1
|
Cangalaya C, Sun W, Stoyanov S, Dunay IR, Dityatev A. Integrity of neural extracellular matrix is required for microglia-mediated synaptic remodeling. Glia 2024; 72:1874-1892. [PMID: 38946065 DOI: 10.1002/glia.24588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 07/02/2024]
Abstract
Microglia continuously remodel synapses, which are embedded in the extracellular matrix (ECM). However, the mechanisms, which govern this process remain elusive. To investigate the influence of the neural ECM in synaptic remodeling by microglia, we disrupted ECM integrity by injection of chondroitinase ABC (ChABC) into the retrosplenial cortex of healthy adult mice. Using in vivo two-photon microscopy we found that ChABC treatment increased microglial branching complexity and ECM phagocytic capacity and decreased spine elimination rate under basal conditions. Moreover, ECM attenuation largely prevented synaptic remodeling following synaptic stress induced by photodamage of single synaptic elements. These changes were associated with less stable and smaller microglial contacts at the synaptic damage sites, diminished deposition of calreticulin and complement proteins C1q and C3 at synapses and impaired expression of microglial CR3 receptor. Thus, our findings provide novel insights into the function of the neural ECM in deposition of complement proteins and synaptic remodeling by microglia.
Collapse
Affiliation(s)
- Carla Cangalaya
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Weilun Sun
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Stoyan Stoyanov
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Alexander Dityatev
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| |
Collapse
|
2
|
Wu S, Shen W, Ge X, Ao F, Zheng Y, Wang Y, Jia X, Mao Y, Luo Y. Advances in Large Gap Peripheral Nerve Injury Repair and Regeneration with Bridging Nerve Guidance Conduits. Macromol Biosci 2023; 23:e2300078. [PMID: 37235853 DOI: 10.1002/mabi.202300078] [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: 02/27/2023] [Revised: 05/10/2023] [Indexed: 05/28/2023]
Abstract
Peripheral nerve injury is a common complication of accidents and diseases. The traditional autologous nerve graft approach remains the gold standard for the treatment of nerve injuries. While sources of autologous nerve grafts are very limited and difficult to obtain. Nerve guidance conduits are widely used in the treatment of peripheral nerve injuries as an alternative to nerve autografts and allografts. However, the development of nerve conduits does not meet the needs of large gap peripheral nerve injury. Functional nerve conduits can provide a good microenvironment for axon elongation and myelin regeneration. Herein, the manufacturing methods and different design types of functional bridging nerve conduits for nerve conduits combined with electrical or magnetic stimulation and loaded with Schwann cells, etc., are summarized. It summarizes the literature and finds that the technical solutions of functional nerve conduits with electrical stimulation, magnetic stimulation and nerve conduits combined with Schwann cells can be used as effective strategies for bridging large gap nerve injury and provide an effective way for the study of large gap nerve injury repair. In addition, functional nerve conduits provide a new way to construct delivery systems for drugs and growth factors in vivo.
Collapse
Affiliation(s)
- Shang Wu
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Wen Shen
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Xuemei Ge
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, 210037, P. R. China
| | - Fen Ao
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Yan Zheng
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Yigang Wang
- Department of Pharmacy, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, Shaanxi, 712000, P. R. China
| | - Xiaoni Jia
- Central Laboratory, Xi'an Mental Health Center, Xi'an, 710061, P. R. China
| | - Yueyang Mao
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Yali Luo
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, 210037, P. R. China
| |
Collapse
|
3
|
Miura T, Yamamoto Y, Funayama E, Ishikawa K, Maeda T. Development of a simultaneous and noninvasive measuring method using high-frame rate videography and motion analysis software for the assessment of facial palsy recovery in a rat model. J Plast Reconstr Aesthet Surg 2023; 82:211-218. [PMID: 37192584 DOI: 10.1016/j.bjps.2023.04.026] [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: 12/11/2022] [Accepted: 04/11/2023] [Indexed: 05/18/2023]
Abstract
BACKGROUND For the development of new therapeutic and reconstructive methods for facial nerve palsy, it is critical to validate them in animal models. This study developed a novel evaluation method using a high-speed camera and motion analysis software for rat facial paralysis models. The validity of the new method was verified using normal rats and rats with facial paralysis. METHODS The whisker movement was recorded using a high-frame video camera. The video files were processed using motion analysis software, and the angular velocities were measured. The score was calculated as the percentage of movement on the side that had palsy compared with the movement on the normal side. Normal rats were used to examine which of the four indices of angular velocity is appropriate for this evaluation method. Using this method, two types of facial nerve palsy models were compared. Furthermore, the three agents that were predicted to promote axon regeneration from previous studies were evaluated. RESULTS The two averages of the protraction and retraction movement velocities of the whiskers were considered as the most appropriate indicators for this new method. Compared with the saline group, all agent groups showed significant differences in the improvement of facial palsy recovery. CONCLUSIONS This method is an evaluation method for the effects of therapeutic intervention for facial nerve paralysis in real time without sacrificing animals.
Collapse
Affiliation(s)
- Takahiro Miura
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo 060-8638, Japan
| | - Yuhei Yamamoto
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo 060-8638, Japan
| | - Emi Funayama
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo 060-8638, Japan
| | - Kosuke Ishikawa
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo 060-8638, Japan
| | - Taku Maeda
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo 060-8638, Japan.
| |
Collapse
|
4
|
Younger DS. Spinal cord motor disorders. HANDBOOK OF CLINICAL NEUROLOGY 2023; 196:3-42. [PMID: 37620076 DOI: 10.1016/b978-0-323-98817-9.00007-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
Spinal cord diseases are frequently devastating due to the precipitous and often permanently debilitating nature of the deficits. Spastic or flaccid paraparesis accompanied by dermatomal and myotomal signatures complementary to the incurred deficits facilitates localization of the insult within the cord. However, laboratory studies often employing disease-specific serology, neuroradiology, neurophysiology, and cerebrospinal fluid analysis aid in the etiologic diagnosis. While many spinal cord diseases are reversible and treatable, especially when recognized early, more than ever, neuroscientists are being called to investigate endogenous mechanisms of neural plasticity. This chapter is a review of the embryology, neuroanatomy, clinical localization, evaluation, and management of adult and childhood spinal cord motor disorders.
Collapse
Affiliation(s)
- David S Younger
- Department of Clinical Medicine and Neuroscience, CUNY School of Medicine, New York, NY, United States; Department of Medicine, Section of Internal Medicine and Neurology, White Plains Hospital, White Plains, NY, United States.
| |
Collapse
|
5
|
Fisher KM, Garner JP, Darian-Smith C. Small sensory spinal lesions that affect hand function in monkeys greatly alter primary afferent and motor neuron connections in the cord. J Comp Neurol 2022; 530:3039-3055. [PMID: 35973735 PMCID: PMC9561953 DOI: 10.1002/cne.25395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/20/2022] [Accepted: 07/15/2022] [Indexed: 11/08/2022]
Abstract
Small sensory spinal injuries induce plasticity across the neuraxis, but little is understood about their effect on segmental connections or motor neuron (MN) function. Here, we begin to address this at two levels. First, we compared afferent input distributions from the skin and muscles of the digits with corresponding MN pools to determine their spatial relationship, in both the normal state and 4-6 months after a unilateral dorsal root/dorsal column lesion (DRL/DCL), affecting digits 1-3. Second, we looked at specific changes to MN inputs and membrane properties that likely impact functional recovery. Monkeys received a targeted unilateral DRL/DCL, and 4-6 months later, cholera toxin subunit B (CT-B) was injected bilaterally into either the distal pads of digits 1-3, or related intrinsic hand muscles, to label inputs to the cord, and corresponding MNs. In controls (unlesioned side), cutaneous and proprioceptive afferents from digits 1-3 showed different distribution patterns but similar rostrocaudal spread within the dorsal horn from C1 to T2. In contrast, MNs were distributed across just two segments (C7-8). Following the lesion, sensory inputs were significantly diminished across all 10 segments, though this did not alter MN distributions. Afferent and monoamine inputs, as well as KCC2 cotransporters, were also significantly altered on the cell membrane of CT-B labeled MNs postlesion. In contrast, inhibitory neurotransmission and perineuronal net integrity were not altered at this prechronic timepoint. Our findings indicate that even a small sensory injury can significantly impact sensory and motor spinal neurons and provide new insight into the complex process of recovery.
Collapse
Affiliation(s)
- Karen M. Fisher
- Department of Comparative Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA94305-5342
| | - Joseph P. Garner
- Department of Comparative Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA94305-5342
| | - Corinna Darian-Smith
- Department of Comparative Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA94305-5342
| |
Collapse
|
6
|
Asthana P, Kumar G, Milanowski LM, Au NPB, Chan SC, Huang J, Feng H, Kwan KM, He J, Chan KWY, Wszolek ZK, Ma CHE. Cerebellar glutamatergic system impacts spontaneous motor recovery by regulating Gria1 expression. NPJ Regen Med 2022; 7:45. [PMID: 36064798 PMCID: PMC9445039 DOI: 10.1038/s41536-022-00243-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 08/12/2022] [Indexed: 11/09/2022] Open
Abstract
Peripheral nerve injury (PNI) often results in spontaneous motor recovery; however, how disrupted cerebellar circuitry affects PNI-associated motor recovery is unknown. Here, we demonstrated disrupted cerebellar circuitry and poor motor recovery in ataxia mice after PNI. This effect was mimicked by deep cerebellar nuclei (DCN) lesion, but not by damaging non-motor area hippocampus. By restoring cerebellar circuitry through DCN stimulation, and reversal of neurotransmitter imbalance using baclofen, ataxia mice achieve full motor recovery after PNI. Mechanistically, elevated glutamate-glutamine level was detected in DCN of ataxia mice by magnetic resonance spectroscopy. Transcriptomic study revealed that Gria1, an ionotropic glutamate receptor, was upregulated in DCN of control mice but failed to be upregulated in ataxia mice after sciatic nerve crush. AAV-mediated overexpression of Gria1 in DCN rescued motor deficits of ataxia mice after PNI. Finally, we found a correlative decrease in human GRIA1 mRNA expression in the cerebellum of patients with ataxia-telangiectasia and spinocerebellar ataxia type 6 patient iPSC-derived Purkinje cells, pointing to the clinical relevance of glutamatergic system. By conducting a large-scale analysis of 9,655,320 patients with ataxia, they failed to recover from carpal tunnel decompression surgery and tibial neuropathy, while aged-match non-ataxia patients fully recovered. Our results provide insight into cerebellar disorders and motor deficits after PNI.
Collapse
Affiliation(s)
- Pallavi Asthana
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR
| | - Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR
| | - Lukasz M Milanowski
- Department of Neurology, Mayo Clinic, Jacksonville, USA.,Department of Neurology, Faculty of Health Science, Medical University of Warsaw, Warsaw, Poland
| | - Ngan Pan Bennett Au
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR
| | - Siu Chung Chan
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR
| | - Jianpan Huang
- Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR
| | - Hemin Feng
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR
| | - Kin Ming Kwan
- School of Life Sciences, Center for Cell and Developmental Biology and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR
| | - Jufang He
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR
| | - Kannie Wai Yan Chan
- Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR.,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, USA
| | | | - Chi Him Eddie Ma
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR.
| |
Collapse
|
7
|
Leite JP, Peixoto-Santos JE. Glia and extracellular matrix molecules: What are their importance for the electrographic and MRI changes in the epileptogenic zone? Epilepsy Behav 2021; 121:106542. [PMID: 31884121 DOI: 10.1016/j.yebeh.2019.106542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 12/22/2022]
Abstract
Glial cells and extracellular matrix (ECM) molecules are crucial for the maintenance of brain homeostasis. Especially because of their actions regarding neurotransmitter and ionic control, and synaptic function, these cells can potentially contribute to the hyperexcitability seen in the epileptogenic, while ECM changes are linked to synaptic reorganization. The present review will explore glial and ECM homeostatic roles and their potential contribution to tissue plasticity. Finally, we will address how glial, and ECM changes in the epileptogenic zone can be seen in magnetic resonance imaging (MRI), pointing out their importance as markers for the extension of the epileptogenic area. This article is part of the Special Issue "NEWroscience 2018".
