS - Université de Strasbourg, Strasbourg, France
| | - Matilde Cordero-Erausquin
- Institut des Neurosciences Cellulaires et Intégrées, CNRS - Université de Strasbourg, Strasbourg, France
| |
Collapse
|
23
|
Inoue T, Takamatsu Y, Okamura M, Maejima H. Ipsilateral BDNF mRNA expression in the motor cortex positively correlates with motor function of the affected forelimb after intracerebral hemorrhage. Brain Res 2021; 1767:147536. [PMID: 34052261 DOI: 10.1016/j.brainres.2021.147536] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/27/2021] [Accepted: 05/20/2021] [Indexed: 01/01/2023]
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stroke that causes major motor impairments. Brain-derived neurotrophic factor (BDNF) is known to have important roles in neuroplasticity and beneficially contributes to stroke recovery. This study aimed to characterize BDNF expression in the motor cortex after ICH and investigate the relationship between cortical BDNF expression and behavioral outcomes using an ICH rat model. Wistar rats were divided into two groups: a SHAM group (n = 7) and an ICH group (n = 8). ICH was induced by the injection of collagenase into the left striatum near the internal capsule. For behavioral assessments, the cylinder test and open field test were performed before surgery and 3 days, 1 week, 2 weeks, and 4 weeks after surgery. Following the behavioral assessments at 4 weeks, BDNF expression in the ipsilateral and contralateral motor cortex was assayed using RT-PCR and ELISA methods. There was no significant difference in either cortical BDNF mRNA or protein expression levels between the SHAM and ICH groups. However, the asymmetry index of BDNF mRNA expression between the ipsilateral and contralateral hemispheres shifted to the ipsilateral hemisphere after ICH. Furthermore, the ipsilateral cortical BDNF mRNA expression level positively correlated with motor function in the affected forelimb after ICH. This study describes for the first time that cortical BDNF mRNA expression is related to post-ICH motor impairment. These results highlight the importance of assessing the interhemispheric laterality of BDNF expression and could help develop novel treatment strategies for BDNF-dependent recovery after ICH.
Collapse
Affiliation(s)
- Takahiro Inoue
- Graduate School of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan; Research Fellow of Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo 102-0083, Japan
| | - Yasuyuki Takamatsu
- Department of Rehabilitation Science, Faculty of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Misato Okamura
- Graduate School of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Hiroshi Maejima
- Department of Rehabilitation Science, Faculty of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
24
|
Ueno M. Restoring neuro-immune circuitry after brain and spinal cord injuries. Int Immunol 2021; 33:311-325. [PMID: 33851981 DOI: 10.1093/intimm/dxab017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/13/2021] [Indexed: 12/17/2022] Open
Abstract
Neuro-immune interactions are essential for our body's defense and homeostasis. Anatomical and physiological analyses have shown that the nervous system comprises multiple pathways that regulate the dynamics and functions of immune cells, which are mainly mediated by the autonomic nervous system and adrenal signals. These are disturbed when the neurons and circuits are damaged by diseases of the central nervous system (CNS). Injuries caused by stroke or trauma often cause immune dysfunction by abrogation of the immune-regulating neural pathways, which leads to an increased risk of infections. Here, I review the structures and functions of the neural pathways connecting the brain and the immune system, and the neurogenic mechanisms of immune dysfunction that emerge after CNS injuries. Recent technological advances in manipulating specific neural circuits have added mechanistic aspects of neuro-immune interactions and their dysfunctions. Understanding the neural bases of immune control and their pathological processes will deepen our knowledge of homeostasis and lead to the development of strategies to cure immune deficiencies observed in various CNS disorders.
Collapse
Affiliation(s)
- Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Niigata 951-8585, Japan
| |
Collapse
|
25
|
Qiu X, Ping S, Kyle M, Chin L, Zhao LR. SCF + G-CSF treatment in the chronic phase of severe TBI enhances axonal sprouting in the spinal cord and synaptic pruning in the hippocampus. Acta Neuropathol Commun 2021; 9:63. [PMID: 33832542 PMCID: PMC8028149 DOI: 10.1186/s40478-021-01160-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 03/17/2021] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of long-term disability in young adults. An evidence-based treatment for TBI recovery, especially in the chronic phase, is not yet available. Using a severe TBI mouse model, we demonstrate that the neurorestorative efficacy of repeated treatments with stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) (SCF + G-CSF) in the chronic phase is superior to SCF + G-CSF single treatment. SCF + G-CSF treatment initiated at 3 months post-TBI enhances contralesional corticospinal tract sprouting into the denervated side of the cervical spinal cord and re-balances the TBI-induced overgrown synapses in the hippocampus by enhancing microglial function of synaptic pruning. These neurorestorative changes are associated with SCF + G-CSF-improved somatosensory-motor function and spatial learning. In the chronic phase of TBI, severe TBI-caused microglial degeneration in the cortex and hippocampus is ameliorated by SCF + G-CSF treatment. These findings reveal the therapeutic potential and possible mechanism of SCF + G-CSF treatment in brain repair during the chronic phase of severe TBI.
Collapse
|
26
|
Choo YJ, Kim JH, Chang MC. The prediction of need of using ankle-foot orthoses in stroke patients based on findings of a transcranial magnetic stimulation study. J Integr Neurosci 2021; 20:119-123. [PMID: 33834698 DOI: 10.31083/j.jin.2021.01.107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/16/2021] [Accepted: 03/10/2021] [Indexed: 11/06/2022] Open
Abstract
Ankle-foot orthoses (AFOs) are widely prescribed for stroke rehabilitation. We investigated the potential of transcranial magnetic stimulation (TMS) at an early stage, after stroke, to predict the need of using AFOs in stroke patients. We recruited 35 patients who could walk with intermittent support of one person or independently 3 months after onset of stroke. The patients included in the study were classified into two groups: a TMS (+) group (n = 10), in which motor-evoked potential (MEP) in the affected tibialis anterior (TA) was present, and a TMS (-) group (n = 25), in which the MEP in the affected TA was absent. Three months after the onset of stroke, we investigated whether patients were using AFOs or not. We also checked the motor function of the affected lower extremity using the Medical Research Council (MRC) scale. After 3 months of onset of stroke in the TMS (+) group, 4 patients (40%) were using an AFO during ambulation. In the TMS (-) group, 21 patients (84%) were using an AFO. The probability of using AFOs in the 2 groups were significantly different. Additionally, 3 months after the onset of stroke, the MRC scores of ankle dorsiflexor power, on the affected side, were significantly higher in the TMS (+) group. Early TMS evaluation of the corticospinal tract to the TA appears to be useful for predicting the need of using AFOs in stroke patients during the recovery phase.
