1
|
Liu P, Hu J, Gao B, Hua Y, Xing Y, Bai Y, Liu N. Constraint-Induced Movement Therapy Promotes Contralesional Red Nucleus Plasticity and Increases Bilateral Motor Cortex-to-Red Nucleus Projections After a Large-Area Stroke. Behav Neurol 2025; 2025:3631524. [PMID: 40166667 PMCID: PMC11955289 DOI: 10.1155/bn/3631524] [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: 02/27/2024] [Revised: 02/20/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
For decades, scientists have explored the patterns of neural network remodeling that occur after a stroke. Several studies have shown that both motor cortexes (MCs) undergo crucial remodeling after cerebral ischemia. However, the mechanism by which corticofugal fibers are remodeled is not well understood. Therefore, this study was aimed at investigating the changes in the bilateral red nucleus (RN) and MC-RN projections during recovery from a large-area stroke in a rat stroke model with or without constraint-induced movement therapy (CIMT). A large-area middle cerebral artery occlusion (MCAO) model was established in rats using the Longa method. CIMT was initiated 7 days after MCAO and continued for 1, 2, or 3 weeks. Rats in the control group underwent spontaneous recovery. Locomotor impairment was evaluated using the CatWalk automated gait analysis system, and overall neurological function was evaluated with the modified neurological severity score. Bilateral MC-RN projections were visualized by labeling fiber tracts with an anterograde tracer. Postsynaptic density 95 (PSD95), growth-associated protein 43 (GAP43), and synaptophysin expression levels in the RN were detected using western blotting and immunohistochemistry. The results showed that CIMT promoted motor recovery after a stroke, increased levels of GAP43 and PSD95 in the contralesional but not ipsilesional RN, and increased projections from the MC to the bilateral RN. Thus, CIMT promotes neuroplasticity after a large-area stroke by stimulating axon outgrowth, improving postsynaptic membrane function in the contralesional RN, and increasing bilateral projections of the MC-RN. These results provide evidence for the therapeutic efficacy of CIMT in restoring motor function and help with understanding RN plasticity after a large-area stroke.
Collapse
Affiliation(s)
- Peile Liu
- Department of Rehabilitation Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jian Hu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Beiyao Gao
- Department of Rehabilitation Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Yan Hua
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Xing
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yulong Bai
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Nan Liu
- Department of Rehabilitation Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
2
|
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
|
3
|
Ríos C, Salgado-Ceballos H, Grijalva I, Morales-Guadarrama A, Diaz-Ruiz A, Olayo R, Morales-Corona J, Olayo MG, Cruz GJ, Mondragón-Lozano R, Alvarez-Mejia L, Orozco-Barrios C, Sánchez-Torres S, Fabela-Sánchez O, Coyoy-Salgado A, Hernández-Godínez B, Ibáñez-Contreras A, Mendez-Armenta M. Demonstration of therapeutic effect of plasma-synthesized polypyrrole/iodine biopolymer in rhesus monkey with complete spinal cord section. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2025; 36:21. [PMID: 39961937 PMCID: PMC11832569 DOI: 10.1007/s10856-025-06862-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 01/10/2025] [Indexed: 02/20/2025]
Abstract
Spinal cord injury (SCI) can cause paralysis, and although multiple therapeutic proposals have been developed in murine models, results have hardly been replicated in humans. As non-human primates (NHP) are more similar to humans than rodents, the current study investigated whether it was possible to reproduce in a NHP, the previously obtained beneficial results by using a plasma-synthesized polypyrrole/iodine (PPy/I) biopolymer, which reduce glial scar formation and inflammatory response and promotes nerve tissue preservation, regenerative processes and functional recovery in rats. In NHPs (Rhesus monkey) with SCI by complete transection (SCT) and with plasma-synthesized PPy/I application (experimental) or without (control), the expression of pro-inflammatory cytokines in blood, preservation of nervous tissue through magnetic resonance imaging and histological and morphometric techniques, regeneration through immunohistochemistry study and functional recovery through clinical examination, were evaluated. Control NHP showed a markedly increased of pro-inflammatory cytokines vs. experimental NHP, which preserved more nerve tissue. At the end of the follow-up, a thinner glial scar in the injured spinal cord was observed in the experimental NHP as well as regenerative nerve processes (NeuN and β-III tubulin expression), while control NHP had a marked glial scar, large cysts and less nerve tissue at the injured zone. Plasma-synthesized PPy/I also reduced the loss of pelvic limb muscle mass and allowed the experimental NHP recovered knee-jerk, withdrawal and plantar reflexes as well as movement in the hind limbs. Since most of the beneficial effects of plasma-synthesized PPy/I previously reported in rats were also observed in the NHP, these preliminary findings make their replication in humans with SCI more likely.
Collapse
Affiliation(s)
- Camilo Ríos
- Research Direction, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, México City, México
| | - Hermelinda Salgado-Ceballos
- Medical Research Unit in Neurological Diseases, Instituto Mexicano del Seguro Social, Mexico City, México.
- Research Center of Proyecto CAMINA A.C., Mexico City, Mexico.
| | - Israel Grijalva
- Medical Research Unit in Neurological Diseases, Instituto Mexicano del Seguro Social, Mexico City, México
- Research Center of Proyecto CAMINA A.C., Mexico City, Mexico
| | - Axayacatl Morales-Guadarrama
- National Center for Research in Imaging and Medical Instrumentation, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
- Division of Basic Sciences and Engineering, Department of Physics, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
| | - Araceli Diaz-Ruiz
- Department of Neurochemistry, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
| | - Roberto Olayo
- Division of Basic Sciences and Engineering, Department of Physics, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
| | - Juan Morales-Corona
- Division of Basic Sciences and Engineering, Department of Physics, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
| | - María G Olayo
- Department of Physics, Instituto Nacional de Investigaciones Nucleares, Estado de México, Mexico
| | - Guillermo J Cruz
- Department of Physics, Instituto Nacional de Investigaciones Nucleares, Estado de México, Mexico
| | - Rodrigo Mondragón-Lozano
- Research Center of Proyecto CAMINA A.C., Mexico City, Mexico
- CONAHCyT-Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
| | - Laura Alvarez-Mejia
- Research Center of Proyecto CAMINA A.C., Mexico City, Mexico
- Division of Basic Sciences and Engineering, Department of Physics, CONAHCyT-Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
| | - Carlos Orozco-Barrios
- Research Center of Proyecto CAMINA A.C., Mexico City, Mexico
- CONAHCyT-Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
| | - Stephanie Sánchez-Torres
- Research Center of Proyecto CAMINA A.C., Mexico City, Mexico
- CONAHCyT-Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
| | - Omar Fabela-Sánchez
- Department of Chemistry Macromolecules and Nanomaterials, CONAHCyT-Centro de Investigación en Química Aplicada, Saltillo, Coahuila, Mexico
| | - Angélica Coyoy-Salgado
- Research Center of Proyecto CAMINA A.C., Mexico City, Mexico
- CONAHCyT-Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
| | | | | | - Marisela Mendez-Armenta
- Department of Neurochemistry, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
| |
Collapse
|
4
|
Gracies J, Alter KE, Biering‐Sørensen B, Dewald JP, Dressler D, Esquenazi A, Franco JH, Jech R, Kaji R, Jin L, Lim EC, Raghavan P, Rosales R, Shalash AS, Simpson DM, Suputtitada A, Vecchio M, Wissel J. Spastic Paresis: A Treatable Movement Disorder. Mov Disord 2025; 40:44-50. [PMID: 39548808 PMCID: PMC11752976 DOI: 10.1002/mds.30038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 11/18/2024] Open
Affiliation(s)
- Jean‐Michel Gracies
- Service de Rééducation Neurolocomotrice, Hôpitaux Universitaires Henri Mondor, Assistance Publique‐Hôpitaux de ParisParisFrance
- UR BIOTN, Université Paris Est Créteil (UPEC)CréteilFrance
| | - Katharine E. Alter
- Mount Washington Pediatric Hospital, An Affiliate of The University of Maryland System and Johns Hopkins Medical InstitutionBaltimoreMarylandUSA
| | - Bo Biering‐Sørensen
- Movement Disorder Clinic, Spasticity Clinic and Neuropathic Pain and CRPS Clinic, Neurological DepartmentCopenhagen University Hospital, RigshospitaletGlostrupDenmark
| | - Julius P.A. Dewald
- Department of Physical Therapy and Human Movement Sciences, Department of Biomedical EngineeringNorthwestern UniversityChicagoIllinoisUSA
| | - Dirk Dressler
- Movement Disorders Section, Department of NeurologyHannover Medical SchoolHannoverGermany
| | - Alberto Esquenazi
- Department of PM&RMoss Rehab Gait and Motion Analysis Laboratory, Elkins Park, Albert Einstein Medical CenterPennsylvaniaUSA
| | - Jorge Hernandez Franco
- Department of RehabilitationNational Institute of Neurology and Neurosurgery MVS, Universidad Nacional Autónoma de MéxicoMexico CityMexico
| | - Robert Jech
- Department of Neurology and Center of Clinical NeuroscienceFirst Faculty of Medicine Charles University and General University HospitalPragueCzech Republic
| | - Ryuji Kaji
- Tokushima University Graduate School of MedicineTokushimaJapan
| | - Lingjing Jin
- Department of NeurologySchool of Medicine, Tongji Hospital and Shanghai Sunshine Rehabilitation Hospital, Tongji University School of MedicineShanghaiChina
| | - Erle C.H. Lim
- Division of NeurologyNational University Health System, National University of SingaporeSingaporeSingapore
| | - Preeti Raghavan
- Department of Physical Medicine and Rehabilitation and Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Raymond Rosales
- Metropolitan Medical Center, Faculty of Medicine and Surgery, University of Santo Tomas Manila and Clinical Neurophysiology and Movement Disorders, St. Luke's Medical CenterQuezon CityPhilippines
| | - Ali S. Shalash
- Ain Shams Movement Disorders Group, Department of NeurologyAin Shams UniversityCairoEgypt
| | - David M. Simpson
- Department of NeurologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Areerat Suputtitada
- Department of Rehabilitation MedicineFaculty of Medicine, Chulalongkorn UniversityBangkokThailand
| | - Michele Vecchio
- Department of Biomedical and Biotechnological SciencesUniversity of Catania, Physical Medicine and Rehabilitation Unit, “AOU Policlinico G. Rodolico”CataniaItaly
| | - Jörg Wissel
- Neurology and Psychosomatic at WittenbergplatzBerlinGermany
- Center of Sports Medicine, University Outpatient ClinicUniversity of PotsdamPotsdamGermany
| |
Collapse
|
5
|
Manesh SB, Kondiles BR, Wheeler S, Liu J, Zhang L, Chernoff C, Duncan GJ, Ramer MS, Tetzlaff W. Compensatory changes after spinal cord injury in a remyelination deficient mouse model. J Neurochem 2025; 169:e16220. [PMID: 39268880 DOI: 10.1111/jnc.16220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024]
Abstract
The development of therapeutic strategies to reduce impairments following spinal cord injury (SCI) motivates an active area of research, because there are no effective therapies. One strategy is to address injury-induced demyelination of spared axons by promoting endogenous or exogenous remyelination. However, previously, we showed that new myelin was not necessary to regain hindlimb stepping following moderate thoracic spinal cord contusion in 3-month-old mice. The present analysis investigated two potential mechanisms by which animals can re-establish locomotion in the absence of remyelination: compensation through intact white matter and conduction through spared axons. We induced a severe contusion injury to reduce the spared white matter rim in the remyelination deficient model, with no differences in recovery between remyelination deficient animals and injured littermate controls. We investigated the nodal properties of the axons at the lesion and found that in the remyelination deficient model, axons express the Nav1.2 voltage-gated sodium channel, a sub-type not typically expressed at mature nodes of Ranvier. In a moderate contusion injury, conduction velocities through the lesions of remyelination deficient animals were similar to those in animals with the capacity to remyelinate after injury. Detailed gait analysis and kinematics reveal subtle differences between remyelination deficient animals and remyelination competent controls, but no worse deficits. It is possible that upregulation of Nav1.2 channels may contribute to establishing conduction through the lesion. This conduction could contribute to compensation and regained motor function in mouse models of SCI. Such compensatory mechanism may have implications for interpreting efficacy results for remyelinating interventions in mice and the development of therapies for improving recovery following SCI.
Collapse
Affiliation(s)
- S B Manesh
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - B R Kondiles
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - S Wheeler
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - J Liu
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - L Zhang
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - C Chernoff
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - G J Duncan
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - M S Ramer
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - W Tetzlaff
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
6
|
Pinosanu LR, Wolff N, Olaru DG, Popa-Wagner A. Stem Cell Treatments in Preclinical Relevant Stroke Models. CURRENT HEALTH SCIENCES JOURNAL 2023; 49:487-494. [PMID: 38559835 PMCID: PMC10976206 DOI: 10.12865/chsj.49.04.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 11/11/2023] [Indexed: 04/04/2024]
Abstract
Since stroke has limited treatment options, an active search for new therapeutic approaches is required. Initial excitement of using cell-based therapies to stimulate recovery processes in the ischemic brain turned into a more measured perspective, acknowledging obstacles related to the unfavorable environments associated in part with aging. Given the predominance of stroke in older populations, evaluating the effectiveness of cell therapies in aged brain environments is essential and clinically relevant. Despite a common perception of the aged brain being resistant to regeneration, recent research with neural precursor cells and bone marrow-derived mesenchymal stem cells indicates that cell-based therapy can promote plasticity and remodeling in the aged rat brain. However, significant differences in the aged brain compared to the young brain, such as expedited progression of ischemic injury to brain infarction, decreased rate of endogenous neurogenesis, and delayed onset of neurological recovery, must be noted. The effectiveness of cell-based therapies may further be connected to age-related comorbidities such as diabetes or hyperlipidemia, potentially leading to maladaptive or impaired brain remodeling. These age-related factors need careful consideration in the clinical application of restorative therapies for stroke.