Collapse
Affiliation(s)
- Joao Pereira Leite
- Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| | - Jose Eduardo Peixoto-Santos
- Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Department of Neurology and Neurosurgery, Paulista School of Medicine, UNIFESP, Sao Paulo, Brazil
| |
Collapse
|
8
|
Wingert JC, Sorg BA. Impact of Perineuronal Nets on Electrophysiology of Parvalbumin Interneurons, Principal Neurons, and Brain Oscillations: A Review. Front Synaptic Neurosci 2021; 13:673210. [PMID: 34040511 PMCID: PMC8141737 DOI: 10.3389/fnsyn.2021.673210] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/14/2021] [Indexed: 12/11/2022] Open
Abstract
Perineuronal nets (PNNs) are specialized extracellular matrix structures that surround specific neurons in the brain and spinal cord, appear during critical periods of development, and restrict plasticity during adulthood. Removal of PNNs can reinstate juvenile-like plasticity or, in cases of PNN removal during early developmental stages, PNN removal extends the critical plasticity period. PNNs surround mainly parvalbumin (PV)-containing, fast-spiking GABAergic interneurons in several brain regions. These inhibitory interneurons profoundly inhibit the network of surrounding neurons via their elaborate contacts with local pyramidal neurons, and they are key contributors to gamma oscillations generated across several brain regions. Among other functions, these gamma oscillations regulate plasticity associated with learning, decision making, attention, cognitive flexibility, and working memory. The detailed mechanisms by which PNN removal increases plasticity are only beginning to be understood. Here, we review the impact of PNN removal on several electrophysiological features of their underlying PV interneurons and nearby pyramidal neurons, including changes in intrinsic and synaptic membrane properties, brain oscillations, and how these changes may alter the integration of memory-related information. Additionally, we review how PNN removal affects plasticity-associated phenomena such as long-term potentiation (LTP), long-term depression (LTD), and paired-pulse ratio (PPR). The results are discussed in the context of the role of PV interneurons in circuit function and how PNN removal alters this function.
Collapse
Affiliation(s)
- Jereme C Wingert
- Program in Neuroscience, Robert S. Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Barbara A Sorg
- Program in Neuroscience, Robert S. Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, United States
| |
Collapse
|
9
|
Atiq Hassan, Nasir N, Muzammil K. Treatment Strategies to Promote Regeneration in Experimental Spinal Cord Injury Models. NEUROCHEM J+ 2021. [DOI: 10.1134/s1819712421010049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
10
|
Hu J, Rodemer W, Zhang G, Jin LQ, Li S, Selzer ME. Chondroitinase ABC Promotes Axon Regeneration and Reduces Retrograde Apoptosis Signaling in Lamprey. Front Cell Dev Biol 2021; 9:653638. [PMID: 33842481 PMCID: PMC8027354 DOI: 10.3389/fcell.2021.653638] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/26/2021] [Indexed: 12/22/2022] Open
Abstract
Paralysis following spinal cord injury (SCI) is due to failure of axonal regeneration. It is believed that axon growth is inhibited by the presence of several types of inhibitory molecules in central nervous system (CNS), including the chondroitin sulfate proteoglycans (CSPGs). Many studies have shown that digestion of CSPGs with chondroitinase ABC (ChABC) can enhance axon growth and functional recovery after SCI. However, due to the complexity of the mammalian CNS, it is still unclear whether this involves true regeneration or only collateral sprouting by uninjured axons, whether it affects the expression of CSPG receptors such as protein tyrosine phosphatase sigma (PTPσ), and whether it influences retrograde neuronal apoptosis after SCI. In the present study, we assessed the roles of CSPGs in the regeneration of spinal-projecting axons from brainstem neurons, and in the process of retrograde neuronal apoptosis. Using the fluorochrome-labeled inhibitor of caspase activity (FLICA) method, apoptotic signaling was seen primarily in those large, individually identified reticulospinal (RS) neurons that are known to be “bad-regenerators.” Compared to uninjured controls, the number of all RS neurons showing polycaspase activity increased significantly at 2, 4, 8, and 11 weeks post-transection (post-TX). ChABC application to a fresh TX site reduced the number of polycaspase-positive RS neurons at 2 and 11 weeks post-TX, and also reduced the number of active caspase 3-positive RS neurons at 4 weeks post-TX, which confirmed the beneficial role of ChABC treatment in retrograde apoptotic signaling. ChABC treatment also greatly promoted axonal regeneration at 10 weeks post-TX. Correspondingly, PTPσ mRNA expression was reduced in the perikaryon. Previously, PTPσ mRNA expression was shown to correlate with neuronal apoptotic signaling at 2 and 10 weeks post-TX. In the present study, this correlation persisted after ChABC treatment, which suggests that PTPσ may be involved more generally in signaling axotomy-induced retrograde neuronal apoptosis. Moreover, ChABC treatment caused Akt activation (pAkt-308) to be greatly enhanced in brain post-TX, which was further confirmed in individually identified RS neurons. Thus, CSPG digestion not only enhances axon regeneration after SCI, but also inhibits retrograde RS neuronal apoptosis signaling, possibly by reducing PTPσ expression and enhancing Akt activation.
Collapse
Affiliation(s)
- Jianli Hu
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - William Rodemer
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Guixin Zhang
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Li-Qing Jin
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Shuxin Li
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Anatomy and Cell Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Michael E Selzer
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
11
|
Carstens KE, Gloss BR, Alexander GM, Dudek SM. Modified adeno-associated virus targets the bacterial enzyme chondroitinase ABC to select mouse neuronal populations in vivo using the Cre-LoxP system. Eur J Neurosci 2020; 53:4005-4015. [PMID: 33220084 PMCID: PMC8137719 DOI: 10.1111/ejn.15050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 01/01/2023]
Abstract
Current methods of experimentally degrading the specialized extracellular matrix (ECM), perineuronal nets (PNNs) have several limitations. Genetic knockout of ECM components typically has only partial effects on PNNs, and knockout of the major ECM component aggrecan is lethal in mice. Direct injection of the chondroitinase ABC (ChABC) enzyme into the mammalian brain is effective at degrading PNNs in vivo but this method typically lacks consistent, localized spatial targeting of PNN degradation. PNNs also regenerate within weeks after a ChABC injection, thus limiting the ability to perform long‐term studies. Previous work has demonstrated that viral delivery of ChABC in mammalian neurons can successfully degrade PNNs for much longer periods, but the effects are similarly diffuse beyond the injection site. In an effort to gain cell‐specific targeting of ChABC, we designed an adeno‐associated virus encoding ChABC under the control of the Cre‐LoxP system. We show that this virus is effective at targeting the synthesis of ChABC to Cre‐expressing mouse neurons in vivo. Although ChABC expression is localized to the Cre‐expressing neurons, we also note that ChABC is apparently trafficked and secreted at projection sites, as was previously reported for the non‐Cre dependent construct. Overall, this method allows for cell‐specific targeting of ChABC and long‐term degradation of PNNs, which will ultimately serve as an effective tool to study the function of cell‐autonomous regulation of PNNs in vivo. This novel approach may also aid in determining whether specific, long‐term PNN loss is an appropriate strategy for treatment of neurodevelopmental disorders associated with PNN pathology.
Collapse
Affiliation(s)
- Kelly E Carstens
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, NC, USA
| | - Bernd R Gloss
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, NC, USA
| | - Georgia M Alexander
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, NC, USA
| | - Serena M Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, NC, USA
| |
Collapse
|
12
|
Warren PM, Andrews MR, Smith M, Bartus K, Bradbury EJ, Verhaagen J, Fawcett JW, Kwok JCF. Secretion of a mammalian chondroitinase ABC aids glial integration at PNS/CNS boundaries. Sci Rep 2020; 10:11262. [PMID: 32647242 PMCID: PMC7347606 DOI: 10.1038/s41598-020-67526-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/26/2020] [Indexed: 12/13/2022] Open
Abstract
Schwann cell grafts support axonal growth following spinal cord injury, but a boundary forms between the implanted cells and host astrocytes. Axons are reluctant to exit the graft tissue in large part due to the surrounding inhibitory environment containing chondroitin sulphate proteoglycans (CSPGs). We use a lentiviral chondroitinase ABC, capable of being secreted from mammalian cells (mChABC), to examine the repercussions of CSPG digestion upon Schwann cell behaviour in vitro. We show that mChABC transduced Schwann cells robustly secrete substantial quantities of the enzyme causing large-scale CSPG digestion, facilitating the migration and adhesion of Schwann cells on inhibitory aggrecan and astrocytic substrates. Importantly, we show that secretion of the engineered enzyme can aid the intermingling of cells at the Schwann cell-astrocyte boundary, enabling growth of neurites over the putative graft/host interface. These data were echoed in vivo. This study demonstrates the profound effect of the enzyme on cellular motility, growth and migration. This provides a cellular mechanism for mChABC induced functional and behavioural recovery shown in in vivo studies. Importantly, we provide in vitro evidence that mChABC gene therapy is equally or more effective at producing these effects as a one-time application of commercially available ChABC.
Collapse
Affiliation(s)
- Philippa M Warren
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, CB2 0PY, UK. .,Wolfson Centre for Age Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Guy's Campus, London Bridge, London, SE1 1UL, UK. .,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 0PY, UK.
| | - Melissa R Andrews
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, CB2 0PY, UK.,Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Marc Smith
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, CB2 0PY, UK
| | - Katalin Bartus
- Wolfson Centre for Age Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Guy's Campus, London Bridge, London, SE1 1UL, UK
| | - Elizabeth J Bradbury
- Wolfson Centre for Age Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Guy's Campus, London Bridge, London, SE1 1UL, UK
| | - Joost Verhaagen
- Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - James W Fawcett
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, CB2 0PY, UK.,Centre for Reconstructive Neuroscience, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic
| | - Jessica C F Kwok
- Centre for Reconstructive Neuroscience, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic.,School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
13
|
Eggers R, de Winter F, Smit L, Luimens M, Muir EM, Bradbury EJ, Tannemaat MR, Verhaagen J. Combining timed GDNF and ChABC gene therapy to promote long-distance regeneration following ventral root avulsion and repair. FASEB J 2020; 34:10605-10622. [PMID: 32543730 DOI: 10.1096/fj.202000559r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/18/2020] [Accepted: 05/27/2020] [Indexed: 12/21/2022]
Abstract
Ventral root avulsion leads to severe motoneuron degeneration and prolonged distal nerve denervation. After a critical period, a state of chronic denervation develops as repair Schwann cells lose their pro-regenerative properties and inhibitory factors such as CSPGs accumulate in the denervated nerve. In rats with ventral root avulsion injuries, we combined timed GDNF gene therapy delivered to the proximal nerve roots with the digestion of inhibitory CSPGs in the distal denervated nerve using sustained lentiviral-mediated chondroitinase ABC (ChABC) enzyme expression. Following reimplantation of lumbar ventral roots, timed GDNF-gene therapy enhanced motoneuron survival up to 45 weeks and improved axonal outgrowth, electrophysiological recovery, and muscle reinnervation. Despite a timed GDNF expression period, a subset of animals displayed axonal coils. Lentiviral delivery of ChABC enabled digestion of inhibitory CSPGs for up to 45 weeks in the chronically denervated nerve. ChABC gene therapy alone did not enhance motoneuron survival, but led to improved muscle reinnervation and modest electrophysiological recovery during later stages of the regeneration process. Combining GDNF treatment with digestion of inhibitory CSPGs did not have a significant synergistic effect. This study suggests a delicate balance exists between treatment duration and concentration in order to achieve therapeutic effects.
