1
|
Sun X, Li L, Huang L, Li Y, Wang L, Wei Q. Harnessing spinal circuit reorganization for targeted functional recovery after spinal cord injury. Neurobiol Dis 2025; 207:106854. [PMID: 40010611 DOI: 10.1016/j.nbd.2025.106854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/13/2025] [Accepted: 02/23/2025] [Indexed: 02/28/2025] Open
Abstract
Spinal cord injury (SCI) disrupts the communication between the brain and spinal cord, resulting in the loss of motor function below the injury site. However, spontaneous structural and functional plasticity occurs in neural circuits after SCI, with unaffected synaptic inputs forming new connections and detour pathways to support recovery. The review discusses various mechanisms of circuit reorganization post-SCI, including supraspinal pathways, spinal interneurons, and spinal central pattern generators. Functional recovery may rely on maintaining a balance between excitatory and inhibitory neural activity, as well as enhancing proprioceptive input, which plays a key role in limb stability. The review emphasizes the importance of endogenous neuronal regeneration, neuromodulation therapies (such as electrical stimulation) and proprioception in SCI treatment. Future research should integrate advanced technologies such as gene targeting, imaging, and single-cell mapping to better understand the mechanisms underpinning SCI recovery, aiming to identify key neuronal subpopulations for targeted reconstruction and enhanced functional recovery. By harnessing spinal circuit reorganization, these efforts hold the potential to pave the way for more precise and effective strategies for functional recovery after SCI.
Collapse
Affiliation(s)
- Xin Sun
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Lijuan Li
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Liyi Huang
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Yangan Li
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Lu Wang
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Quan Wei
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China.
| |
Collapse
|
2
|
Carney BN, Illiano P, Pohl TM, Desu HL, Mudalegundi S, Asencor AI, Jwala S, Ascona MC, Singh PK, Titus DJ, Pazarlar BA, Wang L, Bianchi L, Mikkelsen JD, Atkins CM, Lambertsen KL, Brambilla R. Astroglial TNFR2 signaling regulates hippocampal synaptic function and plasticity in a sex dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.13.643110. [PMID: 40161622 PMCID: PMC11952524 DOI: 10.1101/2025.03.13.643110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Astrocytes participate in synaptic transmission and plasticity through tightly regulated, bidirectional communication with pre- and post-synaptic neurons, as well as microglia and oligodendrocytes. A key component of astrocyte-mediated synaptic regulation is the cytokine tumor necrosis factor (TNF). TNF signals via two cognate receptors, TNFR1 and TNFR2, both expressed in astrocytes. While TNFR1 signaling in astrocytes has been long demonstrated to be necessary for physiological synaptic function, the role of astroglial TNFR2 has never been explored. Here, we demonstrate that astroglial TNFR2 is essential for maintaining hippocampal synaptic function and plasticity in physiological conditions. Indeed, Gfap creERT2 :Tnfrsf1b fl/fl mice with selective ablation of TNFR2 in astrocytes exhibited dysregulated expression of neuronal and glial proteins (e.g., SNARE complex molecules, glutamate receptor subunits, glutamate transporters) essential for hippocampal synaptic transmission and plasticity. Hippocampal astrocytes sorted from Gfap creERT2 :Tnfrsf1b fl/fl mice displayed downregulation of genes and pathways implicated in synaptic plasticity, as well as astrocyte-neuron and astrocyte-oligodendrocyte communication. These alterations were accompanied by increased glial reactivity and impaired astrocyte calcium dynamics, and ultimately translated into functional deficits, specifically impaired long-term potentiation (LTP) and cognitive functions. Notably, male Gfap creERT2 :Tnfrsf1b fl/fl mice exhibited more pronounced hippocampal synaptic and cellular alterations, suggesting sex-dependent differences in astroglial TNFR2 regulation of synaptic function. Together, these findings indicate that TNFR2 signaling in astrocytes is essential for proper astrocyte-neuron communication at the basis of synaptic function, and that this is regulated in a sex-dependent manner.
Collapse
|
3
|
Jia M, Li G, Chen J, Tang X, Zang Y, Yang G, Shi YS, Ma D, Ji M, Yang J. Hippocampal Nogo66-NgR1 signaling activation restricts postsynaptic assembly in aged mice with postoperative neurocognitive disorders. Aging Cell 2025; 24:e14366. [PMID: 39412367 PMCID: PMC11709113 DOI: 10.1111/acel.14366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 09/05/2024] [Accepted: 09/17/2024] [Indexed: 01/11/2025] Open
Abstract
Postoperative neurocognitive disorders (pNCD) are a common neurological complication, especially in elderly following anesthesia and surgery. Yet, the underlying mechanisms of pNCD remain elusive. This study aimed to investigate the molecular mechanisms that compromise synaptic metaplasticity in pNCD development with a focus on the involvement of Nogo-66 receptor 1 (NgR1) in the pathogenesis of pNCD in aged mice. Aged mice subjected to anesthesia and laparotomy surgery exhibited anxiety-like behavior and contextual fear memory impairment. Moreover, the procedure significantly increased NogoA and NgR1 expressions, particularly in the hippocampal CA1 and CA3 regions. This increase led to the depolymerization of F-actin, attributed to the activation of the RhoA-GTPase, resulting in a reduction of dendritic spines and changes in their morphology. Additionally, these changes hindered the efficient postsynaptic delivery of the subunit GluA1 and GluA2 of AMPA receptors (AMPARs), consequently diminishing excitatory neurotransmission in the hippocampus. Importantly, administering the competitive NgR1 antagonist peptide NEP1-40 (Nogo-A extracellular peptide residues 1-40 amino acids of Nogo-66) and Fasudil (a Rho-kinase inhibitor) effectively mitigated synaptic impairments and reversed neurocognitive deficits in aged mice following anesthesia and surgery. Our work indicates that high hippocampal Nogo66-NgR1 signaling disrupts postsynaptic AMPA receptor surface delivery due to F-actin depolymerization in the pathophysiology of pNCD.
Collapse
Affiliation(s)
- Min Jia
- Department of Anaesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Gui‐zhou Li
- Minister of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research CenterNanjing UniversityNanjingChina
| | - Jiang Chen
- Minister of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research CenterNanjing UniversityNanjingChina
| | - Xiao‐hui Tang
- Department of Anaesthesiology and Perioperative MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yan‐yu Zang
- Minister of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research CenterNanjing UniversityNanjingChina
| | - Guo‐lin Yang
- Minister of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research CenterNanjing UniversityNanjingChina
| | - Yun Stone Shi
- Minister of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research CenterNanjing UniversityNanjingChina
| | - Daqing Ma
- Perioperative and Systems Medicine LaboratoryNational Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of MedicineHangzhouChina
- Division of Anaesthetics, Pain Medicine & Intensive Care, Department of Surgery & Cancer, Faculty of MedicineImperial College London, Chelsea & Westminster HospitalLondonUK
| | - Mu‐huo Ji
- Department of AnaesthesiologyThe Second Affiliated Hospital, Nanjing Medical UniversityNanjingChina
| | - Jian‐jun Yang
- Department of Anaesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
4
|
Skinnider MA, Gautier M, Teo AYY, Kathe C, Hutson TH, Laskaratos A, de Coucy A, Regazzi N, Aureli V, James ND, Schneider B, Sofroniew MV, Barraud Q, Bloch J, Anderson MA, Squair JW, Courtine G. Single-cell and spatial atlases of spinal cord injury in the Tabulae Paralytica. Nature 2024; 631:150-163. [PMID: 38898272 DOI: 10.1038/s41586-024-07504-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 05/01/2024] [Indexed: 06/21/2024]
Abstract
Here, we introduce the Tabulae Paralytica-a compilation of four atlases of spinal cord injury (SCI) comprising a single-nucleus transcriptome atlas of half a million cells, a multiome atlas pairing transcriptomic and epigenomic measurements within the same nuclei, and two spatial transcriptomic atlases of the injured spinal cord spanning four spatial and temporal dimensions. We integrated these atlases into a common framework to dissect the molecular logic that governs the responses to injury within the spinal cord1. The Tabulae Paralytica uncovered new biological principles that dictate the consequences of SCI, including conserved and divergent neuronal responses to injury; the priming of specific neuronal subpopulations to upregulate circuit-reorganizing programs after injury; an inverse relationship between neuronal stress responses and the activation of circuit reorganization programs; the necessity of re-establishing a tripartite neuroprotective barrier between immune-privileged and extra-neural environments after SCI and a failure to form this barrier in old mice. We leveraged the Tabulae Paralytica to develop a rejuvenative gene therapy that re-established this tripartite barrier, and restored the natural recovery of walking after paralysis in old mice. The Tabulae Paralytica provides a window into the pathobiology of SCI, while establishing a framework for integrating multimodal, genome-scale measurements in four dimensions to study biology and medicine.
Collapse
Affiliation(s)
- Michael A Skinnider
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Matthieu Gautier
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
| | - Alan Yue Yang Teo
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
| | - Claudia Kathe
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
| | - Thomas H Hutson
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- Wyss Center for Bio and Neuroengineering, Geneva, Switzerland
| | - Achilleas Laskaratos
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
| | - Alexandra de Coucy
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
| | - Nicola Regazzi
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
| | - Viviana Aureli
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- Department of Neurosurgery, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Nicholas D James
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
| | - Bernard Schneider
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Bertarelli Platform for Gene Therapy, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Quentin Barraud
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
| | - Jocelyne Bloch
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
- Department of Neurosurgery, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Mark A Anderson
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland.
- Wyss Center for Bio and Neuroengineering, Geneva, Switzerland.
- Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
| | - Jordan W Squair
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland.
- Department of Neurosurgery, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
| | - Grégoire Courtine
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland.
- Department of Neurosurgery, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
- Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
| |
Collapse
|
5
|
Yang R, Zhang Y, Kang J, Zhang C, Ning B. Chondroitin Sulfate Proteoglycans Revisited: Its Mechanism of Generation and Action for Spinal Cord Injury. Aging Dis 2024; 15:153-168. [PMID: 37307832 PMCID: PMC10796098 DOI: 10.14336/ad.2023.0512] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/12/2023] [Indexed: 06/14/2023] Open
Abstract
Reactive astrocytes (RAs) produce chondroitin sulfate proteoglycans (CSPGs) in large quantities after spinal cord injury (SCI) and inhibit axon regeneration through the Rho-associated protein kinase (ROCK) pathway. However, the mechanism of producing CSPGs by RAs and their roles in other aspects are often overlooked. In recent years, novel generation mechanisms and functions of CSPGs have gradually emerged. Extracellular traps (ETs), a new recently discovered phenomenon in SCI, can promote secondary injury. ETs are released by neutrophils and microglia, which activate astrocytes to produce CSPGs after SCI. CSPGs inhibit axon regeneration and play an important role in regulating inflammation as well as cell migration and differentiation; some of these regulations are beneficial. The current review summarized the process of ET-activated RAs to generate CSPGs at the cellular signaling pathway level. Moreover, the roles of CSPGs in inhibiting axon regeneration, regulating inflammation, and regulating cell migration and differentiation were discussed. Finally, based on the above process, novel potential therapeutic targets were proposed to eliminate the adverse effects of CSPGs.