Collapse
Affiliation(s)
- Leonard Radu Pinosanu
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Nora Wolff
- University of Crete, School of Sciences, Faculty of Medicine, Heraklion, Crete, Greece
| | - Denissa Greta Olaru
- Department of Ophthalmology, University of Medicine and Pharmacy of Craiova, Romania
| | - Aurel Popa-Wagner
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania
| |
Collapse
|
7
|
Nandakumar B, Blumenthal GH, Disse GD, Desmond PC, Ebinu JO, Ricard J, Bethea JR, Moxon KA. Exercise therapy guides cortical reorganization after midthoracic spinal contusion to enhance control of lower thoracic muscles, supporting functional recovery. Exp Neurol 2023; 364:114394. [PMID: 37001630 DOI: 10.1016/j.expneurol.2023.114394] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 03/20/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023]
Abstract
Postural control is critical for locomotion, allowing for gait changes, obstacle avoidance and navigation of rough terrain. A major problem after spinal cord injury (SCI) is regaining the control of balance to prevent falls and further injury. While the circuits for locomotor pattern generation reside in the spinal cord, postural control consists of multiple, complex networks that interact at the spinal, brainstem and cortical levels. After complete SCI, cortical reorganization establishes novel control of trunk musculature that is required for weight-supported stepping. In this study, we examined the impact of exercise therapy on cortical reorganization in the more clinically relevant models of both moderate and severe midthoracic contusion injury in the rat. Results demonstrate that both spontaneous recovery and therapy induced recovery of weight-supported stepping utilize cortical reorganization. Moreover, exercise therapy further improves outcome by enhancing cortical control of lower thoracic muscles enabling improvements in interlimb coordination associated with improved balance that increases weight-supported stepping. The outcome of this study suggest that cortical control of posture is key to functional improvement in locomotion. This information can be used to improve the timing and type of therapy after SCI by considering changes along the entire neural axis.
Collapse
Affiliation(s)
- Bharadwaj Nandakumar
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA; School of Biomedical Engineering Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Gary H Blumenthal
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA; School of Biomedical Engineering Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Gregory D Disse
- Neuroscience Graduate Program, University of California, Davis, CA 95616, USA
| | - Pierce C Desmond
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Julius O Ebinu
- Department of Neurological Surgery, University of California, Davis, CA 95616, USA
| | - Jerome Ricard
- School of Biomedical Engineering Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - John R Bethea
- School of Biomedical Engineering Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Karen A Moxon
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA; Neuroscience Graduate Program, University of California, Davis, CA 95616, USA; Department of Neurological Surgery, University of California, Davis, CA 95616, USA.
| |
Collapse
|
8
|
Boato F, Guan X, Zhu Y, Ryu Y, Voutounou M, Rynne C, Freschlin CR, Zumbo P, Betel D, Matho K, Makarov SN, Wu Z, Son YJ, Nummenmaa A, Huang JZ, Edwards DJ, Zhong J. Activation of MAP2K signaling by genetic engineering or HF-rTMS promotes corticospinal axon sprouting and functional regeneration. Sci Transl Med 2023; 15:eabq6885. [PMID: 36599003 DOI: 10.1126/scitranslmed.abq6885] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Facilitating axon regeneration in the injured central nervous system remains a challenging task. RAF-MAP2K signaling plays a key role in axon elongation during nervous system development. Here, we show that conditional expression of a constitutively kinase-activated BRAF in mature corticospinal neurons elicited the expression of a set of transcription factors previously implicated in the regeneration of zebrafish retinal ganglion cell axons and promoted regeneration and sprouting of corticospinal tract (CST) axons after spinal cord injury in mice. Newly sprouting axon collaterals formed synaptic connections with spinal interneurons, resulting in improved recovery of motor function. Noninvasive suprathreshold high-frequency repetitive transcranial magnetic stimulation (HF-rTMS) activated the BRAF canonical downstream effectors MAP2K1/2 and modulated the expression of a set of regeneration-related transcription factors in a pattern consistent with that induced by BRAF activation. HF-rTMS enabled CST axon regeneration and sprouting, which was abolished in MAP2K1/2 conditional null mice. These data collectively demonstrate a central role of MAP2K signaling in augmenting the growth capacity of mature corticospinal neurons and suggest that HF-rTMS might have potential for treating spinal cord injury by modulating MAP2K signaling.
Collapse
Affiliation(s)
- Francesco Boato
- Molecular Regeneration and Neuroimaging Laboratory, Burke Neurological Institute, White Plains, NY 10605, USA.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Xiaofei Guan
- Molecular Regeneration and Neuroimaging Laboratory, Burke Neurological Institute, White Plains, NY 10605, USA.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yanjie Zhu
- Molecular Regeneration and Neuroimaging Laboratory, Burke Neurological Institute, White Plains, NY 10605, USA.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Youngjae Ryu
- Molecular Regeneration and Neuroimaging Laboratory, Burke Neurological Institute, White Plains, NY 10605, USA.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mariel Voutounou
- Molecular Regeneration and Neuroimaging Laboratory, Burke Neurological Institute, White Plains, NY 10605, USA.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Christopher Rynne
- Molecular Regeneration and Neuroimaging Laboratory, Burke Neurological Institute, White Plains, NY 10605, USA.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Chase R Freschlin
- Molecular Regeneration and Neuroimaging Laboratory, Burke Neurological Institute, White Plains, NY 10605, USA.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Paul Zumbo
- Applied Bioinformatics Core, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Doron Betel
- Applied Bioinformatics Core, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Katie Matho
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Sergey N Makarov
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.,Electrical and Computer Engineering Department, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - Zhuhao Wu
- Icahn School of Medicine at Mount Sinai, New York, NY 10065, USA
| | - Young-Jin Son
- Shriners Hospitals Pediatric Research Center, Temple University, Philadelphia, PA 19140, USA
| | - Aapo Nummenmaa
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Josh Z Huang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.,Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Dylan J Edwards
- Molecular Regeneration and Neuroimaging Laboratory, Burke Neurological Institute, White Plains, NY 10605, USA.,Moss Rehabilitation Research Institute, Elkins Park, PA 19027, USA.,Thomas Jefferson University, Philadelphia, PA 19108, USA.,Exercise Medicine Research Institute, School of Biomedical and Health Sciences, Edith Cowan University, Joondalup 6027, Australia
| | - Jian Zhong
- Molecular Regeneration and Neuroimaging Laboratory, Burke Neurological Institute, White Plains, NY 10605, USA.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
9
|
Anderson MA, Squair JW, Gautier M, Hutson TH, Kathe C, Barraud Q, Bloch J, Courtine G. Natural and targeted circuit reorganization after spinal cord injury. Nat Neurosci 2022; 25:1584-1596. [PMID: 36396975 DOI: 10.1038/s41593-022-01196-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/05/2022] [Indexed: 11/18/2022]
Abstract
A spinal cord injury disrupts communication between the brain and the circuits in the spinal cord that regulate neurological functions. The consequences are permanent paralysis, loss of sensation and debilitating dysautonomia. However, the majority of circuits located above and below the injury remain anatomically intact, and these circuits can reorganize naturally to improve function. In addition, various neuromodulation therapies have tapped into these processes to further augment recovery. Emerging research is illuminating the requirements to reconstitute damaged circuits. Here, we summarize these natural and targeted reorganizations of circuits after a spinal cord injury. We also advocate for new concepts of reorganizing circuits informed by multi-omic single-cell atlases of recovery from injury. These atlases will uncover the molecular logic that governs the selection of 'recovery-organizing' neuronal subpopulations, and are poised to herald a new era in spinal cord medicine.
Collapse
Affiliation(s)
- Mark A Anderson
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland.,Wyss Center for Bio and Neuroengineering, Geneva, Switzerland
| | - Jordan W Squair
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Matthieu Gautier
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Thomas H Hutson
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland.,Wyss Center for Bio and Neuroengineering, Geneva, Switzerland
| | - Claudia Kathe
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Quentin Barraud
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Jocelyne Bloch
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Grégoire Courtine
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland. .,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland. .,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland.
| |
Collapse
|
10
|
Ran N, Li W, Zhang R, Lin C, Zhang J, Wei Z, Li Z, Yuan Z, Wang M, Fan B, Shen W, Li X, Zhou H, Yao X, Kong X, Feng S. Autologous exosome facilitates load and target delivery of bioactive peptides to repair spinal cord injury. Bioact Mater 2022; 25:766-782. [PMID: 37056263 PMCID: PMC10086682 DOI: 10.1016/j.bioactmat.2022.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/03/2022] [Accepted: 07/03/2022] [Indexed: 11/30/2022] Open
Abstract
Spinal cord injury (SCI) causes motor, sensory and automatic impairment due to rarely axon regeneration. Developing effective treatment for SCI in the clinic is extremely challenging because of the restrictive axonal regenerative ability and disconnection of neural elements after injury, as well as the limited systemic drug delivery efficiency caused by blood spinal cord barrier. To develop an effective non-invasive treatment strategy for SCI in clinic, we generated an autologous plasma exosome (AP-EXO) based biological scaffold where AP-EXO was loaded with neuron targeting peptide (RVG) and growth-facilitating peptides (ILP and ISP). This scaffold can be targeted delivered to neurons in the injured area and elicit robust axon regrowth across the lesion core to the levels over 30-fold greater than naïve treatment, thus reestablish the intraspinal circuits and promote motor functional recovery after spinal cord injury in mice. More importantly, in ex vivo, human plasma exosomes (HP-EXO) loaded with combinatory peptides of RVG, ILP and ISP showed safety and no liver and kidney toxicity in the application to nude SCI mice. Combining the efficacy and safety, the AP-EXO-based personalized treatment confers functional recovery after SCI and showed immense promising in biomedical applications in treating SCI. It is helpful to expand the application of combinatory peptides and human plasma derived autologous exosomes in promoting regeneration and recovery upon SCI treatment.
Collapse
Affiliation(s)
- Ning Ran
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wenxiang Li
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Renjie Zhang
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Caorui Lin
- Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Jianping Zhang
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhijian Wei
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Zonghao Li
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhongze Yuan
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Min Wang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Baoyou Fan
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenyuan Shen
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xueying Li
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hengxing Zhou
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xue Yao
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaohong Kong
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Laboratory of Medical Molecular Virology, School of Medicine, Nankai University, Tianjin, China
- Corresponding author. Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Shiqing Feng
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Corresponding author. Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
11
|
Blackmore M, Batsel E, Tsoulfas P. Widening spinal injury research to consider all supraspinal cell types: Why we must and how we can. Exp Neurol 2021; 346:113862. [PMID: 34520726 PMCID: PMC8805209 DOI: 10.1016/j.expneurol.2021.113862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/19/2021] [Accepted: 09/08/2021] [Indexed: 01/05/2023]
Abstract
The supraspinal connectome consists of dozens of neuronal populations that project axons from the brain to the spinal cord to influence a wide range of motor, autonomic, and sensory functions. The complexity and wide distribution of supraspinal neurons present significant technical challenges, leading most spinal cord injury research to focus on a handful of major pathways such as the corticospinal, rubrospinal, and raphespinal. Much less is known about many additional populations that carry information to modulate or compensate for these main pathways, or which carry pre-autonomic and other information of high value to individuals with spinal injury. A confluence of technical developments, however, now enables a whole-connectome study of spinal cord injury. Improved viral labeling, tissue clearing, and automated registration to 3D atlases can quantify supraspinal neurons throughout the murine brain, offering a practical means to track responses to injury and treatment on an unprecedented scale. Here we discuss the need for expanded connectome-wide analyses in spinal injury research, illustrate the potential by discussing a new web-based resource for brain-wide study of supraspinal neurons, and highlight future prospects for connectome analyses.
Collapse
Affiliation(s)
- Murray Blackmore
- Department of Biomedical Sciences, Marquette University, 53201, United States of America.
| | - Elizabeth Batsel
- Department of Biomedical Sciences, Marquette University, 53201, United States of America
| | - Pantelis Tsoulfas
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America
| |
Collapse
|
12
|
Boitnott A, Garcia-Forn M, Ung DC, Niblo K, Mendonca D, Park Y, Flores M, Maxwell S, Ellegood J, Qiu LR, Grice DE, Lerch JP, Rasin MR, Buxbaum JD, Drapeau E, De Rubeis S. Developmental and Behavioral Phenotypes in a Mouse Model of DDX3X Syndrome. Biol Psychiatry 2021; 90:742-755. [PMID: 34344536 PMCID: PMC8571043 DOI: 10.1016/j.biopsych.2021.05.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 05/12/2021] [Accepted: 05/24/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Mutations in the X-linked gene DDX3X account for approximately 2% of intellectual disability in females, often comorbid with behavioral problems, motor deficits, and brain malformations. DDX3X encodes an RNA helicase with emerging functions in corticogenesis and synaptogenesis. METHODS We generated a Ddx3x haploinsufficient mouse (Ddx3x+/- females) with construct validity for DDX3X loss-of-function mutations. We used standardized batteries to assess developmental milestones and adult behaviors, as well as magnetic resonance imaging and immunostaining of cortical projection neurons to capture early postnatal changes in brain development. RESULTS Ddx3x+/- females showed physical, sensory, and motor delays that evolved into behavioral anomalies in adulthood, including hyperactivity, anxiety-like behaviors, cognitive impairments in specific tasks (e.g., contextual fear memory but not novel object recognition memory), and motor deficits. Motor function declined with age but not if mice were previously exposed to behavioral training. Developmental and behavioral changes were associated with a reduction in brain volume, with some regions (e.g., cortex and amygdala) disproportionally affected. Cortical thinning was accompanied by defective cortical lamination, indicating that Ddx3x regulates the balance of glutamatergic neurons in the developing cortex. CONCLUSIONS These data shed new light on the developmental mechanisms driving DDX3X syndrome and support construct and face validity of this novel preclinical mouse model.