Collapse
Affiliation(s)
- Ruben Eggers
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Fred de Winter
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Lotte Smit
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Maruelle Luimens
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Elizabeth M Muir
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Elizabeth J Bradbury
- King's College London, Regeneration Group, Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), London, UK
| | - Martijn R Tannemaat
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands.,Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - Joost Verhaagen
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands.,Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognition Research, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
14
|
Repair strategies for traumatic spinal cord injury, with special emphasis on novel biomaterial-based approaches. Rev Neurol (Paris) 2020; 176:252-260. [PMID: 31982183 DOI: 10.1016/j.neurol.2019.07.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 12/25/2022]
Abstract
As a part of the central nervous system (CNS), the adult mammalian spinal cord displays only very poor ability for self-repair in response to traumatic lesions, which mostly lead to more or less severe, life-long disability. While even adult CNS neurons have a certain plastic potential, their intrinsic regenerative capacity highly varies among different neuronal populations and in the end, regeneration is almost completely inhibited due to extrinsic factors such as glial scar and cystic cavity formation, excessive and persistent inflammation, presence of various inhibitory molecules, and absence of trophic support and of a growth-supportive extracellular matrix structure. In recent years, a number of experimental animal models have been developed to overcome these obstacles. Since all those studies based on a single approach have yielded only relatively modest functional recovery, it is now consensus that different therapeutic approaches will have to be combined to synergistically overcome the multiple barriers to CNS regeneration, especially in humans. In this review, we particularly emphasize the hope raised by the development of novel, implantable biomaterials that should favor the reconstruction of the damaged nervous tissue, and ultimately allow for functional recovery of sensorimotor functions. Since human spinal cord injury pathology depends on the vertebral level and the severity of the traumatic impact, and since the timing of application of the different therapeutic approaches appears very important, we argue that every case will necessitate individual evaluation, and specific adaptation of therapeutic strategies.
Collapse
|
15
|
Souter L, Kwok JCF. Visualization of Perineuronal Nets in Central Nervous System Tissue Sections. Methods Mol Biol 2020; 2043:251-260. [PMID: 31463917 DOI: 10.1007/978-1-4939-9698-8_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The perineuronal net (PNN) is a specialized extracellular matrix structure that surrounds subpopulations of neurons in the central nervous system (CNS). The appearance of PNNs on the cell surface marks the closure of the critical period during development and has been observed to reduce synaptic plasticity. Perineuronal nets comprise hyaluronan, chondroitin sulfate proteoglycans (CSPGs), link proteins, tenascin-R, and other components, some of which are substrates for a disintegrin-like and metalloprotease domain with thrombospondin type 1 motifs (ADAMTS) proteases. There is a high heterogeneity of PNNs in the CNS. Depending on which part of the CNS is studied, the PNNs may be observed surrounding the soma, or both the soma and proximal dendrites. The most robust marker for PNN is a lectin called Wisteria floribunda agglutinin. Here, we describe a method for preparing tissue for visualization of PNNs in CNS.
Collapse
Affiliation(s)
- Luke Souter
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK.,School of Mechanical Engineering, Faculty of Engineering,, University of Leeds, Leeds, UK
| | - Jessica C F Kwok
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK. .,Institute of Experimental Medicine, Czech Academy of Science, Prague 4, Czech Republic.
| |
Collapse
|
16
|
Meyers EC, Kasliwal N, Solorzano BR, Lai E, Bendale G, Berry A, Ganzer PD, Romero-Ortega M, Rennaker RL, Kilgard MP, Hays SA. Enhancing plasticity in central networks improves motor and sensory recovery after nerve damage. Nat Commun 2019; 10:5782. [PMID: 31857587 PMCID: PMC6923364 DOI: 10.1038/s41467-019-13695-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 11/08/2019] [Indexed: 12/11/2022] Open
Abstract
Nerve damage can cause chronic, debilitating problems including loss of motor control and paresthesia, and generates maladaptive neuroplasticity as central networks attempt to compensate for the loss of peripheral connectivity. However, it remains unclear if this is a critical feature responsible for the expression of symptoms. Here, we use brief bursts of closed-loop vagus nerve stimulation (CL-VNS) delivered during rehabilitation to reverse the aberrant central plasticity resulting from forelimb nerve transection. CL-VNS therapy drives extensive synaptic reorganization in central networks paralleled by improved sensorimotor recovery without any observable changes in the nerve or muscle. Depleting cortical acetylcholine blocks the plasticity-enhancing effects of CL-VNS and consequently eliminates recovery, indicating a critical role for brain circuits in recovery. These findings demonstrate that manipulations to enhance central plasticity can improve sensorimotor recovery and define CL-VNS as a readily translatable therapy to restore function after nerve damage. Peripheral nerve damage generates maladaptive neuroplasticity as central networks attempt to compensate for the loss of peripheral connectivity. Here, the authors reverse the aberrant plasticity via vagus nerve stimulation to elicit synaptic reorganization and to improve sensorimotor recovery.
Collapse
Affiliation(s)
- Eric C Meyers
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.
| | - Nimit Kasliwal
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Bleyda R Solorzano
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Elaine Lai
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Geetanjali Bendale
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Abigail Berry
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Patrick D Ganzer
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Mario Romero-Ortega
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.,School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.,Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Robert L Rennaker
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.,School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.,Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Michael P Kilgard
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.,School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.,Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Seth A Hays
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.,School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.,Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| |
Collapse
|
17
|
Rosenzweig ES, Salegio EA, Liang JJ, Weber JL, Weinholtz CA, Brock JH, Moseanko R, Hawbecker S, Pender R, Cruzen CL, Iaci JF, Caggiano AO, Blight AR, Haenzi B, Huie JR, Havton LA, Nout-Lomas YS, Fawcett JW, Ferguson AR, Beattie MS, Bresnahan JC, Tuszynski MH. Chondroitinase improves anatomical and functional outcomes after primate spinal cord injury. Nat Neurosci 2019; 22:1269-1275. [PMID: 31235933 PMCID: PMC6693679 DOI: 10.1038/s41593-019-0424-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 05/10/2019] [Indexed: 01/07/2023]
Abstract
Inhibitory extracellular matrices form around mature neurons as perineuronal nets containing chondroitin sulfate proteoglycans that limit axonal sprouting after CNS injury. The enzyme chondroitinase (Chase) degrades inhibitory chondroitin sulfate proteoglycans and improves axonal sprouting and functional recovery after spinal cord injury in rodents. We evaluated the effects of Chase in rhesus monkeys that had undergone C7 spinal cord hemisection. Four weeks after hemisection, we administered multiple intraparenchymal Chase injections below the lesion, targeting spinal cord circuits that control hand function. Hand function improved significantly in Chase-treated monkeys relative to vehicle-injected controls. Moreover, Chase significantly increased corticospinal axon growth and the number of synapses formed by corticospinal terminals in gray matter caudal to the lesion. No detrimental effects were detected. This approach appears to merit clinical translation in spinal cord injury.
Collapse
Affiliation(s)
- Ephron S Rosenzweig
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Ernesto A Salegio
- California National Primate Research Center, University of California, Davis, Davis, CA, USA
| | - Justine J Liang
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Janet L Weber
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Chase A Weinholtz
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - John H Brock
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
- Veterans Administration Medical Center, La Jolla, CA, USA
| | - Rod Moseanko
- California National Primate Research Center, University of California, Davis, Davis, CA, USA
| | - Stephanie Hawbecker
- California National Primate Research Center, University of California, Davis, Davis, CA, USA
| | - Roger Pender
- California National Primate Research Center, University of California, Davis, Davis, CA, USA
| | - Christina L Cruzen
- California National Primate Research Center, University of California, Davis, Davis, CA, USA
| | | | | | | | | | - J Russell Huie
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA, USA
| | - Leif A Havton
- Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yvette S Nout-Lomas
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | | | - Adam R Ferguson
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA, USA
| | - Michael S Beattie
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA, USA
| | - Jacqueline C Bresnahan
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA, USA
| | - Mark H Tuszynski
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA.
- Veterans Administration Medical Center, La Jolla, CA, USA.
| |
Collapse
|
18
|
Burnside ER, De Winter F, Didangelos A, James ND, Andreica EC, Layard-Horsfall H, Muir EM, Verhaagen J, Bradbury EJ. Immune-evasive gene switch enables regulated delivery of chondroitinase after spinal cord injury. Brain 2019; 141:2362-2381. [PMID: 29912283 PMCID: PMC6061881 DOI: 10.1093/brain/awy158] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 04/22/2018] [Indexed: 12/12/2022] Open
Abstract
Chondroitinase ABC is a promising preclinical therapy that promotes functional neuroplasticity after CNS injury by degrading extracellular matrix inhibitors. Efficient delivery of chondroitinase ABC to the injured mammalian spinal cord can be achieved by viral vector transgene delivery. This approach dramatically modulates injury pathology and restores sensorimotor functions. However, clinical development of this therapy is limited by a lack of ability to exert control over chondroitinase gene expression. Prior experimental gene regulation platforms are likely to be incompatible with the non-resolving adaptive immune response known to occur following spinal cord injury. Therefore, here we apply a novel immune-evasive dual vector system, in which the chondroitinase gene is under a doxycycline inducible regulatory switch, utilizing a chimeric transactivator designed to evade T cell recognition. Using this novel vector system, we demonstrate tight temporal control of chondroitinase ABC gene expression, effectively removing treatment upon removal of doxycycline. This enables a comparison of short and long-term gene therapy paradigms in the treatment of clinically-relevant cervical level contusion injuries in adult rats. We reveal that transient treatment (2.5 weeks) is sufficient to promote improvement in sensory axon conduction and ladder walking performance. However, in tasks requiring skilled reaching and grasping, only long term treatment (8 weeks) leads to significantly improved function, with rats able to accurately grasp and retrieve sugar pellets. The late emergence of skilled hand function indicates enhanced neuroplasticity and connectivity and correlates with increased density of vGlut1+ innervation in spinal cord grey matter, particularly in lamina III–IV above and below the injury. Thus, our novel gene therapy system provides an experimental tool to study temporal effects of extracellular matrix digestion as well as an encouraging step towards generating a safer chondroitinase gene therapy strategy, longer term administration of which increases neuroplasticity and recovery of descending motor control. This preclinical study could have a significant impact for tetraplegic individuals, for whom recovery of hand function is an important determinant of independence, and supports the ongoing development of chondroitinase gene therapy towards clinical application for the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Emily R Burnside
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK
| | - Fred De Winter
- Netherlands Institute for Neuroscience, Laboratory for Neuroregeneration, 1105 BA Amsterdam, The Netherlands
| | - Athanasios Didangelos
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK
| | - Nicholas D James
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK
| | - Elena-Cristina Andreica
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK
| | - Hugo Layard-Horsfall
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK
| | - Elizabeth M Muir
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3EG, UK
| | - Joost Verhaagen
- Netherlands Institute for Neuroscience, Laboratory for Neuroregeneration, 1105 BA Amsterdam, The Netherlands
| | - Elizabeth J Bradbury
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK
| |
Collapse
|
19
|
The roles of perineuronal nets and the perinodal extracellular matrix in neuronal function. Nat Rev Neurosci 2019; 20:451-465. [PMID: 31263252 DOI: 10.1038/s41583-019-0196-3] [Citation(s) in RCA: 288] [Impact Index Per Article: 57.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2019] [Indexed: 01/09/2023]
Abstract
Perineuronal nets (PNNs) are extracellular matrix (ECM) chondroitin sulfate proteoglycan (CSPG)-containing structures that surround the soma and dendrites of various mammalian neuronal cell types. PNNs appear during development around the time that the critical periods for developmental plasticity end and are important for both their onset and closure. A similar structure - the perinodal ECM - surrounds the axonal nodes of Ranvier and appears as myelination is completed, acting as an ion-diffusion barrier that affects axonal conduction speed. Recent work has revealed the importance of PNNs in controlling plasticity in the CNS. Digestion, blocking or removal of PNNs influences functional recovery after a variety of CNS lesions. PNNs have further been shown to be involved in the regulation of memory and have been implicated in a number of psychiatric disorders.