Collapse
Affiliation(s)
- Rui Yang
- Jinan Central Hospital, Shandong University, Jinan, Shandong, China.
| | - Ying Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jianning Kang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ce Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Bin Ning
- Jinan Central Hospital, Shandong University, Jinan, Shandong, China.
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
6
|
Artimagnella O, Mazzon E, Salamone S, Pollastro F, Gugliandolo A, Chiricosta L. Cannabinerol (CBNR) Influences Synaptic Genes Associated with Cytoskeleton and Ion Channels in NSC-34 Cell Line: A Transcriptomic Study. Biomedicines 2024; 12:189. [PMID: 38255294 PMCID: PMC10813620 DOI: 10.3390/biomedicines12010189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Cannabinoids are receiving great attention as a novel approach in the treatment of cognitive and motor disabilities, which characterize neurological disorders. To date, over 100 phytocannabinoids have been extracted from Cannabis sativa, and some of them have shown neuroprotective properties and the capacity to influence synaptic transmission. In this study, we investigated the effects of a less-known phytocannabinoid, cannabinerol (CBNR), on neuronal physiology. Using the NSC-34 motor-neuron-like cell line and next-generation sequencing analysis, we discovered that CBNR influences synaptic genes associated with synapse organization and specialization, including genes related to the cytoskeleton and ion channels. Specifically, the calcium, sodium, and potassium channel subunits (Cacna1b, Cacna1c, Cacnb1, Grin1, Scn8a, Kcnc1, Kcnj9) were upregulated, along with genes related to NMDAR (Agap3, Syngap1) and calcium (Cabp1, Camkv) signaling. Moreover, cytoskeletal and cytoskeleton-associated genes (Actn2, Ina, Trio, Marcks, Bsn, Rtn4, Dgkz, Htt) were also regulated by CBNR. These findings highlight the important role played by CBNR in the regulation of synaptogenesis and synaptic transmission, suggesting the need for further studies to evaluate the neuroprotective role of CBNR in the treatment of synaptic dysfunctions that characterize motor disabilities in many neurological disorders.
Collapse
Affiliation(s)
- Osvaldo Artimagnella
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy (E.M.)
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy (E.M.)
| | - Stefano Salamone
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy; (S.S.); (F.P.)
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy; (S.S.); (F.P.)
| | - Agnese Gugliandolo
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy (E.M.)
| | - Luigi Chiricosta
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy (E.M.)
| |
Collapse
|
7
|
Chambel SS, Cruz CD. Axonal growth inhibitors and their receptors in spinal cord injury: from biology to clinical translation. Neural Regen Res 2023; 18:2573-2581. [PMID: 37449592 DOI: 10.4103/1673-5374.373674] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
Axonal growth inhibitors are released during traumatic injuries to the adult mammalian central nervous system, including after spinal cord injury. These molecules accumulate at the injury site and form a highly inhibitory environment for axonal regeneration. Among these inhibitory molecules, myelin-associated inhibitors, including neurite outgrowth inhibitor A, oligodendrocyte myelin glycoprotein, myelin-associated glycoprotein, chondroitin sulfate proteoglycans and repulsive guidance molecule A are of particular importance. Due to their inhibitory nature, they represent exciting molecular targets to study axonal inhibition and regeneration after central injuries. These molecules are mainly produced by neurons, oligodendrocytes, and astrocytes within the scar and in its immediate vicinity. They exert their effects by binding to specific receptors, localized in the membranes of neurons. Receptors for these inhibitory cues include Nogo receptor 1, leucine-rich repeat, and Ig domain containing 1 and p75 neurotrophin receptor/tumor necrosis factor receptor superfamily member 19 (that form a receptor complex that binds all myelin-associated inhibitors), and also paired immunoglobulin-like receptor B. Chondroitin sulfate proteoglycans and repulsive guidance molecule A bind to Nogo receptor 1, Nogo receptor 3, receptor protein tyrosine phosphatase σ and leucocyte common antigen related phosphatase, and neogenin, respectively. Once activated, these receptors initiate downstream signaling pathways, the most common amongst them being the RhoA/ROCK signaling pathway. These signaling cascades result in actin depolymerization, neurite outgrowth inhibition, and failure to regenerate after spinal cord injury. Currently, there are no approved pharmacological treatments to overcome spinal cord injuries other than physical rehabilitation and management of the array of symptoms brought on by spinal cord injuries. However, several novel therapies aiming to modulate these inhibitory proteins and/or their receptors are under investigation in ongoing clinical trials. Investigation has also been demonstrating that combinatorial therapies of growth inhibitors with other therapies, such as growth factors or stem-cell therapies, produce stronger results and their potential application in the clinics opens new venues in spinal cord injury treatment.
Collapse
Affiliation(s)
- Sílvia Sousa Chambel
- Experimental Biology Unit, Department of Biomedicine, Faculty of Medicine of Porto; Translational NeuroUrology, Instituto de Investigação e Inovação em Saúde-i3S and IBMC, Universidade do Porto, Porto, Portugal
| | - Célia Duarte Cruz
- Experimental Biology Unit, Department of Biomedicine, Faculty of Medicine of Porto; Translational NeuroUrology, Instituto de Investigação e Inovação em Saúde-i3S and IBMC, Universidade do Porto, Porto, Portugal
| |
Collapse
|
8
|
Conti E, Pavone FS, Allegra Mascaro AL. In Vivo Imaging of the Structural Plasticity of Cortical Neurons After Stroke. Methods Mol Biol 2023; 2616:69-81. [PMID: 36715929 DOI: 10.1007/978-1-0716-2926-0_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The comprehension of the finest mechanisms underlying experience-dependent plasticity requires the investigation of neurons and synaptic terminals in the intact brain over prolonged periods of time. Longitudinal two-photon imaging together with the expression of fluorescent proteins enables high-resolution imaging of dendritic spines and axonal varicosities of cortical neurons in vivo. Importantly, the study of the mechanisms of structural reorganization is relevant for a deeper understanding of the pathophysiological mechanisms of neurological diseases such as stroke and for the development of new therapeutic approaches. This protocol describes the principal steps for in vivo investigation of neuronal plasticity both in healthy conditions and after an ischemic lesion. First, we give a description of the surgery to perform a stable cranial window that allows optical access to the mouse brain cortex. Then we explain how to perform longitudinal two-photon imaging of dendrites, axonal branches, and synaptic terminals in the mouse brain cortex in vivo, in order to investigate the plasticity of synaptic terminals and orientation of neuronal processes. Finally, we describe how to induce an ischemic lesion in a target region of the mouse brain cortex through a cranial window by applying the photothrombotic stroke model.
Collapse
Affiliation(s)
- Emilia Conti
- Neuroscience Institute, National Research Council, Pisa, Italy
- European Laboratory for Non-Linear Spectroscopy, Sesto Fiorentino, Italy
| | - Francesco Saverio Pavone
- European Laboratory for Non-Linear Spectroscopy, Sesto Fiorentino, Italy
- Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, Italy
- National Institute of Optics, National Research Council, Sesto Fiorentino, Italy
| | - Anna Letizia Allegra Mascaro
- Neuroscience Institute, National Research Council, Pisa, Italy.
- European Laboratory for Non-Linear Spectroscopy, Sesto Fiorentino, Italy.
| |
Collapse
|
9
|
Kawaguchi Y, Matsubayashi J, Kawakami Y, Nishida R, Kurihara Y, Takei K. LOTUS suppresses amyloid β-induced dendritic spine elimination through the blockade of amyloid β binding to PirB. Mol Med 2022; 28:154. [PMID: 36510132 PMCID: PMC9743548 DOI: 10.1186/s10020-022-00581-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/26/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide but has no effective treatment. Amyloid beta (Aβ) protein, a primary risk factor for AD, accumulates and aggregates in the brain of patients with AD. Paired immunoglobulin-like receptor B (PirB) has been identified as a receptor of Aβ and Aβ-PirB molecular interactions that cause synapse elimination and synaptic dysfunction. PirB deletion has been shown to suppress Aβ-induced synaptic dysfunction and behavioral deficits in AD model mice, implying that PirB mediates Aβ-induced AD pathology. Therefore, inhibiting the Aβ-PirB molecular interaction could be a successful approach for combating AD pathology. We previously showed that lateral olfactory tract usher substance (LOTUS) is an endogenous antagonist of type1 Nogo receptor and PirB and that LOTUS overexpression promotes neuronal regeneration following damage to the central nervous system, including spinal cord injury and ischemic stroke. Therefore, in this study, we investigated whether LOTUS inhibits Aβ-PirB interaction and Aβ-induced dendritic spine elimination. METHODS The inhibitory role of LOTUS against Aβ-PirB (or leukocyte immunoglobulin-like receptor subfamily B member 2: LilrB2) binding was assessed using a ligand-receptor binding assay in Cos7 cells overexpressing PirB and/or LOTUS. We assessed whether LOTUS inhibits Aβ-induced intracellular alterations and synaptotoxicity using immunoblots and spine imaging in a primary cultured hippocampal neuron. RESULTS We found that LOTUS inhibits the binding of Aβ to PirB overexpressed in Cos7 cells. In addition, we found that Aβ-induced dephosphorylation of cofilin and Aβ-induced decrease in post-synaptic density-95 expression were suppressed in cultured hippocampal neurons from LOTUS-overexpressing transgenic (LOTUS-tg) mice compared with that in wild-type mice. Moreover, primary cultured hippocampal neurons from LOTUS-tg mice improved the Aβ-induced decrease in dendritic spine density. Finally, we studied whether human LOTUS protein inhibits Aβ binding to LilrB2, a human homolog of PirB, and found that human LOTUS inhibited the binding of Aβ to LilrB2 in a similar manner. CONCLUSIONS This study implied that LOTUS improved Aβ-induced synapse elimination by suppressing Aβ-PirB interaction in rodents and inhibited Aβ-LilrB2 interaction in humans. Our findings revealed that LOTUS may be a promising therapeutic agent in counteracting Aβ-induced AD pathologies.