Collapse
Affiliation(s)
- Andrea Boitnott
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Marta Garcia-Forn
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dévina C Ung
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kristi Niblo
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Danielle Mendonca
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yeaji Park
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael Flores
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Biology, New York University, College of Arts and Science, New York, NY 10003, USA
| | - Sylvia Maxwell
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Bronx High School of Science, NY 10468, USA
| | - Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, ON M5T 3H7, Canada
| | - Lily R Qiu
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, OX3 9DU, UK
| | - Dorothy E Grice
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jason P Lerch
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, ON M5T 3H7, Canada.,Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, OX3 9DU, UK.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, ON M5T 3H7, Canada
| | - Mladen-Roko Rasin
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Elodie Drapeau
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
13
|
Li G, Zhang B, Sun JH, Shi LY, Huang MY, Huang LJ, Lin ZJ, Lin QY, Lai BQ, Ma YH, Jiang B, Ding Y, Zhang HB, Li MX, Zhu P, Wang YQ, Zeng X, Zeng YS. An NT-3-releasing bioscaffold supports the formation of TrkC-modified neural stem cell-derived neural network tissue with efficacy in repairing spinal cord injury. Bioact Mater 2021; 6:3766-3781. [PMID: 33898877 PMCID: PMC8044869 DOI: 10.1016/j.bioactmat.2021.03.036] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/03/2021] [Accepted: 03/18/2021] [Indexed: 01/01/2023] Open
Abstract
The mechanism underlying neurogenesis during embryonic spinal cord development involves a specific ligand/receptor interaction, which may be help guide neuroengineering to boost stem cell-based neural regeneration for the structural and functional repair of spinal cord injury. Herein, we hypothesized that supplying spinal cord defects with an exogenous neural network in the NT-3/fibroin-coated gelatin sponge (NF-GS) scaffold might improve tissue repair efficacy. To test this, we engineered tropomyosin receptor kinase C (TrkC)-modified neural stem cell (NSC)-derived neural network tissue with robust viability within an NF-GS scaffold. When NSCs were genetically modified to overexpress TrkC, the NT-3 receptor, a functional neuronal population dominated the neural network tissue. The pro-regenerative niche allowed the long-term survival and phenotypic maintenance of the donor neural network tissue for up to 8 weeks in the injured spinal cord. Additionally, host nerve fibers regenerated into the graft, making synaptic connections with the donor neurons. Accordingly, motor function recovery was significantly improved in rats with spinal cord injury (SCI) that received TrkC-modified NSC-derived neural network tissue transplantation. Together, the results suggested that transplantation of the neural network tissue formed in the 3D bioactive scaffold may represent a valuable approach to study and develop therapies for SCI. A NT-3 sustained-release scaffold confers a microenvironment partially simulating the developmental spinal cord. The NT-3 microenvironment boosts neuronal differentiation of TrkC-modified NSCs by interactions between ligand and receptor. TrkC-NSCs is self-organized into a neural network tissue with typical neural excitability in 3D bioactive scaffold in vitro. The grafted neural network tissue can survive and maintain neural property, and improve motor function of paralyzed rats.
Collapse
Affiliation(s)
- Ge Li
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.,Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.,Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China
| | - Bao Zhang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jia-Hui Sun
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Li-Yang Shi
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, 410082, China
| | - Meng-Yao Huang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Li-Jun Huang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zi-Jing Lin
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qiong-Yu Lin
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China
| | - Bi-Qin Lai
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.,Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yuan-Huan Ma
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.,Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China
| | - Bin Jiang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ying Ding
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.,Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China
| | - Hong-Bo Zhang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Miao-Xin Li
- Laboratory of Precision Medical Genomics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China
| | - Ya-Qiong Wang
- Department of Electron Microscope, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiang Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.,Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yuan-Shan Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.,Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
14
|
Suo X, Ding H, Li X, Zhang Y, Liang M, Zhang Y, Yu C, Qin W. Anatomical and functional coupling between the dorsal and ventral attention networks. Neuroimage 2021; 232:117868. [PMID: 33647500 DOI: 10.1016/j.neuroimage.2021.117868] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/21/2021] [Accepted: 02/10/2021] [Indexed: 11/26/2022] Open
Abstract
Studies have indicated that the dorsal attention network (DAN) and the ventral attention network (VAN) functionally interact via several fronto-parietal connector hubs. However, the anatomical connectivity profiles of these connector hubs, and the coupling between the anatomical and functional connectivities of them, are still unknown. In the present study, we found that functional connector hubs anatomically bridged the DAN and VAN based on multimodal magnetic resonance imaging data from the Human Connectome Project (HCP) Consortium and an independent Chinese cohort. The three hubs had unique anatomical connectivity patterns with the attention sub-networks. For each connector hub, the pattern of anatomical connectivity resembled the functional one. Finally, the strength of the anatomical connectivity of these connector hubs was positively associated with the functional connectivity at the group- and individual-levels. Our findings help to better understand the anatomical mechanisms underlying the functional interactions between the DAN and the VAN.
Collapse
Affiliation(s)
- Xinjun Suo
- Department of Radiology, Tianjin Medical University General Hospital, Anshan Road No 154, Heping District, Tianjin 300052, China; Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China; School of Medical Imaging, Tianjin Medical University, Tianjin 300070, China
| | - Hao Ding
- Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China; School of Medical Imaging, Tianjin Medical University, Tianjin 300070, China
| | - Xi Li
- Department of Radiology, Tianjin Medical University General Hospital, Anshan Road No 154, Heping District, Tianjin 300052, China; Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yaodan Zhang
- Department of Radiology, Tianjin Medical University General Hospital, Anshan Road No 154, Heping District, Tianjin 300052, China; Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Meng Liang
- Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China; School of Medical Imaging, Tianjin Medical University, Tianjin 300070, China
| | - Yongqiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chunshui Yu
- Department of Radiology, Tianjin Medical University General Hospital, Anshan Road No 154, Heping District, Tianjin 300052, China; Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China; School of Medical Imaging, Tianjin Medical University, Tianjin 300070, China
| | - Wen Qin
- Department of Radiology, Tianjin Medical University General Hospital, Anshan Road No 154, Heping District, Tianjin 300052, China; Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
15
|
Transneuronal delivery of hyper-interleukin-6 enables functional recovery after severe spinal cord injury in mice. Nat Commun 2021; 12:391. [PMID: 33452250 PMCID: PMC7810685 DOI: 10.1038/s41467-020-20112-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 11/13/2020] [Indexed: 12/27/2022] Open
Abstract
Spinal cord injury (SCI) often causes severe and permanent disabilities due to the regenerative failure of severed axons. Here we report significant locomotor recovery of both hindlimbs after a complete spinal cord crush. This is achieved by the unilateral transduction of cortical motoneurons with an AAV expressing hyper-IL-6 (hIL-6), a potent designer cytokine stimulating JAK/STAT3 signaling and axon regeneration. We find collaterals of these AAV-transduced motoneurons projecting to serotonergic neurons in both sides of the raphe nuclei. Hence, the transduction of cortical neurons facilitates the axonal transport and release of hIL-6 at innervated neurons in the brain stem. Therefore, this transneuronal delivery of hIL-6 promotes the regeneration of corticospinal and raphespinal fibers after injury, with the latter being essential for hIL-6-induced functional recovery. Thus, transneuronal delivery enables regenerative stimulation of neurons in the deep brain stem that are otherwise challenging to access, yet highly relevant for functional recovery after SCI.
Collapse
|
16
|
Blauwblomme T, Demertzi A, Tacchela J, Fillon L, Bourgeois M, Losito E, Eisermann M, Marinazzo D, Raimondo F, Alcauter S, Van De Steen F, Colenbier N, Laureys S, Dangouloff‐Ros V, Naccache L, Boddaert N, Nabbout R. Complete hemispherotomy leads to lateralized functional organization and lower level of consciousness in the isolated hemisphere. Epilepsia Open 2020; 5:537-549. [PMID: 33336125 PMCID: PMC7733653 DOI: 10.1002/epi4.12433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 08/18/2020] [Accepted: 08/22/2020] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE To quantify whole-brain functional organization after complete hemispherotomy, characterizing unexplored plasticity pathways and the conscious level of the dissected hemispheres. METHODS Evaluation with multimodal magnetic resonance imaging in two pediatric patients undergoing right hemispherotomy including complete callosotomy with a perithalamic section. Regional cerebral blood flow and fMRI network connectivity assessed the functional integrity of both hemispheres after surgery. The level of consciousness was tested by means of a support vector machine classifier which compared the intrinsic organization of the dissected hemispheres with those of patients suffering from disorders of consciousness. RESULTS After hemispherotomy, both patients showed typical daily functionality. We found no interhemispheric transfer of functional connectivity in either patient as predicted by the operation. The healthy left hemispheres displayed focal blood hyperperfusion in motor and limbic areas, with preserved network-level organization. Unexpectedly, the disconnected right hemispheres showed sustained network organization despite low regional cerebral blood flow. Subcortically, functional connectivity was increased in the left thalamo-cortical loop and between the cerebelli. One patient further showed unusual ipsilateral right cerebello-cortical connectivity, which was explained by the mediation of the vascular system. The healthy left hemisphere had higher probability to be classified as in a minimally conscious state compared to the isolated right hemisphere. SIGNIFICANCE Complete hemispherotomy leads to a lateralized whole-brain organization, with the remaining hemisphere claiming most of the brain's energetic reserves supported by subcortical structures. Our results further underline the contribution of nonneuronal vascular signals on contralateral connectivity, shedding light on the nature of network organization in the isolated tissue. The disconnected hemisphere is characterized by a level of consciousness which is necessary but insufficient for conscious processing, paving the way for more specific inquiries about its role in awareness in the absence of behavioral output.
Collapse
Affiliation(s)
- Thomas Blauwblomme
- Assistance Publique Hôpitaux de ParisHôpital Necker‐Enfants MaladesParisFrance
- Université de ParisParisFrance
- INSERM U1163Institut ImagineParisFrance
| | - Athena Demertzi
- GIGA‐Consciousness, Physiology of Cognition Research LabGIGA InstituteUniversity of LiègeLiègeBelgium
- INSERMU1127ParisFrance
- Institut du Cerveau et de la Moelle EpinièreHôpital Pitié‐SalpêtrièreParisFrance
| | | | | | - Marie Bourgeois
- Assistance Publique Hôpitaux de ParisHôpital Necker‐Enfants MaladesParisFrance
| | - Emma Losito
- Assistance Publique Hôpitaux de ParisHôpital Necker‐Enfants MaladesParisFrance
| | - Monika Eisermann
- Assistance Publique Hôpitaux de ParisHôpital Necker‐Enfants MaladesParisFrance
| | - Daniele Marinazzo
- Department of Data AnalysisFaculty of Psychological and Educational SciencesUniversity of GhentGhentBelgium
| | - Federico Raimondo
- Institut du Cerveau et de la Moelle EpinièreHôpital Pitié‐SalpêtrièreParisFrance
- GIGA‐Consciousness, Coma Science GroupGIGA InstituteUniversity of LiègeLiègeBelgium
| | - Sarael Alcauter
- Instituto de NeurobiologíaUniversidad Nacional Autónoma de MéxicoQuerétaroMéxico
| | - Frederik Van De Steen
- GIGA‐Consciousness, Physiology of Cognition Research LabGIGA InstituteUniversity of LiègeLiègeBelgium
| | - Nigel Colenbier
- GIGA‐Consciousness, Physiology of Cognition Research LabGIGA InstituteUniversity of LiègeLiègeBelgium
| | - Steven Laureys
- GIGA‐Consciousness, Coma Science GroupGIGA InstituteUniversity of LiègeLiègeBelgium
| | - Volodia Dangouloff‐Ros
- Assistance Publique Hôpitaux de ParisHôpital Necker‐Enfants MaladesParisFrance
- Université de ParisParisFrance
- INSERM U1163Institut ImagineParisFrance
| | - Lionel Naccache
- INSERMU1127ParisFrance
- Institut du Cerveau et de la Moelle EpinièreHôpital Pitié‐SalpêtrièreParisFrance
| | - Nathalie Boddaert
- Assistance Publique Hôpitaux de ParisHôpital Necker‐Enfants MaladesParisFrance
- Université de ParisParisFrance
- INSERM U1163Institut ImagineParisFrance
| | - Rima Nabbout
- Assistance Publique Hôpitaux de ParisHôpital Necker‐Enfants MaladesParisFrance
- Université de ParisParisFrance
- INSERM U1163Institut ImagineParisFrance
| |
Collapse
|
17
|
Margoni M, Poggiali D, Zywicki S, Rubin M, Lazzarotto A, Franciotta S, Anglani MG, Causin F, Rinaldi F, Perini P, Filippi M, Gallo P. Early red nucleus atrophy in relapse-onset multiple sclerosis. Hum Brain Mapp 2020; 42:154-160. [PMID: 33047810 PMCID: PMC7721227 DOI: 10.1002/hbm.25213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/25/2020] [Accepted: 09/14/2020] [Indexed: 12/25/2022] Open
Abstract
No study has investigated red nucleus (RN) atrophy in multiple sclerosis (MS) despite cerebellum and its connections are elective sites of MS‐related pathology. In this study, we explore RN atrophy in early MS phases and its association with cerebellar damage (focal lesions and atrophy) and physical disability. Thirty‐seven relapse‐onset MS (RMS) patients having mean age of 35.6 ± 8.5 (18–56) years and mean disease duration of 1.1 ± 1.5 (0–5) years, and 36 age‐ and sex‐matched healthy controls (HC) were studied. Cerebellar and RN lesions and volumes were analyzed on 3 T‐MRI images. RMS did not differ from HC in cerebellar lobe volumes but significantly differed in both right (107.84 ± 13.95 mm3 vs. 99.37 ± 11.53 mm3, p = .019) and left (109.71 ± 14.94 mm3 vs. 100.47 ± 15.78 mm3, p = .020) RN volumes. Cerebellar white matter lesion volume (WMLV) inversely correlated with both right and left RN volumes (r = −.333, p = .004 and r = −.298, p = .010, respectively), while no correlation was detected between RN volumes and mean cortical thickness, cerebellar gray matter lesion volume, and supratentorial WMLV (right RN: r = −.147, p = .216; left RN: r = −.153, p = .196). Right, but not left, RN volume inversely correlated with midbrain WMLV (r = −.310, p = .008), while no correlation was observed between whole brainstem WMLV and either RN volumes (right RN: r = −.164, p = .164; left RN: r = −.64, p = .588). Finally, left RN volume correlated with vermis VIIb (r = .297, p = .011) and right interposed nucleus (r = .249, p = .034) volumes. We observed RN atrophy in early RMS, likely resulting from anterograde axonal degeneration starting in cerebellar and midbrain WML. RN atrophy seems a promising marker of neurodegeneration and/or cerebellar damage in RMS.