Collapse
Affiliation(s)
- Yoo Jin Choo
- Department of Physical Medicine and Rehabilitation, Yeungnam University College of Medicine, 41020 Daegu, Republic of Korea
| | - Jang-Hwan Kim
- Department of Rehabilitation Technology, Graduate School of Hanseo University, Seosan, 840 92 Chungcheongnam-do, Republic of Korea
| | - Min Cheol Chang
- Department of Physical Medicine and Rehabilitation, Yeungnam University College of Medicine, 41020 Daegu, Republic of Korea
| |
Collapse
|
27
|
Matano Y, Nojiri Y, Nomura M, Masuda A, Moriike Y, Suzuki Y, Umemura K, Nagai N. Repair of brain damage size and recovery of neurological dysfunction after ischemic stroke are different between strains in mice: evaluation using a novel ischemic stroke model. Exp Anim 2021; 70:344-354. [PMID: 33731549 PMCID: PMC8390305 DOI: 10.1538/expanim.20-0182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
In the current study, we established a novel murine ischemic brain damage model using a photochemical reaction to evaluate the recovery of neurological
dysfunction and brain repair reactions. In this model, reproducible damage was induced in the frontal lobe of the cortex, which was accompanied by neurological
dysfunction. Sequential changes in damage size, microglial accumulation, astrocyte activation, and neurological dysfunction were studied in C57BL/6J and BALB/c
mouse strains. Although the initial size of damage was comparable in both strains, the extent of damage was later reduced to a greater extent in C57BL/6J mice
than that in BALB/c mice. In addition, C57BL/6J mice showed later edema clearance until day 7, less microglial accumulation, and relatively more astrocyte
activation on day 7. Neurologic dysfunction was evaluated by three behavioral tests: the von Frey test, the balance beam test, and the tail suspension test. The
behavioral abnormalities evaluated by these tests were remarkable following the induction of damage and recovered by day 21 in both strains. However, the
abnormalities were more prominent and the recovery was later in C57BL/6J mice. These findings demonstrate that our novel ischemic stroke model is useful for
evaluating brain repair reactions and the recovery of neurological dysfunction in mice with different genetic backgrounds. In addition, we found that both the
brain repair reactions and the recovery of neurological dysfunction after comparable ischemic brain damage varied between strains; in that, they both occurred
later in C57BL/6J mice.
Collapse
Affiliation(s)
- Yasuki Matano
- Laboratory of Animal Physiology, Division of Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura, Nagahama, Shiga 526-0829, Japan
| | - Yuuto Nojiri
- Laboratory of Animal Physiology, Division of Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura, Nagahama, Shiga 526-0829, Japan
| | - Mizuki Nomura
- Laboratory of Animal Physiology, Division of Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura, Nagahama, Shiga 526-0829, Japan
| | - Akira Masuda
- Laboratory of Animal Physiology, Division of Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura, Nagahama, Shiga 526-0829, Japan
| | - Yuuki Moriike
- Laboratory of Animal Physiology, Division of Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura, Nagahama, Shiga 526-0829, Japan
| | - Yasuhiro Suzuki
- School of Pharmaceutical Sciences, Faculty of Pharmacy, Ohu University, 31-1 Sankaku-do, Tomita-cho, Kohriyama, Fukushima 963-8611, Japan
| | - Kazuo Umemura
- Department of Pharmacology, Hamamatsu University School of Medicine, 1-20-1 Handa-yama, Higashi-ku, Hamamatsu, Shizuoka 431-3125, Japan
| | - Nobuo Nagai
- Laboratory of Animal Physiology, Division of Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura, Nagahama, Shiga 526-0829, Japan
| |
Collapse
|
28
|
Prolonged deficit of low gamma oscillations in the peri-infarct cortex of mice after stroke. Exp Neurol 2021; 341:113696. [PMID: 33727098 DOI: 10.1016/j.expneurol.2021.113696] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 03/04/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023]
Abstract
Days and weeks after an ischemic stroke, the peri-infarct area adjacent to the necrotic tissue exhibits very intense synaptic reorganization aimed at regaining lost functions. In order to enhance functional recovery, it is important to understand the mechanisms supporting neural repair and neuroplasticity in the cortex surrounding the lesion. Brain oscillations of the local field potential (LFP) are rhythmic fluctuations of neuronal excitability that synchronize neuronal activity to organize information processing and plasticity. Although the oscillatory activity of the brain has been probed after stroke in both animals and humans using electroencephalography (EEG), the latter is ineffective to precisely map the oscillatory changes in the peri-infarct zone where synaptic plasticity potential is high. Here, we worked on the hypothesis that the brain oscillatory system is altered in the surviving peri-infarct cortex, which may slow down the functional repair and reduce the recovery. In order to document the relevance of this hypothesis, oscillatory power was measured at various distances from the necrotic core at 7 and 21 days after a permanent cortical ischemia induced in mice. Delta and theta oscillations remained at a normal power in the peri-infarct cortex, in contrast to low gamma oscillations that displayed a gradual decrease, when approaching the border of the lesion. A broadband increase of power was also observed in the homotopic contralateral sites. Thus, the proximal peri-infarct cortex could become a target of therapeutic interventions applied to correct the oscillatory regimen in order to boost post-stroke functional recovery.
Collapse
|
29
|
Matsuura H, Ohnishi M, Yoshioka Y, Togami Y, Hosomi S, Umemura Y, Ebihara T, Shimizu K, Ogura H, Shimazu T. Original experimental rat model of blast-induced mild traumatic brain injury: a pilot study. Brain Inj 2021; 35:368-381. [PMID: 33455463 DOI: 10.1080/02699052.2020.1861653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Diagnosing blast-induced mild traumatic brain injury (mTBI) is difficult due to minimal imaging findings. This study aimed to establish a rat model of behavioral abnormality caused by blast-induced mTBI and detect new findings for therapeutic intervention. METHODS We used a bench-top blast wave generator with the blast wave exiting through a 20-mm I.D. nozzle aimed at the focused target. The blast wave was directed at the head of male Wistar rats under general anesthesia positioned prone 2.5 cm below the nozzle. Peak shock wave pressure was 646.2 ± 70.3 kPa. RESULTS After blast injury, mTBI rats did not show the findings of brain hemorrhage or contusion macroscopically and on hematoxylin-eosin-stained frozen sections but did show anorexia and weight loss in the early post-injury phase. Behavioral experiments revealed short-term memory impairment at 2 weeks and depression-like behavior at 2 and 6 weeks. Diffusion-weighted ex vivo MRI showed high-intensity areas in layers of the bilateral hippocampus. Immunohistochemical analysis revealed accumulation of reactive microglia and GFAP-positive astrocytes in the same region and loss of NeuN-positive neurons in the hippocampal pyramidal cell layer. CONCLUSIONS This model can reflect the pathophysiology of blast-induced mTBI and could potentially be used to develop therapeutic interventions in the future.
Collapse
Affiliation(s)
- Hiroshi Matsuura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Mitsuo Ohnishi
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshichika Yoshioka
- Center for Information and Neural Networks (Cinet), National Institute of Information and Communications Technology (NICT), and Osaka University, Osaka, Japan
| | - Yuki Togami
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sanae Hosomi
- Division of Trauma and Surgical Critical Care, Osaka General Medical Center, Osaka, Japan
| | - Yutaka Umemura
- Division of Trauma and Surgical Critical Care, Osaka General Medical Center, Osaka, Japan
| | - Takeshi Ebihara
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kentaro Shimizu
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroshi Ogura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takeshi Shimazu
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
30
|
Tsujioka H, Yamashita T. Neural circuit repair after central nervous system injury. Int Immunol 2020; 33:301-309. [PMID: 33270108 DOI: 10.1093/intimm/dxaa077] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/01/2020] [Indexed: 12/24/2022] Open
Abstract
Central nervous system injury often causes lifelong impairment of neural function, because the regenerative ability of axons is limited, making a sharp contrast to the successful regeneration that is seen in the peripheral nervous system. Nevertheless, partial functional recovery is observed, because axonal branches of damaged or undamaged neurons sprout and form novel relaying circuits. Using a lot of animal models such as the spinal cord injury model or the optic nerve injury model, previous studies have identified many factors that promote or inhibit axonal regeneration or sprouting. Molecules in the myelin such as myelin-associated glycoprotein, Nogo-A or oligodendrocyte-myelin glycoprotein, or molecules found in the glial scar such as chondroitin sulfate proteoglycans, activate Ras homolog A (RhoA) signaling, which leads to the collapse of the growth cone and inhibit axonal regeneration. By contrast, axonal regeneration programs can be activated by many molecules such as regeneration-associated transcription factors, cyclic AMP, neurotrophic factors, growth factors, mechanistic target of rapamycin or immune-related molecules. Axonal sprouting and axonal regeneration largely share these mechanisms. For functional recovery, appropriate pruning or suppressing of aberrant sprouting are also important. In contrast to adults, neonates show much higher sprouting ability. Specific cell types, various mouse strains and different species show higher regenerative ability. Studies focusing on these models also identified a lot of molecules that affect the regenerative ability. A deeper understanding of the mechanisms of neural circuit repair will lead to the development of better therapeutic approaches for central nervous system injury.