Collapse
|
20
|
Mukhamedshina Y, Povysheva T, Nikolenko V, Kuznecov M, Rizvanov A, Chelyshev Y. Upregulation of proteoglycans in the perilesion perimeter in ventral horns after spinal cord injury. Neurosci Lett 2019; 704:220-228. [DOI: 10.1016/j.neulet.2019.04.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 03/05/2019] [Accepted: 04/02/2019] [Indexed: 12/18/2022]
|
21
|
Lipachev N, Arnst N, Melnikova A, Jäälinoja H, Kochneva A, Zhigalov A, Kulesskaya N, Aganov AV, Mavlikeev M, Rauvala H, Kiyasov AP, Paveliev M. Quantitative changes in perineuronal nets in development and posttraumatic condition. J Mol Histol 2019; 50:203-216. [PMID: 30903543 DOI: 10.1007/s10735-019-09818-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/11/2019] [Indexed: 11/30/2022]
Abstract
Perineuronal net (PNN) is a highly structured portion of the CNS extracellular matrix (ECM) regulating synaptic plasticity and a range of pathologic conditions including posttraumatic regeneration and epilepsy. Here we studied Wisteria floribunda agglutinin-stained histological sections to quantify the PNN size and enrichment of chondroitin sulfates in mouse brain and spinal cord. Somatosensory cortex sections were examined during the period of PNN establishment at postnatal days 14, 21 and 28. The single cell PNN size and the chondroitin sulfate intensity were quantified for all cortex layers and specifically for the cortical layer IV which has the highest density of PNN-positive neurons. We demonstrate that the chondroitin sulfate proteoglycan staining intensity is increased between P14 and P28 while the PNN size remains unchanged. We then addressed posttraumatic changes of the PNN expression in laminae 6 and 7 of cervical spinal cord following hemisection injury. We demonstrate increase of the chondroitin sulfate content at 1.6-1.8 mm rostrally from the injury site and increase of the density of PNN-bearing cells at 0.4-1.2 mm caudally from the injury site. We further demonstrate decrease of the single cell PNN area at 0.2 mm caudally from the injury site suggesting that the PNN ECM takes part in the posttraumatic tissue rearrangement in the spinal cord. Our results demonstrate new insights on the PNN structure dynamics in the developing and posttraumatic CNS.
Collapse
Affiliation(s)
- Nikita Lipachev
- Neuroscience Center, University of Helsinki, Viikinkaari 4, P.O.Box 56, 00790, Helsinki, Finland.,Institute of Physics, Kazan Federal University, Kazan Kremlyovskaya 16a, Tatarstan, Russia, 420111
| | - Nikita Arnst
- Neuroscience Center, University of Helsinki, Viikinkaari 4, P.O.Box 56, 00790, Helsinki, Finland.,Institut für Biochemie und Biophysik, Friedrich-Schiller-Universität Jena, Hans-Knöll-Str.2, 07745, Jena, Germany
| | - Anastasiia Melnikova
- Neuroscience Center, University of Helsinki, Viikinkaari 4, P.O.Box 56, 00790, Helsinki, Finland.,Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan K.Marx 74, Tatarstan, Russia, 420012
| | - Harri Jäälinoja
- Institute of Biotechnology, University of Helsinki, Viikinkaari 9, P.O.Box 56, 00790, Helsinki, Finland
| | - Anastasiya Kochneva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan K.Marx 74, Tatarstan, Russia, 420012
| | - Alexander Zhigalov
- Neuroscience Center, University of Helsinki, Viikinkaari 4, P.O.Box 56, 00790, Helsinki, Finland
| | - Natalia Kulesskaya
- Neuroscience Center, University of Helsinki, Viikinkaari 4, P.O.Box 56, 00790, Helsinki, Finland
| | - Albert V Aganov
- Institute of Physics, Kazan Federal University, Kazan Kremlyovskaya 16a, Tatarstan, Russia, 420111
| | - Mikhail Mavlikeev
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan K.Marx 74, Tatarstan, Russia, 420012
| | - Heikki Rauvala
- Neuroscience Center, University of Helsinki, Viikinkaari 4, P.O.Box 56, 00790, Helsinki, Finland
| | - Andrey P Kiyasov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan K.Marx 74, Tatarstan, Russia, 420012
| | - Mikhail Paveliev
- Neuroscience Center, University of Helsinki, Viikinkaari 4, P.O.Box 56, 00790, Helsinki, Finland. .,Danish Research Institute of Translational Neuroscience, Aarhus University, Ole Worms Allé 3, 8000, Aarhus C, Denmark.
| |
Collapse
|
22
|
Clemente F, Valle G, Controzzi M, Strauss I, Iberite F, Stieglitz T, Granata G, Rossini PM, Petrini F, Micera S, Cipriani C. Intraneural sensory feedback restores grip force control and motor coordination while using a prosthetic hand. J Neural Eng 2019; 16:026034. [DOI: 10.1088/1741-2552/ab059b] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
23
|
Dyck S, Kataria H, Akbari-Kelachayeh K, Silver J, Karimi-Abdolrezaee S. LAR and PTPσ receptors are negative regulators of oligodendrogenesis and oligodendrocyte integrity in spinal cord injury. Glia 2018; 67:125-145. [PMID: 30394599 DOI: 10.1002/glia.23533] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 12/26/2022]
Abstract
Following spinal cord injury (SCI), the population of mature oligodendrocytes undergoes substantial cell death; promoting their preservation and replacement is a viable strategy for preserving axonal integrity and white matter repair in the injured spinal cord. Dramatic upregulation of matrix chondroitin sulfate proteoglycans (CSPGs) is shown to pose an obstacle to endogenous repair processes, and targeting CSPGs improves functional recovery after SCI. However, the cellular and molecular mechanisms underlying the inhibitory effects of CSPGs remain largely undefined. Modulation of CSPGs specific signaling receptors, leukocyte common antigen-related (LAR), and protein tyrosine phosphatase-sigma (PTPσ) allows us to uncover the role and mechanisms of CSPGs in regulating oligodendrocytes in SCI. Here, utilizing specific functionally blocking peptides in a clinically relevant model of contusive/compressive SCI in the rat, we demonstrate that inhibition of PTPσ and LAR receptors promotes oligodendrogenesis by endogenous precursor cells, attenuates caspase 3-mediated cell death in mature oligodendrocytes, and preserves myelin. In parallel in vitro systems, we have unraveled that CSPGs directly induce apoptosis in populations of neural precursor cells and oligodendrocyte progenitor cells and limit their ability for oligodendrocyte differentiation, maturation, and myelination. These negative effects of CSPGs are mediated through the activation of both LAR and PTPσ receptors and the downstream Rho/ROCK pathway. Thus, we have identified a novel inhibitory role for PTPσ and LAR in regulating oligodendrocyte differentiation and apoptosis in the injured adult spinal cord and a new feasible therapeutic strategy for optimizing endogenous cell replacement following SCI.
Collapse
Affiliation(s)
- Scott Dyck
- Department of Physiology and Pathophysiology, The Regenerative Medicine Program, The Spinal Cord Research Center, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hardeep Kataria
- Department of Physiology and Pathophysiology, The Regenerative Medicine Program, The Spinal Cord Research Center, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Khashayar Akbari-Kelachayeh
- Department of Physiology and Pathophysiology, The Regenerative Medicine Program, The Spinal Cord Research Center, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jerry Silver
- Department of Neuroscience, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, The Regenerative Medicine Program, The Spinal Cord Research Center, University of Manitoba, Winnipeg, Manitoba, Canada.,Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
24
|
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: 513] [Impact Index Per Article: 85.5] [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
|
25
|
Aboseria M, Clemente F, Engels LF, Cipriani C. Discrete Vibro-Tactile Feedback Prevents Object Slippage in Hand Prostheses More Intuitively Than Other Modalities. IEEE Trans Neural Syst Rehabil Eng 2018; 26:1577-1584. [DOI: 10.1109/tnsre.2018.2851617] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
26
|
The Impact of Perineuronal Net Digestion Using Chondroitinase ABC on the Intrinsic Physiology of Cortical Neurons. Neuroscience 2018; 388:23-35. [PMID: 30004010 DOI: 10.1016/j.neuroscience.2018.07.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 06/29/2018] [Accepted: 07/03/2018] [Indexed: 11/21/2022]
Abstract
Perineuronal nets (PNNs) are a form of aggregate Extracellular Matrix (ECM) in the brain. Recent evidence suggests that the postnatal deposition of PNNs may play an active role in regulating neuroplasticity and, potentially, neurological disorders. Observations of high levels of PNN expression around somas, proximal dendrites, and axon initial segments of a subtype of neurons have also led to proposals that PNNs may modulate the intrinsic properties of the neurons they ensheathe. While high levels of PNNs are postnatally expressed throughout the neocortex, it is still unclear how they impact the neuronal physiology of the many classes and subtypes of neurons that exist. In this study, we demonstrate that Chondroitinase ABC digestion of PNNs from acute cortical slices from juvenile mice (P28-35) resulted in neuron-specific impacts on intrinsic physiology. Fast spiking (FS) interneurons showed decreased input resistance, resting membrane potential (RMP), reduced action potential (AP) peaks and altered spontaneous synaptic inputs. Low-Threshold Spiking interneurons showed altered rebound depolarizations and decreased frequency of spontaneous synaptic inputs. Putative excitatory neurons; regular spiking, bursting, and doublet phenotypes did not demonstrate any alterations. Our data indicate that chABC-sensitive PNNs may specifically regulate the intrinsic and synaptic physiology of inhibitory interneurons.
Collapse
|
27
|
Klein JA, Meng L, Zaia J. Deep Sequencing of Complex Proteoglycans: A Novel Strategy for High Coverage and Site-specific Identification of Glycosaminoglycan-linked Peptides. Mol Cell Proteomics 2018; 17:1578-1590. [PMID: 29773674 DOI: 10.1074/mcp.ra118.000766] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/09/2018] [Indexed: 12/17/2022] Open
Abstract
Proteoglycans are distributed in all animal tissues and play critical, multifaceted, physiological roles. Expressed in a spatially and temporally regulated manner, these molecules regulate interactions among growth factors and cell surface receptors and play key roles in basement membranes and other extracellular matrices. Because of the high degree of glycosylation by glycosaminoglycan (GAG), N-glycan and mucin-type O-glycan classes, the peptide sequence coverage of complex proteoglycans is revealed poorly by standard mass spectrometry-based proteomics methods. As a result, there is little information concerning how proteoglycan site specific glycosylation changes during normal and pathological processes. Here, we developed a workflow to improve sequence coverage and identification of glycosylated peptides in proteoglycans. We applied this workflow to the small leucine-rich proteoglycan decorin and three hyalectan proteoglycans: neurocan, brevican, and aggrecan.We characterized glycosylation of these proteoglycans using LC-MS methods easily implemented on instruments widely used in proteomics laboratories. For decorin, we assigned the linker-glycosite and three N-glycosylation sites. For neurocan and brevican, we identified densely glycosylated mucin-like regions in the extended domains. For aggrecan, we identified 50 linker-glycosites and mucin-type O-glycosites in the extended region and N-glycosites in the globular domains, many of which are novel and have not been observed previously. Most importantly, we demonstrate an LC-MS and bioinformatics approach that will enable routine analysis of proteoglycan glycosylation from biological samples to assess their role in pathophysiology.