Collapse
Affiliation(s)
- Yuki Kawaguchi
- grid.268441.d0000 0001 1033 6139Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, 1-7-29 Suehiro-Cho, Tsurumi Ward, Yokohama, 230-0045 Japan
| | - Junpei Matsubayashi
- grid.268441.d0000 0001 1033 6139Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, 1-7-29 Suehiro-Cho, Tsurumi Ward, Yokohama, 230-0045 Japan
| | - Yutaka Kawakami
- grid.268441.d0000 0001 1033 6139Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, 1-7-29 Suehiro-Cho, Tsurumi Ward, Yokohama, 230-0045 Japan ,grid.419280.60000 0004 1763 8916Department of Anesthesiology, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Ryohei Nishida
- grid.268441.d0000 0001 1033 6139Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, 1-7-29 Suehiro-Cho, Tsurumi Ward, Yokohama, 230-0045 Japan
| | - Yuji Kurihara
- grid.268441.d0000 0001 1033 6139Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, 1-7-29 Suehiro-Cho, Tsurumi Ward, Yokohama, 230-0045 Japan ,grid.260433.00000 0001 0728 1069Department of Molecular Biology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Kohtaro Takei
- grid.268441.d0000 0001 1033 6139Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, 1-7-29 Suehiro-Cho, Tsurumi Ward, Yokohama, 230-0045 Japan
| |
Collapse
|
10
|
Development of Neurogenic Detrusor Overactivity after Thoracic Spinal Cord Injury Is Accompanied by Time-Dependent Changes in Lumbosacral Expression of Axonal Growth Regulators. Int J Mol Sci 2022; 23:ijms23158667. [PMID: 35955811 PMCID: PMC9368817 DOI: 10.3390/ijms23158667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 12/04/2022] Open
Abstract
Thoracic spinal cord injury (SCI) results in urinary dysfunction, which majorly affects the quality of life of SCI patients. Abnormal sprouting of lumbosacral bladder afferents plays a crucial role in this condition. Underlying mechanisms may include changes in expression of regulators of axonal growth, including chondroitin sulphate proteoglycans (CSPGs), myelin-associated inhibitors (MAIs) and repulsive guidance molecules, known to be upregulated at the injury site post SCI. Here, we confirmed lumbosacral upregulation of the growth-associated protein GAP43 in SCI animals with bladder dysfunction, indicating the occurrence of axonal sprouting. Neurocan and Phosphacan (CSPGs), as well as Nogo-A (MAI), at the same spinal segments were upregulated 7 days post injury (dpi) but returned to baseline values 28 dpi. In turn, qPCR analysis of the mRNA levels for receptors of those repulsive molecules in dorsal root ganglia (DRG) neurons showed a time-dependent decrease in receptor expression. In vitro assays with DRG neurons from SCI rats demonstrated that exposure to high levels of NGF downregulated the expression of some, but not all, receptors for those regulators of axonal growth. The present results, therefore, show significant molecular changes at the lumbosacral cord and DRGs after thoracic lesion, likely critically involved in neuroplastic events leading to urinary impairment.
Collapse
|
11
|
Ghorbani S, Yong VW. The extracellular matrix as modifier of neuroinflammation and remyelination in multiple sclerosis. Brain 2021; 144:1958-1973. [PMID: 33889940 PMCID: PMC8370400 DOI: 10.1093/brain/awab059] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
Remyelination failure contributes to axonal loss and progression of disability in multiple sclerosis. The failed repair process could be due to ongoing toxic neuroinflammation and to an inhibitory lesion microenvironment that prevents recruitment and/or differentiation of oligodendrocyte progenitor cells into myelin-forming oligodendrocytes. The extracellular matrix molecules deposited into lesions provide both an altered microenvironment that inhibits oligodendrocyte progenitor cells, and a fuel that exacerbates inflammatory responses within lesions. In this review, we discuss the extracellular matrix and where its molecules are normally distributed in an uninjured adult brain, specifically at the basement membranes of cerebral vessels, in perineuronal nets that surround the soma of certain populations of neurons, and in interstitial matrix between neural cells. We then highlight the deposition of different extracellular matrix members in multiple sclerosis lesions, including chondroitin sulphate proteoglycans, collagens, laminins, fibronectin, fibrinogen, thrombospondin and others. We consider reasons behind changes in extracellular matrix components in multiple sclerosis lesions, mainly due to deposition by cells such as reactive astrocytes and microglia/macrophages. We next discuss the consequences of an altered extracellular matrix in multiple sclerosis lesions. Besides impairing oligodendrocyte recruitment, many of the extracellular matrix components elevated in multiple sclerosis lesions are pro-inflammatory and they enhance inflammatory processes through several mechanisms. However, molecules such as thrombospondin-1 may counter inflammatory processes, and laminins appear to favour repair. Overall, we emphasize the crosstalk between the extracellular matrix, immune responses and remyelination in modulating lesions for recovery or worsening. Finally, we review potential therapeutic approaches to target extracellular matrix components to reduce detrimental neuroinflammation and to promote recruitment and maturation of oligodendrocyte lineage cells to enhance remyelination.
Collapse
Affiliation(s)
- Samira Ghorbani
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
12
|
Li H, McLaurin KA, Illenberger JM, Mactutus CF, Booze RM. Microglial HIV-1 Expression: Role in HIV-1 Associated Neurocognitive Disorders. Viruses 2021; 13:924. [PMID: 34067600 PMCID: PMC8155894 DOI: 10.3390/v13050924] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 11/16/2022] Open
Abstract
The persistence of HIV-1 viral reservoirs in the brain, despite treatment with combination antiretroviral therapy (cART), remains a critical roadblock for the development of a novel cure strategy for HIV-1. To enhance our understanding of viral reservoirs, two complementary studies were conducted to (1) evaluate the HIV-1 mRNA distribution pattern and major cell type expressing HIV-1 mRNA in the HIV-1 transgenic (Tg) rat, and (2) validate our findings by developing and critically testing a novel biological system to model active HIV-1 infection in the rat. First, a restricted, region-specific HIV-1 mRNA distribution pattern was observed in the HIV-1 Tg rat. Microglia were the predominant cell type expressing HIV-1 mRNA in the HIV-1 Tg rat. Second, we developed and critically tested a novel biological system to model key aspects of HIV-1 by infusing F344/N control rats with chimeric HIV (EcoHIV). In vitro, primary cultured microglia were treated with EcoHIV revealing prominent expression within 24 h of infection. In vivo, EcoHIV expression was observed seven days after stereotaxic injections. Following EcoHIV infection, microglia were the major cell type expressing HIV-1 mRNA, results that are consistent with observations in the HIV-1 Tg rat. Within eight weeks of infection, EcoHIV rats exhibited neurocognitive impairments and synaptic dysfunction, which may result from activation of the NogoA-NgR3/PirB-RhoA signaling pathway and/or neuroinflammation. Collectively, these studies enhance our understanding of HIV-1 viral reservoirs in the brain and offer a novel biological system to model HIV-associated neurocognitive disorders and associated comorbidities (i.e., drug abuse) in rats.
Collapse
Affiliation(s)
| | | | | | | | - Rosemarie M. Booze
- Department of Psychology, University of South Carolina, Columbia, SC 29208, USA; (H.L.); (K.A.M.); (J.M.I.); (C.F.M.)
| |
Collapse
|
13
|
Stehle JH, Sheng Z, Hausmann L, Bechstein P, Weinmann O, Hernesniemi J, Neimat JS, Schwab ME, Zemmar A. Exercise-induced Nogo-A influences rodent motor learning in a time-dependent manner. PLoS One 2021; 16:e0250743. [PMID: 33951058 PMCID: PMC8099082 DOI: 10.1371/journal.pone.0250743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/13/2021] [Indexed: 11/22/2022] Open
Abstract
The adult, mature central nervous system (CNS) has limited plasticity. Physical exercising can counteract this limitation by inducing plasticity and fostering processes such as learning, memory consolidation and formation. Little is known about the molecular factors that govern these mechanisms, and how they are connected with exercise. In this study, we used immunohistochemical and behavioral analyses to investigate how running wheel exercise affects expression of the neuronal plasticity-inhibiting protein Nogo-A in the rat cortex, and how it influences motor learning in vivo. Following one week of exercise, rats exhibited a decrease in Nogo-A levels, selectively in motor cortex layer 2/3, but not in layer 5. Nogo-A protein levels returned to baseline after two weeks of running wheel exercise. In a skilled motor task (forelimb-reaching), administration of Nogo-A function-blocking antibodies over the course of the first training week led to improved motor learning. By contrast, Nogo-A antibody application over two weeks of training resulted in impaired learning. Our findings imply a bimodal, time-dependent function of Nogo-A in exercise-induced neuronal plasticity: While an activity-induced suppression of the plasticity-inhibiting protein Nogo-A appears initially beneficial for enhanced motor learning, presumably by allowing greater plasticity in establishing novel synaptic connections, this process is not sustained throughout continued exercise. Instead, upregulation of Nogo-A over the course of the second week of running wheel exercise in rats implies that Nogo-A is required for consolidation of acquired motor skills during the delayed memory consolidation process, possibly by inhibiting ongoing neuronal morphological reorganization to stabilize established synaptic pathways. Our findings suggest that Nogo-A downregulation allows leaning to occur, i.e. opens a 'learning window', while its later upregulation stabilizes the learnt engrams. These findings underline the importance of appropriately timing of application of Nogo-A antibodies in future clinical trials that aim to foster memory performance while avoiding adverse effects.