Collapse
Affiliation(s)
- Hiroshi Tsujioka
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.,WPI Immunology Frontier Research Center, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.,WPI Immunology Frontier Research Center, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.,Graduate School of Frontier Bioscience, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.,Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
31
|
TGFβ1 Induces Axonal Outgrowth via ALK5/PKA/SMURF1-Mediated Degradation of RhoA and Stabilization of PAR6. eNeuro 2020; 7:ENEURO.0104-20.2020. [PMID: 32887692 PMCID: PMC7540929 DOI: 10.1523/eneuro.0104-20.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/21/2020] [Accepted: 08/06/2020] [Indexed: 12/11/2022] Open
Abstract
Transforming growth factor (TGF)β1 has repeatedly been associated with axonal regeneration and recovery after injury to the CNS. We found TGFβ1 upregulated in the stroke-denervated mouse spinal cord after ischemic injury to the motor cortex as early as 4 d postinjury (dpi) and persisting up to 28 dpi. Given the potential role of TGFβ1 in structural plasticity and functional recovery after stroke highlighted in several published studies, we investigated its downstream signaling in an in vitro model of neurite outgrowth. We found that in this model, TGFβ1 rescues neurite outgrowth under growth inhibitory conditions via the canonical TGFβR2/ALK5 signaling axis. Thereby, protein kinase A (PKA)-mediated phosphorylation of the E3 ubiquitin ligase SMURF1 induces a switch of its substrate preference from PAR6 to the Ras homolog A (RhoA), in this way enhancing outgrowth on the level of the cytoskeleton. This proposed mechanism of TGFβ1 signaling could underly the observed increase in structural plasticity after stroke in vivo as suggested by the temporal and spatial expression of TGFβ1. In accordance with previous publications, this study corroborates the potential of TGFβ1 and associated signaling cascades as a target for future therapeutic interventions to enhance structural plasticity and functional recovery for stroke patients.
Collapse
|
32
|
Inhibition of HDAC increases BDNF expression and promotes neuronal rewiring and functional recovery after brain injury. Cell Death Dis 2020; 11:655. [PMID: 32811822 PMCID: PMC7434917 DOI: 10.1038/s41419-020-02897-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/17/2022]
Abstract
Brain injury causes serious motor, sensory, and cognitive disabilities. Accumulating evidence has demonstrated that histone deacetylase (HDAC) inhibitors exert neuroprotective effects against various insults to the central nervous system (CNS). In this study, we investigated the effects of the HDAC inhibition on the expression of brain-derived neurotrophic factor (BDNF) and functional recovery after traumatic brain injury (TBI) in mice. Administration of class I HDAC inhibitor increased the number of synaptic boutons in rewiring corticospinal fibers and improved the recovery of motor functions after TBI. Immunohistochemistry results showed that HDAC2 is mainly expressed in the neurons of the mouse spinal cord under normal conditions. After TBI, HDAC2 expression was increased in the spinal cord after 35 days, whereas BDNF expression was decreased after 42 days. Administration of CI-994 increased BDNF expression after TBI. Knockdown of HDAC2 elevated H4K5ac enrichment at the BDNF promoter, which was decreased following TBI. Together, our findings suggest that HDAC inhibition increases expression of neurotrophic factors, and promote neuronal rewiring and functional recovery following TBI.
Collapse
|
33
|
O'Reilly ML, Tom VJ. Neuroimmune System as a Driving Force for Plasticity Following CNS Injury. Front Cell Neurosci 2020; 14:187. [PMID: 32792908 PMCID: PMC7390932 DOI: 10.3389/fncel.2020.00187] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/29/2020] [Indexed: 12/15/2022] Open
Abstract
Following an injury to the central nervous system (CNS), spontaneous plasticity is observed throughout the neuraxis and affects multiple key circuits. Much of this spontaneous plasticity can elicit beneficial and deleterious functional outcomes, depending on the context of plasticity and circuit affected. Injury-induced activation of the neuroimmune system has been proposed to be a major factor in driving this plasticity, as neuroimmune and inflammatory factors have been shown to influence cellular, synaptic, structural, and anatomical plasticity. Here, we will review the mechanisms through which the neuroimmune system mediates plasticity after CNS injury. Understanding the role of specific neuroimmune factors in driving adaptive and maladaptive plasticity may offer valuable therapeutic insight into how to promote adaptive plasticity and/or diminish maladaptive plasticity, respectively.
Collapse
Affiliation(s)
- Micaela L O'Reilly
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Veronica J Tom
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
34
|
Granier C, Schwarting J, Fourli E, Laage-Gaupp F, Hennrich AA, Schmalz A, Jacobi A, Wesolowski M, Conzelmann KK, Bareyre FM. Formation of somatosensory detour circuits mediates functional recovery following dorsal column injury. Sci Rep 2020; 10:10953. [PMID: 32616790 PMCID: PMC7331809 DOI: 10.1038/s41598-020-67866-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 06/15/2020] [Indexed: 11/15/2022] Open
Abstract
Anatomically incomplete spinal cord injuries can be followed by functional recovery mediated, in part, by the formation of intraspinal detour circuits. Here, we show that adult mice recover tactile and proprioceptive function following a unilateral dorsal column lesion. We therefore investigated the basis of this recovery and focused on the plasticity of the dorsal column-medial lemniscus pathway. We show that ascending dorsal root ganglion (DRG) axons branch in the spinal grey matter and substantially increase the number of these collaterals following injury. These sensory fibers exhibit synapsin-positive varicosities, indicating their integration into spinal networks. Using a monosynaptic circuit tracing with rabies viruses injected into the cuneate nucleus, we show the presence of spinal cord neurons that provide a detour pathway to the original target area of DRG axons. Notably the number of contacts between DRG collaterals and those spinal neurons increases by more than 300% after injury. We then characterized these interneurons and showed that the lesion triggers a remodeling of the connectivity pattern. Finally, using re-lesion experiments after initial remodeling of connections, we show that these detour circuits are responsible for the recovery of tactile and proprioceptive function. Taken together our study reveals that detour circuits represent a common blueprint for axonal rewiring after injury.