Collapse
Affiliation(s)
- Monica Margoni
- Multiple Sclerosis Centre of the Veneto Region (CeSMuV), University Hospital of Padua, Padua, Italy.,Padova Neuroscience Centre (PNC), University of Padua, Padua, Italy
| | - Davide Poggiali
- Padova Neuroscience Centre (PNC), University of Padua, Padua, Italy.,Department of Mathematics, University of Padua, Padua, Italy
| | - Sofia Zywicki
- Multiple Sclerosis Centre of the Veneto Region (CeSMuV), University Hospital of Padua, Padua, Italy
| | - Martina Rubin
- Multiple Sclerosis Centre of the Veneto Region (CeSMuV), University Hospital of Padua, Padua, Italy
| | - Andrea Lazzarotto
- Multiple Sclerosis Centre of the Veneto Region (CeSMuV), University Hospital of Padua, Padua, Italy
| | - Silvia Franciotta
- Multiple Sclerosis Centre of the Veneto Region (CeSMuV), University Hospital of Padua, Padua, Italy
| | | | | | - Francesca Rinaldi
- Multiple Sclerosis Centre of the Veneto Region (CeSMuV), University Hospital of Padua, Padua, Italy
| | - Paola Perini
- Multiple Sclerosis Centre of the Veneto Region (CeSMuV), University Hospital of Padua, Padua, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurophysiology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Gallo
- Multiple Sclerosis Centre of the Veneto Region (CeSMuV), University Hospital of Padua, Padua, Italy.,Department of Neurosciences, Medical School, University of Padua, Padua, Italy
| |
Collapse
|
18
|
Tian T, Li X. Applications of tissue clearing in the spinal cord. Eur J Neurosci 2020; 52:4019-4036. [DOI: 10.1111/ejn.14938] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/22/2020] [Accepted: 08/03/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Ting Tian
- Beijing Key Laboratory for Biomaterials and Neural Regeneration School of Biological Science and Medical Engineering Beihang University Beijing China
| | - Xiaoguang Li
- Beijing Key Laboratory for Biomaterials and Neural Regeneration School of Biological Science and Medical Engineering Beihang University Beijing China
- Beijing International Cooperation Bases for Science and Technology on Biomaterials and Neural Regeneration Beijing Advanced Innovation Center for Biomedical Engineering Beihang University Beijing China
- Department of Neurobiology School of Basic Medical Sciences Capital Medical University Beijing China
| |
Collapse
|
19
|
Kutikov AB, Moore SW, Layer RT, Podell PE, Sridhar N, Santamaria AJ, Aimetti AA, Hofstetter CP, Ulich TR, Guest JD. Method and Apparatus for the Automated Delivery of Continuous Neural Stem Cell Trails Into the Spinal Cord of Small and Large Animals. Neurosurgery 2020; 85:560-573. [PMID: 30169668 DOI: 10.1093/neuros/nyy379] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 07/19/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Immature neurons can extend processes after transplantation in adult animals. Neuronal relays can form between injected neural stem cells (NSCs) and surviving neurons, possibly improving recovery after spinal cord injury (SCI). Cell delivery methods of single or multiple bolus injections of concentrated cell suspensions thus far tested in preclinical and clinical experiments are suboptimal for new tract formation. Nonuniform injectate dispersal is often seen due to gravitational cell settling and clumping. Multiple injections have additive risks of hemorrhage, parenchymal damage, and cellular reflux and require additional surgical exposure. The deposition of multiply delivered cells boluses may be uneven and discontinuous. OBJECTIVE To develop an injection apparatus and methodology to deliver continuous cellular trails bridging spinal cord lesions. METHODS We improved the uniformity of cellular trails by formulating NSCs in hyaluronic acid. The TrailmakerTM stereotaxic injection device was automatized to extend a shape memory needle from a single-entry point in the spinal cord longitudinal axis to "pioneer" a new trail space and then retract while depositing an hyaluronic acid-NSC suspension. We conducted testing in a collagen spinal models, and animal testing using human NSCs (hNSCs) in rats and minipigs. RESULTS Continuous surviving trails of hNSCs within rat and minipig naive spinal cords were 12 and 40 mm in length. hNSC trails were delivered across semi-acute contusion injuries in rats. Transplanted hNSCs survived and were able to differentiate into neural lineage cells and astrocytes. CONCLUSION The TrailmakerTM creates longitudinal cellular trails spanning multiple levels from a single-entry point. This may enhance the ability of therapeutics to promote functional relays after SCI.
Collapse
Affiliation(s)
| | - Simon W Moore
- InVivo Therapeutics Corporation, Cambridge, Massachusetts
| | | | | | - Nithya Sridhar
- InVivo Therapeutics Corporation, Cambridge, Massachusetts
| | | | - Alex A Aimetti
- InVivo Therapeutics Corporation, Cambridge, Massachusetts
| | | | - Thomas R Ulich
- InVivo Therapeutics Corporation, Cambridge, Massachusetts
| | - James D Guest
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida.,Department of Neurosurgery, University of Miami, Miami, Florida
| |
Collapse
|
20
|
Garr E, Delamater AR. Chemogenetic inhibition in the dorsal striatum reveals regional specificity of direct and indirect pathway control of action sequencing. Neurobiol Learn Mem 2020; 169:107169. [PMID: 31972244 DOI: 10.1016/j.nlm.2020.107169] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/07/2020] [Accepted: 01/18/2020] [Indexed: 11/17/2022]
Abstract
Animals engage in intricate action sequences that are constructed during instrumental learning. There is broad consensus that the basal ganglia play a crucial role in the formation and fluid performance of action sequences. To investigate the role of the basal ganglia direct and indirect pathways in action sequencing, we virally expressed Cre-dependent Gi-DREADDs in either the dorsomedial (DMS) or dorsolateral (DLS) striatum during and/or after action sequence learning in D1 and D2 Cre rats. Action sequence performance in D1 Cre rats was slowed down early in training when DREADDs were activated in the DMS, but sped up when activated in the DLS. Acquisition of the reinforced sequence was hindered when DREADDs were activated in the DLS of D2 Cre rats. Outcome devaluation tests conducted after training revealed that the goal-directed control of action sequence rates was immune to chemogenetic inhibition-rats suppressed the rate of sequence performance when rewards were devalued. Sequence initiation latencies were generally sensitive to outcome devaluation, except in the case where DREADD activation was removed in D2 Cre rats that previously experienced DREADD activation in the DMS during training. Sequence completion latencies were generally not sensitive to outcome devaluation, except in the case where D1 Cre rats experienced DREADD activation in the DMS during training and test. Collectively, these results suggest that the indirect pathway originating from the DLS is part of a circuit involved in the effective reinforcement of action sequences, while the direct and indirect pathways originating from the DMS contribute to the goal-directed control of sequence completion and initiation, respectively.
Collapse
Affiliation(s)
- Eric Garr
- Graduate Center, City University of New York, United States; Brooklyn College, City University of New York, United States.
| | - Andrew R Delamater
- Graduate Center, City University of New York, United States; Brooklyn College, City University of New York, United States
| |
Collapse
|
21
|
Guo J, Liu J, Wang C, Cao C, Fu L, Han T, Cheng J, Yu C, Qin W. Differential involvement of rubral branches in chronic capsular and pontine stroke. NEUROIMAGE-CLINICAL 2019; 24:102090. [PMID: 31835285 PMCID: PMC6911903 DOI: 10.1016/j.nicl.2019.102090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/06/2019] [Accepted: 11/10/2019] [Indexed: 12/12/2022]
Abstract
Rubral branches were differentially involved in chronic capsular and pontine stroke. The impairment severity of each rubral branch was dependent on lesion locations. The integrity of the rubral branches is related to the severity of motor impairment.
Background and Purpose Early studies have indicated that the cortico-rubro-spinal tracts play important roles in motor dysfunction after stroke. However, the differential involvement of the rubral branches in capsular and pontine stroke, and their associations with the motor impairment are still unknown. Methods The present study recruited 144 chronic stroke patients and 91 normal controls (NC) from three hospitals, including 102 cases with capsular stroke (CS) and 42 cases with pontine stroke (PS). The rubral branches, including bilateral corticorubral tracts (CRT), dentatorubral tracts (DRT), and rubrospinal tracts (RST), and the cortico-spinal tract (CST) were reconstructed based on the dataset of the Human Connectome Project. Group differences in diffusion scalars of each rubral branch were compared, and the associations between the diffusion measures of rubral branches and the Fugl-Meyer assessment (FMA) scores were tested. Results The bilateral CRT of the CS cases showed significantly lower factional anisotropy (FA) than in the NC. The bilateral DRT of the PS cases had lower FA than in the NC. Both CS and PS cases had significantly lower FA of the bilateral RST than the NC. Besides, the stroke patients demonstrated significantly lower FA in bilateral CSTs than the NC. Partial correlation analysis identified significantly positive correlations between the FA of the ipsilesional and CRT and the FMA scores in the CS group, and significantly positive correlations between the FA of the RST bilaterally and the FMA scores in the CS and PS groups. Furthermore, the association between RST integrity and FMA scores still survived after controlling for the effect of the CST. Finally, multiple regression modelling found that rubral tract FA explained 39.2% of the variance in FMA scores for CS patients, and 48.8% of the variance in FMA scores for PS patients. Conclusions The bilateral rubral branches were differentially involved in the chronic capsular and pontine stroke, and the impairment severity of each rubral branch was dependent on lesion locations. The integrity of the rubral branches is related to motor impairment in both the chronic capsular and pontine stroke.
Collapse
Affiliation(s)
- Jun Guo
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Radiology, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Jingchun Liu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Caihong Wang
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Chen Cao
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Lejun Fu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Radiology, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Tong Han
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Chunshui Yu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wen Qin
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
22
|
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
|
23
|
The cortico-rubral and cerebello-rubral pathways are topographically organized within the human red nucleus. Sci Rep 2019; 9:12117. [PMID: 31431648 PMCID: PMC6702172 DOI: 10.1038/s41598-019-48164-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/11/2019] [Indexed: 02/03/2023] Open
Abstract
The Red Nucleus (RN) is a large nucleus located in the ventral midbrain: it is subdivided into a small caudal magnocellular part (mRN) and a large rostral parvocellular part (pRN). These distinct structural regions are part of functionally different networks and show distinctive connectivity features: the mRN is connected to the interposed nucleus, whilst the pRN is mainly connected to dentate nucleus, cortex and inferior olivary complex. Despite functional neuroimaging studies suggest RN involvement in complex motor and higher order functions, the pRN and mRN cannot be distinguished using conventional MRI. Herein, we employ high-quality structural and diffusion MRI data of 100 individuals from the Human Connectome Project repository and constrained spherical deconvolution tractography to perform connectivity-based segmentation of the human RN. In particular, we tracked connections of RN with the inferior olivary complex, the interposed nucleus, the dentate nucleus and the cerebral cortex. We found that the RN can be subdivided according to its connectivity into two clusters: a large ventrolateral one, mainly connected with the cerebral cortex and the inferior olivary complex, and a smaller dorsomedial one, mainly connected with the interposed nucleus. This structural topography strongly reflects the connectivity patterns of pRN and mRN respectively. Structural connectivity-based segmentation could represent a useful tool for the identification of distinct subregions of the human red nucleus on 3T MRI thus allowing a better evaluation of this subcortical structure in healthy and pathological conditions.