Collapse
Affiliation(s)
- Joshua A Klein
- From the ‡Department of Biochemistry, Center for Biomedical Mass Spectrometry.,§Bioinformatics Program Boston University, Boston, Massachusetts 02118
| | - Le Meng
- From the ‡Department of Biochemistry, Center for Biomedical Mass Spectrometry
| | - Joseph Zaia
- From the ‡Department of Biochemistry, Center for Biomedical Mass Spectrometry; .,§Bioinformatics Program Boston University, Boston, Massachusetts 02118
| |
Collapse
|
28
|
Cizkova D, Cubinkova V, Smolek T, Murgoci AN, Danko J, Vdoviakova K, Humenik F, Cizek M, Quanico J, Fournier I, Salzet M. Localized Intrathecal Delivery of Mesenchymal Stromal Cells Conditioned Medium Improves Functional Recovery in a Rat Model of Spinal Cord Injury. Int J Mol Sci 2018; 19:ijms19030870. [PMID: 29543759 PMCID: PMC5877731 DOI: 10.3390/ijms19030870] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/04/2018] [Accepted: 03/09/2018] [Indexed: 12/16/2022] Open
Abstract
It was recently shown that the conditioned medium (CM) of mesenchymal stem cells can enhance viability of neural and glial cell populations. In the present study, we have investigated a cell-free approach via CM from rat bone marrow stromal cells (MScCM) applied intrathecally (IT) for spinal cord injury (SCI) recovery in adult rats. Functional in vitro test on dorsal root ganglion (DRG) primary cultures confirmed biological properties of collected MScCM for production of neurosphere-like structures and axon outgrowth. Afterwards, rats underwent SCI and were treated with IT delivery of MScCM or vehicle at postsurgical Days 1, 5, 9, and 13, and left to survive 10 weeks. Rats that received MScCM showed significantly higher motor function recovery, increase in spared spinal cord tissue, enhanced GAP-43 expression and attenuated inflammation in comparison with vehicle-treated rats. Spared tissue around the lesion site was infiltrated with GAP-43-labeled axons at four weeks that gradually decreased at 10 weeks. Finally, a cytokine array performed on spinal cord extracts after MScCM treatment revealed decreased levels of IL-2, IL-6 and TNFα when compared to vehicle group. In conclusion, our results suggest that molecular cocktail found in MScCM is favorable for final neuroregeneration after SCI.
Collapse
Affiliation(s)
- Dasa Cizkova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 10 Bratislava, Slovakia.
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia.
- Université de Lille, Inserm, U-1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France.
| | - Veronika Cubinkova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 10 Bratislava, Slovakia.
| | - Tomas Smolek
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 10 Bratislava, Slovakia.
| | - Adriana-Natalia Murgoci
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 10 Bratislava, Slovakia.
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia.
- Université de Lille, Inserm, U-1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France.
| | - Jan Danko
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia.
| | - Katarina Vdoviakova
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia.
| | - Filip Humenik
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia.
| | - Milan Cizek
- Department of Epizootology and Parasitology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia.
| | - Jusal Quanico
- Université de Lille, Inserm, U-1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France.
| | - Isabelle Fournier
- Université de Lille, Inserm, U-1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France.
| | - Michel Salzet
- Université de Lille, Inserm, U-1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France.
| |
Collapse
|
29
|
Takeda A, Shuto M, Funakoshi K. Chondroitin Sulfate Expression in Perineuronal Nets After Goldfish Spinal Cord Lesion. Front Cell Neurosci 2018; 12:63. [PMID: 29662439 PMCID: PMC5890146 DOI: 10.3389/fncel.2018.00063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 02/22/2018] [Indexed: 12/03/2022] Open
Abstract
Perineuronal nets (PNNs) surrounding neuronal cell bodies regulate neuronal plasticity during development, but their roles in regeneration are unclear. In the PNNs, chondroitin sulfate (CS) is assumed to be involved in inhibiting contact formation. Here, we examined CS expression in PNNs in the ventral horn of a goldfish hemisected spinal cord in which descending axons regenerate beyond the lesion to connect with distal spinal neurons. In intact fish, chondroitin sulfate A (CS-A)–positive PNNs accounted for 5.0% of HuC/D-immunoreactive neurons, and 48% of choline acetyltransferase (ChAT)-immunoreactive neurons. At 2, 4 and 8 weeks after spinal hemisection, CS-A–positive PNNs accounted for 8.4%–9.9% of HuC/D-immunoreactive neurons, and 50%–60% of ChAT-immunoreactive neurons, which was not significantly different from intact fish. Chondroitin sulfate C (CS-C)–positive PNNs accounted for 6.4% of HuC/D-immunoreactive neuron, and 67% of ChAT-immunoreactive neurons in intact fish. At 2, 4 and 8 weeks after spinal hemisection, CS-C–positive PNNs accounted for 7.9%, 5.5% and 4.3%, respectively, of HuC/D-immunoreactive neurons, and 65%, 52% and 42%, respectively, of ChAT-immunoreactive neurons, demonstrating a significant decrease at 4 and 8 weeks after spinal hemisection. Among ventral horn neurons that received descending axons labeled with tetramethylrhodamine dextran amine (RDA) applied at the level of the first spinal nerve, CS-A–positive PNNs accounted for 53% of HuC/D-immunoreactive neurons. At 2 and 4 weeks after spinal hemisection, CS-A–positive PNNs accounted for 57% and 56% of HuC/D-immunoreactive neurons, which was not significantly different from intact fish. CS-C–positive PNNs, accounted for 48% of HuC/D-immunoreactive neurons that received RDA-labeled axons. At 2 and 4 weeks after spinal hemisection, CS-C–positive PNNs significantly decreased to 22% of the HuC/D-immunoreactive neurons, and by 4 weeks after spinal hemisection they had returned to 47%. These findings suggest that CS expression is maintained in the PNNs after spinal cord lesion, and that the descending axons regenerate to preferentially terminate on neurons not covered with CS-C–positive PNNs. Therefore, CS-C in the PNNs possibly inhibits new contact with descending axons, and plasticity in the spinal neurons might be endowed by downregulation of CS-C in the PNNs in the regeneration process after spinal hemisection in goldfish.
Collapse
Affiliation(s)
- Akihito Takeda
- Department of Neuroanatomy, Yokohama City University School of Medicine, Yokohama, Japan
| | - Masashige Shuto
- Yokohama City University School of Medicine, Yokohama, Japan
| | - Kengo Funakoshi
- Department of Neuroanatomy, Yokohama City University School of Medicine, Yokohama, Japan
| |
Collapse
|
30
|
Carwardine D, Prager J, Neeves J, Muir EM, Uney J, Granger N, Wong LF. Transplantation of canine olfactory ensheathing cells producing chondroitinase ABC promotes chondroitin sulphate proteoglycan digestion and axonal sprouting following spinal cord injury. PLoS One 2017; 12:e0188967. [PMID: 29228020 PMCID: PMC5724818 DOI: 10.1371/journal.pone.0188967] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 11/16/2017] [Indexed: 11/19/2022] Open
Abstract
Olfactory ensheathing cell (OEC) transplantation is a promising strategy for treating spinal cord injury (SCI), as has been demonstrated in experimental SCI models and naturally occurring SCI in dogs. However, the presence of chondroitin sulphate proteoglycans within the extracellular matrix of the glial scar can inhibit efficient axonal repair and limit the therapeutic potential of OECs. Here we have used lentiviral vectors to genetically modify canine OECs to continuously deliver mammalian chondroitinase ABC at the lesion site in order to degrade the inhibitory chondroitin sulphate proteoglycans in a rodent model of spinal cord injury. We demonstrate that these chondroitinase producing canine OECs survived at 4 weeks following transplantation into the spinal cord lesion and effectively digested chondroitin sulphate proteoglycans at the site of injury. There was evidence of sprouting within the corticospinal tract rostral to the lesion and an increase in the number of corticospinal axons caudal to the lesion, suggestive of axonal regeneration. Our results indicate that delivery of the chondroitinase enzyme can be achieved with the genetically modified OECs to increase axon growth following SCI. The combination of these two promising approaches is a potential strategy for promoting neural regeneration following SCI in veterinary practice and human patients.
Collapse
Affiliation(s)
- Darren Carwardine
- School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Jonathan Prager
- School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Jacob Neeves
- School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Elizabeth M. Muir
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - James Uney
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Nicolas Granger
- School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Liang-Fong Wong
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
- * E-mail:
| |
Collapse
|
31
|
van 't Spijker HM, Kwok JCF. A Sweet Talk: The Molecular Systems of Perineuronal Nets in Controlling Neuronal Communication. Front Integr Neurosci 2017; 11:33. [PMID: 29249944 PMCID: PMC5717013 DOI: 10.3389/fnint.2017.00033] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/14/2017] [Indexed: 11/13/2022] Open
Abstract
Perineuronal nets (PNNs) are mesh-like structures, composed of a hierarchical assembly of extracellular matrix molecules in the central nervous system (CNS), ensheathing neurons and regulating plasticity. The mechanism of interactions between PNNs and neurons remain uncharacterized. In this review, we pose the question: how do PNNs regulate communication to and from neurons? We provide an overview of the current knowledge on PNNs with a focus on the cellular interactions. PNNs ensheath a subset of the neuronal population with distinct molecular aspects in different areas of the CNS. PNNs control neuronal communication through molecular interactions involving specific components of the PNNs. This review proposes that the PNNs are an integral part of neurons, crucial for the regulation of plasticity in the CNS.
Collapse
Affiliation(s)
- Heleen M van 't Spijker
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Jessica C F Kwok
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom.,Czech Academy of Sciences, Institute of Experimental Medicine, Centre of Reconstructive Neurosciences, Prague, Czechia
| |
Collapse
|
32
|
Perineuronal Nets Suppress Plasticity of Excitatory Synapses on CA2 Pyramidal Neurons. J Neurosci 2017; 36:6312-20. [PMID: 27277807 DOI: 10.1523/jneurosci.0245-16.2016] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 05/02/2016] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Long-term potentiation of excitatory synapses on pyramidal neurons in the stratum radiatum rarely occurs in hippocampal area CA2. Here, we present evidence that perineuronal nets (PNNs), a specialized extracellular matrix typically localized around inhibitory neurons, also surround mouse CA2 pyramidal neurons and envelop their excitatory synapses. CA2 pyramidal neurons express mRNA transcripts for the major PNN component aggrecan, identifying these neurons as a novel source for PNNs in the hippocampus. We also found that disruption of PNNs allows synaptic potentiation of normally plasticity-resistant excitatory CA2 synapses; thus, PNNs play a role in restricting synaptic plasticity in area CA2. Finally, we found that postnatal development of PNNs on CA2 pyramidal neurons is modified by early-life enrichment, suggesting that the development of circuits containing CA2 excitatory synapses are sensitive to manipulations of the rearing environment. SIGNIFICANCE STATEMENT Perineuronal nets (PNNs) are thought to play a major role in restricting synaptic plasticity during postnatal development, and are altered in several models of neurodevelopmental disorders, such as schizophrenia and Rett syndrome. Although PNNs have been predominantly studied in association with inhibitory neurons throughout the brain, we describe a dense expression of PNNs around excitatory pyramidal neurons in hippocampal area CA2. We also provide insight into a previously unrecognized role for PNNs in restricting plasticity at excitatory synapses and raise the possibility of an early critical period of hippocampal plasticity that may ultimately reveal a key mechanism underlying learning and memory impairments of PNN-associated neurodevelopmental disorders.