Collapse
Affiliation(s)
- Charlène Granier
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany.,Graduate School of Systemic Neurosciences, LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Julian Schwarting
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Evangelia Fourli
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Fabian Laage-Gaupp
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Alexandru A Hennrich
- Max Von Pettenkofer-Institute, Virology, Faculty of Medicine, and Gene Center, LMU Munich, 80336, Munich, Germany
| | - Anja Schmalz
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Anne Jacobi
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Marta Wesolowski
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany.,Graduate School of Systemic Neurosciences, LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Karl Klaus Conzelmann
- Max Von Pettenkofer-Institute, Virology, Faculty of Medicine, and Gene Center, LMU Munich, 80336, Munich, Germany
| | - Florence M Bareyre
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany. .,Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany. .,Munich Cluster of Systems Neurology (SyNergy), 81377, Munich, Germany.
| |
Collapse
|
35
|
Ueno M, Nakamura Y, Nakagawa H, Niehaus JK, Maezawa M, Gu Z, Kumanogoh A, Takebayashi H, Lu QR, Takada M, Yoshida Y. Olig2-Induced Semaphorin Expression Drives Corticospinal Axon Retraction After Spinal Cord Injury. Cereb Cortex 2020; 30:5702-5716. [PMID: 32564090 DOI: 10.1093/cercor/bhaa142] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/02/2020] [Accepted: 05/03/2020] [Indexed: 12/24/2022] Open
Abstract
Axon regeneration is limited in the central nervous system, which hinders the reconstruction of functional circuits following spinal cord injury (SCI). Although various extrinsic molecules to repel axons following SCI have been identified, the role of semaphorins, a major class of axon guidance molecules, has not been thoroughly explored. Here we show that expression of semaphorins, including Sema5a and Sema6d, is elevated after SCI, and genetic deletion of either molecule or their receptors (neuropilin1 and plexinA1, respectively) suppresses axon retraction or dieback in injured corticospinal neurons. We further show that Olig2+ cells are essential for SCI-induced semaphorin expression, and that Olig2 binds to putative enhancer regions of the semaphorin genes. Finally, conditional deletion of Olig2 in the spinal cord reduces the expression of semaphorins, alleviating the axon retraction. These results demonstrate that semaphorins function as axon repellents following SCI, and reveal a novel transcriptional mechanism for controlling semaphorin levels around injured neurons to create zones hostile to axon regrowth.
Collapse
Affiliation(s)
- Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata 951-8585, Japan.,Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), Kawaguchi 332-0012, Japan
| | - Yuka Nakamura
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata 951-8585, Japan.,Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Hiroshi Nakagawa
- Systems Neuroscience Section, Primate Research Institute, Kyoto University, Inuyama 484-8506, Japan.,Department of Molecular Neuroscience, WPI Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan
| | - Jesse K Niehaus
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), Kawaguchi 332-0012, Japan
| | - Mari Maezawa
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Zirong Gu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
| | - Qing Richard Lu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Masahiko Takada
- Systems Neuroscience Section, Primate Research Institute, Kyoto University, Inuyama 484-8506, Japan
| | - Yutaka Yoshida
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Neural Connectivity Development in Physiology and Disease Laboratory, Burke Neurological Institute, White Plains, NY 10605, USA.,Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
36
|
Tanaka T, Ito T, Sumizono M, Ono M, Kato N, Honma S, Ueno M. Combinational Approach of Genetic SHP-1 Suppression and Voluntary Exercise Promotes Corticospinal Tract Sprouting and Motor Recovery Following Brain Injury. Neurorehabil Neural Repair 2020; 34:558-570. [PMID: 32441214 DOI: 10.1177/1545968320921827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background. Brain injury often causes severe motor dysfunction, leading to difficulties with living a self-reliant social life. Injured neural circuits must be reconstructed to restore functions, but the adult brain is limited in its ability to restore neuronal connections. The combination of molecular targeting, which enhances neural plasticity, and rehabilitative motor exercise is an important therapeutic approach to promote neuronal rewiring in the spared circuits and motor recovery. Objective. We tested whether genetic reduction of Src homology 2-containing phosphatase-1 (SHP-1), an inhibitor of brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB) signaling, has synergistic effects with rehabilitative training to promote reorganization of motor circuits and functional recovery in a mouse model of brain injury. Methods. Rewiring of the corticospinal circuit was examined using neuronal tracers following unilateral cortical injury in control mice and in Shp-1 mutant mice subjected to voluntary exercise. Recovery of motor functions was assessed using motor behavior tests. Results. We found that rehabilitative exercise decreased SHP-1 and increased BDNF and TrkB expression in the contralesional motor cortex after the injury. Genetic reduction of SHP-1 and voluntary exercise significantly increased sprouting of corticospinal tract axons and enhanced motor recovery in the impaired forelimb. Conclusions. Our data demonstrate that combining voluntary exercise and SHP-1 suppression promotes motor recovery and neural circuit reorganization after brain injury.
Collapse
Affiliation(s)
- Takashi Tanaka
- Kanazawa Medical University, Kahoku, Ishikawa, Japan.,Kindai University, Osaka-Sayama, Osaka, Japan
| | - Tetsufumi Ito
- Kanazawa Medical University, Kahoku, Ishikawa, Japan
| | - Megumi Sumizono
- Kyushu University of Nursing and Social Welfare, Tamana, Kumamoto, Japan
| | - Munenori Ono
- Kanazawa Medical University, Kahoku, Ishikawa, Japan
| | - Nobuo Kato
- Kanazawa Medical University, Kahoku, Ishikawa, Japan
| | - Satoru Honma
- Kanazawa Medical University, Kahoku, Ishikawa, Japan
| | | |
Collapse
|
37
|
Hiraga SI, Itokazu T, Hoshiko M, Takaya H, Nishibe M, Yamashita T. Microglial depletion under thalamic hemorrhage ameliorates mechanical allodynia and suppresses aberrant axonal sprouting. JCI Insight 2020; 5:131801. [PMID: 32051342 DOI: 10.1172/jci.insight.131801] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 12/30/2019] [Indexed: 01/14/2023] Open
Abstract
Central poststroke pain (CPSP) is one of the neuropathic pain syndromes that can occur following stroke involving the somatosensory system. However, the underlying mechanism of CPSP remains largely unknown. Here, we established a CPSP mouse model by inducing a focal hemorrhage in the thalamic ventrobasal complex and confirmed the development of mechanical allodynia. In this model, microglial activation was observed in the somatosensory cortex, as well as in the injured thalamus. By using a CSF1 receptor inhibitor, we showed that microglial depletion effectively prevented allodynia development in our CPSP model. In the critical phase of allodynia development, c-fos-positive neurons increased in the somatosensory cortex, accompanied by ectopic axonal sprouting of the thalamocortical projection. Furthermore, microglial ablation attenuated both neuronal hyperactivity in the somatosensory cortex and circuit reorganization. These findings suggest that microglia play a crucial role in the development of CPSP pathophysiology by promoting sensory circuit reorganization.
Collapse
Affiliation(s)
- Shin-Ichiro Hiraga
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takahide Itokazu
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka, Japan.,Department of Neuro-Medical Science, Graduate School of Medicine
| | - Maki Hoshiko
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka, Japan
| | - Hironobu Takaya
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Mariko Nishibe
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Office of Strategic Innovative Dentistry, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka, Japan.,Department of Neuro-Medical Science, Graduate School of Medicine.,Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| |
Collapse
|
38
|
Isa T, Mitsuhashi M, Yamaguchi R. Alternative routes for recovery of hand functions after corticospinal tract injury in primates and rodents. Curr Opin Neurol 2019; 32:836-843. [PMID: 31688166 DOI: 10.1097/wco.0000000000000749] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Recent studies on various corticospinal tract (CST) lesions have shown the plastic changes at a variety of motor systems after the lesion. This review provides the alternative routes associated with the motor functional recovery after the CST lesions at various levels in nonhuman primates and rodents. RECENT FINDINGS In the case of the motor cortical lesions, the perilesional area compensates for the lesion. In contrast, sprouting of the corticoreticular tracts was observed after the lesions involving sensorimotor cortical areas. After the internal capsule lesion, sprouting in the cortico-rubral pathway contributes to the recovery. In case of the pyramidal lesion, rubrospinal and reticulospinal tracts play a role of the functional recovery. After the dorsolateral funiculus (DLF) lesion at C4/C5, the indirect pathway via propriospinal tract contributes to the recovery. In case of the hemisection at lower cervical cord, the CST fibers sprouted from the bilateral motor cortex and descended to the contralesional DLF and crossed below the lesion area. SUMMARY The central pathways can change their structure and activity dynamically depending on the lesion sites and size. Revealing the difference of the alternative pathways should be crucial to understand the whole recovery mechanism and develop the further neurorehabilitative treatment.