Collapse
|
24
|
Hellenbrand DJ, Reichl KA, Travis BJ, Filipp ME, Khalil AS, Pulito DJ, Gavigan AV, Maginot ER, Arnold MT, Adler AG, Murphy WL, Hanna AS. Sustained interleukin-10 delivery reduces inflammation and improves motor function after spinal cord injury. J Neuroinflammation 2019; 16:93. [PMID: 31039819 PMCID: PMC6489327 DOI: 10.1186/s12974-019-1479-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 04/09/2019] [Indexed: 12/12/2022] Open
Abstract
Background The anti-inflammatory cytokine interleukin-10 (IL-10) has been explored previously as a treatment method for spinal cord injury (SCI) due to its ability to attenuate pro-inflammatory cytokines and reduce apoptosis. Primary limitations when using systemic injections of IL-10 are that it is rapidly cleared from the injury site and that it does not cross the blood–spinal cord barrier. Objective Here, mineral-coated microparticles (MCMs) were used to obtain a local sustained delivery of IL-10 directly into the injury site after SCI. Methods Female Sprague-Dawley rats were contused at T10 and treated with either an intraperitoneal injection of IL-10, an intramedullary injection of IL-10, or MCMs bound with IL-10 (MCMs+IL-10). After treatment, cytokine levels were measured in the spinal cord, functional testing and electrophysiology were performed, axon tracers were injected into the brainstem and motor cortex, macrophage levels were counted using flow cytometry and immunohistochemistry, and lesion size was measured. Results When treated with MCMs+IL-10, IL-10 was significantly elevated in the injury site and inflammatory cytokines were significantly suppressed, prompting significantly less cells expressing antigens characteristic of inflammatory macrophages and significantly more cells expressing antigens characteristic of earlier stage anti-inflammatory macrophages. Significantly more axons were preserved within the rubrospinal and reticulospinal tracts through the injury site when treated with MCMs+IL-10; however, there was no significant difference in corticospinal tract axons preserved, regardless of treatment group. The rats treated with MCMs+IL-10 were the only group with a significantly higher functional score compared to injured controls 28 days post-contusion. Conclusion These data demonstrate that MCMs can effectively deliver biologically active IL-10 for an extended period of time altering macrophage phenotype and aiding in functional recovery after SCI.
Collapse
Affiliation(s)
- Daniel J Hellenbrand
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA.,Department of Biomedical Engineering, University of Wisconsin, Madison, WI, 53706, USA
| | - Kaitlyn A Reichl
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA.,Department of Biomedical Engineering, University of Wisconsin, Madison, WI, 53706, USA
| | - Benjamin J Travis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Mallory E Filipp
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Andrew S Khalil
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, 53706, USA
| | - Domenic J Pulito
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Ashley V Gavigan
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Elizabeth R Maginot
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Mitchell T Arnold
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Alexander G Adler
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, 53706, USA.,Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, WI, 53705, USA
| | - Amgad S Hanna
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA.
| |
Collapse
|
25
|
Dickson RG, Lall VK, Ichiyama RM. Enhancing plasticity in spinal sensorimotor circuits following injuries to facilitate recovery of motor control. CURRENT OPINION IN PHYSIOLOGY 2019. [DOI: 10.1016/j.cophys.2019.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
26
|
Choi H, Seo KC, Kim TU, Lee SJ, Hyun JK. Repetitive Transcranial Magnetic Stimulation Enhances Recovery in Central Cord Syndrome Patients. Ann Rehabil Med 2019; 43:62-73. [PMID: 30852872 PMCID: PMC6409668 DOI: 10.5535/arm.2019.43.1.62] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 10/05/2018] [Indexed: 12/17/2022] Open
Abstract
Objective To investigate the effect of repetitive transcranial magnetic stimulation (rTMS) on neurological and functional recovery in patients with central cord syndrome (CCS) involving the upper extremities between the treated and non-treated sides of the treated group and whether the outcomes are comparable to that of the untreated control group. Methods Nineteen CCS patients were treated with high-frequency (20 Hz) rTMS over the motor cortex for 5 days. The stimulation side was randomly selected, and all the subjects received conventional occupational therapy during the rTMS-treatment period. Twenty CCS patients who did not receive rTMS were considered as controls. Clinical assessments, including those by the International Standard for Neurological Classification of Spinal Cord Injury, the Jebsen-Taylor Hand Function Test, and the O'Connor Finger Dexterity Test were performed initially and followed up for 1 month after rTMS treatment or 5 weeks after initial assessments. Results The motor scores for upper extremities were increased and the number of improved cases was greater for the treated side in rTMS-treated patients than for the non-treated side in rTMS-treated patients or controls. The improved cases for writing time and score measured on the Jebsen-Taylor Hand Function Test were also significantly greater in number on the rTMS-treated side compared with the non-treated side and controls. There were no adverse effects during rTMS therapy or the follow-up period. Conclusion The results of the application of high-frequency rTMS treatment to CCS patients suggest that rTMS can enhance the motor recovery and functional fine motor task performance of the upper extremities in such individuals.
Collapse
Affiliation(s)
- Hana Choi
- Department of Rehabilitation Medicine, Dankook University College of Medicine, Cheonan, Korea
| | - Kyung Cheon Seo
- Department of Rehabilitation Medicine, Dankook University College of Medicine, Cheonan, Korea
| | - Tae Uk Kim
- Department of Rehabilitation Medicine, Dankook University College of Medicine, Cheonan, Korea
| | - Seong Jae Lee
- Department of Rehabilitation Medicine, Dankook University College of Medicine, Cheonan, Korea
| | - Jung Keun Hyun
- Department of Rehabilitation Medicine, Dankook University College of Medicine, Cheonan, Korea.,Department of Nanobiomedical Science and BK21 Plus NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Korea.,Wiregene Co. Ltd., Cheonan, Korea
| |
Collapse
|
27
|
Yu P, Zhang W, Liu Y, Sheng C, So KF, Zhou L, Zhu H. The effects and potential mechanisms of locomotor training on improvements of functional recovery after spinal cord injury. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 147:199-217. [DOI: 10.1016/bs.irn.2019.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
28
|
Filipp ME, Travis BJ, Henry SS, Idzikowski EC, Magnuson SA, Loh MY, Hellenbrand DJ, Hanna AS. Differences in neuroplasticity after spinal cord injury in varying animal models and humans. Neural Regen Res 2019; 14:7-19. [PMID: 30531063 PMCID: PMC6263009 DOI: 10.4103/1673-5374.243694] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Rats have been the primary model to study the process and underlying mechanisms of recovery after spinal cord injury. Two weeks after a severe spinal cord contusion, rats can regain weight-bearing abilities without therapeutic interventions, as assessed by the Basso, Beattie and Bresnahan locomotor scale. However, many human patients suffer from permanent loss of motor function following spinal cord injury. While rats are the most understood animal model, major differences in sensorimotor pathways between quadrupeds and bipeds need to be considered. Understanding the major differences between the sensorimotor pathways of rats, non-human primates, and humans is a start to improving targets for treatments of human spinal cord injury. This review will discuss the neuroplasticity of the brain and spinal cord after spinal cord injury in rats, non-human primates, and humans. A brief overview of emerging interventions to induce plasticity in humans with spinal cord injury will also be discussed.
Collapse
Affiliation(s)
- Mallory E Filipp
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Benjamin J Travis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Stefanie S Henry
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Emma C Idzikowski
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Sarah A Magnuson
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Megan Yf Loh
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | | | - Amgad S Hanna
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
29
|
Global Connectivity and Function of Descending Spinal Input Revealed by 3D Microscopy and Retrograde Transduction. J Neurosci 2018; 38:10566-10581. [PMID: 30341180 DOI: 10.1523/jneurosci.1196-18.2018] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 10/03/2018] [Accepted: 10/09/2018] [Indexed: 12/20/2022] Open
Abstract
The brain communicates with the spinal cord through numerous axon tracts that arise from discrete nuclei, transmit distinct functions, and often collateralize to facilitate the coordination of descending commands. This complexity presents a major challenge to interpreting functional outcomes from therapies that target supraspinal connectivity after injury or disease, while the wide distribution of supraspinal nuclei complicates the delivery of therapeutics. Here we harness retrograde viral vectors to overcome these challenges. We demonstrate that injection of AAV2-Retro to the cervical spinal cord of adult female mice results in highly efficient transduction of supraspinal populations throughout the brainstem, midbrain, and cortex. Some supraspinal populations, including corticospinal and rubrospinal neurons, were transduced with >90% efficiency, with robust transgene expression within 3 d of injection. In contrast, propriospinal and raphe spinal neurons showed much lower rates of retrograde transduction. Using tissue clearing and light-sheet microscopy we present detailed visualizations of descending axons tracts and create a mesoscopic projectome for the spinal cord. Moreover, chemogenetic silencing of supraspinal neurons with retrograde vectors resulted in complete and reversible forelimb paralysis, illustrating effective modulation of supraspinal function. Retrograde vectors were also highly efficient when injected after spinal injury, highlighting therapeutic potential. These data provide a global view of supraspinal connectivity and illustrate the potential of retrograde vectors to parse the functional contributions of supraspinal inputs.SIGNIFICANCE STATEMENT The complexity of descending inputs to the spinal cord presents a major challenge in efforts deliver therapeutics to widespread supraspinal systems, and to interpret their functional effects. Here we demonstrate highly effective gene delivery to diverse supraspinal nuclei using a retrograde viral approach and combine it with tissue clearing and 3D microscopy to map the descending projectome from brain to spinal cord. These data highlight newly developed retrograde viruses as therapeutic and research tools, while offering new insights into supraspinal connectivity.
Collapse
|
30
|
Qian J, Wu W, Xiong W, Chai Z, Xu XM, Jin X. Longitudinal Optogenetic Motor Mapping Revealed Structural and Functional Impairments and Enhanced Corticorubral Projection after Contusive Spinal Cord Injury in Mice. J Neurotrauma 2018; 36:485-499. [PMID: 29848155 DOI: 10.1089/neu.2018.5713] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Current evaluation of impairment and repair after spinal cord injury (SCI) is largely dependent on behavioral assessment and histological analysis of injured tissue and pathways. Here, we evaluated whether transcranial optogenetic mapping of motor cortex could reflect longitudinal structural and functional damage and recovery after SCI. In Thy1-Channelrhodopsin2 transgenic mice, repeated motor mappings were made by recording optogenetically evoked electromyograms (EMGs) of a hindlimb at baseline and 1 day and 2, 4, and 6 weeks after mild, moderate, and severe spinal cord contusion. Injuries caused initial decreases in EMG amplitude, losses of motor map, and subsequent partial recoveries, all of which corresponded to injury severity. Reductions in map size were positively correlated with motor performance, as measured by Basso Mouse Scale, rota-rod, and grid walk tests, at different time points, as well as with lesion area at spinal cord epicenter at 6 weeks post-SCI. Retrograde tracing with Fluoro-Gold showed decreased numbers of cortico- and rubrospinal neurons, with the latter being negatively correlated with motor map size. Combined retro- and anterograde tracing and immunostaining revealed more neurons activated in red nucleus by cortical stimulation and enhanced corticorubral axons and synapses in red nucleus after SCI. Electrophysiological recordings showed lower threshold and higher amplitude of corticorubral synaptic response after SCI. We conclude that transcranial optogenetic motor mapping is sensitive and efficient for longitudinal evaluation of impairment and plasticity of SCI, and that spinal cord contusion induces stronger anatomical and functional corticorubral connection that may contribute to spontaneous recovery of motor function.
Collapse
Affiliation(s)
- Jun Qian
- 1 Department of Anatomy and Cell Biology & Department of Neurological Surgery, Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana.,2 Department of Spinal Surgery and Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Wu
- 1 Department of Anatomy and Cell Biology & Department of Neurological Surgery, Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| | - Wenhui Xiong
- 1 Department of Anatomy and Cell Biology & Department of Neurological Surgery, Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| | - Zhi Chai
- 3 Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Xiao-Ming Xu
- 1 Department of Anatomy and Cell Biology & Department of Neurological Surgery, Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| | - Xiaoming Jin
- 1 Department of Anatomy and Cell Biology & Department of Neurological Surgery, Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
31
|
Dell'Anno MT, Wang X, Onorati M, Li M, Talpo F, Sekine Y, Ma S, Liu F, Cafferty WBJ, Sestan N, Strittmatter SM. Human neuroepithelial stem cell regional specificity enables spinal cord repair through a relay circuit. Nat Commun 2018; 9:3419. [PMID: 30143638 PMCID: PMC6109094 DOI: 10.1038/s41467-018-05844-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 07/23/2018] [Indexed: 01/18/2023] Open
Abstract
Traumatic spinal cord injury results in persistent disability due to disconnection of surviving neural elements. Neural stem cell transplantation has been proposed as a therapeutic option, but optimal cell type and mechanistic aspects remain poorly defined. Here, we describe robust engraftment into lesioned immunodeficient mice of human neuroepithelial stem cells derived from the developing spinal cord and maintained in self-renewing adherent conditions for long periods. Extensive elongation of both graft and host axons occurs. Improved functional recovery after transplantation depends on neural relay function through the grafted neurons, requires the matching of neural identity to the anatomical site of injury, and is accompanied by expression of specific marker proteins. Thus, human neuroepithelial stem cells may provide an anatomically specific relay function for spinal cord injury recovery. The optimal type or regional origin of stem cells for regenerative applications in the nervous system has not yet been established. Here the authors show that human neuroepithelial stem cells from the developing spinal cord, but not those from the developing cortex, show good host-graft interaction when transplanted to rodent models of spinal cord injury.