Collapse
Affiliation(s)
- Jörg H. Stehle
- Department of Neurosurgery, Henan Provincial People´s Hospital, Henan University People’s Hospital, Henan University School of Medicine, People’s Hospital of Zhengzhou University, Zhengzhou, China
- Dr. Senckenbergische Anatomie, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Zhiyuan Sheng
- Department of Neurosurgery, Henan Provincial People´s Hospital, Henan University People’s Hospital, Henan University School of Medicine, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Laura Hausmann
- Department of Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Philipp Bechstein
- Dr. Senckenbergische Anatomie, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Oliver Weinmann
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Department of Biology and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Juha Hernesniemi
- Department of Neurosurgery, Henan Provincial People´s Hospital, Henan University People’s Hospital, Henan University School of Medicine, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Joseph S. Neimat
- Department of Neurosurgery, University of Louisville, School of Medicine, Louisville, Kentucky, United States of America
| | - Martin E. Schwab
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Department of Biology and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Ajmal Zemmar
- Department of Neurosurgery, Henan Provincial People´s Hospital, Henan University People’s Hospital, Henan University School of Medicine, People’s Hospital of Zhengzhou University, Zhengzhou, China
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Department of Biology and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
14
|
Retinal energy metabolism in health and glaucoma. Prog Retin Eye Res 2020; 81:100881. [PMID: 32712136 DOI: 10.1016/j.preteyeres.2020.100881] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/25/2020] [Accepted: 06/28/2020] [Indexed: 01/17/2023]
Abstract
Energy metabolism refers to the processes by which life transfers energy to do cellular work. The retina's relatively large energy demands make it vulnerable to energy insufficiency. In addition, evolutionary pressures to optimize human vision have been traded against retinal ganglion cell bioenergetic fragility. Details of the metabolic profiles of the different retinal cells remain poorly understood and are challenging to resolve. Detailed immunohistochemical mapping of the energy pathway enzymes and substrate transporters has provided some insights and highlighted interspecies differences. The different spatial metabolic patterns between the vascular and avascular retinas can account for some inconsistent data in the literature. There is a consilience of evidence that at least some individuals with glaucoma have impaired RGC energy metabolism, either due to impaired nutrient supply or intrinsic metabolic perturbations. Bioenergetic-based therapy for glaucoma has a compelling pathophysiological foundation and is supported by recent successes in animal models. Recent demonstrations of visual and electrophysiological neurorecovery in humans with glaucoma is highly encouraging and motivates longer duration trials investigating bioenergetic neuroprotection.
Collapse
|
15
|
Zemmar A, Chen CC, Weinmann O, Kast B, Vajda F, Bozeman J, Isaad N, Zuo Y, Schwab ME. Oligodendrocyte- and Neuron-Specific Nogo-A Restrict Dendritic Branching and Spine Density in the Adult Mouse Motor Cortex. Cereb Cortex 2019; 28:2109-2117. [PMID: 28505229 PMCID: PMC6018724 DOI: 10.1093/cercor/bhx116] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Indexed: 01/27/2023] Open
Abstract
Nogo-A has been well described as a myelin-associated inhibitor of neurite outgrowth and functional neuroregeneration after central nervous system (CNS) injury. Recently, a new role of Nogo-A has been identified as a negative regulator of synaptic plasticity in the uninjured adult CNS. Nogo-A is present in neurons and oligodendrocytes. However, it is yet unclear which of these two pools regulate synaptic plasticity. To address this question we used newly generated mouse lines in which Nogo-A is specifically knocked out in (1) oligodendrocytes (oligoNogo-A KO) or (2) neurons (neuroNogo-A KO). We show that both oligodendrocyte- and neuron-specific Nogo-A KO mice have enhanced dendritic branching and spine densities in layer 2/3 cortical pyramidal neurons. These effects are compartmentalized: neuronal Nogo-A affects proximal dendrites whereas oligodendrocytic Nogo-A affects distal regions. Finally, we used two-photon laser scanning microscopy to measure the spine turnover rate of adult mouse motor cortex layer 5 cells and find that both Nogo-A KO mouse lines show enhanced spine remodeling after 4 days. Our results suggest relevant control functions of glial as well as neuronal Nogo-A for synaptic plasticity and open new possibilities for more selective and targeted plasticity enhancing strategies.
Collapse
Affiliation(s)
- Ajmal Zemmar
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland.,Department of Biology and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland.,Department of Neurosurgery, University Hospital Zurich, University of Zurich, CH-8091, Zurich, Switzerland
| | - Chia-Chien Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Oliver Weinmann
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland.,Department of Biology and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland
| | - Brigitt Kast
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland.,Department of Biology and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland
| | - Flora Vajda
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland.,Department of Biology and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland
| | - James Bozeman
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Noel Isaad
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Yi Zuo
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Martin E Schwab
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland.,Department of Biology and Department of Health Sciences and Technology, ETH Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
16
|
Marin MA, Carmichael ST. Mechanisms of demyelination and remyelination in the young and aged brain following white matter stroke. Neurobiol Dis 2019; 126:5-12. [DOI: 10.1016/j.nbd.2018.07.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/03/2018] [Accepted: 07/18/2018] [Indexed: 01/01/2023] Open
|
17
|
Courtine G, Sofroniew MV. Spinal cord repair: advances in biology and technology. Nat Med 2019; 25:898-908. [PMID: 31160817 DOI: 10.1038/s41591-019-0475-6] [Citation(s) in RCA: 312] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/01/2019] [Indexed: 02/06/2023]
Abstract
Individuals with spinal cord injury (SCI) can face decades with permanent disabilities. Advances in clinical management have decreased morbidity and improved outcomes, but no randomized clinical trial has demonstrated the efficacy of a repair strategy for improving recovery from SCI. Here, we summarize recent advances in biological and engineering strategies to augment neuroplasticity and/or functional recovery in animal models of SCI that are pushing toward clinical translation.
Collapse
Affiliation(s)
- Grégoire Courtine
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Geneva, Switzerland. .,Department of Neurosurgery, University Hospital Lausanne (CHUV), Lausanne, Switzerland.
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
18
|
Tran AP, Warren PM, Silver J. The Biology of Regeneration Failure and Success After Spinal Cord Injury. Physiol Rev 2018. [PMID: 29513146 DOI: 10.1152/physrev.00017.2017] [Citation(s) in RCA: 559] [Impact Index Per Article: 79.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Since no approved therapies to restore mobility and sensation following spinal cord injury (SCI) currently exist, a better understanding of the cellular and molecular mechanisms following SCI that compromise regeneration or neuroplasticity is needed to develop new strategies to promote axonal regrowth and restore function. Physical trauma to the spinal cord results in vascular disruption that, in turn, causes blood-spinal cord barrier rupture leading to hemorrhage and ischemia, followed by rampant local cell death. As subsequent edema and inflammation occur, neuronal and glial necrosis and apoptosis spread well beyond the initial site of impact, ultimately resolving into a cavity surrounded by glial/fibrotic scarring. The glial scar, which stabilizes the spread of secondary injury, also acts as a chronic, physical, and chemo-entrapping barrier that prevents axonal regeneration. Understanding the formative events in glial scarring helps guide strategies towards the development of potential therapies to enhance axon regeneration and functional recovery at both acute and chronic stages following SCI. This review will also discuss the perineuronal net and how chondroitin sulfate proteoglycans (CSPGs) deposited in both the glial scar and net impede axonal outgrowth at the level of the growth cone. We will end the review with a summary of current CSPG-targeting strategies that help to foster axonal regeneration, neuroplasticity/sprouting, and functional recovery following SCI.
Collapse
Affiliation(s)
- Amanda Phuong Tran
- Department of Neurosciences, Case Western Reserve University , Cleveland, Ohio ; and School of Biomedical Sciences, University of Leeds , Leeds , United Kingdom
| | - Philippa Mary Warren
- Department of Neurosciences, Case Western Reserve University , Cleveland, Ohio ; and School of Biomedical Sciences, University of Leeds , Leeds , United Kingdom
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University , Cleveland, Ohio ; and School of Biomedical Sciences, University of Leeds , Leeds , United Kingdom
| |
Collapse
|
19
|
Marin MA, Carmichael ST. Stroke in CNS white matter: Models and mechanisms. Neurosci Lett 2018; 684:193-199. [PMID: 30098384 DOI: 10.1016/j.neulet.2018.07.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/03/2018] [Accepted: 07/30/2018] [Indexed: 12/14/2022]
Abstract
White matter stroke (WMS) is a debilitating disorder, which is characterized by the formation of ischemic lesions along subcortical white matter tracts of the central nervous system. Initial infarction during the early stages of the disease is often asymptomatic and is thus considered a form of silent stroke. However, over time lesions accumulate, resulting in severe cognitive and motor decline of which there are no known therapies. Functional imaging and post mortem analysis of patients demonstrates a loss of oligodendrocytes and the subsequent damage of myelin as a primary hallmark of WMS lesions. Though the adult mammalian brain maintains the capacity to regenerate adult oligodendrocytes, this process is blocked in the infarcted white matter thereby preventing remyelination. Recent evidence suggests that activation of neural circuits via motor training or direct stimulation drives oligodendrogenesis and de novo myelin synthesis, opening a potential avenue for therapy in WMS.
Collapse
Affiliation(s)
- Miguel Alejandro Marin
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 9009, United States.
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 9009, United States.
| |
Collapse
|
20
|
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
|
21
|
Modulation of Receptor Protein Tyrosine Phosphatase Sigma Increases Chondroitin Sulfate Proteoglycan Degradation through Cathepsin B Secretion to Enhance Axon Outgrowth. J Neurosci 2018; 38:5399-5414. [PMID: 29760175 DOI: 10.1523/jneurosci.3214-17.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 05/03/2018] [Accepted: 05/09/2018] [Indexed: 12/22/2022] Open
Abstract
Severed axon tips reform growth cones following spinal cord injury that fail to regenerate, in part, because they become embedded within an inhibitory extracellular matrix. Chondroitin sulfate proteoglycans (CSPGs) are the major axon inhibitory matrix component that is increased within the lesion scar and in perineuronal nets around deafferented neurons. We have recently developed a novel peptide modulator (intracellular sigma peptide) of the cognate receptor of CSPGs, protein tyrosine phosphatase σ (RPTPσ), which has been shown to markedly improve sensorimotor function, micturition, and coordinated locomotor behavior in spinal cord contused rats. However, the mechanism(s) underlying how modulation of RPTPσ mediates axon outgrowth through inhibitory CSPGs remain unclear. Here, we describe how intracellular sigma peptide modulation of RPTPσ induces enhanced protease Cathepsin B activity. Using DRG neurons from female Sprague Dawley rats cultured on an aggrecan/laminin spot assay and a combination of biochemical techniques, we provide evidence suggesting that modulation of RPTPσ regulates secretion of proteases that, in turn, relieves CSPG inhibition through its digestion to allow axon migration though proteoglycan barriers. Understanding the mechanisms underlying RPTPσ modulation elucidates how axon regeneration is impaired by proteoglycans but can then be facilitated following injury.SIGNIFICANCE STATEMENT Following spinal cord injury, chondroitin sulfate proteoglycans (CSPGs) upregulate and potently inhibit axon regeneration and functional recovery. Protein tyrosine phosphatase σ (RPTPσ) has been identified as a critical cognate receptor of CSPGs. We have previously characterized a synthetic peptide (intracellular sigma peptide) that targets the regulatory intracellular domain of the receptor to allow axons to regenerate despite the presence of CSPGs. Here, we have found that one important mechanism by which peptide modulation of the receptor enhances axon outgrowth is through secretion of a protease, Cathepsin B, which enables digestion of CSPGs. This work links protease secretion to the CSPG receptor RPTPσ for the first time with implications for understanding the molecular mechanisms underlying neural regeneration and plasticity.