Collapse
|
33
|
Schultz AJ, Rotterman TM, Dwarakanath A, Alvarez FJ. VGLUT1 synapses and P-boutons on regenerating motoneurons after nerve crush. J Comp Neurol 2017; 525:2876-2889. [PMID: 28543879 DOI: 10.1002/cne.24244] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/03/2017] [Accepted: 05/11/2017] [Indexed: 01/05/2023]
Abstract
Stretch-sensitive Ia afferent monosynaptic connections with motoneurons form the stretch reflex circuit. After nerve transection, Ia afferent synapses and stretch reflexes are permanently lost, even after regeneration and reinnervation of muscle by motor and sensory afferents is completed in the periphery. This loss greatly affects full recovery of motor function. However, after nerve crush, reflex muscle forces during stretch do recover after muscle reinnervation and reportedly exceed 140% baseline values. This difference might be explained by structural preservation after crush of Ia afferent synapses on regenerating motoneurons and decreased presynaptic inhibitory control. We tested these possibilities in rats after crushing the tibial nerve (TN), and using Vesicular GLUtamate Transporter 1 (VGLUT1) and the 65 kDa isoform of glutamic acid-decarboxylase (GAD65) as markers of, respectively, Ia afferent synapses and presynaptic inhibition (P-boutons) on retrogradely labeled motoneurons. We analyzed motoneurons during regeneration (21 days post crush) and after they reinnervate muscle (3 months). The results demonstrate a significant loss of VGLUT1 terminals on dendrites and cell bodies at both 21 days and 3 months post-crush. However, in both cellular compartments, the reductions were small compared to those observed after TN full transection. In addition, we found a significant decrease in the number of GAD65 P-boutons per VGLUT1 terminal and their coverage of VGLUT1 boutons. The results support the hypothesis that better preservation of Ia afferent synapses and a change in presynaptic inhibition could contribute to maintain or even increase the stretch reflex after nerve crush and by difference to nerve transection.
Collapse
Affiliation(s)
- Adam J Schultz
- Department of Physiology, Emory University, Atlanta, Georgia
| | | | | | | |
Collapse
|
34
|
De Luca C, Papa M. Matrix Metalloproteinases, Neural Extracellular Matrix, and Central Nervous System Pathology. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 148:167-202. [PMID: 28662822 DOI: 10.1016/bs.pmbts.2017.04.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The functionality and stability of the central nervous system (CNS) pabulum, called neural extracellular matrix (nECM), is paramount for the maintenance of a healthy network. The loosening or the damage of the scaffold disrupts synaptic transmission with the consequent imbalance of the neurotransmitters, reactive cells invasion, astrocytosis, new matrix deposition, digestion of the previous structure and ultimately, maladaptive plasticity with the loss of neuronal viability. nECM is constantly affected by CNS disorders, particularly in chronic modifying such as neurodegenerative disease, or in acute/subacute with chronic sequelae, like cerebrovascular and inflammatory pathology. Matrix metalloproteinases (MMPs) are the main interfering agent of nECM, guiding the balance of degradation and new deposition of proteins such as proteoglycans and glycoproteins, or glycosaminoglycans, such as hyaluronic acid. Activation of these enzymes is modulated by their physiologic inhibitors, the tissue inhibitors of MMPs or via other proteases inhibitors, as well as genetic or epigenetic up- or downregulation through molecular interaction or receptor activation. The appropriate understanding of the pathways underlying nECM modifications in CNS pathology is probably one of the pivotal future directions to identify the healthy brain network and subsequently design new therapies to interfere with the progression of the CNS disease and eventually find appropriate therapies.
Collapse
Affiliation(s)
- Ciro De Luca
- Laboratory of Neuronal Networks, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Michele Papa
- Laboratory of Neuronal Networks, University of Campania "Luigi Vanvitelli", Naples, Italy; SYSBIO, Centre for Systems Biology, University of Milano-Bicocca, Milano, Italy.
| |
Collapse
|
35
|
Meyers EC, Granja R, Solorzano BR, Romero-Ortega M, Kilgard MP, Rennaker RL, Hays S. Median and ulnar nerve injuries reduce volitional forelimb strength in rats. Muscle Nerve 2017; 56:1149-1154. [PMID: 28120500 DOI: 10.1002/mus.25590] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 01/13/2017] [Accepted: 01/16/2017] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Peripheral nerve injuries (PNI) are among the leading causes of physical disability in the United States. The majority of injuries occur in the upper extremities, and functional recovery is often limited. Robust animal models are critical first steps for developing effective therapies to restore function after PNI. METHODS We developed an automated behavioral assay that provides quantitative measurements of volitional forelimb strength in rats. Multiple forelimb PNI models involving the median and ulnar nerves were used to assess forelimb function for up to 13 weeks postinjury. RESULTS Despite multiple weeks of task-oriented training following injury, rats exhibit significant reductions in multiple quantitative parameters of forelimb function, including maximal pull force and speed of force generation. DISCUSSION This study demonstrates that the isometric pull task is an effective method of evaluating forelimb function following PNI and may aid in development of therapeutic interventions to restore function. Muscle Nerve 56: 1149-1154, 2017.
Collapse
Affiliation(s)
- Eric C Meyers
- The University of Texas at Dallas, Texas Biomedical Device Center, 800 West Campbell Road, Richardson, Texas, 75080-3021, USA.,The University of Texas at Dallas, Erik Jonsson School of Engineering and Computer Science, Richardson, Texas, USA
| | - Rafael Granja
- The University of Texas at Dallas, Texas Biomedical Device Center, 800 West Campbell Road, Richardson, Texas, 75080-3021, USA.,The University of Texas at Dallas, Erik Jonsson School of Engineering and Computer Science, Richardson, Texas, USA
| | - Bleyda R Solorzano
- The University of Texas at Dallas, Texas Biomedical Device Center, 800 West Campbell Road, Richardson, Texas, 75080-3021, USA
| | - Mario Romero-Ortega
- The University of Texas at Dallas, Texas Biomedical Device Center, 800 West Campbell Road, Richardson, Texas, 75080-3021, USA.,The University of Texas at Dallas, Erik Jonsson School of Engineering and Computer Science, Richardson, Texas, USA
| | - Michael P Kilgard
- The University of Texas at Dallas, Texas Biomedical Device Center, 800 West Campbell Road, Richardson, Texas, 75080-3021, USA.,The University of Texas at Dallas, School of Behavioral Brain Sciences, Richardson, Texas, USA
| | - Robert L Rennaker
- The University of Texas at Dallas, Texas Biomedical Device Center, 800 West Campbell Road, Richardson, Texas, 75080-3021, USA.,The University of Texas at Dallas, Erik Jonsson School of Engineering and Computer Science, Richardson, Texas, USA.,The University of Texas at Dallas, School of Behavioral Brain Sciences, Richardson, Texas, USA
| | - Seth Hays
- The University of Texas at Dallas, Texas Biomedical Device Center, 800 West Campbell Road, Richardson, Texas, 75080-3021, USA.,The University of Texas at Dallas, Erik Jonsson School of Engineering and Computer Science, Richardson, Texas, USA
| |
Collapse
|
36
|
Slaker ML, Harkness JH, Sorg BA. A standardized and automated method of perineuronal net analysis using Wisteria floribunda agglutinin staining intensity. IBRO Rep 2016; 1:54-60. [PMID: 28713865 PMCID: PMC5507617 DOI: 10.1016/j.ibror.2016.10.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Perineuronal nets (PNNs) are aggregations of extracellular matrix molecules that are critical for plasticity. Their altered development or changes during adulthood appear to contribute to a wide range of diseases/disorders of the brain. An increasing number of studies examining the contribution of PNN to various behaviors and types of plasticity have analyzed the fluorescence intensity of Wisteria floribunda agglutinin (WFA) as an indirect measure of the maturity of PNNs, with brighter WFA staining corresponding to a more mature PNN and dim WFA staining corresponding to an immature PNN. However, a clearly-defined and unified method for assessing the intensity of PNNs is critical to allow us to make comparisons across studies and to advance our understanding of how PNN plasticity contributes to normal brain function and brain disease states. Here we examined methods of PNN intensity quantification and demonstrate that creating a region of interest around each PNN and subtracting appropriate background is a viable method for PNN intensity quantification that can be automated. This method produces less variability and bias across experiments compared to other published analyses, and this method increases reproducibility and reliability of PNN intensity measures, which is critical for comparisons across studies in this emerging field.
Collapse
Affiliation(s)
- Megan L Slaker
- Department of Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA, 98686
| | - John H Harkness
- Department of Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA, 98686
| | - Barbara A Sorg
- Department of Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA, 98686
| |
Collapse
|
37
|
Dawson G. Quantum dots and potential therapy for Krabbe's disease. J Neurosci Res 2016; 94:1293-303. [PMID: 27638611 PMCID: PMC5027984 DOI: 10.1002/jnr.23805] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 06/08/2016] [Accepted: 06/10/2016] [Indexed: 12/27/2022]
Abstract
Enzyme replacement therapy and substrate reduction therapy have proved useful in reversing many pathological consequences of many nonneural lysosomal storage diseases but have not yet reversed pathology or influenced disease outcome in Krabbe's disease (KD). This Review discusses the relative merits of stem cell therapy, molecular chaperone therapy, gene therapy, substrate reduction therapy, enzyme replacement therapy, and combination therapy. Given the limitations of these approaches, this Review introduces the idea of using tiny, 6-nm, intensely fluorescent quantum dots (QDs) to deliver a cell-penetrating peptide and 6 histidine residue-tagged β-D-galactocerebrosidase across the blood-brain barrier. We can therefore follow the fate of injected material and ensure that all targets are reached and that accumulated material is degraded. Uptake of lysosomal hydrolases is a complex process, and the cell-penetrating peptide JB577 is uniquely able to promote endosomal egress of the QD cargo. This Review further shows that uptake may depend on the charge of the coating of the QD, specifically, that negative charge directs the cargo to neurons. Because KD involves primarily glia, specifically oligodendroglia, we experiment with many coatings and discover a coating (polyethylene glycol 600 amino) that has a positive charge and targets oligodendrocytes. A similar effect is achieved by treating with chondroitinase ABC to degrade the extracellular matrix, indicating that enzyme replacement has several hurdles to overcome before it can become a routine CNS therapy. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Glyn Dawson
- Department of Pediatrics, University of Chicago, Chicago, Illinois.
| |
Collapse
|
38
|
Perineuronal Nets Enhance the Excitability of Fast-Spiking Neurons. eNeuro 2016; 3:eN-NWR-0112-16. [PMID: 27570824 PMCID: PMC4987413 DOI: 10.1523/eneuro.0112-16.2016] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 07/03/2016] [Accepted: 07/06/2016] [Indexed: 12/02/2022] Open
Abstract
Perineuronal nets (PNNs) are specialized complexes of extracellular matrix molecules that surround the somata of fast-spiking neurons throughout the vertebrate brain. PNNs are particularly prevalent throughout the auditory brainstem, which transmits signals with high speed and precision. It is unknown whether PNNs contribute to the fast-spiking ability of the neurons they surround. Whole-cell recordings were made from medial nucleus of the trapezoid body (MNTB) principal neurons in acute brain slices from postnatal day 21 (P21) to P27 mice. PNNs were degraded by incubating slices in chondroitinase ABC (ChABC) and were compared to slices that were treated with a control enzyme (penicillinase). ChABC treatment did not affect the ability of MNTB neurons to fire at up to 1000 Hz when driven by current pulses. However, f–I (frequency–intensity) curves constructed by injecting Gaussian white noise currents superimposed on DC current steps showed that ChABC treatment reduced the gain of spike output. An increase in spike threshold may have contributed to this effect, which is consistent with the observation that spikes in ChABC-treated cells were delayed relative to control-treated cells. In addition, parvalbumin-expressing fast-spiking cortical neurons in >P70 slices that were treated with ChABC also had reduced excitability and gain. The development of PNNs around somata of fast-spiking neurons may be essential for fast and precise sensory transmission and synaptic inhibition in the brain.