Collapse
Affiliation(s)
- Maria Teresa Dell'Anno
- Cellular Neuroscience, Neurodegeneration and Repair (CNNR) Program, Yale School of Medicine, New Haven, CT, 06536, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Xingxing Wang
- Cellular Neuroscience, Neurodegeneration and Repair (CNNR) Program, Yale School of Medicine, New Haven, CT, 06536, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Marco Onorati
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, 56127, Italy.,Department of Neuroscience, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Mingfeng Li
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Francesca Talpo
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Yuichi Sekine
- Cellular Neuroscience, Neurodegeneration and Repair (CNNR) Program, Yale School of Medicine, New Haven, CT, 06536, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Shaojie Ma
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Fuchen Liu
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, 06520, USA
| | | | - Nenad Sestan
- Cellular Neuroscience, Neurodegeneration and Repair (CNNR) Program, Yale School of Medicine, New Haven, CT, 06536, USA.,Department of Neuroscience, Yale School of Medicine, New Haven, CT, 06520, USA.,Department of Genetics, of Psychiatry and of Comparative Medicine, and Yale Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration and Repair (CNNR) Program, Yale School of Medicine, New Haven, CT, 06536, USA. .,Department of Neurology, Yale School of Medicine, New Haven, CT, 06520, USA. .,Department of Neuroscience, Yale School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
32
|
KLF6 and STAT3 co-occupy regulatory DNA and functionally synergize to promote axon growth in CNS neurons. Sci Rep 2018; 8:12565. [PMID: 30135567 PMCID: PMC6105645 DOI: 10.1038/s41598-018-31101-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 08/10/2018] [Indexed: 11/26/2022] Open
Abstract
The failure of axon regeneration in the CNS limits recovery from damage and disease. Members of the KLF family of transcription factors can exert both positive and negative effects on axon regeneration, but the underlying mechanisms are unclear. Here we show that forced expression of KLF6 promotes axon regeneration by corticospinal tract neurons in the injured spinal cord. RNA sequencing identified 454 genes whose expression changed upon forced KLF6 expression in vitro, including sub-networks that were highly enriched for functions relevant to axon extension including cytoskeleton remodeling, lipid synthesis, and bioenergetics. In addition, promoter analysis predicted a functional interaction between KLF6 and a second transcription factor, STAT3, and genome-wide footprinting using ATAC-Seq data confirmed frequent co-occupancy. Co-expression of the two factors yielded a synergistic elevation of neurite growth in vitro. These data clarify the transcriptional control of axon growth and point the way toward novel interventions to promote CNS regeneration.
Collapse
|
33
|
Kovrazhkina EA, Stakhovskaya LV, Razinskaya OD, Serdyuk AV. [Inhibitors of CNS regeneration, their physiological role and participation in pathogenesis of diseases]. Zh Nevrol Psikhiatr Im S S Korsakova 2018; 118:143-149. [PMID: 29927419 DOI: 10.17116/jnevro201811851143] [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: 11/18/2022]
Abstract
The review is devoted to axon growth inhibitors in the CNS, including a physiological role of myelin-associated proteins (Nogo-A, MAG, OMgp) and their involvement in the pathogenesis of various diseases (spinal injuries, stroke, neurodegenerations).
Collapse
Affiliation(s)
- E A Kovrazhkina
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - L V Stakhovskaya
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - O D Razinskaya
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - A V Serdyuk
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
34
|
Local BDNF Delivery to the Injured Cervical Spinal Cord using an Engineered Hydrogel Enhances Diaphragmatic Respiratory Function. J Neurosci 2018; 38:5982-5995. [PMID: 29891731 DOI: 10.1523/jneurosci.3084-17.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 05/16/2018] [Accepted: 05/17/2018] [Indexed: 02/07/2023] Open
Abstract
We developed an innovative biomaterial-based approach to repair the critical neural circuitry that controls diaphragm activation by locally delivering brain-derived neurotrophic factor (BDNF) to injured cervical spinal cord. BDNF can be used to restore respiratory function via a number of potential repair mechanisms; however, widespread BDNF biodistribution resulting from delivery methods such as systemic injection or lumbar puncture can lead to inefficient drug delivery and adverse side effects. As a viable alternative, we developed a novel hydrogel-based system loaded with polysaccharide-BDNF particles self-assembled by electrostatic interactions that can be safely implanted in the intrathecal space for achieving local BDNF delivery with controlled dosing and duration. Implantation of BDNF hydrogel after C4/C5 contusion-type spinal cord injury (SCI) in female rats robustly preserved diaphragm function, as assessed by in vivo recordings of compound muscle action potential and electromyography amplitudes. However, BDNF hydrogel did not decrease lesion size or degeneration of cervical motor neuron soma, suggesting that its therapeutic mechanism of action was not neuroprotection within spinal cord. Interestingly, BDNF hydrogel significantly preserved diaphragm innervation by phrenic motor neurons (PhMNs), as assessed by detailed neuromuscular junction morphological analysis and retrograde PhMN labeling from diaphragm using cholera toxin B. Furthermore, BDNF hydrogel enhanced the serotonergic axon innervation of PhMNs that plays an important role in modulating PhMN excitability. Our findings demonstrate that local BDNF hydrogel delivery is a robustly effective and safe strategy to restore diaphragm function after SCI. In addition, we demonstrate novel therapeutic mechanisms by which BDNF can repair respiratory neural circuitry.SIGNIFICANCE STATEMENT Respiratory compromise is a leading cause of morbidity and mortality following traumatic spinal cord injury (SCI). We used an innovative biomaterial-based drug delivery system in the form of a hydrogel that can be safely injected into the intrathecal space for achieving local delivery of brain-derived neurotrophic factor (BDNF) with controlled dosing and duration, while avoiding side effects associated with other delivery methods. In a clinically relevant rat model of cervical contusion-type SCI, BDNF hydrogel robustly and persistently improved diaphragmatic respiratory function by enhancing phrenic motor neuron (PhMN) innervation of the diaphragm neuromuscular junction and by increasing serotonergic innervation of PhMNs in ventral horn of the cervical spinal cord. These exciting findings demonstrate that local BDNF hydrogel delivery is a safe and robustly effective strategy to maintain respiratory function after cervical SCI.
Collapse
|
35
|
Wen TC, Lall S, Pagnotta C, Markward J, Gupta D, Ratnadurai-Giridharan S, Bucci J, Greenwald L, Klugman M, Hill NJ, Carmel JB. Plasticity in One Hemisphere, Control From Two: Adaptation in Descending Motor Pathways After Unilateral Corticospinal Injury in Neonatal Rats. Front Neural Circuits 2018; 12:28. [PMID: 29706871 PMCID: PMC5906589 DOI: 10.3389/fncir.2018.00028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/23/2018] [Indexed: 11/13/2022] Open
Abstract
After injury to the corticospinal tract (CST) in early development there is large-scale adaptation of descending motor pathways. Some studies suggest the uninjured hemisphere controls the impaired forelimb, while others suggest that the injured hemisphere does; these pathways have never been compared directly. We tested the contribution of each motor cortex to the recovery forelimb function after neonatal injury of the CST. We cut the left pyramid (pyramidotomy) of postnatal day 7 rats, which caused a measurable impairment of the right forelimb. We used pharmacological inactivation of each motor cortex to test its contribution to a skilled reach and supination task. Rats with neonatal pyramidotomy were further impaired by inactivation of motor cortex in both the injured and the uninjured hemispheres, while the forelimb of uninjured rats was impaired only from the contralateral motor cortex. Thus, inactivation demonstrated motor control from each motor cortex. In contrast, physiological and anatomical interrogation of these pathways support adaptations only in the uninjured hemisphere. Intracortical microstimulation of motor cortex in the uninjured hemisphere of rats with neonatal pyramidotomy produced responses from both forelimbs, while stimulation of the injured hemisphere did not elicit responses from either forelimb. Both anterograde and retrograde tracers were used to label corticofugal pathways. There was no increased plasticity from the injured hemisphere, either from cortex to the red nucleus or the red nucleus to the spinal cord. In contrast, there were very strong CST connections to both halves of the spinal cord from the uninjured motor cortex. Retrograde tracing produced maps of each forelimb within the uninjured hemisphere, and these were partly segregated. This suggests that the uninjured hemisphere may encode separate control of the unimpaired and the impaired forelimbs of rats with neonatal pyramidotomy.
Collapse
Affiliation(s)
- Tong-Chun Wen
- Motor Recovery Laboratory, Burke-Cornell Medical Research Institute, White Plains, NY, United States
| | - Sophia Lall
- Motor Recovery Laboratory, Burke-Cornell Medical Research Institute, White Plains, NY, United States
| | - Corey Pagnotta
- Motor Recovery Laboratory, Burke-Cornell Medical Research Institute, White Plains, NY, United States
| | - James Markward
- Motor Recovery Laboratory, Burke-Cornell Medical Research Institute, White Plains, NY, United States
| | - Disha Gupta
- Motor Recovery Laboratory, Burke-Cornell Medical Research Institute, White Plains, NY, United States
| | | | - Jacqueline Bucci
- Motor Recovery Laboratory, Burke-Cornell Medical Research Institute, White Plains, NY, United States
| | - Lucy Greenwald
- Motor Recovery Laboratory, Burke-Cornell Medical Research Institute, White Plains, NY, United States
| | - Madelyn Klugman
- Motor Recovery Laboratory, Burke-Cornell Medical Research Institute, White Plains, NY, United States
| | - N Jeremy Hill
- Motor Recovery Laboratory, Burke-Cornell Medical Research Institute, White Plains, NY, United States
| | - Jason B Carmel
- Motor Recovery Laboratory, Burke-Cornell Medical Research Institute, White Plains, NY, United States.,Departments of Neurology and Pediatrics, Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, United States
| |
Collapse
|
36
|
Sekine Y, Siegel CS, Sekine-Konno T, Cafferty WBJ, Strittmatter SM. The nociceptin receptor inhibits axonal regeneration and recovery from spinal cord injury. Sci Signal 2018; 11:eaao4180. [PMID: 29615517 PMCID: PMC6179440 DOI: 10.1126/scisignal.aao4180] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Axonal growth after traumatic spinal cord injury is limited by endogenous inhibitors, selective blockade of which promotes partial neurological recovery. The partial repair phenotypes suggest that compensatory pathways limit improvement. Gene expression profiles of mice deficient in Ngr1, which encodes a receptor for myelin-associated inhibitors of axonal regeneration such as Nogo, revealed that trauma increased the mRNA expression of ORL1, which encodes the receptor for the opioid-related peptide nociceptin. Endogenous and overexpressed ORL1 coimmunoprecipitated with immature NgR1 protein, and ORL1 enhanced the O-linked glycosylation and surface expression of NgR1 in HEK293T and Neuro2A cells and primary neurons. ORL1 overexpression inhibited cortical neuron axon regeneration independently of NgR1. Furthermore, regeneration was inhibited by an ORL1 agonist and enhanced by the ORL1 antagonist J113397 through a ROCK-dependent mechanism. Mice treated with J113397 after dorsal hemisection of the mid-thoracic spinal cord recovered greater locomotor function and exhibited lumbar raphespinal axon sprouting. These effects were further enhanced by combined Ngr1 deletion and ORL1 inhibition. Thus, ORL1 limits neural repair directly and indirectly by enhancing NgR1 maturation, and ORL1 antagonists enhance recovery from traumatic CNS injuries in wild-type and Ngr1 null mice.
Collapse
Affiliation(s)
- Yuichi Sekine
- Cellular Neuroscience, Neurodegeneration and Repair Program, Interdepartmental Neuroscience Program, Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Chad S Siegel
- Cellular Neuroscience, Neurodegeneration and Repair Program, Interdepartmental Neuroscience Program, Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Tomoko Sekine-Konno
- Cellular Neuroscience, Neurodegeneration and Repair Program, Interdepartmental Neuroscience Program, Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - William B J Cafferty
- Cellular Neuroscience, Neurodegeneration and Repair Program, Interdepartmental Neuroscience Program, Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration and Repair Program, Interdepartmental Neuroscience Program, Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA.
| |
Collapse
|
37
|
Cortico-reticulo-spinal circuit reorganization enables functional recovery after severe spinal cord contusion. Nat Neurosci 2018; 21:576-588. [PMID: 29556028 DOI: 10.1038/s41593-018-0093-5] [Citation(s) in RCA: 208] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 01/15/2018] [Indexed: 12/24/2022]
Abstract
Severe spinal cord contusions interrupt nearly all brain projections to lumbar circuits producing leg movement. Failure of these projections to reorganize leads to permanent paralysis. Here we modeled these injuries in rodents. A severe contusion abolished all motor cortex projections below injury. However, the motor cortex immediately regained adaptive control over the paralyzed legs during electrochemical neuromodulation of lumbar circuits. Glutamatergic reticulospinal neurons with residual projections below the injury relayed the cortical command downstream. Gravity-assisted rehabilitation enabled by the neuromodulation therapy reinforced these reticulospinal projections, rerouting cortical information through this pathway. This circuit reorganization mediated a motor cortex-dependent recovery of natural walking and swimming without requiring neuromodulation. Cortico-reticulo-spinal circuit reorganization may also improve recovery in humans.
Collapse
|
38
|
Richter M, Negro-Demontel ML, Blanco-Ocampo D, Taranto E, Lago N, Peluffo H. Thy1-YFP-H Mice and the Parallel Rod Floor Test to Evaluate Short- and Long-Term Progression of Traumatic Brain Injury. ACTA ACUST UNITED AC 2018; 120:24.1.1-24.1.25. [PMID: 29512144 DOI: 10.1002/cpim.42] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability and is a risk factor for the later development of neuropsychiatric disorders and neurodegenerative diseases. Many models of TBI have been developed, but their further refinement and a more detailed long-term follow-up is needed. We have used the Thy1-YFP-H transgenic mouse line and the parallel rod floor test to produce an unbiased and robust method for the evaluation of the multiple effects of a validated model of controlled cortical injury. This approach reveals short- and long-term progressive changes, including compromised biphasic motor function up to 85 days post-lesion, which correlates with neuronal atrophy, dendrite and spine loss, and long-term axonal pathology evidenced by axon spheroids and fragmentation. Here we present methods for inducing a controlled cortical injury in the Thy1-YFP-H transgenic mouse line and for evaluating the resulting deficits in the parallel rod floor test. This technique constitutes a new, unbiased, and robust method for the evaluation of motor and behavioral alterations after TBI. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Monique Richter
- Neurodegeneration Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Current Address: Roche Diagnostics GmbH, Penzberg, Germany
| | - María Luciana Negro-Demontel
- Neuroinflammation and Gene Therapy Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Department of Histology and Embryology, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Daniela Blanco-Ocampo
- Department of Physiopathology, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Eliseo Taranto
- Department of Physiopathology, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Natalia Lago
- Neuroinflammation and Gene Therapy Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Hugo Peluffo
- Neuroinflammation and Gene Therapy Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Department of Histology and Embryology, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
39
|
Identification of Intrinsic Axon Growth Modulators for Intact CNS Neurons after Injury. Cell Rep 2017; 18:2687-2701. [PMID: 28297672 PMCID: PMC5389739 DOI: 10.1016/j.celrep.2017.02.058] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 12/19/2016] [Accepted: 02/16/2017] [Indexed: 12/28/2022] Open
Abstract
Functional deficits persist after spinal cord injury (SCI) because axons in the adult mammalian central nervous system (CNS) fail to regenerate. However, modest levels of spontaneous functional recovery are typically observed after trauma and are thought to be mediated by the plasticity of intact circuitry. The mechanisms underlying intact circuit plasticity are not delineated. Here, we characterize the in vivo transcriptome of sprouting intact neurons from Ngr1 null mice after partial SCI. We identify the lysophosphatidic acid signaling modulators LPPR1 and LPAR1 as intrinsic axon growth modulators for intact corticospinal motor neurons after adjacent injury. Furthermore, in vivo LPAR1 inhibition or LPPR1 overexpression enhances sprouting of intact corticospinal tract axons and yields greater functional recovery after unilateral brainstem lesion in wild-type mice. Thus, the transcriptional profile of injury-induced sprouting of intact neurons reveals targets for therapeutic enhancement of axon growth initiation and new synapse formation.