Collapse
|
22
|
Abstract
The inability to recover functions lost after severe spinal cord injury has been recognized for millennia and was first attributed to a failure of spinal cord neural regeneration over 100 years ago. The last forty years have seen intense research into achieving such regeneration, but in spite of conceptual advances and many reports announcing successful interventions, progress has been slow and often controversial. Here, I examine consequential advances and setbacks, and critically consider assumptions underlying certain approaches. I argue that expanding mechanistic knowledge about multiple forms of neural regeneration, why they fail and how they can restore function will resolve conceptual contentions and push the field forward.
Collapse
|
23
|
PTEN expression in astrocytic processes after spinal cord injury. Mol Cell Neurosci 2018; 88:231-239. [DOI: 10.1016/j.mcn.2018.02.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/12/2018] [Accepted: 02/13/2018] [Indexed: 11/21/2022] Open
|
24
|
Iobbi C, Korte M, Zagrebelsky M. Nogo-66 Restricts Synaptic Strengthening via Lingo1 and the ROCK2-Cofilin Pathway to Control Actin Dynamics. Cereb Cortex 2018; 27:2779-2792. [PMID: 27166169 DOI: 10.1093/cercor/bhw122] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nogo-A restricts long-term potentiation (LTP) at the Schaffer collateral-CA1 pathway in the adult hippocampus via 2 extracellular domains: Nogo-A-Δ20 and Nogo-66. Nogo-66 signals via Nogo Receptor 1 (NgR1) to regulate synaptic function. Whether the NgR1 coreceptors Lingo1 and p75NTR are involved in the signaling in this context is still not known. Moreover, the intracellular cascade mediating the activity of Nogo-66 in restricting LTP is unexplored. We combine electrophysiology and biochemistry in acute hippocampal slices and demonstrate that a loss of function for Lingo1 results in a significant increase in LTP levels at the Schaffer collateral-CA1 pathway, and that Lingo1 is the NgR1 coreceptor mediating the role of Nogo-66 in restricting LTP. Our data show that p75NTR is not involved in mediating the Nogo-66 effect on LTP. Moreover, loss of function for p75NTR and NgR1 equally attenuate LTD, suggesting that p75NTR might mediate the NgR1-dependent regulation of LTD, independently of Nogo-66. Finally, our results indicate that Nogo-66 signaling limits LTP via the ROCK2-Cofilin pathway to control the dynamics of the actin cytoskeleton. The present results elucidate the signaling pathway activated by Nogo-66 to control LTP and contribute to the understanding of how Nogo-A stabilizes the neural circuits to limit activity-dependent plasticity events in the mature hippocampus.
Collapse
Affiliation(s)
- Cristina Iobbi
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, 38106, Braunschweig, Germany
| | - Martin Korte
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, 38106, Braunschweig, Germany.,Helmholtz Centre for Infection Research, AG NIND, 38124, Braunschweig, Germany
| | - Marta Zagrebelsky
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, 38106, Braunschweig, Germany
| |
Collapse
|
25
|
Smedfors G, Olson L, Karlsson TE. A Nogo-Like Signaling Perspective from Birth to Adulthood and in Old Age: Brain Expression Patterns of Ligands, Receptors and Modulators. Front Mol Neurosci 2018. [PMID: 29520216 PMCID: PMC5827527 DOI: 10.3389/fnmol.2018.00042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
An appropriate strength of Nogo-like signaling is important to maintain synaptic homeostasis in the CNS. Disturbances have been associated with schizophrenia, MS and other diseases. Blocking Nogo-like signaling may improve recovery after spinal cord injury, stroke and traumatic brain injury. To understand the interacting roles of an increasing number of ligands, receptors and modulators engaged in Nogo-like signaling, the transcriptional activity of these genes in the same brain areas from birth to old age in the normal brain is needed. Thus, we have quantitatively mapped the innate expression of 11 important genes engaged in Nogo-like signaling. Using in situ hybridization, we located and measured the amount of mRNA encoding Nogo-A, OMgp, NgR1, NgR2, NgR3, Lingo-1, Troy, Olfactomedin, LgI1, ADAM22, and MAG, in 18 different brain areas at six different ages (P0, 1, 2, 4, 14, and 104 weeks). We show gene- and area-specific activities and how the genes undergo dynamic regulation during postnatal development and become stable during adulthood. Hippocampal areas underwent the largest changes over time. We only found differences between individual cortical areas in Troy and MAG. Subcortical areas presented the largest inter-regional differences; lateral and basolateral amygdala had markedly higher expression than other subcortical areas. The widespread differences and unique expression patterns of the different genes involved in Nogo-like signaling suggest that the functional complexes could look vastly different in different areas.
Collapse
Affiliation(s)
| | - Lars Olson
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | | |
Collapse
|
26
|
MT3-MMP Promotes Excitatory Synapse Formation by Promoting Nogo-66 Receptor Ectodomain Shedding. J Neurosci 2017; 38:518-529. [PMID: 29196321 DOI: 10.1523/jneurosci.0962-17.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 10/23/2017] [Accepted: 11/20/2017] [Indexed: 12/14/2022] Open
Abstract
Cell-surface molecules are dynamically regulated at the synapse to assemble and disassemble adhesive contacts that are important for synaptogenesis and for tuning synaptic transmission. Metalloproteinases dynamically regulate cellular behaviors through the processing of cell surface molecules. In the present study, we evaluated the role of membrane-type metalloproteinases (MT-MMPs) in excitatory synaptogenesis. We find that MT3-MMP and MT5-MMP are broadly expressed in the mouse cerebral cortex and that MT3-MMP loss-of-function interferes with excitatory synapse development in dissociated cortical neurons and in vivo We identify Nogo-66 receptor (NgR1) as an MT3-MMP substrate that is required for MT3-MMP-dependent synapse formation. Introduction of the shed ectodomain of NgR1 is sufficient to accelerate excitatory synapse formation in dissociated cortical neurons and in vivo Together, our findings support a role for MT3-MMP-dependent shedding of NgR1 in regulating excitatory synapse development.SIGNIFICANCE STATEMENT In this study, we identify MT3-MMP, a membrane-bound zinc protease, to be necessary for the development of excitatory synapses in cortical neurons. We identify Nogo-66 receptors (NgR1) as a downstream target of MT3-MMP proteolytic activity. Furthermore, processing of surface NgR1 by MT3-MMP generates a soluble ectodomain fragment that accelerates the formation of excitatory synapses. We propose that MT3-MMP activity and NgR1 shedding could stimulate circuitry remodeling in the adult brain and enhance functional connectivity after brain injury.
Collapse
|
27
|
Zhao Y, Sivaji S, Chiang MC, Ali H, Zukowski M, Ali S, Kennedy B, Sklyar A, Cheng A, Guo Z, Reed AK, Kodali R, Borowski J, Frost G, Beukema P, Wills ZP. Amyloid Beta Peptides Block New Synapse Assembly by Nogo Receptor-Mediated Inhibition of T-Type Calcium Channels. Neuron 2017; 96:355-372.e6. [PMID: 29024660 DOI: 10.1016/j.neuron.2017.09.041] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 07/27/2017] [Accepted: 09/22/2017] [Indexed: 01/07/2023]
Abstract
Compelling evidence links amyloid beta (Aβ) peptide accumulation in the brains of Alzheimer's disease (AD) patients with the emergence of learning and memory deficits, yet a clear understanding of the events that drive this synaptic pathology are lacking. We present evidence that neurons exposed to Aβ are unable to form new synapses, resulting in learning deficits in vivo. We demonstrate the Nogo receptor family (NgR1-3) acts as Aβ receptors mediating an inhibition of synapse assembly, plasticity, and learning. Live imaging studies reveal Aβ activates NgRs on the dendritic shaft of neurons, triggering an inhibition of calcium signaling. We define T-type calcium channels as a target of Aβ-NgR signaling, mediating Aβ's inhibitory effects on calcium, synapse assembly, plasticity, and learning. These studies highlight deficits in new synapse assembly as a potential initiator of cognitive pathology in AD, and pinpoint calcium dysregulation mediated by NgRs and T-type channels as key components. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Yanjun Zhao
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Sivaprakash Sivaji
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Michael C Chiang
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Haadi Ali
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Monica Zukowski
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Sareen Ali
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Bryan Kennedy
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Alex Sklyar
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Alice Cheng
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Zihan Guo
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Alexander K Reed
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Ravindra Kodali
- Department of Structural Biology and Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jennifer Borowski
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Georgia Frost
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Patrick Beukema
- Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Zachary P Wills
- Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| |
Collapse
|
28
|
Pronker MF, Tas RP, Vlieg HC, Janssen BJC. Nogo Receptor crystal structures with a native disulfide pattern suggest a novel mode of self-interaction. ACTA CRYSTALLOGRAPHICA SECTION D-STRUCTURAL BIOLOGY 2017; 73:860-876. [DOI: 10.1107/s2059798317013791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 09/25/2017] [Indexed: 11/10/2022]
Abstract
The Nogo Receptor (NgR) is a glycophosphatidylinositol-anchored cell-surface protein and is a receptor for three myelin-associated inhibitors of regeneration: myelin-associated glycoprotein, Nogo66 and oligodendrocyte myelin glycoprotein. In combination with different co-receptors, NgR mediates signalling that reduces neuronal plasticity. The available structures of the NgR ligand-binding leucine-rich repeat (LRR) domain have an artificial disulfide pattern owing to truncated C-terminal construct boundaries. NgR has previously been shown to self-associateviaits LRR domain, but the structural basis of this interaction remains elusive. Here, crystal structures of the NgR LRR with a longer C-terminal segment and a native disulfide pattern are presented. An additional C-terminal loop proximal to the C-terminal LRR cap is stabilized by two newly formed disulfide bonds, but is otherwise mostly unstructured in the absence of any stabilizing interactions. NgR crystallized in six unique crystal forms, three of which share a crystal-packing interface. NgR crystal-packing interfaces from all eight unique crystal forms are compared in order to explore how NgR could self-interact on the neuronal plasma membrane.