Collapse
|
39
|
Caught in the Net: Perineuronal Nets and Addiction. Neural Plast 2016; 2016:7538208. [PMID: 26904301 PMCID: PMC4745418 DOI: 10.1155/2016/7538208] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/23/2015] [Indexed: 12/14/2022] Open
Abstract
Exposure to drugs of abuse induces plasticity in the brain and creates persistent drug-related memories. These changes in plasticity and persistent drug memories are believed to produce aberrant motivation and reinforcement contributing to addiction. Most studies have explored the effect drugs of abuse have on pre- and postsynaptic cells and astrocytes; however, more recently, attention has shifted to explore the effect these drugs have on the extracellular matrix (ECM). Within the ECM are unique structures arranged in a net-like manner, surrounding a subset of neurons called perineuronal nets (PNNs). This review focuses on drug-induced changes in PNNs, the molecules that regulate PNNs, and the expression of PNNs within brain circuitry mediating motivation, reward, and reinforcement as it pertains to addiction.
Collapse
|
40
|
Distribution of N-Acetylgalactosamine-Positive Perineuronal Nets in the Macaque Brain: Anatomy and Implications. Neural Plast 2016; 2016:6021428. [PMID: 26881119 PMCID: PMC4735937 DOI: 10.1155/2016/6021428] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/17/2015] [Accepted: 08/26/2015] [Indexed: 11/17/2022] Open
Abstract
Perineuronal nets (PNNs) are extracellular molecules that form around neurons near the end of critical periods during development. They surround neuronal cell bodies and proximal dendrites. PNNs inhibit the formation of new connections and may concentrate around rapidly firing inhibitory interneurons. Previous work characterized the important role of perineuronal nets in plasticity in the visual system, amygdala, and spinal cord of rats. In this study, we use immunohistochemistry to survey the distribution of perineuronal nets in representative areas of the primate brain. We also document changes in PNN prevalence in these areas in animals of different ages. We found that PNNs are most prevalent in the cerebellar nuclei, surrounding >90% of the neurons there. They are much less prevalent in cerebral cortex, surrounding less than 10% of neurons in every area that we examined. The incidence of perineuronal nets around parvalbumin-positive neurons (putative fast-spiking interneurons) varies considerably between different areas in the brain. Our survey indicates that the presence of PNNs may not have a simple relationship with neural plasticity and may serve multiple functions in the central nervous system.
Collapse
|
41
|
Zhai X, Sun C, Rong P, Li S, McCabe MF, Wang X, Mao J, Wang S. A Correlative Relationship Between Chronic Pain and Insulin Resistance in Zucker Fatty Rats: Role of Downregulation of Insulin Receptors. THE JOURNAL OF PAIN 2015; 17:404-13. [PMID: 26705975 DOI: 10.1016/j.jpain.2015.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 11/29/2015] [Accepted: 12/07/2015] [Indexed: 02/08/2023]
Abstract
UNLABELLED Epidemiological studies and meta-analyses report a strong relationship between chronic pain and abnormalities in glucose metabolism, but the exact relationship between chronic pain and insulin resistance in type 2 diabetes (T2D) remains unknown. Using a model of neuropathic thermal and tactile hypersensitivity induced by chronic constriction injury (CCI) of the sciatic nerve in Zucker Diabetic Fatty (ZDF) and Zucker Lean (ZL) littermates, we compared the recovery period of hypersensitivity and the progression of T2D and studied the possible involvement of insulin receptors (IRs) in the comorbidity of these 2 conditions. We found that the nociceptive thresholds to thermal and mechanical stimulation in naive ZDF rats were lower than in ZL littermates at 6 weeks of age. Although ZDF and ZL rats developed thermal and tactile hypersensitivity after CCI, it took a longer time nociceptive sensitivity to be restored in ZDF rats. Nerve injury accelerated the progression of T2D in ZDF rats, shown by an earlier onset of hyperglycemia, more severe hyperinsulinemia, and a higher concentration of glycosylated hemoglobin Alc 6 weeks after CCI, compared with those in naive ZDF and ZL rats. IR-immunoreactive cells were located across the central nervous system and skeletal muscles. In the central nervous system, IR coexpressed with a neuronal marker (neuronal nuclei) but not a glial marker (glial fibrillary acidic protein). There was a low level of IR expression in skeletal muscles of naive ZDF rats. In contrast, CCI reduced the IR expression in skeletal muscles as well as the ipsilateral spinal cord, primarily in the dorsal horn. In conclusion, our data suggest that the relationship between insulin resistance and chronic pain in ZDF rats is bidirectional and an impaired IR signaling system might be implicated in this reciprocal relationship. PERSPECTIVE Nerve injuries in genetically susceptible individuals might accelerate the development of insulin resistance as in T2D. A downregulated expression of IRs in the skeletal muscle innervated by the injured nerve is one of the underlying mechanisms.
Collapse
Affiliation(s)
- Xu Zhai
- Department of Anatomy, Xinxiang Medical University, Xinxiang, Henan Province, China; Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chunli Sun
- Department of Anatomy, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Peijing Rong
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shaoyuan Li
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Michael F McCabe
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Xing Wang
- Department of Anatomy, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Jianren Mao
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shuxing Wang
- Guangdong Institute of Applied Biological Resources, Guangzhou, China; Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Guangzhou, China.
| |
Collapse
|
42
|
Karus M, Ulc A, Ehrlich M, Czopka T, Hennen E, Fischer J, Mizhorova M, Qamar N, Brüstle O, Faissner A. Regulation of oligodendrocyte precursor maintenance by chondroitin sulphate glycosaminoglycans. Glia 2015; 64:270-86. [PMID: 26454153 DOI: 10.1002/glia.22928] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 09/16/2015] [Indexed: 01/06/2023]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) have been proven to inhibit morphological maturation of oligodendrocytes as well as their myelination capabilities. Yet, it remained unclear, whether CSPGs and/or their respective chondroitin sulfate glycosaminoglycan (CS-GAG) side chains also regulate the oligodendrocyte lineage progression. Here, we initially show that CS-GAGs detected by the monoclonal antibody 473HD are expressed by primary rat NG2-positive oligodendrocyte precursor cells (OPCs) and O4-positive immature oligodendrocytes. CS-GAGs become down-regulated with ongoing oligodendrocyte differentiation. Enzymatic removal of the CS-GAG chains by the bacterial enzyme Chondroitinase ABC (ChABC) promoted spontaneous differentiation of proliferating rat OPCs toward O4-positive immature oligodendrocytes. Upon forced differentiation, the enzymatic removal of the CS-GAGs accelerated oligodendrocyte differentiation toward both MBP-positive and membrane forming oligodendrocytes. These processes were attenuated on enriched CSPG fractions, mainly consisting of Phosphacan/RPTPβ/ζ and to less extent of Brevican and NG2. To qualify CS-GAGs as universal regulators of oligodendrocyte biology, we finally tested the effect of CS-GAG removal on OPCs from different sources such as mouse cortical oligospheres, mouse spinal cord neurospheres, and most importantly human-induced pluripotent stem cell-derived radial glia-like neural precursor cells. For all culture systems used, we observed a similar inhibitory effect of CS-GAGs on oligodendrocyte differentiation. In conclusion, this study clearly suggests an important fundamental principle for complex CS-GAGs to regulate the oligodendrocyte lineage progression. Moreover, the use of ChABC in order to promote oligodendrocyte differentiation toward myelin gene expressing cells might be an applicable therapeutic option to enhance white matter repair.
Collapse
Affiliation(s)
- Michael Karus
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr-University Bochum, Bochum, Germany.,Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, Bonn, Germany
| | - Annika Ulc
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Marc Ehrlich
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Tim Czopka
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr-University Bochum, Bochum, Germany
| | - Eva Hennen
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr-University Bochum, Bochum, Germany
| | - Julia Fischer
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, Bonn, Germany
| | - Marija Mizhorova
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, Bonn, Germany
| | - Naila Qamar
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, Bonn, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
43
|
O'Daly A, Rohde C, Brushart T. The topographic specificity of muscle reinnervation predicts function. Eur J Neurosci 2015; 43:443-50. [PMID: 26332647 DOI: 10.1111/ejn.13058] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 08/10/2015] [Accepted: 08/24/2015] [Indexed: 11/30/2022]
Abstract
Functional testing has assumed a progressively dominant role in validating the success of experimental nerve repair. Results obtained in one model, however, cannot predict the results in others because they reflect the coordinated interaction of several muscles across multiple joints. As a result, many combinations of topographically correct and incorrect muscle reinnervation could produce the same result. We have developed a binary model in which elbow flexors and extensors are reinnervated, and elbow flexion and extension are the functions tested. The musculocutaneous and radial nerves of Lister-Hooded rats were subjected to axonotmetic injuries that produced increasing degrees of axonal misdirection at the site of injury ranging from simple crush to transection and rotational offset of proximal and distal stumps. Elbow function was tested with a device that requires coordinated elbow extension to reach sugar pellets and flexion to return them to the mouth. After 12 weeks of regeneration, motoneurons projecting to the distal musculocutaneous nerve were retrogradely labelled with WGA-Ruby and scored regarding their location within musculocutaneous or radial motoneuron pools. The severity of axonal misdirection resulting from the initial surgery was mirrored by progressive degrees of inappropriate reinnervation of the musculocutaneous nerve by radial nerve axons. The specificity of reinnervation predicted elbow function (r = 0.72), whereas the number of motoneurons regenerating did not. This model is thus well suited to study the interaction of regeneration specificity and function across a single joint, and to produce data that can be generalized more broadly than those obtained from more complex models.
Collapse
Affiliation(s)
- Andres O'Daly
- Department of Orthopaedic Surgery, Johns Hopkins School of Medicine, 601 N. Caroline St., Baltimore, MD, 21287, USA
| | - Charles Rohde
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Thomas Brushart
- Department of Orthopaedic Surgery, Johns Hopkins School of Medicine, 601 N. Caroline St., Baltimore, MD, 21287, USA
| |
Collapse
|
44
|
Oohashi T, Edamatsu M, Bekku Y, Carulli D. The hyaluronan and proteoglycan link proteins: Organizers of the brain extracellular matrix and key molecules for neuronal function and plasticity. Exp Neurol 2015; 274:134-44. [PMID: 26387938 DOI: 10.1016/j.expneurol.2015.09.010] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 09/11/2015] [Accepted: 09/17/2015] [Indexed: 02/06/2023]
Abstract
The hyaluronan and proteoglycanbinding link protein (Hapln) is a key molecule in the formation and control of hyaluronan-based condensed perineuronal matrix in the adult brain. This review summarizes the recent advances in understanding the role of Haplns in the formation and control of two distinct types of perineuronal matrices, one for "classical" PNN and the other for the specialized extracellular matrix (ECM) at the node of Ranvier in the central nervous system (CNS). We introduce the structural components of each ECM organization including the basic concept of supramolecular structure named "HLT model". We furthermore summarize the developmental and physiological role of perineuronal ECMs from the studies of Haplns and related molecules. Finally, we also discuss the potential mechanism modulating PNNs in the adult CNS. This layer of organized matrices may exert a direct effect via core protein or sugar moiety from the structure or by acting as a binding site for biologically active molecules, which are important for neuronal plasticity and saltatory conduction.
Collapse
Affiliation(s)
- Toshitaka Oohashi
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Midori Edamatsu
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Yoko Bekku
- NYU Neuroscience Institute, New York University Langone Medical Center, 522 First Avenue, New York, NY 10016, USA
| | - Daniela Carulli
- Department of Neuroscience, Neuroscience Institute of Turin (NIT), Neuroscience Institute Cavalieri-Ottolenghi (NICO), University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| |
Collapse
|
45
|
Grulova I, Slovinska L, Blaško J, Devaux S, Wisztorski M, Salzet M, Fournier I, Kryukov O, Cohen S, Cizkova D. Delivery of Alginate Scaffold Releasing Two Trophic Factors for Spinal Cord Injury Repair. Sci Rep 2015; 5:13702. [PMID: 26348665 PMCID: PMC4562265 DOI: 10.1038/srep13702] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/04/2015] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) has been implicated in neural cell loss and consequently functional motor and sensory impairment. In this study, we propose an alginate -based neurobridge enriched with/without trophic growth factors (GFs) that can be utilized as a therapeutic approach for spinal cord repair. The bioavailability of key GFs, such as Epidermal Growth factor (EGF) and basic Fibroblast Growth Factor (bFGF) released from injected alginate biomaterial to the central lesion site significantly enhanced the sparing of spinal cord tissue and increased the number of surviving neurons (choline acetyltransferase positive motoneurons) and sensory fibres. In addition, we document enhanced outgrowth of corticospinal tract axons and presence of blood vessels at the central lesion. Tissue proteomics was performed at 3, 7 and 10 days after SCI in rats indicated the presence of anti-inflammatory factors in segments above the central lesion site, whereas in segments below, neurite outgrowth factors, inflammatory cytokines and chondroitin sulfate proteoglycan of the lectican protein family were overexpressed. Collectively, based on our data, we confirm that functional recovery was significantly improved in SCI groups receiving alginate scaffold with affinity-bound growth factors (ALG +GFs), compared to SCI animals without biomaterial treatment.