Collapse
|
40
|
Okabe N, Himi N, Maruyama-Nakamura E, Hayashi N, Narita K, Miyamoto O. Rehabilitative skilled forelimb training enhances axonal remodeling in the corticospinal pathway but not the brainstem-spinal pathways after photothrombotic stroke in the primary motor cortex. PLoS One 2017; 12:e0187413. [PMID: 29095902 PMCID: PMC5667818 DOI: 10.1371/journal.pone.0187413] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/19/2017] [Indexed: 12/17/2022] Open
Abstract
Task-specific rehabilitative training is commonly used for chronic stroke patients. Axonal remodeling is believed to be one mechanism underlying rehabilitation-induced functional recovery, and significant roles of the corticospinal pathway have previously been demonstrated. Brainstem-spinal pathways, as well as the corticospinal tract, have been suggested to contribute to skilled motor function and functional recovery after brain injury. However, whether axonal remodeling in the brainstem-spinal pathways is a critical component for rehabilitation-induced functional recovery is not known. In this study, rats were subjected to photothrombotic stroke in the caudal forelimb area of the primary motor cortex and received rehabilitative training with a skilled forelimb reaching task for 4 weeks. After completion of the rehabilitative training, the retrograde tracer Fast blue was injected into the contralesional lower cervical spinal cord. Fast blue-positive cells were counted in 32 brain areas located in the cerebral cortex, hypothalamus, midbrain, pons, and medulla oblongata. Rehabilitative training improved motor performance in the skilled forelimb reaching task but not in the cylinder test, ladder walk test, or staircase test, indicating that rehabilitative skilled forelimb training induced task-specific recovery. In the histological analysis, rehabilitative training significantly increased the number of Fast blue-positive neurons in the ipsilesional rostral forelimb area and secondary sensory cortex. However, rehabilitative training did not alter the number of Fast blue-positive neurons in any areas of the brainstem. These results indicate that rehabilitative skilled forelimb training enhances axonal remodeling selectively in the corticospinal pathway, which suggests a critical role of cortical plasticity, rather than brainstem plasticity, in task-specific recovery after subtotal motor cortex destruction.
Collapse
Affiliation(s)
- Naohiko Okabe
- Second Department of Physiology, Kawasaki Medical School, Matsushima, Kurashiki City, Okayama, Japan
- * E-mail:
| | - Naoyuki Himi
- Second Department of Physiology, Kawasaki Medical School, Matsushima, Kurashiki City, Okayama, Japan
| | - Emi Maruyama-Nakamura
- Second Department of Physiology, Kawasaki Medical School, Matsushima, Kurashiki City, Okayama, Japan
| | - Norito Hayashi
- Second Department of Physiology, Kawasaki Medical School, Matsushima, Kurashiki City, Okayama, Japan
| | - Kazuhiko Narita
- Second Department of Physiology, Kawasaki Medical School, Matsushima, Kurashiki City, Okayama, Japan
| | - Osamu Miyamoto
- Second Department of Physiology, Kawasaki Medical School, Matsushima, Kurashiki City, Okayama, Japan
| |
Collapse
|
41
|
Optogenetic Interrogation of Functional Synapse Formation by Corticospinal Tract Axons in the Injured Spinal Cord. J Neurosci 2017; 36:5877-90. [PMID: 27225775 DOI: 10.1523/jneurosci.4203-15.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 04/08/2016] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED To restore function after injury to the CNS, axons must be stimulated to extend into denervated territory and, critically, must form functional synapses with appropriate targets. We showed previously that forced overexpression of the transcription factor Sox11 increases axon growth by corticospinal tract (CST) neurons after spinal injury. However, behavioral outcomes were not improved, raising the question of whether the newly sprouted axons are able to form functional synapses. Here we developed an optogenetic strategy, paired with single-unit extracellular recordings, to assess the ability of Sox11-stimulated CST axons to functionally integrate in the circuitry of the cervical spinal cord. Initial time course experiments established the expression and function of virally expressed Channelrhodopsin (ChR2) in CST cell bodies and in axon terminals in cervical spinal cord. Pyramidotomies were performed in adult mice to deprive the left side of the spinal cord of CST input, and the right CST was treated with adeno-associated virus (AAV)-Sox11 or AAV-EBFP control, along with AAV-ChR2. As expected, Sox11 treatment caused robust midline crossing of CST axons into previously denervated left spinal cord. Clear postsynaptic responses resulted from optogenetic activation of CST terminals, demonstrating the ability of Sox11-stimulated axons to form functional synapses. Mapping of the distribution of CST-evoked spinal activity revealed overall similarity between intact and newly innervated spinal tissue. These data demonstrate the formation of functional synapses by Sox11-stimulated CST axons without significant behavioral benefit, suggesting that new synapses may be mistargeted or otherwise impaired in the ability to coordinate functional output. SIGNIFICANCE STATEMENT As continued progress is made in promoting the regeneration of CNS axons, questions of synaptic integration are increasingly prominent. Demonstrating direct synaptic integration by regenerated axons and distinguishing its function from indirect relay circuits and target field plasticity have presented technical challenges. Here we force the overexpression of Sox11 to stimulate the growth of corticospinal tract axons in the cervical spinal cord and then use specific optogenetic activation to assess their ability to directly drive postsynaptic activity in spinal cord neurons. By confirming successful synaptic integration, these data illustrate a novel optogenetic-based strategy to monitor and optimize functional reconnection by newly sprouted axons in the injured CNS.
Collapse
|
42
|
Mosberger AC, Miehlbradt JC, Bjelopoljak N, Schneider MP, Wahl AS, Ineichen BV, Gullo M, Schwab ME. Axotomized Corticospinal Neurons Increase Supra-Lesional Innervation and Remain Crucial for Skilled Reaching after Bilateral Pyramidotomy. Cereb Cortex 2017; 28:625-643. [DOI: 10.1093/cercor/bhw405] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 12/20/2016] [Indexed: 12/11/2022] Open
|
43
|
Dell'Anno MT, Strittmatter SM. Rewiring the spinal cord: Direct and indirect strategies. Neurosci Lett 2016; 652:25-34. [PMID: 28007647 DOI: 10.1016/j.neulet.2016.12.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/15/2016] [Accepted: 12/02/2016] [Indexed: 12/23/2022]
Abstract
Spinal cord injury is currently incurable. Treatment is limited to minimizing secondary complications and maximizing residual function by rehabilitation. Neurologic recovery is prevented by the poor intrinsic regenerative capacity of neurons in the adult central nervous system and by the presence of growth inhibitors in the adult brain and spinal cord. Here we identify three approaches to rewire the spinal cord after injury: axonal regeneration (direct endogenous reconnection), axonal sprouting (indirect endogenous reconnection) and neural stem cell transplantation (indirect exogenous reconnection). Regeneration and sprouting of axonal fibers can be both enhanced through the neutralization of myelin- and extracellular matrix-associated inhibitors described in the first part of this review. Alternatively, in the second part we focus on the formation of a novel circuit through the grafting of neural stem cells in the lesion site. Transplanted neural stem cells differentiate in vivo into neurons and glial cells which form an intermediate station between the rostral and caudal segment of the recipient spinal cord. In particular, here we describe how neural stem cells-derived neurons are endowed with the ability to extend long-distance axons to regain the transmission of motor and sensory information.
Collapse
Affiliation(s)
- Maria Teresa Dell'Anno
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Stephen M Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT 06536, USA.
| |
Collapse
|
44
|
Serradj N, Agger SF, Hollis ER. Corticospinal circuit plasticity in motor rehabilitation from spinal cord injury. Neurosci Lett 2016; 652:94-104. [PMID: 27939980 DOI: 10.1016/j.neulet.2016.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/29/2016] [Accepted: 12/02/2016] [Indexed: 11/18/2022]
Abstract
Restoring corticospinal function after spinal cord injury is a significant challenge as the corticospinal tract elicits no substantive, spontaneous regeneration, and its interruption leaves a permanent deficit. The corticospinal circuit serves multiple motor and sensory functions within the mammalian nervous system as the direct link between isocortex and spinal cord. Maturation of the corticospinal circuit involves the refinement of projections within the spinal cord and a subsequent refinement of motor maps within the cortex. The plasticity of these cortical motor maps mirrors the acquisition of skilled motor learning, and both the maps and motor skills are disrupted following injury to the corticospinal tract. The motor cortex exhibits the capacity to incorporate changes in corticospinal projections induced by both spontaneous and therapeutic-mediated plasticity of corticospinal axons through appropriate rehabilitation. An understanding of the mechanisms of corticospinal plasticity in motor learning will undoubtedly help inform strategies to improve motor rehabilitation after spinal cord injury.
Collapse
Affiliation(s)
- Najet Serradj
- Burke Medical Research Institute, White Plains, New York, NY 10605, United States
| | - Sydney F Agger
- Burke Medical Research Institute, White Plains, New York, NY 10605, United States
| | - Edmund R Hollis
- Burke Medical Research Institute, White Plains, New York, NY 10605, United States; Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, United States.
| |
Collapse
|
45
|
Re-Establishment of Cortical Motor Output Maps and Spontaneous Functional Recovery via Spared Dorsolaterally Projecting Corticospinal Neurons after Dorsal Column Spinal Cord Injury in Adult Mice. J Neurosci 2016; 36:4080-92. [PMID: 27053214 DOI: 10.1523/jneurosci.3386-15.2016] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 02/25/2016] [Indexed: 01/08/2023] Open
Abstract
UNLABELLED Motor cortical plasticity contributes to spontaneous recovery after incomplete spinal cord injury (SCI), but the pathways underlying this remain poorly understood. We performed optogenetic mapping of motor cortex in channelrhodopsin-2 expressing mice to assess the capacity of the cortex to re-establish motor output longitudinally after a C3/C4 dorsal column SCI that bilaterally ablated the dorsal corticospinal tract (CST) containing ∼96% of corticospinal fibers but spared ∼3% of CST fibers that project via the dorsolateral funiculus. Optogenetic mapping revealed extensive early deficits, but eventual reestablishment of motor cortical output maps to the limbs at the same latency as preoperatively by 4 weeks after injury. Analysis of skilled locomotion on the horizontal ladder revealed early deficits followed by partial spontaneous recovery by 6 weeks after injury. To dissociate between the contributions of injured dorsal projecting versus spared dorsolateral projecting corticospinal neurons, we established a transient silencing approach to inactivate spared dorsolaterally projecting corticospinal neurons specifically by injecting adeno-associated virus (AAV)-expressing Cre-dependent DREADD (designer receptor exclusively activated by designer drug) receptor hM4Di in sensorimotor cortex and AAV-expressing Cre in C7/C8 dorsolateral funiculus. Transient silencing uninjured dorsolaterally projecting corticospinal neurons via activation of the inhibitory DREADD receptor hM4Di abrogated spontaneous recovery and resulted in a greater change in skilled locomotion than in control uninjured mice using the same silencing approach. These data demonstrate the pivotal role of a minor dorsolateral corticospinal pathway in mediating spontaneous recovery after SCI and support a focus on spared corticospinal neurons as a target for therapy. SIGNIFICANCE STATEMENT Spontaneous recovery can occur after incomplete spinal cord injury (SCI), but the pathways underlying this remain poorly understood. We performed optogenetic mapping of motor cortex after a cervical SCI that interrupts most corticospinal transmission but results in partial recovery on a horizontal ladder task of sensorimotor function. We demonstrate that the motor cortex can reestablish output to the limbs longitudinally. To dissociate the roles of injured and uninjured corticospinal neurons in mediating recovery, we transiently silenced the minor dorsolateral corticospinal pathway spared by our injury. This abrogated spontaneous recovery and resulted in a greater change in skilled locomotion than in uninjured mice using the same approach. Therefore, uninjured corticospinal neurons substantiate remarkable motor cortical plasticity and partial recovery after SCI.