Collapse
|
29
|
Ferrari L, Scuvera G, Tucci A, Bianchessi D, Rusconi F, Menni F, Battaglioli E, Milani D, Riva P. Identification of an atypical microdeletion generating the RNF135-SUZ12 chimeric gene and causing a position effect in an NF1 patient with overgrowth. Hum Genet 2017; 136:1329-1339. [PMID: 28776093 DOI: 10.1007/s00439-017-1832-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/26/2017] [Indexed: 02/02/2023]
Abstract
Neurofibromatosis type I (NF1) microdeletion syndrome, which is present in 4-11% of NF1 patients, is associated with a severe phenotype as it is caused by the deletion of NF1 and other genes in the 17q11.2 region. The variable expressivity of the disease makes it challenging to establish genotype-phenotype correlations, which also affects prognosis and counselling. We here describe a 3-year-old NF1 patient with an atypical deletion and a complex phenotype. The patient showed overgrowth, café au lait spots, inguinal freckling, and neurological abnormalities. The extent of the deletion was determined by means of array comparative genomic hybridisation, and its breakpoints were isolated by means of long-range polymerase chain reaction. Sequence analysis of the deletion junction fragment revealed the occurrence of an Alu-mediated recombination that led to the generation of a chimeric gene consisting of three exons of RNF135 and eleven exons of SUZ12. Interestingly, the deletion shares a common RNF135-centred region with another deletion described in a non-NF1 patient with overgrowth. In comparison with the normal RNF135 allele, the chimeric transcript was 350-fold over-expressed in peripheral blood, and the ADAP2 gene located upstream of RNF135 was also up-regulated. In line with this, the deletion causes the loss of a chromatin TD boundary, which entails the aberrant adoption of distal cis-acting regulatory elements. These findings suggest that RNF135 haploinsufficiency is related to overgrowth in patients with NF1 microdeletion syndrome and, for the first time, strongly indicate a position effect that warrants further genotype-phenotype correlation studies to investigate the possible existence of previously unknown pathogenic mechanisms.
Collapse
Affiliation(s)
- Luca Ferrari
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Via Viotti 3/5, 20133, Milan, Italy
| | - Giulietta Scuvera
- Dipartimento Donna-Bambino-Neonato, UOSD Pediatria ad Alta Intensità di Cura, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via della Commenda 9, 20122, Milan, Italy
| | - Arianna Tucci
- Dipartimento Donna-Bambino-Neonato, UOSD Pediatria ad Alta Intensità di Cura, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via della Commenda 9, 20122, Milan, Italy
| | - Donatella Bianchessi
- Dipartimento di Neuro-Oncologia Molecolare, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Francesco Rusconi
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Via Viotti 3/5, 20133, Milan, Italy
| | - Francesca Menni
- Dipartimento Donna-Bambino-Neonato, UOSD Pediatria ad Alta Intensità di Cura, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via della Commenda 9, 20122, Milan, Italy
| | - Elena Battaglioli
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Via Viotti 3/5, 20133, Milan, Italy
| | - Donatella Milani
- Dipartimento Donna-Bambino-Neonato, UOSD Pediatria ad Alta Intensità di Cura, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via della Commenda 9, 20122, Milan, Italy.
| | - Paola Riva
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Via Viotti 3/5, 20133, Milan, Italy.
| |
Collapse
|
30
|
Kempf A, Boda E, Kwok JC, Fritz R, Grande V, Kaelin AM, Ristic Z, Schmandke A, Schmandke A, Tews B, Fawcett JW, Pertz O, Buffo A, Schwab ME. Control of Cell Shape, Neurite Outgrowth, and Migration by a Nogo-A/HSPG Interaction. Dev Cell 2017; 43:24-34.e5. [DOI: 10.1016/j.devcel.2017.08.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 07/04/2017] [Accepted: 08/21/2017] [Indexed: 11/16/2022]
|
31
|
Abstract
Spinal cord injury (SCI) lesions present diverse challenges for repair strategies. Anatomically complete injuries require restoration of neural connectivity across lesions. Anatomically incomplete injuries may benefit from augmentation of spontaneous circuit reorganization. Here, we review SCI cell biology, which varies considerably across three different lesion-related tissue compartments: (a) non-neural lesion core, (b) astrocyte scar border, and (c) surrounding spared but reactive neural tissue. After SCI, axon growth and circuit reorganization are determined by neuron-cell-autonomous mechanisms and by interactions among neurons, glia, and immune and other cells. These interactions are shaped by both the presence and the absence of growth-modulating molecules, which vary markedly in different lesion compartments. The emerging understanding of how SCI cell biology differs across lesion compartments is fundamental to developing rationally targeted repair strategies.
Collapse
|
32
|
Abstract
Lattice-like structures known as perineuronal nets (PNNs) are key components of the extracellular matrix (ECM). Once fully crystallized by adulthood, they are largely stable throughout life. Contrary to previous reports that PNNs inhibit processes involving plasticity, here we report that the dynamic regulation of PNN expression in the adult auditory cortex is vital for fear learning and consolidation in response to pure tones. Specifically, after first confirming the necessity of auditory cortical activity for fear learning and consolidation, we observed that mRNA levels of key proteoglycan components of PNNs were enhanced 4 hr after fear conditioning but were no longer different from the control groups 24 hr later. A similar pattern of regulation was observed in numbers of cells surrounded by PNNs and area occupied by them in the auditory cortex. Finally, the removal of auditory cortex PNNs resulted in a deficit in fear learning and consolidation.
Collapse
|
33
|
Karlsson TE, Wellfelt K, Olson L. Spatiotemporal and Long Lasting Modulation of 11 Key Nogo Signaling Genes in Response to Strong Neuroexcitation. Front Mol Neurosci 2017; 10:94. [PMID: 28442990 PMCID: PMC5386981 DOI: 10.3389/fnmol.2017.00094] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/20/2017] [Indexed: 12/13/2022] Open
Abstract
Inhibition of nerve growth and plasticity in the CNS is to a large part mediated by Nogo-like signaling, now encompassing a plethora of ligands, receptors, co-receptors and modulators. Here we describe the distribution and levels of mRNA encoding 11 key genes involved in Nogo-like signaling (Nogo-A, Oligodendrocyte-Myelin glycoprotein (OMgp), Nogo receptor 1 (NgR1), NgR2, NgR3, Lingo-1, TNF receptor orphan Y (Troy), Olfactomedin, Lateral olfactory tract usher substance (Lotus) and membrane-type matrix metalloproteinase-3 (MT3-MPP)), as well as BDNF and GAPDH. Expression was analyzed in nine different brain areas before, and at eight time points during the first 3 days after a strong neuroexcitatory stimulation, caused by one kainic acid injection. A temporo-spatial pattern of orderly transcriptional regulations emerges that strengthens the role of Nogo-signaling mechanisms for synaptic plasticity in synchrony with transcriptional increases of BDNF mRNA. For most Nogo-type signaling genes, the largest alterations of mRNA levels occur in the dentate gyrus, with marked alterations also in the CA1 region. Changes occurred somewhat later in several areas of the cerebral cortex. The detailed spatio-temporal pattern of mRNA presence and kainic acid-induced transcriptional response is gene-specific. We reveal that several different gene alterations combine to decrease (and later increase) Nogo-like signaling, as expected to allow structural plasticity responses. Other genes are altered in the opposite direction, suggesting that the system prepares in advance in order to rapidly restore balance. However, the fact that Lingo-1 shows a seemingly opposite, plasticity inhibiting response to kainic acid (strong increase of mRNA in the dentate gyrus), may instead suggest a plasticity-enhancing intracellular function of this presumed NgR1 co-receptor.
Collapse
Affiliation(s)
| | - Katrin Wellfelt
- Department of Neuroscience, Karolinska InstitutetStockholm, Sweden
| | - Lars Olson
- Department of Neuroscience, Karolinska InstitutetStockholm, Sweden
| |
Collapse
|
34
|
Kehrer-Sawatzki H, Mautner VF, Cooper DN. Emerging genotype-phenotype relationships in patients with large NF1 deletions. Hum Genet 2017; 136:349-376. [PMID: 28213670 PMCID: PMC5370280 DOI: 10.1007/s00439-017-1766-y] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 02/08/2017] [Indexed: 02/07/2023]
Abstract
The most frequent recurring mutations in neurofibromatosis type 1 (NF1) are large deletions encompassing the NF1 gene and its flanking regions (NF1 microdeletions). The majority of these deletions encompass 1.4-Mb and are associated with the loss of 14 protein-coding genes and four microRNA genes. Patients with germline type-1 NF1 microdeletions frequently exhibit dysmorphic facial features, overgrowth/tall-for-age stature, significant delay in cognitive development, large hands and feet, hyperflexibility of joints and muscular hypotonia. Such patients also display significantly more cardiovascular anomalies as compared with patients without large deletions and often exhibit increased numbers of subcutaneous, plexiform and spinal neurofibromas as compared with the general NF1 population. Further, an extremely high burden of internal neurofibromas, characterised by >3000 ml tumour volume, is encountered significantly, more frequently, in non-mosaic NF1 microdeletion patients than in NF1 patients lacking such deletions. NF1 microdeletion patients also have an increased risk of malignant peripheral nerve sheath tumours (MPNSTs); their lifetime MPNST risk is 16-26%, rather higher than that of NF1 patients with intragenic NF1 mutations (8-13%). NF1 microdeletion patients, therefore, represent a high-risk group for the development of MPNSTs, tumours which are very aggressive and difficult to treat. Co-deletion of the SUZ12 gene in addition to NF1 further increases the MPNST risk in NF1 microdeletion patients. Here, we summarise current knowledge about genotype-phenotype relationships in NF1 microdeletion patients and discuss the potential role of the genes located within the NF1 microdeletion interval whose haploinsufficiency may contribute to the more severe clinical phenotype.
Collapse
Affiliation(s)
| | - Victor-Felix Mautner
- Department of Neurology, University Hospital Hamburg Eppendorf, 20246, Hamburg, Germany
| | - David N Cooper
- Institute of Medical Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| |
Collapse
|
35
|
Abstract
Most of the current therapies, as well as many of the clinical trials, for multiple sclerosis (MS) target the inflammatory autoimmune processes, but less than 20% of all clinical trials investigate potential therapies for the chronic progressive disease stage of MS. The latter is responsible for the steadily increasing disability in many patients, and there is an urgent need for novel therapies that protect nervous system tissue and enhance axonal growth and/or remyelination. As outlined in this review, solid pre-clinical data suggest neutralization of the neurite outgrowth inhibitor Nogo-A as a potential new way to achieve both axonal and myelin repair. Several phase I clinical studies with anti-Nogo-A antibodies have been conducted in different disease paradigms including MS and spinal cord injury. Data from spinal cord injury and amyotrophic lateral sclerosis (ALS) trials accredit a good safety profile of high doses of anti-Nogo-A antibodies administered intravenously or intrathecally. An antibody against a Nogo receptor subunit, leucine rich repeat and immunoglobulin-like domain-containing protein 1 (LINGO-1), was recently shown to improve outcome in patients with acute optic neuritis in a phase II study. Nogo-A-suppressing antibodies could be novel drug candidates for the relapsing as well as the progressive MS disease stage. In this review, we summarize the available pre-clinical and clinical evidence on Nogo-A and elucidate the potential of Nogo-A-antibodies as a therapy for progressive MS.