Collapse
Affiliation(s)
- Tadashi Isa
- Department of Neuroscience, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Human Brain Research Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Mitsuhashi
- Department of Neuroscience, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Reona Yamaguchi
- Department of Neuroscience, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| |
Collapse
|
39
|
PKCγ promotes axonal remodeling in the cortico-spinal tract via GSK3β/β-catenin signaling after traumatic brain injury. Sci Rep 2019; 9:17078. [PMID: 31745212 PMCID: PMC6863826 DOI: 10.1038/s41598-019-53225-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 07/22/2019] [Indexed: 12/27/2022] Open
Abstract
Traumatic brain injury (TBI) is a common cause of death and disability. Enhancing the midline-crossing of the contralateral corticospinal tract (CST) to the denervated side of spinal cord facilitates functional recovery after TBI. Activation of the gamma isoform of PKC (PKCγ) in contralateral CST implicates its roles in promoting CST remodeling after TBI. In this study, we deployed loss and gain of function strategies in N2a cells and primary cortical neurons in vitro, and demonstrated that PKCγ is not only important but necessary for neuronal differentiation, neurite outgrowth and axonal branching but not for axonal extension. Mechanically, through the phosphorylation of GSK3β, PKCγ stabilizes the expression of cytosolic β-catenin and increase GAP43 expression, thus promoting axonal outgrowth. Further, rAAV2/9-mediated delivery of constitutive PKCγ in the corticospinal tract after unilateral TBI in vivo additionally showed that specifically delivery of active PKCγ mutant to cortical neuron promotes midline crossing of corticospinal fibers from the uninjured side to the denervated cervical spinal cord. This PKCγ-mediated injury response promoted sensorimotor functional recovery. In conclusion, PKCγ mediates stability of β-catenin through the phosphorylation of GSK3β to facilitate neuronal differentiation, neurite outgrowth and axonal branching, and PKCγ maybe a novel therapeutic target for physiological and functional recovery after TBI.
Collapse
|
40
|
Ueno R, Takase H, Suenaga J, Kishimoto M, Kurihara Y, Takei K, Kawahara N, Yamamoto T. Axonal regeneration and functional recovery driven by endogenous Nogo receptor antagonist LOTUS in a rat model of unilateral pyramidotomy. Exp Neurol 2019; 323:113068. [PMID: 31629859 DOI: 10.1016/j.expneurol.2019.113068] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 12/26/2022]
Abstract
The adult mammalian central nervous system (CNS) rarely recovers from injury. Myelin fragments contain axonal growth inhibitors that limit axonal regeneration, thus playing a major role in determining neural recovery. Nogo receptor-1 (NgR1) and its ligands are among the inhibitors that limit axonal regeneration. It has been previously shown that the endogenous protein, lateral olfactory tract usher substance (LOTUS), antagonizes NgR1-mediated signaling and accelerates neuronal plasticity after spinal cord injury and cerebral ischemia in mice. However, it remained unclear whether LOTUS-mediated reorganization of descending motor pathways in the adult brain is physiologically functional and contributes to functional recovery. Here, we generated LOTUS-overexpressing transgenic (LOTUS-Tg) rats to investigate the role of LOTUS in neuronal function after damage. After unilateral pyramidotomy, motor function in LOTUS-Tg rats recovered significantly compared to that in wild-type animals. In a retrograde tracing study, labeled axons spanning from the impaired side of the cervical spinal cord to the unlesioned hemisphere of the red nucleus and sensorimotor cortex were increased in LOTUS-Tg rats. Anterograde tracing from the unlesioned cortex also revealed enhanced ipsilateral connectivity to the impaired side of the cervical spinal cord in LOTUS-Tg rats. Moreover, electrophysiological analysis showed that contralesional cortex stimulation significantly increased ipsilateral forelimb movement in LOTUS-Tg rats, which was consistent with the histological findings. According to these data, LOTUS overexpression accelerates ipsilateral projection from the unlesioned cortex and promotes functional recovery after unilateral pyramidotomy. LOTUS could be a future therapeutic option for CNS injury.
Collapse
Affiliation(s)
- Ryu Ueno
- Department of Neurosurgery, Yokohama City University, Yokohama, Japan.
| | - Hajime Takase
- Department of Neurosurgery, Yokohama City University, Yokohama, Japan.
| | - Jun Suenaga
- Department of Neurosurgery, Yokohama City University, Yokohama, Japan.
| | - Masao Kishimoto
- Department of Neurosurgery, Yokohama City University, Yokohama, Japan.
| | - Yuji Kurihara
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan.
| | - Kohtaro Takei
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan.
| | - Nobutaka Kawahara
- Department of Neurosurgery, Yokohama City University, Yokohama, Japan
| | - Tetsuya Yamamoto
- Department of Neurosurgery, Yokohama City University, Yokohama, Japan.
| |
Collapse
|
41
|
Ohne H, Takahashi M, Satomi K, Hasegawa A, Takeuchi T, Sato S, Ichimura S. Mechanism of forelimb motor function restoration in rats with cervical spinal cord hemisection-neuroanatomical validation. IBRO Rep 2019; 7:10-25. [PMID: 31431931 PMCID: PMC6581651 DOI: 10.1016/j.ibror.2019.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/05/2019] [Indexed: 11/13/2022] Open
Abstract
Purpose The purpose of this study is neuroanatomical validation of forelimb motor function restoration in rats with cervical spinal cord injury. Materials and methods We used eight cervical hemisected rats and eight normal rats. We cut in half the C3/4 cervical spinal cord of 18-weeks-old normal rats. We used 24-weeks-old rats that had reached a nearly steady state of forelimb motor function after the hemisection (Hemisection group). Normal 24-week-old rats were used as Control group. To evaluate the corticospinal tracts, neuro-tracing by biotynirated dextran-amine (BDA) was used. BDA was injected into the damaged side of the cerebral primary motor cortex. In order to quantitatively analyze the specimen, we recorded a site where nerve fibers appear in each specimen in the image analysis (1) and defined the increase rate of immunostaining area using ImageJ in the image analysis (2). Based on the evaluation in the image analysis (1) and the image analysis (2), the Hemisection group and the Control group were compared. Results In the image analysis (1), a region with robust appearance of aberrant nerve fibers was observed in the cephalad side of the Hemisection site in Hemisection group than Control group. In the spinal cord caudal to the hemisection, such region was generally more in Hemisection group, however, disappeared or reduced appearance was observed in some regions. In the image analysis (2), no statistical significant difference was noted in each level. Conclusion There is a high probability that these aberrant nerve fibers beyond the midline could be involved in forelimb motor function restoration in rats with cervical cord hemisection.