Collapse
|
46
|
Causal Link between the Cortico-Rubral Pathway and Functional Recovery through Forced Impaired Limb Use in Rats with Stroke. J Neurosci 2016; 36:455-67. [PMID: 26758837 DOI: 10.1523/jneurosci.2399-15.2016] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED Intensive rehabilitation is believed to induce use-dependent plasticity in the injured nervous system; however, its causal relationship to functional recovery is unclear. Here, we performed systematic analysis of the effects of forced use of an impaired forelimb on the recovery of rats after lesioning the internal capsule with intracerebral hemorrhage (ICH). Forced limb use (FLU) group rats exhibited better recovery of skilled forelimb functions and their cortical motor area with forelimb representation was restored and enlarged on the ipsilesional side. In addition, abundant axonal sprouting from the reemerged forelimb area was found in the ipsilateral red nucleus after FLU. To test the causal relationship between the plasticity in the cortico-rubral pathway and recovery, loss-of-function experiments were conducted using a double-viral vector technique, which induces selective blockade of the target pathway. Blockade of the cortico-rubral tract resulted in deficits of the recovered forelimb function in FLU group rats. These findings suggest that the cortico-rubral pathway is a substrate for recovery induced by intensive rehabilitation after ICH. SIGNIFICANCE STATEMENT The research aimed at determining the causal linkage between reorganization of the motor pathway induced by intensive rehabilitative training and recovery after stroke. We clarified the expansion of the forelimb representation area of the ipsilesional motor cortex by forced impaired forelimb use (FLU) after lesioning the internal capsule with intracerebral hemorrhaging (ICH) in rats. Anterograde tracing showed robust axonal sprouting from the forelimb area to the red nucleus in response to FLU. Selective blockade of the cortico-rubral pathway by the novel double-viral vector technique clearly revealed that the increased cortico-rubral axonal projections had causal linkage to the recovery of reaching movements induced by FLU. Our data demonstrate that the cortico-rubral pathway is responsible for the effect of intensive limb use.
Collapse
|
47
|
Fink KL, Cafferty WBJ. Reorganization of Intact Descending Motor Circuits to Replace Lost Connections After Injury. Neurotherapeutics 2016; 13:370-81. [PMID: 26846379 PMCID: PMC4824020 DOI: 10.1007/s13311-016-0422-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Neurons have a limited capacity to regenerate in the adult central nervous system (CNS). The inability of damaged axons to re-establish original circuits results in permanent functional impairment after spinal cord injury (SCI). Despite abortive regeneration of axotomized CNS neurons, limited spontaneous recovery of motor function emerges after partial SCI in humans and experimental rodent models of SCI. It is hypothesized that this spontaneous functional recovery is the result of the reorganization of descending motor pathways spared by the injury, suggesting that plasticity of intact circuits is a potent alternative conduit to enhance functional recovery after SCI. In support of this hypothesis, several studies have shown that after unilateral corticospinal tract (CST) lesion (unilateral pyramidotomy), the intact CST functionally sprouts into the denervated side of the spinal cord. Furthermore, pharmacologic and genetic methods that enhance the intrinsic growth capacity of adult neurons or block extracellular growth inhibitors are effective at significantly enhancing intact CST reorganization and recovery of motor function. Owing to its importance in controlling fine motor behavior in primates, the CST is the most widely studied descending motor pathway; however, additional studies in rodents have shown that plasticity within other spared descending motor pathways, including the rubrospinal tract, raphespinal tract, and reticulospinal tract, can also result in restoration of function after incomplete SCI. Identifying the molecular mechanisms that drive plasticity within intact circuits is crucial in developing novel, potent, and specific therapeutics to restore function after SCI. In this review we discuss the evidence supporting a focus on exploring the capacity of intact motor circuits to functionally repair the damaged CNS after SCI.
Collapse
Affiliation(s)
- Kathren L Fink
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - William B J Cafferty
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
48
|
Ganzer PD, Meyers EC, Sloan AM, Maliakkal R, Ruiz A, Kilgard MP, Robert LR. Awake behaving electrophysiological correlates of forelimb hyperreflexia, weakness and disrupted muscular synchronization following cervical spinal cord injury in the rat. Behav Brain Res 2016; 307:100-11. [PMID: 27033345 DOI: 10.1016/j.bbr.2016.03.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/22/2016] [Accepted: 03/26/2016] [Indexed: 01/22/2023]
Abstract
Spinal cord injury usually occurs at the level of the cervical spine and results in profound impairment of forelimb function. In this study, we recorded awake behaving intramuscular electromyography (EMG) from the biceps and triceps muscles of the impaired forelimb during volitional and reflexive forelimb movements before and after unilateral cervical spinal cord injury (cSCI) in rats. C5/C6 hemicontusion reduced volitional forelimb strength by more than 50% despite weekly rehabilitation for one month post-injury. Triceps EMG during volitional strength assessment was reduced by more than 60% following injury, indicating reduced descending drive. Biceps EMG during reflexive withdrawal from a thermal stimulus was increased by 500% following injury, indicating flexor withdrawal hyperreflexia. The reduction in volitional forelimb strength was significantly correlated with volitional and reflexive biceps EMG activity. Our results support the hypothesis that biceps hyperreflexia and descending volitional drive both significantly contribute to forelimb strength deficits after cSCI and provide new insight into dynamic muscular dysfunction after cSCI. The use of multiple automated quantitative measures of forelimb dysfunction in the rodent cSCI model will likely aid the search for effective regenerative, pharmacological, and neuroprosthetic treatments for spinal cord injury.
Collapse
Affiliation(s)
- Patrick Daniel Ganzer
- The University of Texas at Dallas, Texas Biomedical Device Center, 800 West Campbell Road, Richardson, TX 75080, United States; The University of Texas at Dallas, Erik Jonsson School of Engineering and Computer Science, 800 West Campbell Road, Richardson, TX 75080, United States.
| | - Eric Christopher Meyers
- The University of Texas at Dallas, Texas Biomedical Device Center, 800 West Campbell Road, Richardson, TX 75080, United States; The University of Texas at Dallas, Erik Jonsson School of Engineering and Computer Science, 800 West Campbell Road, Richardson, TX 75080, United States.
| | - Andrew Michael Sloan
- The University of Texas at Dallas, Texas Biomedical Device Center, 800 West Campbell Road, Richardson, TX 75080, United States; The University of Texas at Dallas, Erik Jonsson School of Engineering and Computer Science, 800 West Campbell Road, Richardson, TX 75080, United States.
| | - Reshma Maliakkal
- The University of Texas at Dallas, School of Behavioral Brain Sciences, 800 West Campbell Road, GR41, Richardson, TX 75080, United States.
| | - Andrea Ruiz
- The University of Texas at Dallas, Texas Biomedical Device Center, 800 West Campbell Road, Richardson, TX 75080, United States; The University of Texas at Dallas, School of Behavioral Brain Sciences, 800 West Campbell Road, GR41, Richardson, TX 75080, United States.
| | - Michael Paul Kilgard
- The University of Texas at Dallas, Texas Biomedical Device Center, 800 West Campbell Road, Richardson, TX 75080, United States; The University of Texas at Dallas, School of Behavioral Brain Sciences, 800 West Campbell Road, GR41, Richardson, TX 75080, United States.
| | - LeMoine Rennaker Robert
- The University of Texas at Dallas, Texas Biomedical Device Center, 800 West Campbell Road, Richardson, TX 75080, United States; The University of Texas at Dallas, School of Behavioral Brain Sciences, 800 West Campbell Road, GR41, Richardson, TX 75080, United States; The University of Texas at Dallas, Erik Jonsson School of Engineering and Computer Science, 800 West Campbell Road, Richardson, TX 75080, United States.
| |
Collapse
|
49
|
López AJ, Kramár E, Matheos DP, White AO, Kwapis J, Vogel-Ciernia A, Sakata K, Espinoza M, Wood MA. Promoter-Specific Effects of DREADD Modulation on Hippocampal Synaptic Plasticity and Memory Formation. J Neurosci 2016; 36:3588-99. [PMID: 27013687 PMCID: PMC4804014 DOI: 10.1523/jneurosci.3682-15.2016] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 01/20/2016] [Accepted: 02/15/2016] [Indexed: 11/21/2022] Open
Abstract
Designer receptors exclusively activated by designer drug (DREADDs) are a novel tool with the potential to bidirectionally drive cellular, circuit, and ultimately, behavioral changes. We used DREADDs to evaluate memory formation in a hippocampus-dependent task in mice and effects on synaptic physiology in the dorsal hippocampus. We expressed neuron-specific (hSyn promoter) DREADDs that were either excitatory (HM3D) or inhibitory (HM4D) in the dorsal hippocampus. As predicted, hSyn-HM3D was able to transform a subthreshold learning event into long-term memory (LTM), and hSyn-HM4D completely impaired LTM formation. Surprisingly, the opposite was observed during experiments examining the effects on hippocampal long-term potentiation (LTP). hSyn-HM3D impaired LTP and hSyn-HM4D facilitated LTP. Follow-up experiments indicated that the hSyn-HM3D-mediated depression of fEPSP appears to be driven by presynaptic activation of inhibitory currents, whereas the hSyn-HM4D-mediated increase of fEPSP is induced by a reduction in GABAA receptor function. To determine whether these observations were promoter specific, we next examined the effects of using the CaMKIIα promoter that limits expression to forebrain excitatory neurons. CaMKIIα-HM3D in the dorsal hippocampus led to the transformation of a subthreshold learning event into LTM, whereas CaMKIIα-HM4D blocked LTM formation. Consistent with these findings, baseline synaptic transmission and LTP was increased in CaMKIIα-HM3D hippocampal slices, whereas slices from CaMKIIα-HM4D mice produced expected decreases in baseline synaptic transmission and LTP. Together, these experiments further demonstrate DREADDs as being a robust and reliable means of modulating neuronal function to manipulate long-term changes in behavior, while providing evidence for specific dissociations between LTM and LTP. SIGNIFICANCE STATEMENT This study evaluates the efficacy of designer receptors exclusively activated by designer drug (DREADDs) as a means of bidirectionally modulating the hippocampus in not only a hippocampus-dependent task but also in hippocampal synaptic plasticity. This is the first study to evaluate the effects of DREADD-mediated inhibition and excitation in hippocampal long-term potentiation. More specifically, this study evaluates the effect of promoter-specific expression of DREADD viruses in a heterogenic cell population, which revealed surprising effects of different promoters. With chemogenetics becoming a more ubiquitous tool throughout studies investigating circuit-specific function, these data are of broad interest to the neuroscientific community because we have shown that promoter-specific effects can drastically alter synaptic function within a specific region, without parallel changes at the level of behavior.
Collapse
Affiliation(s)
- Alberto J López
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697, UC Irvine Center for Addiction Neuroscience, and
| | - Enikö Kramár
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697, UC Irvine Center for Addiction Neuroscience, and
| | - Dina P Matheos
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697, UC Irvine Center for Addiction Neuroscience, and
| | - André O White
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697, UC Irvine Center for Addiction Neuroscience, and
| | - Janine Kwapis
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697, UC Irvine Center for Addiction Neuroscience, and
| | - Annie Vogel-Ciernia
- Davis M.I.N.D. Institute, University of California, Davis, Davis, California 96516
| | - Keith Sakata
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697, UC Irvine Center for Addiction Neuroscience, and
| | - Monica Espinoza
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697, UC Irvine Center for Addiction Neuroscience, and
| | - Marcelo A Wood
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697, UC Irvine Center for Addiction Neuroscience, and
| |
Collapse
|
50
|
Comprehensive Corticospinal Labeling with mu-crystallin Transgene Reveals Axon Regeneration after Spinal Cord Trauma in ngr1-/- Mice. J Neurosci 2016; 35:15403-18. [PMID: 26586827 DOI: 10.1523/jneurosci.3165-15.2015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Spinal cord injury interrupts descending motor tracts and creates persistent functional deficits due to the absence of spontaneous axon regeneration. Of descending pathways, the corticospinal tract (CST) is thought to be the most critical for voluntary function in primates. Even with multiple tracer injections and genetic tools, the CST is visualized to only a minor degree in experimental studies. Here, we identify and validate the mu-crystallin (crym) gene as a high-fidelity marker of the CST. In transgenic mice expressing green fluorescent protein (GFP) under crym regulatory elements (crym-GFP), comprehensive and near complete CST labeling is achieved throughout the spinal cord. Bilateral pyramidotomy eliminated the 17,000 GFP-positive CST axons that were reproducibly labeled in brainstem from the spinal cord. We show that CST tracing with crym-GFP is 10-fold more efficient than tracing with biotinylated dextran amine (BDA). Using crym-GFP, we reevaluated the CST in mice lacking nogo receptor 1 (NgR1), a protein implicated in limiting neural repair. The number and trajectory of CST axons in ngr1(-/-) mice without injury was indistinguishable from ngr1(+/+) mice. After dorsal hemisection in the midthoracic cord, CST axons did not significantly regenerate in ngr1(+/+) mice, but an average of 162 of the 6000 labeled thoracic CST axons (2.68%) regenerated >100 μm past the lesion site in crym-GFP ngr1(-/-) mice. Although traditional BDA tracing cannot reliably visualize regenerating ngr1(-/-) CST axons, their regenerative course is clear with crym-GFP. Therefore the crym-GFP transgenic mouse is a useful tool for studies of CST anatomy in experimental studies of motor pathways. SIGNIFICANCE STATEMENT Axon regeneration fails in the adult CNS, resulting in permanent functional deficits. Traditionally, inefficient extrinsic tracers such a biotinylated dextran amine (BDA) are used to label regenerating fibers after therapeutic intervention. We introduce crym-green fluorescent protein (GFP) transgenic mice as a comprehensive and specific tool with which to study the primary descending motor tract, the corticospinal tract (CST). CST labeling with crym-GFP is 10 times more efficient compared with BDA. The enhanced sensitivity afforded by crym-GFP revealed significant CST regeneration in NgR1 knock-out mice. Therefore, crym-GFP can be used as a standardized tool for future CST spinal cord injury studies.
Collapse
|