Collapse
|
36
|
Insights from extracellular matrix studies in the hypothalamus: structural variations of perineuronal nets and discovering a new perifornical area of the anterior hypothalamus. Anat Sci Int 2016; 92:18-24. [PMID: 27714583 DOI: 10.1007/s12565-016-0375-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 09/26/2016] [Indexed: 12/18/2022]
Abstract
The hypothalamus controls metabolism, stress responses, and instinctive behaviors for individual survival and species preservation. Recent studies suggest that hypothalamic neurons retain plasticity throughout adulthood, which enables these neurons to respond to various kinds of changes in environment, nutrients, and fluctuating hormones. One of the mechanisms underlying the regulation of neural plasticity is the formation of a stable extracellular matrix (ECM) structure called perineuronal nets (PNNs). PNNs are large aggregates of heterogeneous ECM molecules such as chondroitin sulfate proteoglycans (CSPGs), hyaluronan, their link proteins, and tenascin-R. PNNs surround the cell body and proximal dendrites of a subset of neurons and limit adult neural plasticity. This review describes the CSPG-based ECM, including the PNNs, with a special focus on the hypothalamus of mice. We first provide an overview of PNNs in terms of their structure, molecular components, and functions, most of which have been demonstrated by extrahypothalamic studies. Second, we show the presence or absence of PNNs within individual hypothalamic regions and then describe non-PNN-formed ECM containing CSPGs that can be observed in particular hypothalamic regions. Finally, we will introduce a newly identified mouse hypothalamic area that we named the perifornical area of the anterior hypothalamus (PeFAH), which contains a cluster of PNN-positive neurons. PeFAH neurons express enkephalin and have bidirectional connections with the lateral septum. The anterior hypothalamus and lateral septum are thought to regulate defensive behaviors; therefore, the PeFAH neurons and PNNs around them could be involved in the regulation of defensive behaviors.
Collapse
|
37
|
Korte M, Schmitz D. Cellular and System Biology of Memory: Timing, Molecules, and Beyond. Physiol Rev 2016; 96:647-93. [PMID: 26960344 DOI: 10.1152/physrev.00010.2015] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The storage of information in the mammalian nervous systems is dependent on a delicate balance between change and stability of neuronal networks. The induction and maintenance of processes that lead to changes in synaptic strength to a multistep process which can lead to long-lasting changes, which starts and ends with a highly choreographed and perfectly timed dance of molecules in different cell types of the central nervous system. This is accompanied by synchronization of specific networks, resulting in the generation of characteristic "macroscopic" rhythmic electrical fields, whose characteristic frequencies correspond to certain activity and information-processing states of the brain. Molecular events and macroscopic fields influence each other reciprocally. We review here cellular processes of synaptic plasticity, particularly functional and structural changes, and focus on timing events that are important for the initial memory acquisition, as well as mechanisms of short- and long-term memory storage. Then, we cover the importance of epigenetic events on the long-time range. Furthermore, we consider how brain rhythms at the network level participate in processes of information storage and by what means they participating in it. Finally, we examine memory consolidation at the system level during processes of sleep.
Collapse
Affiliation(s)
- Martin Korte
- Zoological Institute, Division of Cellular Neurobiology, Braunschweig, Germany; Helmholtz Centre for Infection Research, AG NIND, Braunschweig, Germany; and Neuroscience Research Centre, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Dietmar Schmitz
- Zoological Institute, Division of Cellular Neurobiology, Braunschweig, Germany; Helmholtz Centre for Infection Research, AG NIND, Braunschweig, Germany; and Neuroscience Research Centre, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
38
|
Korecka J, Levy S, Isacson O. In vivo modeling of neuronal function, axonal impairment and connectivity in neurodegenerative and neuropsychiatric disorders using induced pluripotent stem cells. Mol Cell Neurosci 2016; 73:3-12. [DOI: 10.1016/j.mcn.2015.12.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 12/04/2015] [Accepted: 12/08/2015] [Indexed: 02/07/2023] Open
|
39
|
Astrocyte scar formation aids central nervous system axon regeneration. Nature 2016; 532:195-200. [PMID: 27027288 DOI: 10.1038/nature17623] [Citation(s) in RCA: 1309] [Impact Index Per Article: 145.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/26/2016] [Indexed: 12/20/2022]
Abstract
Transected axons fail to regrow in the mature central nervous system. Astrocytic scars are widely regarded as causal in this failure. Here, using three genetically targeted loss-of-function manipulations in adult mice, we show that preventing astrocyte scar formation, attenuating scar-forming astrocytes, or ablating chronic astrocytic scars all failed to result in spontaneous regrowth of transected corticospinal, sensory or serotonergic axons through severe spinal cord injury (SCI) lesions. By contrast, sustained local delivery via hydrogel depots of required axon-specific growth factors not present in SCI lesions, plus growth-activating priming injuries, stimulated robust, laminin-dependent sensory axon regrowth past scar-forming astrocytes and inhibitory molecules in SCI lesions. Preventing astrocytic scar formation significantly reduced this stimulated axon regrowth. RNA sequencing revealed that astrocytes and non-astrocyte cells in SCI lesions express multiple axon-growth-supporting molecules. Our findings show that contrary to the prevailing dogma, astrocyte scar formation aids rather than prevents central nervous system axon regeneration.
Collapse
|
40
|
Karlsson TE, Smedfors G, Brodin ATS, Åberg E, Mattsson A, Högbeck I, Wellfelt K, Josephson A, Brené S, Olson L. NgR1: A Tunable Sensor Regulating Memory Formation, Synaptic, and Dendritic Plasticity. Cereb Cortex 2016; 26:1804-17. [PMID: 26838771 PMCID: PMC4785958 DOI: 10.1093/cercor/bhw007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Nogo receptor 1 (NgR1) is expressed in forebrain neurons and mediates nerve growth inhibition in response to Nogo and other ligands. Neuronal activity downregulates NgR1 and the inability to downregulate NgR1 impairs long-term memory. We investigated behavior in a serial behavioral paradigm in mice that overexpress or lack NgR1, finding impaired locomotor behavior and recognition memory in mice lacking NgR1 and impaired sequential spatial learning in NgR1 overexpressing mice. We also investigated a role for NgR1 in drug-mediated sensitization and found that repeated cocaine exposure caused stronger locomotor responses but limited development of stereotypies in NgR1 overexpressing mice. This suggests that NgR1-regulated synaptic plasticity is needed to develop stereotypies. Ex vivo magnetic resonance imaging and diffusion tensor imaging analyses of NgR1 overexpressing brains did not reveal any major alterations. NgR1 overexpression resulted in significantly reduced density of mature spines and dendritic complexity. NgR1 overexpression also altered cocaine-induced effects on spine plasticity. Our results show that NgR1 is a negative regulator of both structural synaptic plasticity and dendritic complexity in a brain region-specific manner, and highlight anterior cingulate cortex as a key area for memory-related plasticity.
Collapse
Affiliation(s)
- Tobias E Karlsson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Alvin T S Brodin
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Elin Åberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden Present address: AstraZeneca R&D, AstraZeneca Translational Science Centre at Science for Life Laboratory, Solna, Sweden
| | - Anna Mattsson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden Present address: The Swedish Council on Health Technology Assessment, Stockholm, Sweden
| | - Isabelle Högbeck
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Katrin Wellfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anna Josephson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Stefan Brené
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| | - Lars Olson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
41
|
Blocking the Nogo-A Signaling Pathway to Promote Regeneration and Plasticity After Spinal Cord Injury and Stroke. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
42
|
ZHONG JIANBIN, LI XIE, WAN LIMEI, CHEN ZHIBANG, ZHONG SIMIN, XIAO SONGHUA, YAN ZHENGWEN. Knockdown of NogoA prevents MPP+-induced neurotoxicity in PC12 cells via the mTOR/STAT3 signaling pathway. Mol Med Rep 2015; 13:1427-33. [DOI: 10.3892/mmr.2015.4637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 11/17/2015] [Indexed: 11/06/2022] Open
|
43
|
Peng WS, Qi C, Zhang H, Gao ML, Wang H, Ren F, Li XQ. Distribution of paired immunoglobulin-like receptor B in the nervous system related to regeneration difficulties after unilateral lumbar spinal cord injury. Neural Regen Res 2015; 10:1139-46. [PMID: 26330840 PMCID: PMC4541248 DOI: 10.4103/1673-5374.160111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2015] [Indexed: 12/21/2022] Open
Abstract
Paired immunoglobulin-like receptor B (PirB) is a functional receptor of myelin-associated inhibitors for axonal regeneration and synaptic plasticity in the central nervous system, and thus suppresses nerve regeneration. The regulatory effect of PirB on injured nerves has received a lot of attention. To better understand nerve regeneration inability after spinal cord injury, this study aimed to investigate the distribution of PirB (via immunofluorescence) in the central nervous system and peripheral nervous system 10 days after injury. Immunoreactivity for PirB increased in the dorsal root ganglia, sciatic nerves, and spinal cord segments. In the dorsal root ganglia and sciatic nerves, PirB was mainly distributed along neuronal and axonal membranes. PirB was found to exhibit a diffuse, intricate distribution in the dorsal and ventral regions. Immunoreactivity for PirB was enhanced in some cortical neurons located in the bilateral precentral gyri. Overall, the findings suggest a pattern of PirB immunoreactivity in the nervous system after unilateral spinal transection injury, and also indicate that PirB may suppress repair after injury.
Collapse
Affiliation(s)
- Wan-Shu Peng
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Chao Qi
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Hong Zhang
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Mei-Ling Gao
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Hong Wang
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Fei Ren
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Xia-Qing Li
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
44
|
Jones TA, Adkins DL. Motor System Reorganization After Stroke: Stimulating and Training Toward Perfection. Physiology (Bethesda) 2015; 30:358-70. [PMID: 26328881 PMCID: PMC4556825 DOI: 10.1152/physiol.00014.2015] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Stroke instigates regenerative responses that reorganize connectivity patterns among surviving neurons. The new connectivity patterns can be suboptimal for behavioral function. This review summarizes current knowledge on post-stroke motor system reorganization and emerging strategies for shaping it with manipulations of behavior and cortical activity to improve functional outcome.