Collapse
Affiliation(s)
- Hideaki Ohne
- Department of Orthopaedic Surgery, Kyorin University School of Medicine, 6-20-2 Shinkawa Mitaka, Tokyo, 181-8611, Japan
| | - Masahito Takahashi
- Department of Orthopaedic Surgery, Kyorin University School of Medicine, 6-20-2 Shinkawa Mitaka, Tokyo, 181-8611, Japan
| | - Kazuhiko Satomi
- Orthopaedic Surgery, Kugayama Hospital, 2-14-20 Kitakarasuyama Setagaya, Tokyo, 157-0061, Japan
| | - Atsushi Hasegawa
- Department of Orthopaedic Surgery, Kyorin University School of Medicine, 6-20-2 Shinkawa Mitaka, Tokyo, 181-8611, Japan
| | - Takumi Takeuchi
- Department of Orthopaedic Surgery, Kyorin University School of Medicine, 6-20-2 Shinkawa Mitaka, Tokyo, 181-8611, Japan
| | - Shunsuke Sato
- Department of Orthopaedic Surgery, Kyorin University School of Medicine, 6-20-2 Shinkawa Mitaka, Tokyo, 181-8611, Japan
| | - Shoichi Ichimura
- Department of Orthopaedic Surgery, Kyorin University School of Medicine, 6-20-2 Shinkawa Mitaka, Tokyo, 181-8611, Japan
| |
Collapse
|
42
|
Bradley PM, Denecke CK, Aljovic A, Schmalz A, Kerschensteiner M, Bareyre FM. Corticospinal circuit remodeling after central nervous system injury is dependent on neuronal activity. J Exp Med 2019; 216:2503-2514. [PMID: 31391209 PMCID: PMC6829605 DOI: 10.1084/jem.20181406] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 03/10/2019] [Accepted: 07/17/2019] [Indexed: 11/04/2022] Open
Abstract
The remodeling of supraspinal axonal circuits mediates functional recovery after spinal cord injury. This process critically depends on the selection of appropriate synaptic connections between cortical projection and spinal relay neurons. To unravel the principles that guide this target selection, we used genetic and chemogenetic tools to modulate NMDA receptor (NMDAR) integrity and function, CREB-mediated transcription, and neuronal firing of relay neurons during injury-induced corticospinal remodeling. We show that NMDAR signaling and CREB-mediated transcription maintain nascent corticospinal tract (CST)-relay neuron contacts. These activity-dependent signals act during a defined period of circuit remodeling and do not affect mature or uninjured circuits. Furthermore, chemogenetic modulation of relay neuron activity reveals that the regrowing CST axons select their postsynaptic partners in a competitive manner and that preventing such activity-dependent shaping of corticospinal circuits limits motor recovery after spinal cord injury.
Collapse
Affiliation(s)
- Peter M Bradley
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Medical Faculty, Ludwig-Maximilians University Munich, Martinsried, Germany
| | - Carmen K Denecke
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Medical Faculty, Ludwig-Maximilians University Munich, Martinsried, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians University Munich, Martinsried, Germany
| | - Almir Aljovic
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Medical Faculty, Ludwig-Maximilians University Munich, Martinsried, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians University Munich, Martinsried, Germany
| | - Anja Schmalz
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Medical Faculty, Ludwig-Maximilians University Munich, Martinsried, Germany
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Medical Faculty, Ludwig-Maximilians University Munich, Martinsried, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Florence M Bareyre
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany .,Biomedical Center, Medical Faculty, Ludwig-Maximilians University Munich, Martinsried, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
43
|
Tsujioka H, Yamashita T. Comparison of gene expression profile of the spinal cord of sprouting-capable neonatal and sprouting-incapable adult mice. BMC Genomics 2019; 20:619. [PMID: 31362699 PMCID: PMC6668129 DOI: 10.1186/s12864-019-5974-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022] Open
Abstract
Background The regenerative ability of severed axons in the central nervous system is limited in mammals. However, after central nervous system injury, neural function is partially recovered by the formation of a compensatory neural circuit. In a mouse pyramidotomy model, axonal sprouting of the intact side of the corticospinal tract is observed in the spinal cord, and the axons make new synapses with the denervated side of propriospinal neurons. Moreover, this sprouting ability is enhanced in neonatal mice compared to that in adult mice. Myelin-associated molecules in the spinal cord or intrinsic factors in corticospinal neurons have been investigated in previous studies, but the factors that determine elevated sprouting ability in neonatal mice are not fully understood. Further, in the early phase after pyramidotomy, glial responses are observed in the spinal cord. To elucidate the basal difference in the spinal cord, we compared gene expression profiles of entire C4–7 cervical cord tissues between neonatal (injured at postnatal day 7) and adult (injured at 8 weeks of age) mice by RNA-sequencing. We also tried to identify discordant gene expression changes that might inhibit axonal sprouting in adult mice at the early phase (3 days) after pyramidotomy. Results A comparison of neonatal and adult sham groups revealed remarkable basal differences in the spinal cord, such as active neural circuit formation, cell proliferation, the development of myelination, and an immature immune system in neonatal mice compared to that observed in adult mice. Some inflammation-related genes were selectively expressed in adult mice after pyramidotomy, implying the possibility that these genes might be related to the low sprouting ability in adult mice. Conclusions This study provides useful information regarding the basal difference between neonatal and adult spinal cords and the possible differential response after pyramidotomy, both of which are necessary to understand why sprouting ability is increased in neonatal mice compared to that in adult mice. Electronic supplementary material The online version of this article (10.1186/s12864-019-5974-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hiroshi Tsujioka
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan. .,WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan. .,Graduate School of Frontier Bioscience, Osaka University, Osaka, Japan. .,Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Osaka, Japan.
| |
Collapse
|
44
|
The Spinal Transcriptome after Cortical Stroke: In Search of Molecular Factors Regulating Spontaneous Recovery in the Spinal Cord. J Neurosci 2019; 39:4714-4726. [PMID: 30962276 PMCID: PMC6561692 DOI: 10.1523/jneurosci.2571-18.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 03/22/2019] [Accepted: 03/28/2019] [Indexed: 11/21/2022] Open
Abstract
In response to cortical stroke and unilateral corticospinal tract degeneration, compensatory sprouting of spared corticospinal fibers is associated with recovery of skilled movement in rodents. To date, little is known about the molecular mechanisms orchestrating this spontaneous rewiring. In this study, we provide insights into the molecular changes in the spinal cord tissue after large ischemic cortical injury in adult female mice, with a focus on factors that might influence the reinnervation process by contralesional corticospinal neurons. We mapped the area of cervical gray matter reinnervation by sprouting contralesional corticospinal axons after unilateral photothrombotic stroke of the motor cortex in mice using anterograde tracing. The mRNA profile of this reinnervation area was analyzed using whole-genome sequencing to identify differentially expressed genes at selected time points during the recovery process. Bioinformatic analysis revealed two phases of processes: early after stroke (4–7 d post-injury), the spinal transcriptome is characterized by inflammatory processes, including phagocytic processes as well as complement cascade activation. Microglia are specifically activated in the denervated corticospinal projection fields in this early phase. In a later phase (28–42 d post-injury), biological processes include tissue repair pathways with upregulated genes related to neurite outgrowth. Thus, the stroke-denervated spinal gray matter, in particular its intermediate laminae, represents a growth-promoting environment for sprouting corticospinal fibers originating from the contralesional motor cortex. This dataset provides a solid starting point for future studies addressing key elements of the post-stroke recovery process, with the goal to improve neuroregenerative treatment options for stroke patients. SIGNIFICANCE STATEMENT We show that the molecular changes in the spinal cord target tissue of the stroke-affected corticospinal tract are mainly defined by two phases: an early inflammatory phase during which microglia are specifically activated in the target area of reinnervating corticospinal motor neurons; and a late phase during which growth-promoting factors are upregulated which can influence the sprouting response, arborization, and synapse formation. By defining for the first time the endogenous molecular machinery in the stroke-denervated cervical spinal gray matter with a focus on promotors of axon growth through the growth-inhibitory adult CNS, this study will serve as a basis to address novel neuroregenerative treatment options for chronic stroke patients.