Collapse
Affiliation(s)
- I Grulova
- Institute of Neurobiology, Center of Excellence for Brain Research, Department of Regenerative Medicine and Stem Cell Therapy, Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Kosice, Slovakia
| | - L Slovinska
- Institute of Neurobiology, Center of Excellence for Brain Research, Department of Regenerative Medicine and Stem Cell Therapy, Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Kosice, Slovakia
| | - J Blaško
- Institute of Neurobiology, Center of Excellence for Brain Research, Department of Regenerative Medicine and Stem Cell Therapy, Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Kosice, Slovakia
| | - S Devaux
- Institute of Neurobiology, Center of Excellence for Brain Research, Department of Regenerative Medicine and Stem Cell Therapy, Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Kosice, Slovakia.,Laboratoire PRISM: Protéomique, Réponse Inflammatoire, Spectrométrie de Masse, INSERM U1192, Bât SN3, 1er étage, Université de Lille 1, F-59655 Villeneuve d'Ascq, France
| | - M Wisztorski
- Laboratoire PRISM: Protéomique, Réponse Inflammatoire, Spectrométrie de Masse, INSERM U1192, Bât SN3, 1er étage, Université de Lille 1, F-59655 Villeneuve d'Ascq, France
| | - M Salzet
- Laboratoire PRISM: Protéomique, Réponse Inflammatoire, Spectrométrie de Masse, INSERM U1192, Bât SN3, 1er étage, Université de Lille 1, F-59655 Villeneuve d'Ascq, France
| | - I Fournier
- Laboratoire PRISM: Protéomique, Réponse Inflammatoire, Spectrométrie de Masse, INSERM U1192, Bât SN3, 1er étage, Université de Lille 1, F-59655 Villeneuve d'Ascq, France
| | - O Kryukov
- The Center of Regenerative Medicine and Stem Cell Research and The Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - S Cohen
- The Center of Regenerative Medicine and Stem Cell Research and The Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - D Cizkova
- Institute of Neurobiology, Center of Excellence for Brain Research, Department of Regenerative Medicine and Stem Cell Therapy, Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Kosice, Slovakia.,Laboratoire PRISM: Protéomique, Réponse Inflammatoire, Spectrométrie de Masse, INSERM U1192, Bât SN3, 1er étage, Université de Lille 1, F-59655 Villeneuve d'Ascq, France
| |
Collapse
|
46
|
Modifications of perineuronal nets and remodelling of excitatory and inhibitory afferents during vestibular compensation in the adult mouse. Brain Struct Funct 2015; 221:3193-209. [PMID: 26264050 DOI: 10.1007/s00429-015-1095-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 07/27/2015] [Indexed: 12/13/2022]
Abstract
Perineuronal nets (PNNs) are aggregates of extracellular matrix molecules surrounding several types of neurons in the adult CNS, which contribute to stabilising neuronal connections. Interestingly, a reduction of PNN number and staining intensity has been observed in conditions associated with plasticity in the adult brain. However, it is not known whether spontaneous PNN changes are functional to plasticity and repair after injury. To address this issue, we investigated PNN expression in the vestibular nuclei of the adult mouse during vestibular compensation, namely the resolution of motor deficits resulting from a unilateral peripheral vestibular lesion. After unilateral labyrinthectomy, we found that PNN number and staining intensity were strongly attenuated in the lateral vestibular nucleus on both sides, in parallel with remodelling of excitatory and inhibitory afferents. Moreover, PNNs were completely restored when vestibular deficits of the mice were abated. Interestingly, in mice with genetically reduced PNNs, vestibular compensation was accelerated. Overall, these results strongly suggest that temporal tuning of PNN expression may be crucial for vestibular compensation.
Collapse
|
47
|
Smith PD, Coulson-Thomas VJ, Foscarin S, Kwok JCF, Fawcett JW. "GAG-ing with the neuron": The role of glycosaminoglycan patterning in the central nervous system. Exp Neurol 2015; 274:100-14. [PMID: 26277685 DOI: 10.1016/j.expneurol.2015.08.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/17/2015] [Accepted: 08/06/2015] [Indexed: 01/17/2023]
Abstract
Proteoglycans (PGs) are a diverse family of proteins that consist of one or more glycosaminoglycan (GAG) chains, covalently linked to a core protein. PGs are major components of the extracellular matrix (ECM) and play critical roles in development, normal function and damage-response of the central nervous system (CNS). GAGs are classified based on their disaccharide subunits, into the following major groups: chondroitin sulfate (CS), heparan sulfate (HS), heparin (HEP), dermatan sulfate (DS), keratan sulfate (KS) and hyaluronic acid (HA). All except HA are modified by sulfation, giving GAG chains specific charged structures and binding properties. While significant neuroscience research has focused on the role of one PG family member, chondroitin sulfate proteoglycan (CSPG), there is ample evidence in support of a role for the other PGs in regulating CNS function in normal and pathological conditions. This review discusses the role of all the identified PG family members (CS, HS, HEP, DS, KS and HA) in normal CNS function and in the context of pathology. Understanding the pleiotropic roles of these molecules in the CNS may open the door to novel therapeutic strategies for a number of neurological conditions.
Collapse
Affiliation(s)
- Patrice D Smith
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK; Department of Neuroscience, Carleton University, Ottawa, ON, Canada.
| | - Vivien J Coulson-Thomas
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | - Simona Foscarin
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | - Jessica C F Kwok
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | - James W Fawcett
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK.
| |
Collapse
|
48
|
Walters R, Medintz IL, Delehanty JB, Stewart MH, Susumu K, Huston AL, Dawson PE, Dawson G. The Role of Negative Charge in the Delivery of Quantum Dots to Neurons. ASN Neuro 2015; 7:7/4/1759091415592389. [PMID: 26243591 PMCID: PMC4550297 DOI: 10.1177/1759091415592389] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Despite our extensive knowledge of the structure of negatively charged cell surface proteoglycans and sialoglycoconjugates in the brain, we have little understanding of how their negative charge contributes to brain function. We have previously shown that intensely photoluminescent 9-nm diameter quantum dots (QDs) with a CdSe core, a ZnS shell, and a negatively charged compact molecular ligand coating (CL4) selectively target neurons rather than glia. We now provide an explanation for this selective neuronal delivery. In this study, we compared three zwitterionic QD coatings differing only in their regions of positive or negative charge, as well as a positively charged (NH2) polyethylene glycol (PEG) coat, for their ability to deliver the cell-membrane-penetrating chaperone lipopeptide JB577 (WG(Palmitoyl)VKIKKP9G2H6) to individual cells in neonatal rat hippocampal slices. We confirm both that preferential uptake in neurons, and the lack of uptake in glia, is strongly associated with having a region of greater negative charge on the QD coating. In addition, the role of negatively charged chondroitin sulfate of the extracellular matrix (ECM) in restricting uptake was further suggested by digesting neonatal rat hippocampal slices with chondroitinase ABC and showing increased uptake of QDs by oligodendrocytes. Treatment still did not affect uptake in astrocytes or microglia. Finally, the future potential of using QDs as vehicles for trafficking proteins into cells continues to show promise, as we show that by administering a histidine-tagged green fluorescent protein (eGFP-His6) to hippocampal slices, we can observe neuronal uptake of GFP.
Collapse
Affiliation(s)
- Ryan Walters
- Committee on Neurobiology, University of Chicago, IL, USA
| | - Igor L Medintz
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC, USA
| | - James B Delehanty
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC, USA
| | - Michael H Stewart
- Optical Sciences Division, Code 5611, U.S. Naval Research Laboratory, Washington, DC, USA
| | - Kimihiro Susumu
- Optical Sciences Division, Code 5611, U.S. Naval Research Laboratory, Washington, DC, USA
| | - Alan L Huston
- Optical Sciences Division, Code 5611, U.S. Naval Research Laboratory, Washington, DC, USA
| | | | - Glyn Dawson
- Committee on Neurobiology, University of Chicago, IL, USA Departments of Pediatrics, Biochemistry and Molecular Biology, University of Chicago, IL, USA
| |
Collapse
|
49
|
Molecular interactions between chondroitin-dermatan sulfate and growth factors/receptors/matrix proteins. Curr Opin Struct Biol 2015; 34:35-42. [PMID: 26164146 DOI: 10.1016/j.sbi.2015.06.004] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 06/12/2015] [Accepted: 06/19/2015] [Indexed: 02/08/2023]
Abstract
Recent functional studies on chondroitin sulfate-dermatan sulfate (CS-DS) demonstrated its indispensable roles in various biological events including brain development and cancer. CS-DS proteoglycans exert their physiological activity through interactions with specific proteins including growth factors, cell surface receptors, and matrix proteins. The characterization of these interactions is essential for regulating the biological functions of CS-DS proteoglycans. Although amino acid sequences on the bioactive proteins required for these interactions have already been elucidated, the specific saccharide sequences involved in the binding of CS-DS to target proteins have not yet been sufficiently identified. In this review, recent findings are described on the interaction between CS-DS and some proteins which are especially involved in the central nervous system and cancer development/metastasis.
Collapse
|
50
|
Dyck SM, Karimi-Abdolrezaee S. Chondroitin sulfate proteoglycans: Key modulators in the developing and pathologic central nervous system. Exp Neurol 2015; 269:169-87. [PMID: 25900055 DOI: 10.1016/j.expneurol.2015.04.006] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 04/11/2015] [Accepted: 04/14/2015] [Indexed: 12/15/2022]
Abstract
Chondroitin Sulfate Proteoglycans (CSPGs) are a major component of the extracellular matrix in the central nervous system (CNS) and play critical role in the development and pathophysiology of the brain and spinal cord. Developmentally, CSPGs provide guidance cues for growth cones and contribute to the formation of neuronal boundaries in the developing CNS. Their presence in perineuronal nets plays a crucial role in the maturation of synapses and closure of critical periods by limiting synaptic plasticity. Following injury to the CNS, CSPGs are dramatically upregulated by reactive glia which form a glial scar around the lesion site. Increased level of CSPGs is a hallmark of all CNS injuries and has been shown to limit axonal plasticity, regeneration, remyelination, and conduction after injury. Additionally, CSPGs create a non-permissive milieu for cell replacement activities by limiting cell migration, survival and differentiation. Mounting evidence is currently shedding light on the potential benefits of manipulating CSPGs in combination with other therapeutic strategies to promote spinal cord repair and regeneration. Moreover, the recent discovery of multiple receptors for CSPGs provides new therapeutic targets for targeted interventions in blocking the inhibitory properties of CSPGs following injury. Here, we will provide an in depth discussion on the impact of CSPGs in normal and pathological CNS. We will also review the recent preclinical therapies that have been developed to target CSPGs in the injured CNS.
Collapse
Affiliation(s)
- Scott M Dyck
- Regenerative Medicine Program, Department of Physiology and the Spinal Cord Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Regenerative Medicine Program, Department of Physiology and the Spinal Cord Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|