Collapse
Affiliation(s)
- Theresa A Jones
- Psychology Department, Neuroscience Institute, University of Texas at Austin, Austin, Texas; and
| | - DeAnna L Adkins
- Neurosciences Department, and Health Sciences & Research Department, Colleges of Medicine & Health Professions, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
45
|
Zemmar A, Kast B, Lussi K, Luft AR, Schwab ME. Acquisition of a High-precision Skilled Forelimb Reaching Task in Rats. J Vis Exp 2015:e53010. [PMID: 26131653 DOI: 10.3791/53010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Movements are the main measurable output of central nervous system function. Developing behavioral paradigms that allow detailed analysis of motor learning and execution is of critical importance in order to understand the principles and processes that underlie motor function. Here we present a paradigm to study movement acquisition within a daily session of training (within-session) representing the fast learning component and primary acquisition as well as skilled motor learning over several training sessions (between-session) representing the slow learning component and consolidation of the learned task. This behavioral paradigm increases the degree of difficulty and complexity of the motor skill task due to two features: First, the animal realigns its body prior to each pellet retrieval forcing renewed orientation and preventing movement execution from the same angle. Second, pellets are grasped from a vertical post that matches the diameter of the pellet and is placed in front of the cage. This requires a precise grasp for successful pellet retrieval and thus prevents simple pulling of the pellet towards the animal. In combination with novel genetics, imaging and electrophysiological technologies, this behavioral method will aid to understand the morphological, anatomical and molecular underpinnings of motor learning and memory.
Collapse
Affiliation(s)
- Ajmal Zemmar
- Brain Research Institute, University of Zurich; Department of Biology and Department of Health Sciences and Technology, ETH Zurich;
| | - Brigitte Kast
- Brain Research Institute, University of Zurich; Department of Biology and Department of Health Sciences and Technology, ETH Zurich
| | - Karin Lussi
- Brain Research Institute, University of Zurich; Department of Biology and Department of Health Sciences and Technology, ETH Zurich
| | - Andreas R Luft
- Clinical Neurorehabilitation, Department of Neurology, University of Zurich & University Hospital Zurich
| | - Martin E Schwab
- Brain Research Institute, University of Zurich; Department of Biology and Department of Health Sciences and Technology, ETH Zurich;
| |
Collapse
|
46
|
Dickson HM, Wilbur A, Reinke AA, Young MA, Vojtek AB. Targeted inhibition of the Shroom3-Rho kinase protein-protein interaction circumvents Nogo66 to promote axon outgrowth. BMC Neurosci 2015; 16:34. [PMID: 26077244 PMCID: PMC4467669 DOI: 10.1186/s12868-015-0171-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 06/03/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Inhibitory molecules in the adult central nervous system, including NogoA, impede neural repair by blocking axon outgrowth. The actin-myosin regulatory protein Shroom3 directly interacts with Rho kinase and conveys axon outgrowth inhibitory signals from Nogo66, a C-terminal inhibitory domain of NogoA. The purpose of this study was to identify small molecules that block the Shroom3-Rho kinase protein-protein interaction as a means to modulate NogoA signaling and, in the longer term, enhance axon outgrowth during neural repair. RESULTS A high throughput screen for inhibitors of the Shroom3-Rho kinase protein-protein interaction identified CCG-17444 (Chem ID: 2816053). CCG-17444 inhibits the Shroom3-Rho kinase interaction in vitro with micromolar potency. This compound acts through an irreversible, covalent mechanism of action, targeting Shroom3 Cys1816 to inhibit the Shroom3-Rho kinase protein-protein interaction. Inhibition of the Shroom3-Rho kinase protein-protein interaction with CCG-17444 counteracts the inhibitory action of Nogo66 and enhances neurite outgrowth. CONCLUSIONS This study identifies a small molecule inhibitor of the Shroom3-Rho kinase protein-protein interaction that circumvents the inhibitory action of Nogo66 in neurons. Identification of a small molecule compound that blocks the Shroom3-Rho kinase protein-protein interaction provides a first step towards a potential new strategy for enhancing neural repair.
Collapse
Affiliation(s)
- Heather M Dickson
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Amanda Wilbur
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Ashley A Reinke
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Mathew A Young
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Anne B Vojtek
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
47
|
Baldwin KT, Giger RJ. Insights into the physiological role of CNS regeneration inhibitors. Front Mol Neurosci 2015; 8:23. [PMID: 26113809 PMCID: PMC4462676 DOI: 10.3389/fnmol.2015.00023] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/26/2015] [Indexed: 12/14/2022] Open
Abstract
The growth inhibitory nature of injured adult mammalian central nervous system (CNS) tissue constitutes a major barrier to robust axonal outgrowth and functional recovery following trauma or disease. Prototypic CNS regeneration inhibitors are broadly expressed in the healthy and injured brain and spinal cord and include myelin-associated glycoprotein (MAG), the reticulon family member NogoA, oligodendrocyte myelin glycoprotein (OMgp), and chondroitin sulfate proteoglycans (CSPGs). These structurally diverse molecules strongly inhibit neurite outgrowth in vitro, and have been most extensively studied in the context of nervous system injury in vivo. The physiological role of CNS regeneration inhibitors in the naïve, or uninjured, CNS remains less well understood, but has received growing attention in recent years and is the focus of this review. CNS regeneration inhibitors regulate myelin development and axon stability, consolidate neuronal structure shaped by experience, and limit activity-dependent modification of synaptic strength. Altered function of CNS regeneration inhibitors is associated with neuropsychiatric disorders, suggesting crucial roles in brain development and health.
Collapse
Affiliation(s)
- Katherine T Baldwin
- Department of Cell and Developmental Biology, University of Michigan School of Medicine Ann Arbor, MI, USA ; Cellular and Molecular Biology Graduate Program, University of Michigan School of Medicine Ann Arbor, MI, USA
| | - Roman J Giger
- Department of Cell and Developmental Biology, University of Michigan School of Medicine Ann Arbor, MI, USA ; Department of Neurology, University of Michigan School of Medicine Ann Arbor, MI, USA
| |
Collapse
|
48
|
Frantz MG, Kast RJ, Dorton HM, Chapman KS, McGee AW. Nogo Receptor 1 Limits Ocular Dominance Plasticity but not Turnover of Axonal Boutons in a Model of Amblyopia. Cereb Cortex 2015; 26:1975-85. [PMID: 25662716 DOI: 10.1093/cercor/bhv014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The formation and stability of dendritic spines on excitatory cortical neurons are correlated with adult visual plasticity, yet how the formation, loss, and stability of postsynaptic spines register with that of presynaptic axonal varicosities is unknown. Monocular deprivation has been demonstrated to increase the rate of formation of dendritic spines in visual cortex. However, we find that monocular deprivation does not alter the dynamics of intracortical axonal boutons in visual cortex of either adult wild-type (WT) mice or adult NgR1 mutant (ngr1-/-) mice that retain critical period visual plasticity. Restoring normal vision for a week following long-term monocular deprivation (LTMD), a model of amblyopia, partially restores ocular dominance (OD) in WT and ngr1-/- mice but does not alter the formation or stability of axonal boutons. Both WT and ngr1-/- mice displayed a rapid return of normal OD within 8 days after LTMD as measured with optical imaging of intrinsic signals. In contrast, single-unit recordings revealed that ngr1-/- exhibited greater recovery of OD by 8 days post-LTMD. Our findings support a model of structural plasticity in which changes in synaptic connectivity are largely postsynaptic. In contrast, axonal boutons appear to be stable during changes in cortical circuit function.
Collapse
Affiliation(s)
- Michael G Frantz
- Developmental Neuroscience Program, Saban Research Institute, Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | - Ryan J Kast
- Developmental Neuroscience Program, Saban Research Institute, Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | - Hilary M Dorton
- Developmental Neuroscience Program, Saban Research Institute, Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | - Katherine S Chapman
- Developmental Neuroscience Program, Saban Research Institute, Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | - Aaron W McGee
- Developmental Neuroscience Program, Saban Research Institute, Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| |
Collapse
|
49
|
Lynch AM, Cleveland M, Prinjha R, Kumar U, Stubbs R, Wuerthner J. Non-clinical development of ozanezumab: a humanised antibody targeting the amino terminus of neurite outgrowth inhibitor A (Nogo-A). Toxicol Res (Camb) 2015. [DOI: 10.1039/c5tx00179j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Ozanezumab (GSK1223249) is a humanised, Fc-disabled, monoclonal antibody (mAb) which targets the amino terminus of Neurite Outgrowth Inhibitor A (Nogo-A) which is currently being developed for the treatment of amyotrophic lateral sclerosis (ALS).
Collapse
|
50
|
Duan Y, Wang SH, Song J, Mironova Y, Ming GL, Kolodkin AL, Giger RJ. Semaphorin 5A inhibits synaptogenesis in early postnatal- and adult-born hippocampal dentate granule cells. eLife 2014; 3. [PMID: 25313870 PMCID: PMC4236683 DOI: 10.7554/elife.04390] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 10/13/2014] [Indexed: 12/20/2022] Open
Abstract
Human SEMAPHORIN 5A (SEMA5A) is an autism susceptibility gene; however, its function in brain development is unknown. In this study, we show that mouse Sema5A negatively regulates synaptogenesis in early, developmentally born, hippocampal dentate granule cells (GCs). Sema5A is strongly expressed by GCs and regulates dendritic spine density in a cell-autonomous manner. In the adult mouse brain, newly born Sema5A-/- GCs show an increase in dendritic spine density and increased AMPA-type synaptic responses. Sema5A signals through PlexinA2 co-expressed by GCs, and the PlexinA2-RasGAP activity is necessary to suppress spinogenesis. Like Sema5A-/- mutants, PlexinA2-/- mice show an increase in GC glutamatergic synapses, and we show that Sema5A and PlexinA2 genetically interact with respect to GC spine phenotypes. Sema5A-/- mice display deficits in social interaction, a hallmark of autism-spectrum-disorders. These experiments identify novel intra-dendritic Sema5A/PlexinA2 interactions that inhibit excitatory synapse formation in developmentally born and adult-born GCs, and they provide support for SEMA5A contributions to autism-spectrum-disorders.
Collapse
Affiliation(s)
- Yuntao Duan
- Department of Cell and Developmental Biology, University of Michigan School of Medicine, Ann Arbor, United States
| | - Shih-Hsiu Wang
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Juan Song
- Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Yevgeniya Mironova
- Department of Cell and Developmental Biology, University of Michigan School of Medicine, Ann Arbor, United States
| | - Guo-li Ming
- Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Alex L Kolodkin
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Roman J Giger
- Department of Cell and Developmental Biology, University of Michigan School of Medicine, Ann Arbor, United States
| |
Collapse
|