Collapse
|
45
|
Hosomi S, Koyama Y, Watabe T, Ohnishi M, Ogura H, Yamashita T, Shimazu T. Myeloid-Derived Suppressor Cells Infiltrate the Brain and Suppress Neuroinflammation in a Mouse Model of Focal Traumatic Brain Injury. Neuroscience 2019; 406:457-466. [PMID: 30880103 DOI: 10.1016/j.neuroscience.2019.03.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 12/22/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) have strong immunosuppressive characteristics, which allow them to limit inflammation and facilitate wound healing and recovery. Although MDSCs are a newly-determined cell type that is gaining attention in the immunology field, their neuroimmunological characteristics remain unstudied. In this study, we explored the suppressive role of MDSCs in cerebral inflammatory reactions after focal traumatic brain injury (TBI) using in vivo imaging. Through morphological, functional, and phenotypic analyses we determined that CD11b+/Gr-1+ cells infiltrating the contusion area are MDSCs. MDSCs are among the first responders to tissue injury, responding even prior to microglial activation. Positron emission tomography imaging of translocator protein results suggest that infiltrating MDSCs suppress neuronal inflammation and interact with resident immune cells, like microglia, following focal TBI.
Collapse
Affiliation(s)
- Sanae Hosomi
- Department of Traumatology and Acute Critical Medicine, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.
| | - Yoshihisa Koyama
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.
| | - Tadashi Watabe
- Positron Emission Tomography Molecular Imaging Center, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.
| | - Mitsuo Ohnishi
- Department of Traumatology and Acute Critical Medicine, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.
| | - Hiroshi Ogura
- Department of Traumatology and Acute Critical Medicine, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.
| | - Takeshi Shimazu
- Department of Traumatology and Acute Critical Medicine, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
46
|
Neuropilin-1-mediated pruning of corticospinal tract fibers is required for motor recovery after spinal cord injury. Cell Death Dis 2019; 10:67. [PMID: 30683854 PMCID: PMC6347615 DOI: 10.1038/s41419-019-1338-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/25/2018] [Accepted: 01/07/2019] [Indexed: 11/30/2022]
Abstract
Following incomplete spinal cord injury (SCI), reorganization of the corticospinal tract (CST) contributes to spontaneous motor recovery. Axotomized CST fibers form collaterals and make synapses with interneurons, followed by pruning of excess fibers. Although axonal pruning is involved in refinement of neural circuits, its molecular mechanisms and functional roles remain poorly understood. To address these questions, we performed dorsal hemisections of mouse thoracic spinal cord. We observed that Neuropilin-1 (Nrp1) mRNA was upregulated in layer 5 pyramidal neurons in the motor cortex 14 days after SCI, when the pruning occurred. Nrp1 knockdown using adeno-associated virus (AAV) vector encoding Nrp1 shRNA in the hindlimb motor area impaired the pruning of collaterals after SCI. Nrp1 knockout by injecting AAV vector encoding Cre recombinase into Nrp1 floxed mice also suppressed axonal pruning. Propriospinal neurons, interneurons that connect CST and motoneurons, expressed Semaphorin 3A (Sema3A), the ligand of Nrp1. Furthermore, the genetic deletion of Nrp1 specifically in the hindlimb motor area suppressed the recovery of skilled movement at 21 and 28 days after SCI. The present findings demonstrate that the pruning of collaterals mediated by Nrp1 is required for motor recovery after SCI, and suggest that refinement of the neuronal network facilitates motor recovery.
Collapse
|
47
|
Abe Y, Honsho M, Itoh R, Kawaguchi R, Fujitani M, Fujiwara K, Hirokane M, Matsuzaki T, Nakayama K, Ohgi R, Marutani T, Nakayama KI, Yamashita T, Fujiki Y. Peroxisome biogenesis deficiency attenuates the BDNF-TrkB pathway-mediated development of the cerebellum. Life Sci Alliance 2018; 1:e201800062. [PMID: 30519675 PMCID: PMC6277683 DOI: 10.26508/lsa.201800062] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 11/08/2018] [Accepted: 11/08/2018] [Indexed: 01/22/2023] Open
Abstract
Peroxisome biogenesis disorders (PBDs) manifest as neurological deficits in the central nervous system, including neuronal migration defects and abnormal cerebellum development. However, the mechanisms underlying pathogenesis remain enigmatic. Here, to investigate how peroxisome deficiency causes neurological defects of PBDs, we established a new PBD model mouse defective in peroxisome assembly factor Pex14p, termed Pex14 ΔC/ΔC mouse. Pex14 ΔC/ΔC mouse manifests a severe symptom such as disorganization of cortical laminar structure and dies shortly after birth, although peroxisomal biogenesis and metabolism are partially defective. The Pex14 ΔC/ΔC mouse also shows malformation of the cerebellum including the impaired dendritic development of Purkinje cells. Moreover, extracellular signal-regulated kinase and AKT signaling are attenuated in this mutant mouse by an elevated level of brain-derived neurotrophic factor (BDNF) together with the enhanced expression of TrkB-T1, a dominant-negative isoform of the BDNF receptor. Our results suggest that dysregulation of the BDNF-TrkB pathway, an essential signaling for cerebellar morphogenesis, gives rise to the pathogenesis of the cerebellum in PBDs.
Collapse
Affiliation(s)
- Yuichi Abe
- Division of Organelle Homeostasis, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Masanori Honsho
- Division of Organelle Homeostasis, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Ryota Itoh
- Graduate School of Systems Life Sciences and Department of Biology, Faculty of Sciences, Kyushu University Graduate School, Fukuoka, Japan
| | - Ryoko Kawaguchi
- Graduate School of Systems Life Sciences and Department of Biology, Faculty of Sciences, Kyushu University Graduate School, Fukuoka, Japan
| | - Masashi Fujitani
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kazushirou Fujiwara
- Graduate School of Systems Life Sciences and Department of Biology, Faculty of Sciences, Kyushu University Graduate School, Fukuoka, Japan
| | - Masaaki Hirokane
- Graduate School of Systems Life Sciences and Department of Biology, Faculty of Sciences, Kyushu University Graduate School, Fukuoka, Japan
| | - Takashi Matsuzaki
- Graduate School of Systems Life Sciences and Department of Biology, Faculty of Sciences, Kyushu University Graduate School, Fukuoka, Japan
| | - Keiko Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.,Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryohei Ohgi
- Graduate School of Systems Life Sciences and Department of Biology, Faculty of Sciences, Kyushu University Graduate School, Fukuoka, Japan
| | - Toshihiro Marutani
- Graduate School of Systems Life Sciences and Department of Biology, Faculty of Sciences, Kyushu University Graduate School, Fukuoka, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan.,Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Tokyo, Japan
| | - Yukio Fujiki
- Division of Organelle Homeostasis, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
48
|
Meves JM, Geoffroy CG, Kim ND, Kim JJ, Zheng B. Oligodendrocytic but not neuronal Nogo restricts corticospinal axon sprouting after CNS injury. Exp Neurol 2018; 309:32-43. [PMID: 30055160 PMCID: PMC6139267 DOI: 10.1016/j.expneurol.2018.07.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/24/2018] [Accepted: 07/24/2018] [Indexed: 12/20/2022]
Abstract
Recovery from injury to the central nervous system (CNS) is limited in the mammalian adult. Nonetheless, some degree of spontaneous recovery occurs after partial CNS injury. Compensatory axonal growth from uninjured neurons, termed sprouting, contributes to this naturally occurring recovery process and can be modulated by molecular intervention. Extensive studies have depicted a long-held hypothesis that oligodendrocyte-derived Nogo restricts axonal sprouting and functional recovery after CNS injury. However, cell type-specific function of Nogo in compensatory sprouting, spinal axon repair or functional recovery after CNS injury has not been reported. Here we present data showing that inducible, cell type-specific deletion of Nogo from oligodendrocytes led to a ~50% increase in the compensatory sprouting of corticospinal tract (CST) axons in the cervical spinal cord after unilateral pyramidotomy in mice. In contrast to a previously proposed growth-promoting role of neuronal Nogo in the optic nerve, deleting neuronal Nogo did not significantly affect CST axon sprouting in the spinal cord. Sprouting axons were associated with the expression of synaptic marker VGLUT1 in both the oligodendrocytic Nogo deletion and control mice. However, we did not detect any functional improvement in fine motor control associated with the increased sprouting in oligodendrocytic Nogo deletion mice. These data show for the first time with genetic evidence that Nogo specifically expressed by oligodendrocytes restricts compensatory sprouting after CNS injury, supporting a longstanding but heretofore untested hypothesis. While implicating a focus on sprouting as a repair mechanism in the translational potential of targeting the myelin inhibitory pathway, our study illustrates the challenge to harness enhanced structural plasticity for functional improvement.
Collapse
Affiliation(s)
- Jessica M Meves
- Neurosciences Graduate Program, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Neurosciences, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Cédric G Geoffroy
- Department of Neurosciences, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Noah D Kim
- Department of Neurosciences, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Joseph J Kim
- Department of Neurosciences, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Binhai Zheng
- Neurosciences Graduate Program, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Neurosciences, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
49
|
Hosomi S, Watabe T, Mori Y, Koyama Y, Adachi S, Hoshi N, Ohnishi M, Ogura H, Yoshioka Y, Hatazawa J, Yamashita T, Shimazu T. Inflammatory projections after focal brain injury trigger neuronal network disruption: An 18F-DPA714 PET study in mice. NEUROIMAGE-CLINICAL 2018; 20:946-954. [PMID: 30312938 PMCID: PMC6178196 DOI: 10.1016/j.nicl.2018.09.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/05/2018] [Accepted: 09/28/2018] [Indexed: 11/12/2022]
Abstract
Due to the heterogeneous pathology of traumatic brain injury (TBI), the exact mechanism of how initial brain damage leads to chronic inflammation and its effects on the whole brain remain unclear. Here, we report on long-term neuroinflammation, remote from the initial injury site, even after subsiding of the original inflammatory response, in a focal TBI mouse model. The use of translocator protein-positron emission tomography in conjunction with specialised magnetic resonance imaging modalities enabled us to visualize “previously undetected areas” of spreading inflammation after focal cortical injury. These clinically available modalities further revealed the pathophysiology of thalamic neuronal degeneration occurring as resident microglia sense damage to corticothalamic neuronal tracts and become activated. The resulting microglial activation plays a major role in prolonged inflammatory processes, which are deleterious to the thalamic network. In light of the association of this mechanism with neuronal tracts, we propose it can be termed “brain injury related inflammatory projection”. Our findings on multiple spatial and temporal scales provide insight into the chronic inflammation present in neurodegenerative diseases after TBI. TSPO-PET tomography enables the assessment of longitudinal neuronal inflammation Inflammatory responses at the cortical injury site diminish after about 1 week The ipsilateral thalamus exhibits remote neuroinflammation for up to 14 weeks Microglial activation is associated with remote chronic degeneration Inflammation expands to remote sites via damaged cortico-thalamic projections
Collapse
Affiliation(s)
- Sanae Hosomi
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamada-oka, Suita-shi, Osaka 565-0871, Japan.
| | - Tadashi Watabe
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, 2-15 Yamada-oka, Suita-shi, Osaka 565-0871, Japan; Medical Imaging Centre for Translational Research, Osaka University Graduate School of Medicine, 2-15 Yamada-oka, Suita-shi, Osaka 565-0871, Japan
| | - Yuki Mori
- Centre for Information and Neural Networks (CiNet), National Institute of Information and Communications Technology (NICT) and Osaka University, 1-4 Yamada-oka, Suita-shi, Osaka 565-0871, Japan
| | - Yoshihisa Koyama
- Department of Molecular Neuroscience, Osaka University Graduate School of Medicine, 2-15 Yamada-oka, Suita-shi, Osaka 565-0871, Japan
| | - Soichiro Adachi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 5-2 Kusunoki-cho 7, Chuo-ku, Kobe-shi, Hyougo 650-0017, Japan
| | - Namiko Hoshi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 5-2 Kusunoki-cho 7, Chuo-ku, Kobe-shi, Hyougo 650-0017, Japan
| | - Mitsuo Ohnishi
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamada-oka, Suita-shi, Osaka 565-0871, Japan
| | - Hiroshi Ogura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamada-oka, Suita-shi, Osaka 565-0871, Japan
| | - Yoshichika Yoshioka
- Centre for Information and Neural Networks (CiNet), National Institute of Information and Communications Technology (NICT) and Osaka University, 1-4 Yamada-oka, Suita-shi, Osaka 565-0871, Japan
| | - Jun Hatazawa
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, 2-15 Yamada-oka, Suita-shi, Osaka 565-0871, Japan; Medical Imaging Centre for Translational Research, Osaka University Graduate School of Medicine, 2-15 Yamada-oka, Suita-shi, Osaka 565-0871, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Osaka University Graduate School of Medicine, 2-15 Yamada-oka, Suita-shi, Osaka 565-0871, Japan
| | - Takeshi Shimazu
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamada-oka, Suita-shi, Osaka 565-0871, Japan
| |
Collapse
|
50
|
Wiersma AM, Winship IR. In vivo Use of Dextran-based Anterograde Cortical Tracers to Assess the Integrity of the Cortical Spinal Tract. Bio Protoc 2018; 8:e2862. [PMID: 34285978 DOI: 10.21769/bioprotoc.2862] [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: 03/18/2018] [Revised: 05/08/2018] [Accepted: 05/14/2018] [Indexed: 11/02/2022] Open
Abstract
When injected into the motor cortex of rats, anterograde tracers label fibers of the associated descending corticospinal tract (CST) that originate from pyramidal neurons in the tracer-injected cortex. These fibers can be assessed at the level of the spinal cord to determine the integrity of the descending CST and the spatial distribution of axons in the spinal grey matter. Here we provide detailed methods on the minimally invasive stereotaxic injection of anterograde tracers into the forelimb sensorimotor representation in the rat cortex. In addition, we detail the fixing and processing of spinal tissue for assessment of CST integrity and branching into spinal grey matter.
Collapse
Affiliation(s)
- Anna M Wiersma
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Ian R Winship
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|